Next Article in Journal
Predictors of Corporal Punishment during the COVID-19 Pandemic
Previous Article in Journal
The Impact of Prenatal Alcohol Exposure on the Autonomic Nervous System and Cardiovascular System in Rats in a Sex-Specific Manner
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Metabolic-Associated Steatotic Liver Disease (MASLD): A New Term for a More Appropriate Therapy in Pediatrics?

by
Mosca Antonella
*,
Andrea Pietrobattista
and
Giuseppe Maggiore
Hepatology and Liver Transplant Unit, ERN RARE LIVER, Bambino Gesù Children’s Hospital, Istituto di ricerca, 00165 Rome, Italy
*
Author to whom correspondence should be addressed.
Pediatr. Rep. 2024, 16(2), 288-299; https://doi.org/10.3390/pediatric16020025
Submission received: 15 January 2024 / Accepted: 15 March 2024 / Published: 10 April 2024
The term “non-alcoholic fatty liver disease” (NAFLD) has been, for a long time, used to describe the spectrum of liver lesions encompassing steatosis, steatohepatitis (NASH), and steatotic cirrhosis. A form of chronic liver disease, it is prevalent worldwide, affecting more than 30% of the global population [1]. Its histological classification has been further expanded by various scoring systems concerning the degrees of steatosis, disease activity, and fibrosis [2].
In 2020, a proposal was made to change this terminology by introducing the term “metabolic dysfunction-associated fatty liver disease” (MAFLD) to replace NAFLD. While some have accepted this new terminology, concerns have been raised about the mixing of etiologies [3]. One area of particular concern was the potential negative impact of changes in diagnostic criteria for the disease in terms of biomarkers and therapeutic development. For this reason, in 2023, an expert committee used an interactive approach involving intentional sampling to generate a large global panel for this Delphi study. Based on publication records and research on fatty liver disease, the co-chairs identified 31 experts in clinical care (liver, diabetes, obesity, and nutrition), public health, and patient representation, who collectively formed the lead author group. From this consensus, it has been decided that “steatotic liver disease” remains a general term for “fatty liver disease”, encompassing the different etiologies. The term “steatohepatitis” has been deemed an important pathophysiological concept that should be retained, but “non-alcoholic steatohepatitis” (NASH) should be replaced with the term “metabolic dysfunction-associated steatohepatitis” (MASH). There was consensus to change the definition by acknowledging the presence of at least 5 out of 140 cardio-metabolic risk factors. Those with no metabolic parameters and no known cause were thought to have cryptogenic steatotic liver disease [4].
The new definitions definitively link the pathophysiology of fatty liver with metabolic dysfunction and insulin resistance, reinforcing the role of cardiometabolic risk factors associated with “metabolic dysfunction-associated steatosis liver disease”—MASLD [5].
MASLD is characterized by liver damage associated with metabolic dysfunction, lobular inflammation, ballooning, and fibrosis [6]. This condition may progress to cirrhosis and possibly to hepatocellular carcinoma (HCC). To date, a multitude of genetic, epigenetic, and environmental MASLD modifiers have been reported, especially in the perinatal period and in the first years of life. However, diagnosis is often delayed due to its non-specific clinical manifestation, with its discovery commonly occurring during routine examinations.
MASLD, however, could lead to new scenarios for diagnosis on the pediatric level; even for children, the presence of five simple components of metabolic syndrome persists, including central or general obesity. Moreover, in this consensus, a re-evaluation of the definitions of steatohepatitis in the pediatric setting would be advantageous for clinicians’ therapeutic approach. On the other hand, the use of the term “metabolic” in the nomenclature could create confusion in the pediatric context since inborn errors of metabolism are referred to as “metabolic liver disease” [4,5].
A recent small pediatric study showed that the new MASLD criteria performed better than that for NAFLD in selecting children with obesity and higher cardio-metabolic risk, including the risk of kidney damage. Patients with MASLD showed an overall worse cardiovascular and metabolic risk profile than their counterparts with NAFLD, as supported by the presence and significant increase in HOMA-IR, uric acid, and TG/HDL-c ratio values and a higher percentage of kidney injury [7].
Approximately 20–30% of adult patients with MASLD develop MASH leading to liver cirrhosis and, rarely, HCC. Metabolic syndrome and its components such as obesity, impaired glucose metabolism, hypertension, and dyslipidemia are all closely associated with insulin resistance.
1
The Pathogenesis of MASLD
The pathogenesis of MASLD remains largely unknown, but, today, the fields of study are different.
1.1 Lipogenesis: Adipose tissue is a metabolically active endocrine organ that causes the release of proinflammatory cytokines (TNF-α and Interleukin (IL)-6), whereas beneficial adipokines, such as adiponectin, become suppressed [8]. This situation leads to the development of peripheral insulin resistance with hyperinsulinemia and increased fatty acid delivery into hepatocytes. The disruption of normal insulin signaling in the hepatocyte and the increased abundance of fatty acids leads to disordered lipid metabolism with over-activation of de novo lipogenesis (DNL) transcriptional factors, causing more fatty acid and glucose products to be shunted into these lipogenetic pathways. Beta-oxidation in the mitochondria is also inhibited, as well as very-low-density lipoprotein (VLDL) packaging and export, leading to the build-up of triglycerides in the hepatocytes. Uncontrolled and incomplete lipid oxidation, oxidative stress, and protein response activation are explained as two well-characterized pathways that promote cell death in NASH [9].
