Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
Artif Intell Gastroenterol. Jun 28, 2022; 3(3): 80-87
Published online Jun 28, 2022. doi: 10.35712/aig.v3.i3.80
Machine learning approaches using blood biomarkers in non-alcoholic fatty liver diseases
Randhall B Carteri, Department of Nutrition, Methodist University Center - IPA, Porto Alegre 90420-060, Rio Grande do Sul, Brazil
Randhall B Carteri, Department of Health and Behaviour, Catholic University of Pelotas, Pelotas 96015-560, Rio Grande do Sul, Brazil
Mateus Grellert, Department of Informatics and Statistics, Federal University of Santa Catarina, Florianópolis 88040-900, Santa Catarina, Brazil
Daniela Luisa Borba, Sabrina Alves Fernandes, Postgraduate Program in Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
Claudio Augusto Marroni, Department of Gastroenterology and Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
ORCID number: Randhall B Carteri (0000-0003-4124-9470); Mateus Grellert (0000-0003-0600-7054); Daniela Luisa Borba ((0000-0002- 2141-3993); Claudio Augusto Marroni (0000-0002-1718-6548); Sabrina Alves Fernandes (0000-0001-8504-603X).
Author contributions: Carteri RB organized the structuring of the article in the order of the subheadings covered; Grellert M and Borba DL contributed to manuscript writing; Maroni CA and Fernandes SA supervised and revised the manuscript; All authors contributed to the writing of the article and review of the scientific article.
Conflict-of-interest statement: All authors declare that there are no conflicts of interest related to this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Sabrina Alves Fernandes, PhD, Research Scientist, Teacher, Postgraduate Program in Hepatology, Federal University of Health Sciences of Porto Alegre, Sarmento Leite Street, 245 - Centro Histórico, Porto Alegre 90050-170, Rio Grande do Sul, Brazil. sabrinaafernandes@gmail.com
Received: December 31, 2021
Peer-review started: December 31, 2021
First decision: March 28, 2022
Revised: April 15, 2022
Accepted: May 8, 2022
Article in press: May 8, 2022
Published online: June 28, 2022

Abstract

The prevalence of nonalcoholic fatty liver disease (NAFLD) is an important public health concern. Early diagnosis of NAFLD and potential progression to nonalcoholic steatohepatitis (NASH), could reduce the further advance of the disease, and improve patient outcomes. Aiming to support patient diagnostic and predict specific outcomes, the interest in artificial intelligence (AI) methods in hepatology has dramatically increased, especially with the application of less-invasive biomarkers. In this review, our objective was twofold: Firstly, we presented the most frequent blood biomarkers in NAFLD and NASH and secondly, we reviewed recent literature regarding the use of machine learning (ML) methods to predict NAFLD and NASH in large cohorts. Strikingly, these studies provide insights into ML application in NAFLD patients' prognostics and ranked blood biomarkers are able to provide a recognizable signature allowing cost-effective NAFLD prediction and also differentiating NASH patients. Future studies should consider the limitations in the current literature and expand the application of these algorithms in different populations, fortifying an already promising tool in medical science.

Key Words: Artificial intelligence, Liver diseases, Healthcare, Hepatology, Prognosis, Diagnostics

Core Tip: The ability of machine learning approaches to process multiple variables, map linear and nonlinear interactions, ranking the most important features, in addition to the capability of building accurate prediction models, sets a future direction to its application in complex diseases such as nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Future studies should consider the limitations in the current literature and expand the application of these algorithms in different populations, fortifying an already promising tool in medical science.



INTRODUCTION

Nonalcoholic fatty liver disease (NAFLD) affects an expressive part of the population worldwide and is a major cause of liver-disease related morbidity[1]. The most common cause of death in NAFLD patients is related to cardiovascular diseases, which is partially explained by the presence of metabolic comorbidities, such as obesity, type 2 diabetes, dyslipidemia, and hypertension[2]. Recently, there was concordance that the term NAFLD cannot represent the multisystemic metabolic disruption associated with the disease, resulting in the novel term MAFLD - metabolic associated fatty liver disease. Moreover, MAFLD considers the hepatic manifestation of a multimodal disease that is heterogeneous in its causes, symptoms, progression, and outcomes[3]. Nevertheless, the progression of liver fibrosis could lead to Nonalcoholic steatohepatitis (NASH), a condition characterized by histological lobular inflammation and hepatocyte ballooning[2]. Hence, detecting possible elements related to a worse prognosis in these conditions in the early stages of the disease could improve the treatment and its efficiency. Considering the significance of advanced fibrosis in NAFLD patients, differentiating NASH from steatosis is vital, reinforcing the need for cost-effective methods for risk stratification in this population[4]. Although liver biopsy is widely considered the gold standard in liver diseases investigation, it is also invasive, expensive, and prone to sampling error. In this context, the use of non-invasive biomarkers gains considerable importance[5].

