Skip to main content
Erschienen in: BMC Medical Imaging 1/2022

Open Access 01.12.2022 | Research

A comparative study between deep learning and radiomics models in grading liver tumors using hepatobiliary phase contrast-enhanced MR images

verfasst von: Lixin Du, Jianpeng Yuan, Meng Gan, Zhigang Li, Pan Wang, Zujun Hou, Cong Wang

Erschienen in: BMC Medical Imaging | Ausgabe 1/2022

Abstract

Purpose

To compare a deep learning model with a radiomics model in differentiating high-grade (LR-3, LR-4, LR-5) liver imaging reporting and data system (LI-RADS) liver tumors from low-grade (LR-1, LR-2) LI-RADS tumors based on the contrast-enhanced magnetic resonance images.

Methods

Magnetic resonance imaging scans of 361 suspected hepatocellular carcinoma patients were retrospectively reviewed. Lesion volume segmentation was manually performed by two radiologists, resulting in 426 lesions from the training set and 83 lesions from the test set. The radiomics model was constructed using a support vector machine (SVM) with pre-defined features, which was first selected using Chi-square test, followed by refining using binary least absolute shrinkage and selection operator (LASSO) regression. The deep learning model was established based on the DenseNet. Performance of the models was quantified by area under the receiver-operating characteristic curve (AUC), accuracy, sensitivity, specificity and F1-score.

Results

A set of 8 most informative features was selected from 1049 features to train the SVM classifier. The AUCs of the radiomics model were 0.857 (95% confidence interval [CI] 0.816–0.888) for the training set and 0.879 (95% CI 0.779–0.935) for the test set. The deep learning method achieved AUCs of 0.838 (95% CI 0.799–0.871) for the training set and 0.717 (95% CI 0.601–0.814) for the test set. The performance difference between these two models was assessed by t-test, which showed the results in both training and test sets were statistically significant.

Conclusion

The deep learning based model can be trained end-to-end with little extra domain knowledge, while the radiomics model requires complex feature selection. However, this process makes the radiomics model achieve better performance in this study with smaller computational cost and more potential on model interpretability.
Hinweise
Lixin Du, Jianpeng Yuan and Meng Gan have contributed equally to this work.

Publisher's Note

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

Introduction

Hepatocellular carcinoma (HCC) is a common type of cancer and a leading cause of cancer death worldwide [1]. The prognosis of HCC depends largely on the stage of the tumor. Multiphasic computed tomography (CT) and magnetic resonance imaging (MRI) have been endorsed as first-line modalities for non-invasive diagnosis and staging of liver tumors [2]. A number of studies have been conducted on the relationship between imaging features with HCC histopathologic grading [36], and the liver imaging reporting and data system (LI-RADS) is a widely-used technique to provide standardized criteria for performing, interpreting, and reporting multiphase CT and MRI exams for HCC diagnosis [7].
Recently, with the flourishing of radiomics techniques in tumor analysis [812], there is increasing interest in the development of imaging-based system for automated liver tumor grading [1317], where machine learning methods were employed to find potential pattern from images in differentiation of liver tumor grade. A popular technique as employed in these studies to build the computer-aided systems is the method of deep learning [18, 19], which adds more layers into conventional neural network architecture and represents data in a hierarchical form to capture the complexity in the data. In spite of good performance as reported, deep learning-based systems in medical imaging application have been of debate in terms of size of dataset, computational cost and model interpretability [2022].
In general, deep learning is a subdomain of machine learning, which includes supervised learning and unsupervised learning, and usually consists of two key components: features and classifiers. Common features include image spatial domain feature such as texture information, tumor shape characteristics, image intensity statistics, or image feature in a transform domain such as wavelet feature. Primary classifiers include decision tree, naive Bayes, neural network, support vector machine (SVM), etc. The basic classifiers can be ensembled to arrive at a classifier with improved performance, as represented by random forest and AdaBoost. Different machine learning methods have different advantages and disadvantages. Interested readers can refer to [23, 24] for a recent review on the application of machine learning to medical imaging.
In cancer imaging, it is unlikely to have a dataset of scale in applications such as face recognition or internet-based image recognition, hence, it is of interest to build systems taking into account the intrinsic property of medical imaging data and choosing machine learning algorithm with less requirement on the amount of data. This study attempted to compare a conventional radiomics model based on SVM with respect to a deep learning based model using DenseNet [25] for liver tumor grading.

Materials and methods

The workflow of this study was illustrated in Fig. 1, as detailed in the following.

Study participants

This retrospective study was approved by the Ethical Committee of Shenzhen Longhua District Central Hospital. A total of 462 patients with suspected HCC who underwent examination between June 2016 and June 2021 were reviewed and included in this study. Inclusion criteria were: (1) patients with clinical reports; (2) patients with contrast enhanced MRI (CE-MRI) examination; (3) staging of liver tumors confirmed by radiologists; (4) no history of other types of tumor. Exclusion criteria were: (1) incomplete clinical results; (2) incomplete CE-MRI scan or unsatisfactory image quality due to patient movement.