1.2 Oxidative Stress: In the presence of high amounts of fatty acids in hepatocytes, oxidative stress occurs due to high levels of reactive oxygen/nitrogen species (ROS/RNS) and lipid peroxidation that are generated during the metabolism of free fatty acids in microsomes, peroxisomes, and in the mitochondria [10]. The peroxidation of plasma and intracellular membranes may cause direct cell necrosis or apoptosis, while the ROS-induced expression of Fas ligand on hepatocytes may induce cell death.
1.3 Gut microbiota and Cytokines: An altered microbiome (“dysbiosis”) can contribute to liver damage. Human studies have documented a fecal microbiome signature characterized by increased Proteobacteria and Bacteroidetes along with a decrease in Firmicutes in patients with obesity and NASH [11]. Early studies have suggested that the intestinal microbiota is responsible for the synthesis of various hepatotoxic bacterial substances (e.g., ammonia, phenols, and ethanol). The main bacterial product involved in the pathogenesis of NASH/NAFLD is LPS, an active component of bacterial endotoxin. The endogenous production of LPS due to bacterial death induces its translocation through the intestinal capillaries thanks to a TLR4-dependent mechanism [12]. LPS, through a complex process of association with LPS-binding protein and CD14, activates TLR4 located on different inflammatory cells, increasing the expression of target genes involved in the synthesis of inflammatory cytokines such as TNF-α, IL-1, and IL-6, thus promoting insulin resistance, hepatic steatosis, hepatic inflammation, and fibrogenesis [13].
1.4 Genetics: Single nucleotide polymorphisms (SNPs) in the genes involved in lipid metabolism (Lipin 1—LPIN1, patatin-like phospholipase domain containing-3—PNPLA3), oxidative stress (superoxide dismutase 2—SOD2), insulin signaling (insulin receptor substrate-1—IRS-1), and fibrogenesis (Kruppel-like factor 6—KLF6) have been associated with severe NASH, but a very interesting interaction has recently been reported between genetic risk factors (PNPLA3 I148M) and the severity of steatosis and fibrosis [14]. PNPLA3 is a member of the patatin-like phospholipase family. Several studies have established that the presence of PNPLA3-GG/GC is associated with an increased risk of advanced fibrosis among patients with a variety of liver diseases and is an independent risk factor for HCC among patients with NASH [15].
2
Diagnosis
The proposed diagnostic criteria for MAFLD are based on the evidence of fatty liver disease, which can be detected by blood biomarkers, imaging techniques, or liver histology. In recent years, several non-invasive screening and diagnostic assessments have been developed for the evaluation of NAFLD.
2.1 Laboratory tests. ALT has long been accepted as an accurate indicator of liver damage and inflammation; it has been commonly used in disease monitoring and early-stage clinical trials for NAFLD. NASPGHAN recommends the assessment of ALT levels in children over 10 years of age with a BMI ≥ the 85th percentile as a screening measure for NAFLD [16]. Numerous studies have shown that the biochemical parameters of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), associated with hemoglobin A1c and the homeostatic assessment model (HOMA)-IR, could provide good prediction indices of NAFLD in patients with metabolic syndrome or even in the general population [17].
Over the past 20 years, non-invasive biomarkers have been developed for the detection of steatosis and fibrosis in patients with NAFLD. In clinical practice, the NAFLD Fibrosis Score (NFS), Fibrosis-4 Index (FIB-4), and AST to Platelet Ratio Index (APRI), work well to rule out advanced fibrosis–cirrhosis and, therefore, could be used as a first-line classification to identify patients at a low risk of advanced fibrosis. However, it may be too late to take action to reverse fibrosis. Thus, the ability to detect simple steatosis and steatohepatitis early and non-invasively in patients with fatty liver is critical to preventing disease progression [18].
A systematic review showed that FIB-4 predicted advanced fibrosis more accurately compared to the NFS, APRI, and BARD (a score that considers the BMI, AST/ALT ratio, and diabetes). Another meta-analysis concluded that among the non-invasive simple blood scores, the NFS and FIB-4 offer the best diagnostic performance to detect advanced fibrosis in an adult population [19].
The Hepamet Fibrosis Score (HFS) was developed and validated by Ampuero et al. and is one of the most recent non-invasive fibrosis scores; it performs better than FIB-4 and NFS for the diagnosis of advanced fibrosis. The HFS was developed using data from nearly 2500 patients from five countries (Spain, France, Italy, Cuba, and China), comprising various ethnicities (Caucasian, Latino, and Asian populations) and different rates of baseline characteristics (diabetes, obesity, prevalence of fibrosis) [20]. Recently, Prasoppokakorn et al. reported the Fibrosis-8 (FIB-8) score [the FIB-4 variables plus BMI, albumin/globulin ratio, serum gamma glutamyl transpeptidase (GGT) level, and presence of T2DM] for the diagnosis of significant fibrosis. FIB-8 has been shown to have a sensitivity and specificity of 92.4% and 67.5%, respectively [21].