The interest in artificial intelligence (AI) methods in different medical specialties, including hepatology, has dramatically increased during the last decade[6]. Advances in technology and data acquisition have simplified the collection and storage of large data sets with long time series, leading to increasingly varied fields of application, including biomedical areas. In this context, large-volume data mining evaluations had been showing promising results in recent clinical studies using machine learning methods[7-9]. More specifically, supervised machine learning (SML), can automatically detect patterns in existing training data and then use the detected patterns to predict future data[6]. Rather than considering differences between groups (as traditional statistical comparisons do), SML methods address individual differences, classifying individuals in ways that contribute to the clinical decision-making process.

The commonly late diagnosis of liver disorders contributes to suboptimal treatment and poor results. More specifically, as the prevalence of NAFLD is an important public health concern, early diagnosis of NAFLD and potential progression to NASH, could reduce the further advance of the disease, and improve patient outcomes. Using SML methods allows for collecting patient data and identifying their profile regarding the risk of developing comorbidities associated with liver damage, such as the development of metabolic syndrome or even predicting the patient's prognosis. Several recent reviews highlighted the application of artificial intelligence in hepatology, while broadly discussing how different approaches present potential applications in several areas of hepatology[10-12]. However, specific discussion of machine learning approaches using cost-effective biomarkers could help to guide future studies towards the improvement of NAFLD diagnosis. Therefore, the objective of this mini-review is to discuss the application of SML approaches using biomarkers for the diagnosis of NAFLD and the prediction of NASH presence.

BLOOD BIOMARKERS IN NAFLD

Biomarkers are a "defined characteristic that is measured as an indicator of normal biological processes, pathogenic processes or responses to an exposure or intervention”. This includes a plethora of possible assessments commonly investigated in NAFLD, such as blood profile, imaging (histological/radiographic) exams, specific anthropometric characteristics (body composition), and also phase angle derived from bioimpedance[13]. Noteworthy, blood biomarkers are a less invasive approach from a biological point of view and could complement imaging techniques to improve disease monitoring. In clinical settings, liver biopsy is the diagnostic gold standard for NAFLD, allowing the assessment of lipid content, inflammation, hepatocellular ballooning, and fibrotic alterations, which can also determine NASH diagnostics[14]. However, non-invasive techniques provide limited inflammation and hepatocellular ballooning determination, making objective biomarker panels for the assessment and monitoring of NAFLD or NASH a current challenge[14,15].