Data acquisition

All scans were performed using a 3.0T MRI scanner (Skyra, Siemens, Germany) with a sixteen-channel phase array coil that covered the entire liver. Routine MRI protocols included a respiratory-triggered fat-suppressed T1-weighted dual-echo sequence (repetition time [TR] 4.5 ms, echo time [TE] 1.29 ms and 2.52 ms, slice thickness 3 mm, gap of slice 0.6 mm, field of view [FOV] 380 mm \(\times\) 340 mm, matrix 195 \(\times\)320), a respiratory-triggered fat-suppressed T2-weighted fast spin-echo sequence (TR 3000 ms, TE 84 ms, slice thickness 5 mm, gap of slice 1 mm, FOV 380 mm \(\times\) 380 mm, matrix 320 \(\times\) 320) and a diffusion-weighted sequence (b values 0, 400, 1000 s/\(\text {mm}^2\), TR 3500 ms, TE 59 ms, slice thickness 5 mm, gap of slice 1mm, FOV 380 mm \(\times\) 340 mm, matrix 108 \(\times\) 128). Contrast agents (Gadolinium-ethoxybenzyl-diethylenetriamine pentaacetic acid [Gd-EOB-DTPA]; Primovist, Bayer) were administered at rate of 2 mL/sec and 0.1 mmol gadolinium per kilogram followed by 20 ml saline infusion. The early hepatic arterial phase was omitted. The post-contrast scan was performed at 30 s (arterial phase), 60 s (portal venous phase), 180 s (transitional phase) and 20 mins (hepatobiliary phase) after the injection of contrast agent using T1-weighted 3D gradient echo sequence with fat saturation and volumetric interpolated breath-hold examination and the parameters were: slice thickness 6 mm, TR 3.12 ms, TE 1.51 ms, matrix 290\(\times\) 290, flip angle \(10^\circ\).

Image segmentation and LI-RADS grading

The images in hepatobiliary phase were employed for analysis in this study. Segmentation of images into volume of interest (VOI) that covered lesions was performed manually by two radiologists (with 6 and 10 years of experience) using ITK-SNAP (version 3.8.0), while blinded to histopathological results. Each case was first drawn by one radiologist and then reviewed by the other one to ensure high-quality final segmentation results.
Then, these two radiologists independently analyzed all magnetic resonance (MR) images for assessing major and ancillary features, and assigned an LI-RADS category for each lesion according to the LI-RADS v2018 criteria [7]. Disagreements regarding the LI-RADS categorization were resolved by consensus with a senior abdominal radiologist with over 25 years of liver imaging experience. According to the LI-RADS diagnostic algorithm, a liver lesion is assigned a LI-RADS (LR) category (LR1 to LR5) to reflect the likelihood of being HCC. LR-1 and LR-2 are definitely benign and probably benign, respectively. LR-3 indicates intermediate probability of HCC, LR-4 indicates probable HCC, and LR-5 indicates definite HCC. In this study, the lesions are classified as low-grade tumor (LR-1 and LR-2) and high-grade tumor (LR-3, LR-4 and LR-5) for the following analysis. Samples of images from patients and the corresponding segmentation and grading results were shown in Fig. 2.

Radiomics model construction

Radiomics feature extraction

Radiomics feature extraction based on the VOIs was performed using Pyradiomics software following the IBSI recommendation [26]. The VOIs were resampled with obtained isotropic voxels (\(1 \times 1 \times 1\) mm). Normalization was performed by subtracting the mean value from each voxel and dividing by the standard deviation. A fixed bin width of 25 was used to compute textural features. Moreover, a Laplacian of Gaussian (LoG) filter (sigma values 1.5 mm and 2.5 mm) and wavelet decompositions (start level = 0, level = 1, wavelet = ”coif1”) were employed to accentuate textural differences in the VOIs. The extracted features included first-order features, shape-based features and higher-order texture features.

Feature selection

Feature reduction was performed using MATLAB (R2019b, Mathworks, Natick, USA). Chi-square test [27] was firstly used for univariate analysis, followed by binary least absolute shrinkage and selection operator (LASSO) regression [28] for multivariate analysis to achieve a proper combination of these radiomic features. p-value less than 0.1 was considered statistically significant in the Chi-square test.

Building of radiomics model

SVM [29] was employed to construct the model for distinguishing low-grade and high-grade liver tumors. In model training, 10-fold cross-validation was used for algorithm hyperparameter tuning based on the training set. During the process, we tested two kernels (linear and Gaussian) and four box constraint parameters C (1, 5, 10, 100). The parameter combination with the highest area under the receiver-operating characteristic curve (AUC) value was selected to build the final SVM model.

Deep learning model construction

Data preprocessing

The VOIs were resampled to an isotropic resolution of voxel size (\(1 \times 1 \times 1\) \(\text {mm}^3\)) and the intensity was rescaled to the range of 0 and 1. For each VOI, the volumes were zero-padded to the same size \(100 \times 100 \times 100\) as the input for the deep networks, which covers the whole tumor region.

Model architectures

We employed the DenseNet for this task, which connects each layer to every other layer in a feed-forward fashion [25]. The DenseNet has several compelling advantages: it alleviates the vanishing-gradient problem, strengthens feature propagation, encourages feature reuse, and substantially reduces the number of parameters [25]. The network outputs a probability number ranging from 0 to 1 for each liver lesion, with a number closer to 1 indicating a higher probability that the lesion is high-grade liver tumor, whereas a number closer to 0 indicates a greater probability of being a low-grade liver tumor.

Training of the deep network

The DenseNet was implemented in Pytorch. Training of the network was performed on a 15 GB Nvidia A16 GPU using CUDA 11.1 with cuDNN v8.
The network was trained end-to-end using the Adam optimizer with Nesterov momentum of 0.9. An initial learning rate of \(1 \times 10^{-4}\) was applied and was decayed by a factor of 10 if the validation loss failed to improve over ten consecutive epochs. Training was performed in batches of 16 randomly chosen samples at each iteration. After going through the entire training set, an epoch was finished, and 50 epochs were needed to accomplish the training. Finally, the model with the lowest validation loss was employed, which was used to measure the diagnostic performance of the network based on the test dataset.
During the training process, data augmentation was used to improve the network robustness. The data augmentation techniques used in this study included random rotation (range = 10 degrees), random shear (range = 0.05), random shift (range = 0.05) and random zoom (range = 0.05). Except for the random rotation range, the other parameters represent a fraction relative to the size of the corresponding dimension. When we feed images to the network during training, these augmentation techniques randomly operate on the input to provide images under different conditions, thereby improving network performance.