In the pediatric setting, the use of these scores is less frequent; a study showed that Hepamet and APRI scores perform better than the NFS and FIB-4 for the identification of significant fibrosis in patients with NAFLD but that they do not have PPVs high enough to be considered a diagnostic tool. In 173 (60.4%) adolescents presenting with fibrosis on histological analysis, the APRI and Hepamet had significant accuracy (p < 0.001) in distinguishing subjects with fibrosis ≥ 1, while the NFS and FIB-4 did not. The APRI had a positive predictive value (PPV) of 62.77% and Hepamet had a PPV of 63.24%. Therefore, the early detection of fibrosis in NAFLD can significantly reduce the use of liver biopsy, which is not entirely without complications in children [22].
2.2 Serum biomarkers of fibrosis. To date, we can classify specific biomarkers of fibrosis according to their molecular structure:
(1)
Collagens (procollagen I and III, released in serum during deposition and remodeling of the extracellular matrix; collagen IV, released during the degradation and remodeling of the extracellular matrix).
(2)
Glycoproteins and polysaccharides (hyaluronic acid, laminin, tenascin, YKL-40).
(3)
Collagenases and their inhibitors; metalloproteinases (MMPs) and their inhibitors (TIMPs).
(4)
Cytokines (TNF-a, IL-6, IL-8, IL-1B, and IL-10) [23].
Recently, a new panel, ELF, has been defined, which includes hyaluronic acid, type III amino terminal procollagen propeptide (PIIINP), and tissue inhibitor of metalloproteinases-1 (TIMP-1) and recommended for the determination of fibrosis [24]. ELF demonstrated good diagnostic accuracy in detecting advanced fibrosis, with a mean sensitivity and specificity of 65% and 86%, respectively, using the higher threshold of 9.8, based on a systematic review and meta-analysis by Vali et al. [25].
Hyaluronic acid (HA) is a glycosaminoglycan synthesized by hepatic stellate cells and degraded by hepatic sinusoidal cells and is a component of the extracellular matrix. In patients with chronic liver disease, especially in those with cirrhosis, its presence is an indication of the impaired functioning of hepatic sinusoidal cells and reflects an increase in the fibrogenic process.
PRO-C3, a neo-epitope-specific competitive enzyme-linked immunosorbent assay (ELISA) for PIIINP, is a relatively new direct marker of active fibrogenesis, mostly in patients with chronic hepatitis C [26]. Recent results support the use of PRO-C3 as an effective candidate biomarker for the non-invasive assessment of liver fibrosis in NAFLD. Subsequently, an algorithm called ADAPT, which incorporates PRO-C3 along with age, the presence of diabetes, and platelet count, was developed to detect advanced fibrosis in a multinational retrospective study (Australia, UK, and Japan) with 431 biopsy-proven NAFLD patients. ADAPT exhibited a negative predictive value of 96.6%, which was superior to PRO-C3 as a stand-alone marker and to existing simple fibrosis scores, namely, APRI, FIB-4, and NFS [27].
In the pediatric setting, in a study of 204 children/adolescents with NAFLD, children with NASH had higher PIIINP plasma levels and APRI and FIB-4 scores than those without NASH (p < 0.001). However, the PIIINP levels had a much better diagnostic performance and accuracy than the APRI and FIB-4 scores in predicting the stage of liver fibrosis. The PIIINP levels correlated with the total NAFLD activity score (NAS) and its constituent components (p < 0.0001). The risk of NASH or F ≥ 2 fibrosis increases progressively with increasing PIIINP levels (p < 0.0001). For every 3.6 ng/mL increase in PIIINP levels, the likelihood of having F ≥ 2 fibrosis increased approximately 14-fold (OR 14.1, 95% CI 5.50–35.8, p < 0.0001) [28].
In a number of studies, it has been shown that hepatic steatosis associated with obesity is characterized by the increased production of inflammatory cytokines by hepatocytes, which, in turn, is secondary to NF-kB stimulation, with the activation of Kupffer cells and hepatic and systemic insulin resistance. The cytokines involved are capable of producing all the classic histological features of NASH, including hepatocyte necrosis/apoptosis (TNF alpha, TGF-beta), neutrophil chemotaxis (IL-8), hepatic stellate cell activation (HSC) (TNF-alpha, TGF-beta) and Mallory body formation (TGF-beta) [29].
Another marker is fibroblast growth factor 21 (FGF-21). In fact, data generated from both adult and pediatric cohorts demonstrated that the circulating FGF-21 levels were positively correlated with NAFLD and NASH [30].
In a study of 203 obese adolescents, the authors showed that FGF-21 as a biomarker could improve the accuracy of identifying obese children with dyslipidemia and insulin resistance as well as high-grade fatty liver disease [31]. FGF-21 levels can predict the onset of simple steatosis with an accuracy of 0.661. These findings suggest that FGF-21 may have a role in identifying and monitoring the onset and prognosis of patients with NAFLD [32].
In summary, all clinical scores based on simple serum biomarkers of fibrosis are inexpensive, reproducible, and have a high sensitivity to rule out advanced fibrosis. In contrast, fibrosis-specific serum biomarkers are better at identifying patients with significant fibrosis and advanced fibrosis, but are generally more expensive and not routinely available. In addition, most of the non-invasive serum biomarkers for fibrosis have been developed and validated in the secondary and tertiary settings with a considerably higher prevalence of advanced fibrosis than in the general population, thus limiting their applicability in the primary care setting.