Nevertheless, abnormal liver function is often initially identified by nonspecific hepatocellular damage through elevations in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in addition to alkaline phosphatase and gamma-glutamyl transferase (GGT)[16]. However, ALT and AST can present normal levels while GGT can present a 1.5 - fold elevation, and this response does not reflect hepatic inflammation, fibrosis, or patient metabolic risks[17,18]. Recently, cytokeratin (CK)-18 gained attention as a more specific approach for hepatocyte apoptosis since CK-18 is a major intermediate filament protein cleaved by caspases creating fragments during the apoptotic processes[19]. Assays of CK-18 fragments provide moderate accuracy due to high variability between cut-offs and respective diagnostic accuracy among studies[19]. More specifically, M30 measures caspase-cleaved CK18 produced during apoptosis, and M65 measures the total levels of (both cleaved and intact) CK18[20]. The CK-18 fragments could independently predict NAFLD severity and detect the presence of NASH with a specificity close to 90%[21,22]. In a large and heterogeneous cohort, the blood concentration of CK-18 fragments of patients with NAFLD was higher when compared with healthy volunteers and correlated to several biomarkers of liver damage and steatosis[22]. Moreover, several "biomarker panels" to grade NAFLD patients’ steatosis and fibrosis through specific scores comprise different biomarker combinations, summarized in Table 1. Notably, the FibroTest, Fibrometer, Hepascore, and Enhanced Liver Fibrosis scores are patented and commercially available panels. Nevertheless, most of the biomarker panels for the diagnosis of NAFLD and NASH, lack validation in specific cohorts, such as bariatric patients and patients with varying ethnicities[23,24]. Further, recent evidence reinforces that a combination of different commonly assessed blood-based biomarkers in addition to direct fibrogenesis markers can provide higher diagnostic accuracy in detecting advanced fibrosis when compared to current protocols. The study of Vilar-Gomez et al[25], reviewed the diagnostic accuracy of several blood-based biomarkers, suggesting an algorithm to diagnose NAFLD patients at risk of fibrosis development. Additionally, the European guidelines recommend the combination of different tests to assess NAFLD, stating that the Fibrometer is a non-invasive alternative to liver biopsy, albeit the guidelines are not clear regarding which specific version of the FibroMeter is preferred[26]. Also, the commercially available biomarker panels and other complementary methods are not accessible for most health services, justifying the search for alternative approaches[25].

Table 1 Blood biomarker panels for nonalcoholic fatty liver disease and nonalcoholic steatohepatitis.
Blood biomarker panels for steatosis
PanelPatientAnthropometryBlood biomarkers
FLI-BMI, Waist circumferenceGGT and TG
HSIPresence of DMBMIAST:ASL
SteatotestSexBMIALT, GGT, TG, A2M, ApoA1, haptoglobin, bilirubin,cholesterol, and glucose
LAPSexWaist circumferenceTG
IONSexWaist to hip ratioALT, TG
NAFLD LFSPresence of DM and MS-AST:ALT, Insulin
Blood biomarker panels for fibrosis
PanelPatientAnthropometryBlood biomarkers
APRI--Platelet count, AST
FIB-4Age-Platelet count, AST, ALT
FibroTestAge, sexBMIGGT, A2M, ApoA1, haptoglobin, and total bilirubin
Fibrometer AgeBody weightPlatelet count, AST, ALT, glucose, ferritin
ELF--Hyaluronic acid, PIIINP and TIMP-1
HepascoreAge, sex-GGT, Hyaluronic acid, PIIINP and TIMP-1
BARDPresence of DMBMIAST:ALT
NFSAge, sex, Presence of DM-Platelet count, AST:ALT, Albumin

The validation study by Wu et al[27] compared different panels of biomarkers in 417 NAFLD patients (156 with advanced fibrosis), showing that when predicting liver fibrosis scores Fibrosis-4 (FIB-4), NAFLD Fibrosis Score (NFS), AST to Platelet Ratio Index (APRI) and BARD score (BARD), it is possible to obtain a prediction of moderate fibrosis based on the receptor operator area under the curve (AUROC; 0.724, 0.671 and 0.609, respectively). The authors argued that FIB-4 and NFS performed better compared to both APRI and BARD scores, which resulted in high false-positive rates. Importantly, this study evaluated NAFLD patients based on the new definition of MAFLD, highlighting that the investigated biomarker panels provided poor performance in this setting[27]. In conclusion, the fact that the aforementioned biomarkers come from different types of procedures makes it hard for human experts to jointly analyze all this information, which motivates the use of machine learning techniques. These models can work with different types of data and discovering the relationship between them to obtain a better prediction.