Model evaluation

The effectiveness of the model was assessed on the independent test set. Receiver operating characteristic (ROC) curve and metrics including precision, recall and F1-score were used to assess the diagnostic performance [30, 31]. Difference between models was evaluated using t-test and p value less than 0.05 was considered statistically significant.
ROC curve for DenseNet can be calculated based on the value generated by the final sigmoid activate function, which can be regarded as a probabilistic output. As for the SVM, the output can be expressed as Eq. (1),
$$\begin{aligned} y = \text {sgn} (f({{\textbf {x}}})) = \text {sgn} (h({{\textbf {x}}}) + b) \end{aligned}$$
(1)
where sgn indicates the sign function. \(h({{\textbf {x}}})\) is defined in Eq. (2), where K is the kernel function [32].
$$\begin{aligned} h({{\textbf {x}}}) = \sum _i y_i \alpha _i K({{\textbf {x}}}_i, {{\textbf {x}}}) \end{aligned}$$
(2)
In this study, we mapped the unthresholded outputs \(f({{\textbf {x}}})\) into probabilities using an additional sigmoid function with the strategy proposed by Platt [32]. With the output probabilities, the ROC curve for SVM can be achieved.
Table 1
Characteristics of included patients
Item
Training
Test
Patient
293
68
Lesion
426
83
Low-grade
236
40
   LR-1
111
19
   LR-2
125
21
High-grade
190
43
   LR-3
22
10
   LR-4
35
7
   LR-5
133
26
Table 2
Metrics for the proposed model on different data sets
Model
Data set
Accuracy
Precision
Recall
F1-score
AUC (95% CI)
Radiomics
Training
0.798
0.757
0.805
0.781
0.857 (0.816–0.888)
Test
0.759
0.717
0.884
0.792
0.879 (0.779–0.935)
Deep
Training
0.770
0.775
0.815
0.794
0.838 (0.799–0.871)
Test
0.659
0.617
0.690
0.652
0.717 (0.601–0.814)
AUC area under the curve, CI confidence interval

Results

Study participants

Among the 462 patients reviewed, 101 patients were excluded and 361 patients formed the study cohort (267 males and 94 females, age with mean 48.6 years and range 22 to 77 years). These HCC patients were divided into the training and the test set, which included 293 and 68 patients, respectively. From the training set, 426 lesions were segmented, and 83 lesions were from the test set. These lesions were graded based on LI-RADS v2018 and detailed patient demographics were listed in Table 1.

Feature extraction and selection for the radiomics model

A total of 1049 features were extracted for each VOI. After univariate analysis, 645 features were regarded as statistically significant (\(p < 0.1\)). The LASSO regression demonstrated that 8 features were effective indicators for discriminating low-grade and high-grade lesions, as illustrated in Fig. 3.

Performance and comparisons of the two models

In this study, the SVM model uses the linear kernel with box constraint parameter C set to 5. The ROC curves of the radiomics model for the training and the test set were shown in Fig. 4a. The AUCs in differentiating low-grade from high-grade liver tumors were 0.857 (95% confidence interval [CI]: 0.816–0.888) for the training set, 0.879 (95% CI 0.779–0.935) for the test set. As for the deep learning model, the ROC curves were shown in Fig. 4b. The AUCs were 0.838 (95% CI 0.799–0.871) for the training set and 0.717 (95% CI 0.601–0.814) for the test set. More detailed metrics to quantify the model performance were listed in Table 2. The two models attained similar performance in differential diagnosis for the training set where the radiomics model only achieves slightly higher AUC than the deep learning model. As for the test set, the radiomics model shows much better performance, indicating the robustness of the system. The t-test shows that the results in both the training and the test set were statistically significant (p < 0.001), which further confirmed a better performance of the radiomics model compared to the deep learning model.