2.3 Imaging tests. Fatty liver disease is often diagnosed incidentally via imaging checks such as abdominal ultrasound, CT scan, or magnetic resonance imaging (MRI). The most common imaging method for diagnosis is abdominal ultrasound, which is easily accessible and can demonstrate the infiltration of fat into the liver. However, when steatosis is less than 30%, the sensitivity is significantly reduced. An alternative diagnostic method is MRI, which is highly sensitive for small amounts of isolated steatosis. Magnetic resonance spectroscopy (MRS) measures proton signals as a function of their resonant frequency to separate the signal fractions of fat and water. MRS can detect small amounts of liver fat and is considered the most accurate non-invasive method of quantifying liver fat, but this modality is not readily available or convenient [33].
More recently, ultrasound-based measurements of liver stiffness can be integrated into conventional ultrasound devices such as acoustic resonance forced pulse imaging (ARFI) and shear wave elastography (SWE) or obtained using a dedicated device, most commonly VCTE, commercially available as the FibroScan. ARFI and SWE use high-frequency ultrasonic pulses to generate fine waves and require the operator to define a region of interest and obtain a series of liver stiffness measurements. A limited number of studies of SWE and ARFI in patients with NAFLD has demonstrated very good diagnostic accuracy for advanced fibrosis [34].
3
Therapy
The treatment of MAFLD, in addition to diet and exercise, is based on the use of omega-3 fatty acids. It has been demonstrated well that polyunsaturated fatty acids of the omega-3 series, eicosapentaenoic acid and DHA, improve hepatic lipid metabolism and adipose tissue function and act as anti-inflammatory agents. A randomized study conducted in a population of 60 overweight or obese children with NAFLD tested the efficacy of DHA (250 mg/day and 500 mg/day compared to placebo) on liver fat content as assessed by ultrasound. Both dosages were able to improve steatosis, triglycerides, and ALT levels, while the dosage of DHA 250 mg/day was able to improve the histological parameters of NAFLD [35].
In a randomized, double-blind, placebo-controlled trial, the effect of DHA (500 mg) with vitamin D (800 IU) was tested in obese children with NAFLD and vitamin D deficiency. The treatment with DHA and vitamin D reduced the NAS score, HSC activation, and fibrillar collagen content. In addition, triglycerides, ALT, and IR decreased after treatment [36]. We recently carried out a randomized, double-blind, placebo-controlled trial to test the efficacy and safety of a mixture of vitamin E and hydroxytyrosol, an olive oil phenol, in adolescents with NAFLD. Patients, randomized, received either two capsules combining 7.5 mg hydroxytyrosol and 10 mg vitamin E per day or a placebo. After 4 months, the children in the treated arm showed a decrease in IR, triglyceride levels, and oxidative stress and inflammation parameters [37]. A new Phase 3 trial in pediatric NASH is currently investigating obeticholic acid, and has shown a ≥stage 1 improvement in liver fibrosis compared to the control (23% vs. 12%). However, the frequent presence of itching as a side effect (51%), the increase in serum cholesterol requiring statin therapy, and increased number of hepatobiliary events seem to outweigh the benefits. The final results of this study are yet to be published at the time of writing this article (NCT02548351) [38]. All the trials conducted on pediatric NAFLD/NASH patients to date are reported in Table 1.
In a prospective study, 101 adults with biopsy-proven NASH were put on a low-calorie diet and then randomly assigned to pioglitazone for 18 months, and the results showed that 58% achieved a two-point reduction ≥ steatosis without worsening fibrosis compared to 17% for those on placebo and 51% exhibited resolution of NASH compared to 19% for those on placebo. Liraglutide and Dulaglutide have both shown efficacy in studies investigating their use. More recently, a study conducted a 72-week, double-blind, placebo-controlled, phase II study of biopsy-confirmed NASH on Semaglutide demonstrated the resolution of NASH in 36–59% of the treated subjects compared to 17% of the controls [39].
In conclusion, lifestyle modifications remain the frontline intervention. With the increasing burden of cirrhotic disease and transplantation in adulthood, additional pharmaceutical intervention could also play a future role in the management of MAFLD in some children having to use adult medications as early as adolescence.