ARTIFICIAL INTELLIGENCE APPLICATION IN NAFLD

Briefly, AI is an umbrella term, referring to a structured utilization of software and algorithms that analyze a wide range of data, ultimately simulating human cognition and intelligence[6]. Machine learning (ML) is one of the subdisciplines of AI, focusing on learning from data and associating specific patterns with different outcomes. An important advantage of ML techniques is that they allow the modeling of complex problems that depend on multiple input variables, justifying the application of ML methods to potentially fill several gaps in the study of complex diseases, such as NAFLD[6]. This is especially important in the case of NAFLD, which is closely related to metabolic disturbances associated with obesity and metabolic syndrome[28]. Given its complexity, NAFLD presents in different forms, from simple asymptomatic lipid accumulation to symptomatic non-alcoholic steatohepatitis (NASH) characterized by several factors, including steatosis, hepatocellular ballooning, lobular inflammation, and often fibrosis[28]. Machine learning methods are becoming increasingly popular, which has also motivated an increase in the complexity of these models. Particularly, deep learning (DL) models, like convolutional neural networks (CNN), showed promising results in hepatology, especially with high-resolution data such as images and spectrograms[29]. Likewise, CNN models encompass several layers that involve operations like convolution, pooling, and nonlinear activations, making their decisions difficult to understand. Therefore, they represent black-box models, as opposed to interpretable (white-box) techniques, such as regression/decision trees and Bayesian networks[30,31]. Hence, ML could identify patients at risk and guide clinical treatments, whilst considering that the clinical manifestations of NAFLD appear in advanced disease status and the availability and cost of screening methods for the clinicians. Also, ML can help to rank and categorize specific biomarkers and help to elaborate specific "disease signatures", contributing not only to clinical diagnostics, but also provide mechanistic insights for the study of the disease and the development of specific treatments.

MACHINE LEARNING APPROACHES USING BLOOD BIOMARKERS IN HEPATOLOGY

As stated above, the interest in using AI approaches to support clinical decision-making processes in hepatology has increased, albeit current literature is still scarce. Table 2 summarizes the specific studies addressing NAFLD and NASH classification. Initially, the study of Sowa et al[32] showed no differences in the investigated biomarkers (ALT, AST, and apoptotic signaling) between patients with a fibrosis score of 1 or 2. However, combining these parameters using random forests (RF) reached 79% accuracy in fibrosis prediction with a sensitivity of more than 60% and specificity of 77%. Moreover, RF identified the cell death markers M30 and M65 as more important for the decision than the classic liver parameters. Similarly, Yip et al[33] built a model to predict steatosis in a study including 922 individuals with assessment for NAFLD. The four models developed presented good diagnostic precision for steatosis (AUROC was 0.87-0.9), albeit the authors claimed that the “NAFLD ridge score” offered the best balance between efficacy and simplicity. This model included six parameters (serum triglycerides, alanine aminotransferase, high-density lipoprotein cholesterol, hemoglobin A1c, white cell count, and the presence of hypertension) that are routinely available for individuals undergoing medical checkups, and it does not require anthropometric measures, which are not always available. Although there is evident feasibility of the NAFLD ridge score to screen individuals, it still needs additional validation in other ethnicities. The study of Ma et al[34], investigated the predictive power for NAFLD of eleven machine learning techniques, demonstrating that the Bayesian network model had the best performance, revealing that the five most discriminating features (based on information gain scores) to be weight, TG, ALT, GGT, and serum uric acid levels. Thus, in practice, users could focus on these features. Furthermore, Canbay et al[35] compared different scores for the non-invasive detection of NASH. Briefly, using an ensemble feature selection approach for biomarker selection, the authors built a logistic regression model and validated in an independent study cohort of 122 patients. The logistic regression model generated from age, GGT, hemoglobin A1c, M30, and adiponectin had a strong correlation with the non-alcoholic steatohepatitis activity score and demonstrated reasonable performance to discriminate between NAFL and NASH. Likewise, Liu et al[36] performed a retrospective cross-sectional study on 15315 Chinese subjects, where 5878 patients presented NAFLD. The biomarker ranking indicated the body mass index as the most valuable indicator to predict NAFLD, followed by waist circumference, triglycerides, waist-to-height ratio, and alanine aminotransferase. Notably, among seven machine learning models, the extreme gradient boosting (XGBoost) model demonstrated the best prediction ability. Similarly, the XGBoost also presented the highest AUC (0.93), accuracy (0.94), and sensitivity value (0.90) in the study of Pei et al[37], comparing different models for predicting fatty liver Disease risk in 3419 participants, of which 845 had diagnostic confirmation. Importantly, regarding the biomarkers, uric acid, body mass index, and triglycerides were the most decisive risk factors for the ML models, whilst high-density lipoprotein and hemoglobin also counted as important risk factors for prediction. Strikingly, these studies provide insights into ML application in a complex context such as NAFLD patients' prognostics. Notably, while there are investigations using AI techniques and common biomarkers to predict NAFLD and NASH, approaches using AI and novel proposed biomarkers are scarce. For instance, a recent meta-analysis showed that CK-18 is the only marker for NASH presenting external validation, with an AUROC of 0.82[38]. Conversely, a large study conducted by the multicenter NASH Clinical Research Network demonstrated that the addition of routinely available clinical-laboratory parameters to CK-18 measurement did not significantly improve its diagnostic performance[22]. However, it remains unknown whether the use of AI techniques combining different biomarkers in a large and diverse cohort could provide different results. Taken together, the data suggests that ranked blood biomarkers can provide a recognizable signature allowing cost-effective NAFLD prediction and also differentiating NASH patients.