Discussion

This study compared a deep learning model with a radiomics model in differentiating high-grade liver tumors from low-grade ones. The result showed that the two models attained close performance in the training set with similar metric values (Table 2) and ROC curves (Fig. 4), which indicated that both methods could find effective manifolds for describing the training data space. However, the radiomics model performs much better in the test set. One reason could be that the deep network was trained based on the whole volumes of size \(100 \times 100 \times 100\), which includes intensities of 1,000,000 voxels regarded as features. Although 426 lesions were employed for training, the data size was still small for the large input features and the complex architecture, which limited the generalization ability of the model. On the contrary, the radiomics model was established using only 8 selected radiomics features, and the intrinsic manifold was much easier to model than the original volumes, thus resulting in a more robust predicting system.
In this study, the SVM is used to build the radiomics model. The SVM is effective in high dimensional space and was widely used in the field of medical image diagnosis [9, 10]. Besides, it uses a subset of training points in the decision function, which makes it memory efficient. The deep learning model is established using DenseNet, which won the best paper award in CVPR2017 [25]. DenseNet has several compelling advantages: they alleviate the vanishing-gradient problem, strengthen feature propagation, encourage feature reuse, and substantially reduce the number of parameters [25]. As a result, these two representative methods were employed in this study to provide convincing comparisons.
The requirement of sample size in deep learning was studied in [33] and it was pointed out that for single hidden-layer feedforward neural networks with number of units k, number of weights d and classification error \(\epsilon\) (\(0 < \epsilon \le 1/8\)), the required number of training images m could be estimated as \(m \le O(d/\epsilon , \log _2(k/\epsilon ))\). In practice, the architecture of deep networks involves many hidden layers, leading to large number of parameters needed to be estimated, which in turn increases the requirement on the size of training data. The method of deep learning model in this study was implemented using data augmentation to alleviate this issue, attaining AUC of 0.838 (0.799–0.871) and 0.717 (0.601–0.814) in liver tumor grading for the training and the test set. Zhou et al. [34] proposed a deep neural network by combining the squeeze-and-excitation networks in a three-dimensional densely connected convolutional network, and validated the method in a dataset consisting of 213 HCC patients of contrast-enhanced MR images, attaining AUC of 0.83, which is close to the result of the deep network implemented in this study. Nevertheless, If the training samples are from a data distribution that is very different from the one met in the real world, then the network’s generalization performance will be lower than expected [35].
Another major problem associated with deep learning is the issue of black box, as the complicated hierarchical representation of training data makes the prediction very difficult to understand and interpret [36]. How to enhance the interpretability of machine learning system has become a young but rapidly growing body of research in the domain of artificial intelligence (AI), and a lot of research efforts have been dedicated to this direction, including the explainable AI program launched by the defense advanced research projects agency (DARPA). Majority of these efforts focus on enhancing the transparency of machine learning systems through visualizing features learned in the training process and providing insight into the working of deep neural networks. Interested readers can refer to [37] for a recent survey on interpretable machine learning. Nevertheless, the research on interpretable deep learning remains in an early stage. Neurons might learn features without equivalent human concepts, and visualized features do not mean that the underlying reasoning mechanism of deep neural nets would be disclosed explicitly.
By comparison, conventional machine learning methods could be much easier to understand. For example, a decision tree establishes the classification based on a set of “if/else” rules, which is very interpretable. An SVM model represents different classes in a hyperplane in multidimensional space and divides the datasets into classes through maximizing the margin of hyperplane. Methods have been developed to facilitate the interpretability of SVMs by visualizing results as nomograms, such as the nomogram method using decomposable kernels in SVMs [38], the nomogram representation by replacing the lines by color bars with colors offering the same interpretation as the length of the lines in conventional nomograms [39]. In addition, non-image features such as clinical findings could easily be integrated into conventional machine learning systems, which is particularly important in the development of medical AI solution. SVM is very flexible in handling multiple continuous and categorical variables.
One more advantage of conventional machine learning methods is the cost of computation, which is generally more cost-saving in conventional models than in deep-learning methods. In this study, training the SVM classifier took less than 5 s in MATLAB with a personal computer (Intel Core i7-6700 CPU, 16 GB RAM). In comparison, achieving a well trained DenseNet took about 1 h using the 15 GB Nvidia GPU. The high efficiency of SVM-based radiomics model could be attributed to the use of hand-crafted features, whereas the end-to-end deep learning strategy saved human intervention at the cost of computational resources.
The VOI that covered lesions was obtained by manual segmentation. The procedure is performed by two radiologists. Each case was first drawn by one radiologist and then reviewed by the other one to reduce subjectivity. An alternative approach is using the STAPLE [40] tool that can produce a consolidated reference between different operators. Although two radiologists were invited to ensure a high-quality result, limitations of manual delineation still exist. As reported in [41], manual segmentation is labor-intensive, time-consuming, and not always feasible for radiomics analysis requiring huge datasets. Additionally, manual segmentation is subject to inter- and intra-observer variability [42]. One solution is using semi-automatic delineation algorithms, such as region growing or thresholding. These methods may be less precise than manual segmentation, but are more efficient with better reproducibility.
This study was limited in the design of experiment which included only the contrast-enhanced MR images, and did not incorporate other information such as other imaging modalities, or clinical findings. As the present study aimed to compare the efficacy between deep learning models and conventional machine learning methods, the focus of experiment design was to ensure a fair comparison rather than to arrive at an optimal liver tumor grading solution. As discussed in [43], HCC examinations should include late hepatic arterial, portal venous, and, at about 3–5 min, delayed phase acquisitions. In addition, liver tissue microenvironment could be quantitatively derived using dynamic contrast-enhanced imaging [44], and it would be straightforward to integrate such information in the framework of the SVM-based radiomics model.
In conclusion, this study compared a deep learning model with respect to the radiomics model in differentiating high-grade liver tumors from low-grade tumors, and showed that simple machine learning method could attain better performance than the deep learning method in liver tumor grading, with much smaller computational cost. As machine learning problems in medical imaging are featured by limited number of patient data and high requirement on interpretability of intelligent solutions, it would be recommended to explore the issue with conventional and more understandable machine learning methods.

Acknowledgements

Not applicable.