Author Contributions

M.A. and G.M. conceptualization, formal analysis, writing of the original draft, and editing. A.P. contributed to conceptualization, formal analysis, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Han, S.K.; Baik, S.K.; Kim, M.Y. Non-alcoholic fatty liver disease: Definition and subtypes. Clin. Mol. Hepatol. 2023, 29, S5–S16. [Google Scholar] [CrossRef] [PubMed]
  2. Nascimbeni, F.; Ballestri, S.; Machado, M.V.; Mantovani, A.; Cortez-Pinto, H.; Targher, G.; Lonardo, A. Clinical relevance of liver histopathology and different histological classifications of NASH in adults. Expert Rev. Gastroenterol. Hepatol. 2018, 12, 351–367. [Google Scholar] [CrossRef] [PubMed]
  3. Eslam, M.; Sanyal, A.J.; George, J.; International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef] [PubMed]
  4. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; NAFLD Nomenclature consensus group. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J. Hepatol. 2023, 79, 1542–1556. [Google Scholar] [CrossRef] [PubMed]
  5. Rinella, M.E.; Sookoian, S. From NAFLD to MASLD: Updated Naming and Diagnosis Criteria for Fatty Liver Disease. J. Lipid Res. 2024, 65, 100485. [Google Scholar] [CrossRef] [PubMed]
  6. Ilagan-Ying, Y.C.; Banini, B.A.; Do, A.; Lam, R.; Lim, J.K. Screening, Diagnosis, and Staging of Non-Alcoholic Fatty Liver Disease (NAFLD): Application of Society Guidelines to Clinical Practice. Curr. Gastroenterol. Rep. 2023, 25, 213–224. [Google Scholar] [CrossRef] [PubMed]
  7. De, A.; Bhagat, N.; Mehta, M.; Taneja, S.; Duseja, A. Metabolic dysfunction-associated steatotic liver disease (MASLD) definition is better than MAFLD criteria for lean patients with NAFLD. J. Hepatol. 2023, 80, e61–e62. [Google Scholar] [CrossRef] [PubMed]
  8. Duan, Y.; Pan, X.; Luo, J.; Xiao, X.; Li, J.; Bestman, P.L.; Luo, M. Association of Inflammatory Cytokines With Non-Alcoholic Fatty Liver Disease. Front. Immunol. 2022, 13, 880298. [Google Scholar] [CrossRef] [PubMed]
  9. Geng, Y.; Faber, K.N.; de Meijer, V.E.; Blokzijl, H.; Moshage, H. How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol. Int. 2021, 15, 21–35. [Google Scholar] [CrossRef]
  10. Mann, J.P.; Raponi, M.; Nobili, V. Clinical implications of understanding the association between oxidative stress and pediatric NAFLD. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 371–382. [Google Scholar] [CrossRef]
  11. Del Chierico, F.; Nobili, V.; Vernocchi, P.; Russo, A.; De Stefanis, C.; Gnani, D.; Furlanello, C.; Zandonà, A.; Paci, P.; Capuani, G.; et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 2017, 65, 451–464. [Google Scholar] [CrossRef] [PubMed]
  12. Khanmohammadi, S.; Kuchay, M.S. Toll-like receptors and metabolic (dysfunction)-associated fatty liver disease. Pharmacol. Res. 2022, 185, 106507. [Google Scholar] [CrossRef] [PubMed]
  13. Nobili, V.; Alisi, A.; Mosca, A.; Della Corte, C.; Veraldi, S.; De Vito, R.; De Stefanis, C.; D’Oria, V.; Jahnel, J.; Zohrer, E.; et al. Hepatic farnesoid X receptor protein level and circulating fibroblast growth factor 19 concentration in children with NAFLD. Liver Int. 2018, 38, 342–349. [Google Scholar] [CrossRef] [PubMed]
  14. Carpino, G.; Pastori, D.; Baratta, F.; Overi, D.; Labbadia, G.; Polimeni, L.; Di Costanzo, A.; Pannitteri, G.; Carnevale, R.; Del Ben, M.; et al. PNPLA3 variant and portal/periportal histological pattern in patients with biopsy-proven non-alcoholic fatty liver disease: A possible role for oxidative stress. Sci. Rep. 2017, 7, 15756. [Google Scholar] [CrossRef] [PubMed]
  15. Vespasiani-Gentilucci, U.; Gallo, P.; Porcari, A.; Carotti, S.; Galati, G.; Piccioni, L.; De Vincentis, A.; Dell’unto, C.; Vorini, F.; Morini, S.; et al. The PNPLA3 rs738409 C > G polymorphism is associated with the risk of progression to cirrhosis in NAFLD patients. Scand. J. Gastroenterol. 2016, 51, 967–973. [Google Scholar] [CrossRef] [PubMed]
  16. Vos, M.B.; Abrams, S.H.; Barlow, S.E.; Caprio, S.; Daniels, S.R.; Kohli, R.; Mouzaki, M.; Sathya, P.; Schwimmer, J.B.; Sundaram, S.S.; et al. NASPGHAN Clinical Practice Guideline for the Diagnosis and Treatment of Nonalcoholic Fatty Liver Disease in Children: Recommendations from the Expert Committee on NAFLD (ECON) and the North American Society of Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN). J. Pediatr. Gastroenterol. Nutr. 2017, 64, 319–334. [Google Scholar] [CrossRef] [PubMed]
  17. Ajmera, V.; Loomba, R. Imaging biomarkers of NAFLD, NASH, and fibrosis. Mol. Metab. 2021, 50, 101167. [Google Scholar] [CrossRef] [PubMed]