Table 2 Machine learning studies in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis patients.
Ref.
Patients
Investigated biomarker
Model with best performance
Results
Sowa et al[32], 2013126 patientsAlanine aminotransferase; Aspartate aminotransferase; M30; M60; Hyaluronic acidRandon forest79% Accuracy in fibrosis prediction; 60% sensitivity; 77% specificity
Yip et al[33], 2017922 patientsAlanine aminotransferase; High-density lipoprotein cholesterol; Triglycerides; HbA1c; White blood cells; HypertensionRidge score88% Accuracy in steatosis prediction; 92% sensitivity; 90% specificity
Ma et al[34], 201810.508 patients; 2522 NAFLD patientsAge; Sex; Body mass index; Alanine aminotransferase; Aspartate aminotransferase; Alkaline phosphatase; Gamma-glutamyl transpeptidase; Triglycerides; Blood urea nitrogen; Bilirubin; Cholesterol; Creatinine; Fasting glucose; Uric acidBayesian network model83% Accuracy in NAFLD prediction; 68% sensitivity; 94% specificity
Canbay et al[35], 2019164 patients; 122 (validation)Age; HbA1c; Gamma-glutamyl transpeptidase; M30; AdiponectinLogistic regression70% Accuracy in separate NAFLD and NASH
Liu et al[36], 202115.315 patients5878 with NAFLD Body mass index; Waist circumference; Waist-to-height ratio; Alanine aminotransferase; Fasting blood glucose; Gamma-glutamyl transpeptidase; Very-low-density lipoprotein cholesterol; Low-density lipoprotein cholesterol; High-density lipoprotein cholesterol; Systolic blood pressure; Alkaline phosphatase; Diastolic blood pressureXGBoost model79% Accuracy in NAFLD prediction; 61% sensitivity; 90% specificity
Pei et al[37], 20213.419 patients; 845 with fat liver diseasesAge; Height; Hemoglobin; Aspartate aminotransferase; Glucose; Uric acid; Low-density lipoprotein; Alpha-fetoprotein; Triglycerides; High-density lipoprotein; Carcinoembryonic antigenXGBoost model94% accuracy of prediction; 90% sensitivity; 95% specificity
CURRENT CHALLENGES IN SML APPROACHES IN HEPATOLOGY

The term "AI-Chasm" describes the gap between developing and testing an algorithm and the definitive application of the algorithm in clinical practice[39]. Unequivocally, the AI application in medical sciences is auspicious, and current literature is shading light on a plethora of potential applications; however, many challenges for SML approaches using biomarkers in hepatology still await scrutiny.

Firstly, the collection, curation, and preprocessing of patient data is a major concern, since SML methods are data-driven[10]. Notably, the cited studies in this mini-review provide relatively small data from specific populations which could lead to sampling bias whilst limiting the generalization of the obtained results. Further, data collection should be standardized and precise, but should also be monitored for privacy and data security breaches. Secondly, as recently discussed by Quinn et al[40], one of the main aspects of concern in future studies is the understanding that transdisciplinary approaches require cooperation to build a conceptually appropriate framework while also focusing on evaluating the performance of SML algorithms in terms of clinical endpoints and not just predictive accuracy. In addition to these technical challenges, there is also an increasing demand for transparency concerning the predictions of these models, especially in areas that have no computing background. For instance, healthcare professionals and other stakeholders that can benefit from these solutions are still reluctant to the idea of employing these methods, evidencing the necessity of educational programs aimed to explicit information about the involved decision processes. Nevertheless, the field of explainable AI has emerged to address these issues, with the purpose of creating ML techniques that produce explainable models while maintaining a high level of learning performance, enabling humans to understand and trust the predictions to support their decisions[41].