Declarations

This study followed the Helsinki Declaration and was approved by the Ethical Committee of Shenzhen Longhua District Central Hospital. The need to obtain the informed consent was waived by the Ethical Committee because of de-identifcation data involving no potential risk to patients and no link between the patients and researchers.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca Cancer J Clin. 2021;71(3):209–49.CrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca Cancer J Clin. 2021;71(3):209–49.CrossRef
2.
Zurück zum Zitat Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–2.CrossRef Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–2.CrossRef
3.
Zurück zum Zitat Nasu K, Kuroki Y, Tsukamoto T, Nakajima H, Mori K, Minami M. Diffusion-weighted imaging of surgically resected hepatocellular carcinoma: imaging characteristics and relationship among signal intensity, apparent diffusion coefficient, and histopathologic grade. Am J Roentgenol. 2009;193(2):438–44.CrossRef Nasu K, Kuroki Y, Tsukamoto T, Nakajima H, Mori K, Minami M. Diffusion-weighted imaging of surgically resected hepatocellular carcinoma: imaging characteristics and relationship among signal intensity, apparent diffusion coefficient, and histopathologic grade. Am J Roentgenol. 2009;193(2):438–44.CrossRef
4.
Zurück zum Zitat Kitao A, Zen Y, Matsui O, Gabata T, Kobayashi S, Koda W, Kozaka K, Yoneda N, Yamashita T, Kaneko S, Nakanuma Y. Hepatocellular carcinoma: signal intensity at gadoxetic acid-enhanced mr imaging-correlation with molecular transporters and histopathologic features. Radiology. 2010;256(3):817–26.CrossRef Kitao A, Zen Y, Matsui O, Gabata T, Kobayashi S, Koda W, Kozaka K, Yoneda N, Yamashita T, Kaneko S, Nakanuma Y. Hepatocellular carcinoma: signal intensity at gadoxetic acid-enhanced mr imaging-correlation with molecular transporters and histopathologic features. Radiology. 2010;256(3):817–26.CrossRef
5.
Zurück zum Zitat Kogita S, Imai Y, Okada M, Kim T, Onishi H, Takamura M, Fukuda K, Igura T, Sawai Y, Morimoto O, Hori M, Nagano H, Wakasa K, Hayashi N, Murakami T. Gd-eob-dtpa-enhanced magnetic resonance images of hepatocellular carcinoma: correlation with histological grading and portal blood flow. Eur Radiol. 2010;20(10):2405–13.CrossRef Kogita S, Imai Y, Okada M, Kim T, Onishi H, Takamura M, Fukuda K, Igura T, Sawai Y, Morimoto O, Hori M, Nagano H, Wakasa K, Hayashi N, Murakami T. Gd-eob-dtpa-enhanced magnetic resonance images of hepatocellular carcinoma: correlation with histological grading and portal blood flow. Eur Radiol. 2010;20(10):2405–13.CrossRef
6.
Zurück zum Zitat Becker AS, Barth BK, Marquez PH, Donati OF, Ulbrich EJ, Karlo C, Reiner CS, Fischer MA. Increased interreader agreement in diagnosis of hepatocellular carcinoma using an adapted li-rads algorithm. Eur J Radiol. 2017;86:33–40.CrossRef Becker AS, Barth BK, Marquez PH, Donati OF, Ulbrich EJ, Karlo C, Reiner CS, Fischer MA. Increased interreader agreement in diagnosis of hepatocellular carcinoma using an adapted li-rads algorithm. Eur J Radiol. 2017;86:33–40.CrossRef
7.
Zurück zum Zitat Chernyak V, Fowler KJ, Kamaya A, Kielar AZ, Elsayes KM, Bashir MR, Kono Y, Do RK, Mitchell DG, Singal AG, Tang A, Sirlin CB. Liver imaging reporting and data system (li-rads) version 2018: imaging of hepatocellular carcinoma in at-risk patients. Radiology. 2018;289(3):816–30.CrossRef Chernyak V, Fowler KJ, Kamaya A, Kielar AZ, Elsayes KM, Bashir MR, Kono Y, Do RK, Mitchell DG, Singal AG, Tang A, Sirlin CB. Liver imaging reporting and data system (li-rads) version 2018: imaging of hepatocellular carcinoma in at-risk patients. Radiology. 2018;289(3):816–30.CrossRef
8.
Zurück zum Zitat Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology. 2016;278(2):563–77.CrossRef Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology. 2016;278(2):563–77.CrossRef
9.
Zurück zum Zitat Wang XY, Dai SF, Wang Q, Chai XF, Xian JF. Investigation of mri-based radiomics model in differentiation between sinonasal primary lymphomas and squamous cell carcinomas. Jpn J Radiol. 2021;39(8):755–62.CrossRef Wang XY, Dai SF, Wang Q, Chai XF, Xian JF. Investigation of mri-based radiomics model in differentiation between sinonasal primary lymphomas and squamous cell carcinomas. Jpn J Radiol. 2021;39(8):755–62.CrossRef
10.
Zurück zum Zitat Granata V, Fusco R, De Muzio F, Cutolo C, Setola SV, Grassi R, Grassi F, Ottaiano A, Nasti G, Tatangelo F, Pilone V, Miele V, Brunese MC, Izzo F, Petrillo A. Radiomics textural features by mr imaging to assess clinical outcomes following liver resection in colorectal liver metastases. Radiol Med. 2022;127(5):461–70.CrossRef Granata V, Fusco R, De Muzio F, Cutolo C, Setola SV, Grassi R, Grassi F, Ottaiano A, Nasti G, Tatangelo F, Pilone V, Miele V, Brunese MC, Izzo F, Petrillo A. Radiomics textural features by mr imaging to assess clinical outcomes following liver resection in colorectal liver metastases. Radiol Med. 2022;127(5):461–70.CrossRef
11.