  18. Chandra Kumar, C.V.; Skantha, R.; Chan, W.K. Non-invasive assessment of metabolic dysfunction-associated fatty liver disease. Ther. Adv. Endocrinol. Metab. 2022, 13, 20420188221139614. [Google Scholar] [CrossRef] [PubMed]
  19. Lee, J.; Vali, Y.; Boursier, J.; Spijker, R.; Anstee, Q.M.; Bossuyt, P.M.; Zafarmand, M.H. Prognostic accuracy of FIB-4, NAFLD fibrosis score and APRI for NAFLD-related events: A systematic review. Liver Int. 2020, 41, 261–270. [Google Scholar] [CrossRef]
  20. Ampuero, J.; Pais, R.; Aller, R.; Gallego-Durán, R.; Crespo, J.; García-Monzón, C.; Boursier, J.; Vilar, E.; Petta, S.; Zheng, M.-H.; et al. Development and Validation of Hepamet Fibrosis Scoring System-A Simple, Noninvasive Test to Identify Patients With Nonalcoholic Fatty Liver Disease With Advanced Fibrosis. Clin. Gastroenterol. Hepatol. 2019, 18, 216–225.e5. [Google Scholar] [CrossRef]
  21. Prasoppokakorn, T.; Chan, W.-K.; Wong, V.W.-S.; Pitisuttithum, P.; Mahadeva, S.; Mustapha, N.R.N.; Wong, G.L.-H.; Leung, H.H.-W.; Sripongpun, P.; Treeprasertsuk, S. Validation model of fibrosis-8 index score to predict significant fibrosis among patients with nonalcoholic fatty liver disease. World J. Gastroenterol. 2022, 28, 1563–1573. [Google Scholar] [CrossRef]
  22. Mosca, A.; Della Volpe, L.; Alisi, A.; Veraldi, S.; Francalanci, P.; Maggiore, G. Non-Invasive Diagnostic Test for Advanced Fibrosis in Adolescents With Non-Alcoholic Fatty Liver Disease. Front. Pediatr. 2022, 10, 885576. [Google Scholar] [CrossRef] [PubMed]
  23. Loomba, R.; Adams, L.A. Advances in non-invasive assessment of hepatic fibrosis. Gut 2020, 69, 1343–1352. [Google Scholar] [CrossRef]
  24. Dellavance, A.; Fernandes, F.; Shimabokuro, N.; Latini, F.; Baldo, D.; Barreto, J.A.; Marvulle, V.; Andrade, L.E.; Ferraz, M.L. Enhanced liver fibrosis (ELF) score: Analytical performance and distribution range in a large cohort of blood donors. Clin. Chim. Acta 2016, 461, 151–155. [Google Scholar] [CrossRef] [PubMed]
  25. Vali, Y.; Lee, J.; Boursier, J.; Spijker, R.; Löffler, J.; Verheij, J.; Brosnan, M.J.; Böcskei, Z.; Anstee, Q.M.; LITMUS Systematic Review Team; et al. Enhanced liver fibrosis test for the non-invasive diagnosis of fibrosis in patients with NAFLD: A systematic review and meta-analysis. J. Hepatol. 2020, 73, 252–262. [Google Scholar] [CrossRef] [PubMed]
  26. Genovese, F.; Rasmussen, D.G.K.; Karsdal, M.A.; Jesky, M.; Ferro, C.; Fenton, A.; Cockwell, P. Imbalanced turnover of collagen type III is associated with disease progression and mortality in high-risk chronic kidney disease patients. Clin. Kidney J. 2020, 14, 593–601. [Google Scholar] [CrossRef]
  27. Daniels, S.J.; Leeming, D.J.; Eslam, M.; Hashem, A.M.; Nielsen, M.J.; Krag, A.; Karsdal, M.A.; Grove, J.I.; Neil Guha, I.; Kawaguchi, T.; et al. ADAPT: An Algorithm Incorporating PRO-C3 Accurately Identifies Patients with NAFLD and Advanced Fibrosis. Hepatology 2019, 69, 1075–1086. [Google Scholar] [CrossRef] [PubMed]
  28. Mosca, A.; Mantovani, A.; Crudele, A.; Panera, N.; Comparcola, D.; De Vito, R.; Bianchi, M.; Byrne, C.D.; Targher, G.; Alisi, A. Higher Levels of Plasma Hyaluronic Acid and N-terminal Propeptide of Type III Procollagen Are Associated With Lower Kidney Function in Children With Non-alcoholic Fatty Liver Disease. Front. Pediatr. 2022, 10, 917714. [Google Scholar] [CrossRef]
  29. Myint, M.; Oppedisano, F.; De Giorgi, V.; Kim, B.M.; Marincola, F.M.; Alter, H.J.; Nesci, S. Inflammatory signaling in NASH driven by hepatocyte mitochondrial dysfunctions. J. Transl. Med. 2023, 21, 757. [Google Scholar] [CrossRef]
  30. Crudele, L.; Garcia-Irigoyen, O.; Cariello, M.; Piglionica, M.; Scialpi, N.; Florio, M.; Piazzolla, G.; Suppressa, P.; Sabbà, C.; Gadaleta, R.M.; et al. Total serum FGF-21 levels positively relate to visceral adiposity differently from its functional intact form. Front. Endocrinol. 2023, 14, 1159127. [Google Scholar] [CrossRef]
  31. Hua, M.C.; Huang, J.L.; Hu, C.C.; Yao, T.C.; Lai, M.W. Including Fibroblast Growth Factor-21 in Combined Biomarker Panels Improves Predictions of Liver Steatosis Severity in Children. Front. Pediatr. 2019, 7, 420. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, G.; Li, H.; Fang, Q.; Zhang, J.; Zhang, M.; Zhang, L.; Wu, L.; Hou, X.; Lu, J.; Bao, Y.; et al. Complementary Role of Fibroblast Growth Factor 21 and Cytokeratin 18 in Monitoring the Different Stages of Nonalcoholic Fatty Liver Disease. Sci. Rep. 2017, 7, 5095. [Google Scholar] [CrossRef] [PubMed]