CONCLUSION

Recent advances in the field of biosciences applying machine learning algorithms resulted in promising results for the diagnosis of disease and biomarker study. The main idea is that SML could overcome the limitations of common statistical techniques. For instance, SML identifies data patterns for classification, considering multiple features at once, allowing the ranking and selection of the available blood biomarkers related to disease pathogenesis for the prediction of NAFLD or NASH, minimizing potential errors between the predicted values and the real data. Although the cited studies provide promising results, there are specific limitations that future studies should reduce. For example, most of the studies involved the Chinese population, and these algorithms still need additional validation in heterogeneous populations. The strong association between NAFLD and metabolic syndrome, obesity, and alcohol consumption may be a confounding factor in previous studies, and the application of these methods in diabetic patients with and without NAFLD could shed light on the influence of specific treatments on the performance of these ML methods. Nevertheless, the ability of ML approaches to process multiple variables, map linear and nonlinear interactions, and rank the most important features, in addition to the capability of building accurate prediction models, sets a future direction to its application in complex diseases, including NAFLD and NASH. Future studies should consider the limitations in the current literature and expand the application of these algorithms in different populations, fortifying an already promising tool in medical science.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Brazil

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): E

P-Reviewer: Cengiz M, Turkey; Gadekallu TR, India; Masarone M, Italy; Portius D, Germany S-Editor: Liu JH L-Editor: Filipodia P-Editor: Liu JH