Zurück zum Zitat Wang YT, Luo SY, Jin GH, Fu RD, Yu ZF, Zhang J. Preoperative clinical-radiomics nomogram for microvascular invasion prediction in hepatocellular carcinoma using f-18-fdg pet/ct. BMC Med Imaging. 2022;22(1):66.CrossRef Wang YT, Luo SY, Jin GH, Fu RD, Yu ZF, Zhang J. Preoperative clinical-radiomics nomogram for microvascular invasion prediction in hepatocellular carcinoma using f-18-fdg pet/ct. BMC Med Imaging. 2022;22(1):66.CrossRef
12.
Zurück zum Zitat Li CQ, Zheng X, Guo HL, Cheng MQ, Huang Y, Xie XY, Lu MD, Kuang M, Wang W, Chen LD. Differentiation between combined hepatocellular carcinoma and hepatocellular carcinoma: comparison of diagnostic performance between ultrasomics-based model and ceus li-rads v2017. BMC Med Imaging. 2022;22(1):66. Li CQ, Zheng X, Guo HL, Cheng MQ, Huang Y, Xie XY, Lu MD, Kuang M, Wang W, Chen LD. Differentiation between combined hepatocellular carcinoma and hepatocellular carcinoma: comparison of diagnostic performance between ultrasomics-based model and ceus li-rads v2017. BMC Med Imaging. 2022;22(1):66.
13.
Zurück zum Zitat Bevilacqua V, Brunetti A, Trotta GF, Carnimeo L, Marino F, Alberotanza V, Scardapane A. A deep learning approach for hepatocellular carcinoma grading. Int J Comput Vis Image Process. 2017;7(2):1–18.CrossRef Bevilacqua V, Brunetti A, Trotta GF, Carnimeo L, Marino F, Alberotanza V, Scardapane A. A deep learning approach for hepatocellular carcinoma grading. Int J Comput Vis Image Process. 2017;7(2):1–18.CrossRef
14.
Zurück zum Zitat Mao B, Zhang LZ, Ning PG, Ding F, Wu FT, Lu GR, Geng YY, Ma JD. Preoperative prediction for pathological grade of hepatocellular carcinoma via machine learning-based radiomics. Eur Radiol. 2020;30(12):6924–32.CrossRef Mao B, Zhang LZ, Ning PG, Ding F, Wu FT, Lu GR, Geng YY, Ma JD. Preoperative prediction for pathological grade of hepatocellular carcinoma via machine learning-based radiomics. Eur Radiol. 2020;30(12):6924–32.CrossRef
15.
Zurück zum Zitat Lewis S, Hectors S, Taouli B. Radiomics of hepatocellular carcinoma. Abdom Radiol. 2021;46(1):111–23.CrossRef Lewis S, Hectors S, Taouli B. Radiomics of hepatocellular carcinoma. Abdom Radiol. 2021;46(1):111–23.CrossRef
16.
Zurück zum Zitat Liu QP, Yang KL, Xu X, Liu XS, Qu JR, Zhang YD. Radiomics analysis of pretreatment mri in predicting tumor response and outcome in hepatocellular carcinoma with transarterial chemoembolization: a two-center collaborative study. Abdom Radiol. 2022;47(2):651–63.CrossRef Liu QP, Yang KL, Xu X, Liu XS, Qu JR, Zhang YD. Radiomics analysis of pretreatment mri in predicting tumor response and outcome in hepatocellular carcinoma with transarterial chemoembolization: a two-center collaborative study. Abdom Radiol. 2022;47(2):651–63.CrossRef
17.
Zurück zum Zitat Hu P, Chen LY, Zhong YY, Lin YD, Yu XJ, Hu X, Tao XW, Lin SS, Niu TY, Chen R, Wu X, Sun JH. Effects of slice thickness on ct radiomics features and models for staging liver fibrosis caused by chronic liver disease. Jpn J Radiol. 2022;6:66. Hu P, Chen LY, Zhong YY, Lin YD, Yu XJ, Hu X, Tao XW, Lin SS, Niu TY, Chen R, Wu X, Sun JH. Effects of slice thickness on ct radiomics features and models for staging liver fibrosis caused by chronic liver disease. Jpn J Radiol. 2022;6:66.
18.
Zurück zum Zitat Hinton GE, Osindero S, Teh YW. A fast learning algorithm for deep belief nets. Neural Comput. 2006;18(7):1527–54.CrossRef Hinton GE, Osindero S, Teh YW. A fast learning algorithm for deep belief nets. Neural Comput. 2006;18(7):1527–54.CrossRef
19.
Zurück zum Zitat Gao WY, Wang WT, Song DJ, Yang C, Zhu K, Zeng MS, Rao SX, Wang MN. A predictive model integrating deep and radiomics features based on gadobenate dimeglumine-enhanced mri for postoperative early recurrence of hepatocellular carcinoma. Radiol Med. 2022;127(3):259–71.CrossRef Gao WY, Wang WT, Song DJ, Yang C, Zhu K, Zeng MS, Rao SX, Wang MN. A predictive model integrating deep and radiomics features based on gadobenate dimeglumine-enhanced mri for postoperative early recurrence of hepatocellular carcinoma. Radiol Med. 2022;127(3):259–71.CrossRef
20.
Zurück zum Zitat Samala RK, Chan HP, Hadjiiski L, Helvie MA, Richter CD, Cha KH. Breast cancer diagnosis in digital breast tomosynthesis: effects of training sample size on multi-stage transfer learning using deep neural nets. IEEE Trans Med Imaging. 2019;38(3):686–96.CrossRef Samala RK, Chan HP, Hadjiiski L, Helvie MA, Richter CD, Cha KH. Breast cancer diagnosis in digital breast tomosynthesis: effects of training sample size on multi-stage transfer learning using deep neural nets. IEEE Trans Med Imaging. 2019;38(3):686–96.CrossRef
21.
Zurück zum Zitat Balki I, Amirabadi A, Levman J, Martel AL, Emersic Z, Meden B, Garcia-Pedrero A, Ramirez SC, Kong DH, Moody AR, Tyrrell PN. Sample-size determination methodologies for machine learning in medical imaging research: a systematic review. Can Assoc Radiol J J De L Assoc Canadienne Des Radiologistes. 2019;70(4):344–53.CrossRef Balki I, Amirabadi A, Levman J, Martel AL, Emersic Z, Meden B, Garcia-Pedrero A, Ramirez SC, Kong DH, Moody AR, Tyrrell PN. Sample-size determination methodologies for machine learning in medical imaging research: a systematic review. Can Assoc Radiol J J De L Assoc Canadienne Des Radiologistes. 2019;70(4):344–53.CrossRef