  33. Jang, W.; Song, J.S. Non-Invasive Imaging Methods to Evaluate Non-Alcoholic Fatty Liver Disease with Fat Quantification: A Review. Diagnostics 2023, 13, 1852. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, Y.N.; Fowler, K.J.; Ozturk, A.; Potu, C.K.; Louie, A.L.; Montes, V.; Henderson, W.C.; Wang, K.; Andre, M.P.; Samir, A.E.; et al. Liver fibrosis imaging: A clinical review of ultrasound and magnetic resonance elastography. J. Magn. Reson. Imaging 2019, 51, 25–42. [Google Scholar] [CrossRef] [PubMed]
  35. Della Corte, C.; Mosca, A.; Ionata, A.; Nobili, V. Docosahexaenoic Acid and Its Role in G-Protein-Coupled Receptor 120 Activation in Children Affected by Nonalcoholic Fatty Liver Disease. Endocr. Dev. 2016, 30, 29–36. [Google Scholar] [CrossRef] [PubMed]
  36. Della Corte, C.; Carpino, G.; De Vito, R.; De Stefanis, C.; Alisi, A.; Cianfarani, S.; Overi, D.; Mosca, A.; Stronati, L.; Cucchiara, S.; et al. Docosahexanoic Acid Plus Vitamin D Treatment Improves Features of NAFLD in Children with Serum Vitamin D Deficiency: Results from a Single Centre Trial. PLoS ONE 2016, 11, e0168216. [Google Scholar] [CrossRef] [PubMed]
  37. Nobili, V.; Alisi, A.; Mosca, A.; Crudele, A.; Zaffina, S.; Denaro, M.; Smeriglio, A.; Trombetta, D. The Antioxidant Effects of Hydroxytyrosol and Vitamin E on Pediatric Nonalcoholic Fatty Liver Disease, in a Clinical Trial: A New Treatment? Antioxid. Redox Signal. 2019, 31, 127–133. [Google Scholar] [CrossRef] [PubMed]
  38. Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: Interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2019, 394, 2184–2196, Erratum in Lancet 2020, 396, 312; Erratum in Lancet 2021, 397, 2336. [Google Scholar] [CrossRef]
  39. Rupasinghe, K.; Hind, J.; Hegarty, R. Updates in Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) in Children. J. Pediatr. Gastroenterol. Nutr. 2023, 77, 583–591. [Google Scholar] [CrossRef]
Table 1. Therapeutic trials in pediatric NAFLD (ClinicalTrials.gov search results 1 February 2024, https://classic.clinicaltrials.gov).
Table 1. Therapeutic trials in pediatric NAFLD (ClinicalTrials.gov search results 1 February 2024, https://classic.clinicaltrials.gov).
NCT NumberTitleStatusStudy ResultsConditionsInterventionsLocations
1NCT01529268Cysteamine Bitartrate Delayed-Release for the Treatment of NAFLD in ChildrenCompletedHas resultsNon-alcoholic fatty liver disease (NAFLD)-Drug: DR cysteamine bitartrate capsule
-Other: DR cysteamine bitartrate placebo
University of California, San Diego, San Diego, California, United States
2NCT01913470Study of Losartan in the Treatment of NAFLD in ChildrenCompletedHas resultsNAFLDDrug: LosartanEmory University/Children’s Healthcare of Atlanta, Atlanta, Georgia, United States
3NCT00063635Treatment of Nonalcoholic Fatty Liver Disease in Children
(TONIC)
CompletedHas resultsFatty liver-Drug: Metformin
-Dietary supplement: vitamin E
-Drug: matching placebo
-University of California, San Diego, San Diego, California, United States
-University of California, San Francisco, San Francisco, California, United States
4NCT02134522The Role of Obstructive Sleep Apnea in Children With Fatty Liver DiseaseTerminatedHas resultsNon-alcoholic fatty liver diseaseDevice: continuous positive airway pressure (CPAP).Yale University, New Haven, Connecticut, United States
5NCT04415112Mediterranean Diet Treatment for NAFLDCompletedNo results available-Mediterranean diet-Behavioral: Mediterranean diet
-Behavioral: low-fat diet
Ulas Emre Akbulut, Antalya, Turkey
6NCT02842567Hydroxytyrosol and Vitamin E in Pediatric NASHCompletedNo results availableNAFLD-Drug: Hydroxytyrosol plus vitamin E
-Drug: placebo
-Hepatometabolic Department, Bambino Gesù Children’s Hospital, Rome, Italy
7NCT01934777Efficacy and Tolerance of Treatment With DHA,
Choline and Vitamin E in Children With Non-alcoholic Steatohepatitis
CompletedNo results available-Fatty liver
-Liver fibrosis
-Drug: Docosahexaenoic Acid plus vitamin E plus choline
Drug: placebo pearls
Bambino Gesù Hospital and Research Institute, Rome, Rome, Italy, Italy
8NCT01285362Fish Oil and Nonalcoholic Fatty Liver Disease (NAFLD) StudyCompletedHas results-Non-alcoholic fatty liver disease-Drug: fish oil supplementation
-Drug: placebo supplementation
-Irving Clinical Research Center (GCRC) at Columbia
University Medical Center, New York, New York, United States
9NCT02258126Effect of Exercise on Hepatic Fat in Overweight ChildrenCompletedNo results available-Non-alcoholic fatty liver disease
-Obesity
-Metabolic syndrome
-Other: multidisciplinary intervention program-Pediatric Endocrinology Unit of the University Hospital of Araba (HUA), Vitoria-Gasteiz, Araba, Spain
10NCT00655018Effect of Vitamin E on Pediatric Nonalcoholic Fatty Liver
Disease (NAFLD)
CompletedNo results available-Inflammation
-Fibrosis
-Insulin resistance
-Dietary supplement: vitamin treatment
(alpha tocopherol plus ascorbic acid)