References
1.  Nd AM. Non-Alcoholic Fatty Liver Disease, an Overview. Integr Med (Encinitas). 2019;18:42-49.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65:1038-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1490]  [Cited by in F6Publishing: 1706]  [Article Influence: 213.3]  [Reference Citation Analysis (1)]
3.  Geier A, Tiniakos D, Denk H, Trauner M. From the origin of NASH to the future of metabolic fatty liver disease. Gut. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 36]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
4.  Juanola O, Martínez-López S, Francés R, Gómez-Hurtado I. Non-Alcoholic Fatty Liver Disease: Metabolic, Genetic, Epigenetic and Environmental Risk Factors. Int J Environ Res Public Health. 2021;18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 87]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
5.  Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, Harrison SA, Brunt EM, Sanyal AJ. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018;67:328-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3544]  [Cited by in F6Publishing: 4072]  [Article Influence: 678.7]  [Reference Citation Analysis (7)]
6.  Aggarwal P, Alkhouri N. Artificial Intelligence in Nonalcoholic Fatty Liver Disease: A New Frontier in Diagnosis and Treatment. Clin Liver Dis (Hoboken). 2021;17:392-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
7.  Bertsimas D, Kung J, Trichakis N, Wang Y, Hirose R, Vagefi PA. Development and validation of an optimized prediction of mortality for candidates awaiting liver transplantation. Am J Transplant. 2019;19:1109-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
8.  Shousha HI, Awad AH, Omran DA, Elnegouly MM, Mabrouk M. Data Mining and Machine Learning Algorithms Using IL28B Genotype and Biochemical Markers Best Predicted Advanced Liver Fibrosis in Chronic Hepatitis C. Jpn J Infect Dis. 2018;71:51-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
9.  Konerman MA, Lu D, Zhang Y, Thomson M, Zhu J, Verma A, Liu B, Talaat N, Balis U, Higgins PDR, Lok ASF, Waljee AK. Assessing risk of fibrosis progression and liver-related clinical outcomes among patients with both early stage and advanced chronic hepatitis C. PLoS One. 2017;12:e0187344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
10.  Spann A, Yasodhara A, Kang J, Watt K, Wang B, Goldenberg A, Bhat M. Applying Machine Learning in Liver Disease and Transplantation: A Comprehensive Review. Hepatology. 2020;71:1093-1105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 78]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
11.  Balsano C, Alisi A, Brunetto MR, Invernizzi P, Burra P, Piscaglia F; Special Interest Group (SIG) Artificial Intelligence and Liver Diseases;  Italian Association for the Study of the Liver (AISF). The application of artificial intelligence in hepatology: A systematic review. Dig Liver Dis. 2022;54:299-308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
12.  Su TH, Wu CH, Kao JH. Artificial intelligence in precision medicine in hepatology. J Gastroenterol Hepatol. 2021;36:569-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
13.  Califf RM. Biomarker definitions and their applications. Exp Biol Med (Maywood). 2018;243:213-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 576]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
14.  Younossi ZM, Loomba R, Anstee QM, Rinella ME, Bugianesi E, Marchesini G, Neuschwander-Tetri BA, Serfaty L, Negro F, Caldwell SH, Ratziu V, Corey KE, Friedman SL, Abdelmalek MF, Harrison SA, Sanyal AJ, Lavine JE, Mathurin P, Charlton MR, Goodman ZD, Chalasani NP, Kowdley KV, George J, Lindor K. Diagnostic modalities for nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, and associated fibrosis. Hepatology. 2018;68:349-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 250]  [Article Influence: 41.7]  [Reference Citation Analysis (0)]
15.  Castera L, Friedrich-Rust M, Loomba R. Noninvasive Assessment of Liver Disease in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2019;156:1264-1281.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 619]  [Cited by in F6Publishing: 783]  [Article Influence: 156.6]  [Reference Citation Analysis (0)]
16.  Neuman MG, Cohen LB, Nanau RM. Biomarkers in nonalcoholic fatty liver disease. Can J Gastroenterol Hepatol. 2014;28:607-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 106]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
17.  Fracanzani AL, Valenti L, Bugianesi E, Andreoletti M, Colli A, Vanni E, Bertelli C, Fatta E, Bignamini D, Marchesini G, Fargion S. Risk of severe liver disease in nonalcoholic fatty liver disease with normal aminotransferase levels: a role for insulin resistance and diabetes. Hepatology. 2008;48:792-798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 441]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
18.  Mofrad P, Contos MJ, Haque M, Sargeant C, Fisher RA, Luketic VA, Sterling RK, Shiffman ML, Stravitz RT, Sanyal AJ. Clinical and histologic spectrum of nonalcoholic fatty liver disease associated with normal ALT values. Hepatology. 2003;37:1286-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 787]  [Cited by in F6Publishing: 766]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
19.  He L, Deng L, Zhang Q, Guo J, Zhou J, Song W, Yuan F. Diagnostic Value of CK-18, FGF-21, and Related Biomarker Panel in Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Biomed Res Int. 2017;2017:9729107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
20.  de Haas EC, di Pietro A, Simpson KL, Meijer C, Suurmeijer AJ, Lancashire LJ, Cummings J, de Jong S, de Vries EG, Dive C, Gietema JA. Clinical evaluation of M30 and M65 ELISA cell death assays as circulating biomarkers in a drug-sensitive tumor, testicular cancer. Neoplasia. 2008;10:1041-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