22.
Zurück zum Zitat Lundervold AS, Lundervold A. An overview of deep learning in medical imaging focusing on mri. Zeitschrift Fur Medizinische Physik. 2019;29(2):102–27.CrossRef Lundervold AS, Lundervold A. An overview of deep learning in medical imaging focusing on mri. Zeitschrift Fur Medizinische Physik. 2019;29(2):102–27.CrossRef
23.
Zurück zum Zitat Erickson BJ, Korfiatis P, Akkus Z, Kline TL. Machine learning for medical imaging(1). Radiographics. 2017;37(2):505–15.CrossRef Erickson BJ, Korfiatis P, Akkus Z, Kline TL. Machine learning for medical imaging(1). Radiographics. 2017;37(2):505–15.CrossRef
24.
Zurück zum Zitat Giger ML. Machine learning in medical imaging. J Am Coll Radiol. 2018;15(3):512–20.CrossRef Giger ML. Machine learning in medical imaging. J Am Coll Radiol. 2018;15(3):512–20.CrossRef
25.
Zurück zum Zitat Huang, G., Liu, Z., van der Maaten, L., Weinberger, K.Q.: Densely connected convolutional networks. In: 30th IEEE Conference on Computer Vision and Pattern Recognition (Cvpr 2017), pp. 2261–2269 (2017) Huang, G., Liu, Z., van der Maaten, L., Weinberger, K.Q.: Densely connected convolutional networks. In: 30th IEEE Conference on Computer Vision and Pattern Recognition (Cvpr 2017), pp. 2261–2269 (2017)
26.
Zurück zum Zitat Yan BC, Li Y, Hua F, Zhang GF, Feng F, Sun MH, Lin GW, Qiang JW. Radiologists with mri-based radiomics aids to predict the pelvic lymph node metastasis in endometrial cancer: a multicenter study. Eur Radiol. 2021;31(1):411–22.CrossRef Yan BC, Li Y, Hua F, Zhang GF, Feng F, Sun MH, Lin GW, Qiang JW. Radiologists with mri-based radiomics aids to predict the pelvic lymph node metastasis in endometrial cancer: a multicenter study. Eur Radiol. 2021;31(1):411–22.CrossRef
27.
Zurück zum Zitat Qi YF, Zhang ST, Wei JW, Zhang GY, Lei J, Yan WG, Xiao Y, Yan S, Xue HD, Feng F, Sun H, Tian J, Jin ZY. Multiparametric mri-based radiomics for prostate cancer screening with psa in 4–10 ng/ml to reduce unnecessary biopsies. J Magn Resonan Imaging. 2020;51(6):1890–9.CrossRef Qi YF, Zhang ST, Wei JW, Zhang GY, Lei J, Yan WG, Xiao Y, Yan S, Xue HD, Feng F, Sun H, Tian J, Jin ZY. Multiparametric mri-based radiomics for prostate cancer screening with psa in 4–10 ng/ml to reduce unnecessary biopsies. J Magn Resonan Imaging. 2020;51(6):1890–9.CrossRef
28.
Zurück zum Zitat Luo Y, Mei DD, Gong JS, Zuo M, Guo XJ. Multiparametric mri-based radiomics nomogram for predicting lymphovascular space invasion in endometrial carcinoma. J Magn Resonan Imaging. 2020;52(4):1257–62.CrossRef Luo Y, Mei DD, Gong JS, Zuo M, Guo XJ. Multiparametric mri-based radiomics nomogram for predicting lymphovascular space invasion in endometrial carcinoma. J Magn Resonan Imaging. 2020;52(4):1257–62.CrossRef
29.
Zurück zum Zitat Cheng S, Shi H, Lu M, Wang C, Duan S, Xu Q, Shi H. Radiomics analysis for predicting malignant potential of intraductal papillary mucinous neoplasms of the pancreas: comparison of ct and mri. Acad Radiol. 2022;29(3):367–75.CrossRef Cheng S, Shi H, Lu M, Wang C, Duan S, Xu Q, Shi H. Radiomics analysis for predicting malignant potential of intraductal papillary mucinous neoplasms of the pancreas: comparison of ct and mri. Acad Radiol. 2022;29(3):367–75.CrossRef
30.
Zurück zum Zitat Guo Y, Song Q, Jiang M, Guo Y, Xu P, Zhang Y, Fu C-C, Fang Q, Zeng M, Yao X. Histological subtypes classification of lung cancers on ct images using 3d deep learning and radiomics. Acad Radiol. 2021;28(9):258–66.CrossRef Guo Y, Song Q, Jiang M, Guo Y, Xu P, Zhang Y, Fu C-C, Fang Q, Zeng M, Yao X. Histological subtypes classification of lung cancers on ct images using 3d deep learning and radiomics. Acad Radiol. 2021;28(9):258–66.CrossRef
31.
Zurück zum Zitat Han YX, Wang TZ, Wu P, Zhang H, Chen HH, Yang C. Meningiomas: preoperative predictive histopathological grading based on radiomics of mri. Magn Resonan Imaging. 2021;77:36–43.CrossRef Han YX, Wang TZ, Wu P, Zhang H, Chen HH, Yang C. Meningiomas: preoperative predictive histopathological grading based on radiomics of mri. Magn Resonan Imaging. 2021;77:36–43.CrossRef
32.
Zurück zum Zitat Platt J. Probabilistic outputs for support vector machines and comparisons to regularized likelihood methods. Adv Large Margin Classif. 1999;10(3):61–74. Platt J. Probabilistic outputs for support vector machines and comparisons to regularized likelihood methods. Adv Large Margin Classif. 1999;10(3):61–74.
33.
Zurück zum Zitat Hitzl W, Reitsamer HA, Hornykewycz K, Mistlberger A, Grabner G. Application of discriminant, classification tree and neural network analysis to differentiate between potential glaucoma suspects with and without visual field defects. J Theor Med. 2003;5(3–4):161–70.CrossRef Hitzl W, Reitsamer HA, Hornykewycz K, Mistlberger A, Grabner G. Application of discriminant, classification tree and neural network analysis to differentiate between potential glaucoma suspects with and without visual field defects. J Theor Med. 2003;5(3–4):161–70.CrossRef
34.