-Dietary supplement: placebo
-Dept. Of HepatoGastoEnterology and Nutrition, Liver Unit, Rome, Italy
11NCT01547910Effect of Supplementation of Fish Oil on Non-alcoholic
Fatty Liver Disease in Children
CompletedNo results availableNon-alcoholic fatty liver diseaseDietary supplement: fish oilChildren’s Memorial Health Institute, Poland
12NCT01553500Glucomannan Effects on Children With Non-alcoholic Fatty Liver DiseaseCompletedNo Results Available-Metabolic syndrome
-Non-alcoholic fatty liver disease
-Insulin resistance
-Dietary supplement: glucomannan
-Behavioral: lifestyle intervention
-Bambino Gesù Children’s Hospital and Research Institute, Rome, Italy
13NCT00823277Metabolic Syndrome and Gen-polymorphs Influence on
Weightloss Among Children in Treatment for Overweight
CompletedNo results available-Childhood Obesity
-NAFLD (Non-alcoholic fatty liver disease)
-Other: chronic care multidisciplinary intervention of childhood obesity-The Children’s Obesity Clinic, Paediatric Department,
University Hospital Holbaek, Region Zealand,
University of Copenhagen, Holbaek, Denmark
14NCT01556113Genetic Effect on Omega 3 Fatty Acids for the Treatment of
Fatty Liver Disease
CompletedNo results available-Non-alcoholic fatty liver disease
-Steatohepatitis
-Hypertriglyceridemia
-Other: Omega diet-Yale School of Medicine, New Haven, Connecticut, United States
15NCT02117700Fatty Liver Disease in Obese ChildrenCompletedNo results available-Obesity
-Non-alcoholic fatty liver disease
-Cardiovascular disease
-Dietary supplement: N-acetyl cysteine 600 mg once/day
-Dietary supplement: N-acetyl cysteine
600 mg twice/day
-Other: placebo twice/day
-Nemours Children’s Clinic/Alfred I duPont Hospital, Jacksonville, Florida, United States
16NCT02644239Impact of Ketogenic Diet on Lipoproteins in Refractory EpilepsyUnknown statusNo results available-Epilepsy
-Cardiovascular disease
-Non-alcoholic fatty Liver disease
-Quality of life
Dietary supplement: ketogenic dietNagila Raquel Teixeira Damasceno, Sao Paulo, SP, Brazil
17NCT02098317DHA and Vitamin D in Children With Biopsy-proven NAFLDCompletedNo results available-NAFLD
-Non-alcoholic steatohepatitis (NASH)
-Drug: DHA plus vitamin D
-Drug: placebo
Bambino Gesù Children Hospital, Rome, Italy
18NCT05073588Effect of Indo-Mediterranean Diet on Hepatic Steatosis and
Fibrosis in NAFLD Children
CompletedNo results availableNon-alcoholic fatty liver disease-Other: Indo-Mediterranean diet
-Other: calorie-restricted diet
-Institute of liver and biliary sciences, Delhi, India
19NCT03883607Elafibranor, PK and Safety in Children and Adolescents
8 to 17 Years of Age With Non Alcoholic Steatohepatitis (NASH)
TerminatedHas resultsNon-alcoholic steatohepatitis-Drug: Elafibranor 80 mg
-Drug: Elafibranor 120 mg
-University of California, San Diego, California, United States
-Columbia University, New York, New York, United States
20NCT00885313Effects of Docosahexaenoic Acid (DHA) on Children With
Nonalcoholic Fatty Liver Disease (NAFLD)
CompletedNo results available-Fatty liver
-Liver fibrosis
-Obesity
-Drug: DHA250
-Drug: DHA500
-Drug: PLACEBO
Behavioral: lifestyle intervention
Bambino Gesù Hospital and Research Institute, Rome,
Italy
21NCT04281121Omega 3 Supplementation in Children With Non Alcoholic Fatty LiverCompletedNo results availableFatty liverDietary supplement: omega 3 fatty acidsPediatrics hospital Ain shams University, Cairo, Egypt
22NCT02548351Randomized Global Phase 3 Study to Evaluate the Impact on NASH With Fibrosis of Obeticholic Acid Treatment (REGENERATE)TerminatedNo results availableNon-alcoholic steatohepatitis (NASH)Drug: Obeticholic acid
Drug: placebo
The University of Alabama at Birmingham, Birmingham, Alabama, United States
Digestive Health Specialists of the Southeast, Dothan, Alabama, United States
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Antonella, M.; Pietrobattista, A.; Maggiore, G. Metabolic-Associated Steatotic Liver Disease (MASLD): A New Term for a More Appropriate Therapy in Pediatrics? Pediatr. Rep. 2024, 16, 288-299. https://doi.org/10.3390/pediatric16020025

AMA Style

Antonella M, Pietrobattista A, Maggiore G. Metabolic-Associated Steatotic Liver Disease (MASLD): A New Term for a More Appropriate Therapy in Pediatrics? Pediatric Reports. 2024; 16(2):288-299. https://doi.org/10.3390/pediatric16020025

Chicago/Turabian Style

Antonella, Mosca, Andrea Pietrobattista, and Giuseppe Maggiore. 2024. "Metabolic-Associated Steatotic Liver Disease (MASLD): A New Term for a More Appropriate Therapy in Pediatrics?" Pediatric Reports 16, no. 2: 288-299. https://doi.org/10.3390/pediatric16020025

Article Metrics

Back to TopTop