21.  Tamimi TI, Elgouhari HM, Alkhouri N, Yerian LM, Berk MP, Lopez R, Schauer PR, Zein NN, Feldstein AE. An apoptosis panel for nonalcoholic steatohepatitis diagnosis. J Hepatol. 2011;54:1224-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 127]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
22.  Feldstein AE, Wieckowska A, Lopez AR, Liu YC, Zein NN, McCullough AJ. Cytokeratin-18 fragment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: a multicenter validation study. Hepatology. 2009;50:1072-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 498]  [Cited by in F6Publishing: 484]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
23.  Fallatah HI. Noninvasive Biomarkers of Liver Fibrosis: An Overview. Adv Hepatol. 2014;357287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
24.  Soresi M, Giannitrapani L, Cervello M, Licata A, Montalto G. Non invasive tools for the diagnosis of liver cirrhosis. World J Gastroenterol. 2014;20:18131-18150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 61]  [Cited by in F6Publishing: 57]  [Article Influence: 5.7]  [Reference Citation Analysis (1)]
25.  Vilar-Gomez E, Chalasani N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J Hepatol. 2018;68:305-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 331]  [Cited by in F6Publishing: 378]  [Article Influence: 63.0]  [Reference Citation Analysis (0)]
26.  European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD);  European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64:1388-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2290]  [Cited by in F6Publishing: 2694]  [Article Influence: 336.8]  [Reference Citation Analysis (2)]
27.  Wu YL, Kumar R, Wang MF, Singh M, Huang JF, Zhu YY, Lin S. Validation of conventional non-invasive fibrosis scoring systems in patients with metabolic associated fatty liver disease. World J Gastroenterol. 2021;27:5753-5763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 18]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
28.  Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62:S47-S64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1516]  [Cited by in F6Publishing: 1764]  [Article Influence: 196.0]  [Reference Citation Analysis (0)]
29.  LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36149]  [Cited by in F6Publishing: 17282]  [Article Influence: 1920.2]  [Reference Citation Analysis (0)]
30.  Ribeiro E, Uhl A, Wimmer G, Häfner M. Exploring Deep Learning and Transfer Learning for Colonic Polyp Classification. Comput Math Methods Med. 2016;2016:6584725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
31.  Bernal J, Tajkbaksh N, Sanchez FJ, Matuszewski BJ, Hao Chen, Lequan Yu, Angermann Q, Romain O, Rustad B, Balasingham I, Pogorelov K, Sungbin Choi, Debard Q, Maier-Hein L, Speidel S, Stoyanov D, Brandao P, Cordova H, Sanchez-Montes C, Gurudu SR, Fernandez-Esparrach G, Dray X, Jianming Liang, Histace A. Comparative Validation of Polyp Detection Methods in Video Colonoscopy: Results From the MICCAI 2015 Endoscopic Vision Challenge. IEEE Trans Med Imaging. 2017;36:1231-1249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 156]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
32.  Sowa JP, Heider D, Bechmann LP, Gerken G, Hoffmann D, Canbay A. Novel algorithm for non-invasive assessment of fibrosis in NAFLD. PLoS One. 2013;8:e62439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (1)]
33.  Yip TC, Ma AJ, Wong VW, Tse YK, Chan HL, Yuen PC, Wong GL. Laboratory parameter-based machine learning model for excluding non-alcoholic fatty liver disease (NAFLD) in the general population. Aliment Pharmacol Ther. 2017;46:447-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 109]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
34.  Ma H, Xu CF, Shen Z, Yu CH, Li YM. Application of Machine Learning Techniques for Clinical Predictive Modeling: A Cross-Sectional Study on Nonalcoholic Fatty Liver Disease in China. Biomed Res Int. 2018;2018:4304376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
35.  Canbay A, Kälsch J, Neumann U, Rau M, Hohenester S, Baba HA, Rust C, Geier A, Heider D, Sowa JP. Non-invasive assessment of NAFLD as systemic disease-A machine learning perspective. PLoS One. 2019;14:e0214436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
36.  Liu YX, Liu X, Cen C, Li X, Liu JM, Ming ZY, Yu SF, Tang XF, Zhou L, Yu J, Huang KJ, Zheng SS. Comparison and development of advanced machine learning tools to predict nonalcoholic fatty liver disease: An extended study. Hepatobiliary Pancreat Dis Int. 2021;20:409-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
37.  Pei X, Deng Q, Liu Z, Yan X, Sun W. Machine Learning Algorithms for Predicting Fatty Liver Disease. Ann Nutr Metab. 2021;77:38-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (1)]
38.  Musso G, Gambino R, Cassader M, Pagano G. Meta-analysis: natural history of non-alcoholic fatty liver disease (NAFLD) and diagnostic accuracy of non-invasive tests for liver disease severity. Ann Med. 2011;43:617-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 886]  [Cited by in F6Publishing: 854]  [Article Influence: 65.7]  [Reference Citation Analysis (0)]
39.  Keane PA, Topol EJ. With an eye to AI and autonomous diagnosis. NPJ Digit Med. 2018;1:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 122]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
40.  Quinn TP, Senadeera M, Jacobs S, Coghlan S, Le V. Trust and medical AI: the challenges we face and the expertise needed to overcome them. J Am Med Inform Assoc. 2021;28:890-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
41.  Barredo Arrieta A, Díaz-Rodríguez N, Del Ser J, Bennetot A, Tabik S, Barbado A, Garcia S, Gil-Lopez S, Molina D, Benjamins R, Chatila R, Herrera F. Explainable Artificial Intelligence (XAI): Concepts, taxonomies, opportunities and challenges toward responsible AI. Information Fusion. 2020;82.  [PubMed]  [DOI]  [Cited in This Article: ]