Zurück zum Zitat Zhou Q, Zhou ZY, Chen CM, Fan GH, Chen GQ, Heng HY, Ji JS, Dai YK. Grading of hepatocellular carcinoma using 3d se-densenet in dynamic enhanced mr images. Comput Biol Med. 2019;107:47–57.CrossRef Zhou Q, Zhou ZY, Chen CM, Fan GH, Chen GQ, Heng HY, Ji JS, Dai YK. Grading of hepatocellular carcinoma using 3d se-densenet in dynamic enhanced mr images. Comput Biol Med. 2019;107:47–57.CrossRef
35.
Zurück zum Zitat Zech JR, Badgeley MA, Liu M, Costa AB, Titano JJ, Oermann EK. Variable generalization performance of a deep learning model to detect pneumonia in chest radiographs: a cross-sectional study. PLoS Med. 2018;15(11):66.CrossRef Zech JR, Badgeley MA, Liu M, Costa AB, Titano JJ, Oermann EK. Variable generalization performance of a deep learning model to detect pneumonia in chest radiographs: a cross-sectional study. PLoS Med. 2018;15(11):66.CrossRef
36.
Zurück zum Zitat Castelvecchi D. Can we open the black box of ai? Nature. 2016;538:20–3.CrossRef Castelvecchi D. Can we open the black box of ai? Nature. 2016;538:20–3.CrossRef
37.
Zurück zum Zitat Hohman F, Kahng M, Pienta R, Chau DH. Visual analytics in deep learning: an interrogative survey for the next frontiers. IEEE Trans Vis Comput Graph. 2019;25(8):2674–93.CrossRef Hohman F, Kahng M, Pienta R, Chau DH. Visual analytics in deep learning: an interrogative survey for the next frontiers. IEEE Trans Vis Comput Graph. 2019;25(8):2674–93.CrossRef
38.
Zurück zum Zitat Jakulin A, Možina M, Demšar J, Bratko I, Zupan B. Nomograms for visualizing support vector machines. In: KDD’05. New York: Association for Computing Machinery; 2005. p. 108–17. Jakulin A, Možina M, Demšar J, Bratko I, Zupan B. Nomograms for visualizing support vector machines. In: KDD’05. New York: Association for Computing Machinery; 2005. p. 108–17.
39.
Zurück zum Zitat Van Belle V, Van Calster B, Van Huffel S, Suykens JAK, Lisboa P. Explaining support vector machines: a color based nomogram. PLoS ONE. 2016;11(10):66. Van Belle V, Van Calster B, Van Huffel S, Suykens JAK, Lisboa P. Explaining support vector machines: a color based nomogram. PLoS ONE. 2016;11(10):66.
40.
Zurück zum Zitat Warfield SK, Zou KH, Wells WM. Simultaneous truth and performance level estimation (staple): an algorithm for the validation of image segmentation. IEEE Trans Med Imaging. 2004;23(7):903–21.CrossRef Warfield SK, Zou KH, Wells WM. Simultaneous truth and performance level estimation (staple): an algorithm for the validation of image segmentation. IEEE Trans Med Imaging. 2004;23(7):903–21.CrossRef
41.
Zurück zum Zitat Stefano A, Leal A, Richiusa S, Trang P, Comelli A, Benfante V, Cosentino S, Sabini MG, Tuttolomondo A, Altieri R, Certo F, Barbagallo GMV, Ippolito M, Russo G. Robustness of pet radiomics features: impact of co-registration with mri. Appl Sci. 2021;11(21):10170.CrossRef Stefano A, Leal A, Richiusa S, Trang P, Comelli A, Benfante V, Cosentino S, Sabini MG, Tuttolomondo A, Altieri R, Certo F, Barbagallo GMV, Ippolito M, Russo G. Robustness of pet radiomics features: impact of co-registration with mri. Appl Sci. 2021;11(21):10170.CrossRef
42.
Zurück zum Zitat Alongi P, Stefano A, Comelli A, Laudicella R, Scalisi S, Arnone G, Barone S, Spada M, Purpura P, Bartolotta T, Midiri M, Lagalla R, Russo G. Radiomics analysis of 18f-choline pet/ct in the prediction of disease outcome in high-risk prostate cancer: an explorative study on machine learning feature classification in 94 patients. Eur Radiol. 2021;31(7):4595–605.CrossRef Alongi P, Stefano A, Comelli A, Laudicella R, Scalisi S, Arnone G, Barone S, Spada M, Purpura P, Bartolotta T, Midiri M, Lagalla R, Russo G. Radiomics analysis of 18f-choline pet/ct in the prediction of disease outcome in high-risk prostate cancer: an explorative study on machine learning feature classification in 94 patients. Eur Radiol. 2021;31(7):4595–605.CrossRef
43.
Zurück zum Zitat 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. Radiology. 2014;273(1):30–50.CrossRef 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. Radiology. 2014;273(1):30–50.CrossRef
44.
Zurück zum Zitat Koh TS, Thng CH, Lee PS, Hartono S, Rumpel H, Goh BC, Bisdas S. Hepatic metastases: in vivo assessment of perfusion parameters at dynamic contrast-enhanced mr imaging with dual-input two-compartment tracer kinetics model. Radiology. 2008;249(1):307–20.CrossRef Koh TS, Thng CH, Lee PS, Hartono S, Rumpel H, Goh BC, Bisdas S. Hepatic metastases: in vivo assessment of perfusion parameters at dynamic contrast-enhanced mr imaging with dual-input two-compartment tracer kinetics model. Radiology. 2008;249(1):307–20.CrossRef
Metadaten
Titel
A comparative study between deep learning and radiomics models in grading liver tumors using hepatobiliary phase contrast-enhanced MR images
verfasst von
Lixin Du
Jianpeng Yuan
Meng Gan
Zhigang Li
Pan Wang
Zujun Hou
Cong Wang
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Medical Imaging / Ausgabe 1/2022
Elektronische ISSN: 1471-2342
DOI
https://doi.org/10.1186/s12880-022-00946-8

Weitere Artikel der Ausgabe 1/2022

BMC Medical Imaging 1/2022 Zur Ausgabe

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärztinnen und Psychotherapeuten.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Update Radiologie

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