Next Article in Journal
Donate or Not to Donate—Willingness to Donate and Accept Donor Human Milk
Previous Article in Journal
Anti-Thrombotic Effects of Coprinus comatus Fibrinolytic Enzyme in Zebrafish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pediatric Familial Hypercholesterolemia: Targeting Intestinal Absorption and Other Therapeutic Strategies

by
Konstantinos Arvanitakis
1,2,
Elena Chatzikalil
3,4,
Christina Antza
5,
Christos Topalidis
6,
Georgios Kalopitas
1,2,
Elena Solomou
7,
Vasilios Kotsis
5,
Georgios Germanidis
1,2,
Theocharis Koufakis
8 and
Michael Doumas
8,*
1
Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636 Thessaloniki, Greece
2
Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
3
First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece
4
“Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
5
Third Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Papageorgiou Hospital, 56403 Thessaloniki, Greece
6
Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
7
Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
8
Second Propaedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(14), 2357; https://doi.org/10.3390/nu17142357
Submission received: 3 June 2025 / Revised: 11 July 2025 / Accepted: 16 July 2025 / Published: 18 July 2025

Abstract

Familial hypercholesterolemia (FH) is a genetic disorder marked by significantly elevated levels of low-density lipoprotein cholesterol (LDL-C) since childhood, substantially increasing the risk of premature atherosclerosis and cardiovascular disease. While dysfunction of hepatic LDL-C receptors is the main underlying cause, the gastrointestinal tract plays a key role in cholesterol homeostasis and represents an important therapeutic target. Inhibition of intestinal cholesterol absorption has emerged as an effective strategy in the management of pediatric FH, particularly in patients for whom statins may not be the ideal first-line treatment. Ezetimibe, an inhibitor of the Niemann-Pick C1-like 1 (NPC1L1) protein, has been shown to reduce LDL-C levels in children with FH, with a greater efficacy observed when used in combination with statins. Bile acid sequestrants also enhance cholesterol excretion but are often limited by gastrointestinal side effects, while dietary interventions, such as phytosterol supplementation and fiber-enriched diets, provide additional benefits in lowering LDL-C and are generally well tolerated. Emerging therapies, including microbiota-targeted strategies and novel cholesterol absorption inhibitors, show promise for expanding future treatment options. This review explores the mechanisms of intestinal cholesterol absorption and their relevance to pediatric FH. We examine key pathways, including dietary cholesterol uptake through NPC1L1, bile acid reabsorption, and cholesterol efflux mediated by ATP-binding cassette transporters, while also discussing clinical and experimental evidence on pharmacological and dietary interventions that modulate these pathways. A deeper understanding of cholesterol metabolism, the emerging role of the gut microbiota, and innovative therapeutic agents can support the development of more effective and personalized approaches to the treatment of children with FH.

1. Introduction

Cholesterol is a lipid-soluble cyclopentane polyhydrophene derivative of eukaryotic cells, which possesses crucial structural and functional properties [1]. It represents a basic component of the phospholipid bilayer part of the eukaryotic cell membrane, a synthetic precursor of bioactive molecules (e.g., steroid hormones, lipid-soluble vitamins) via oxidative cleavage of side chains, and a crucial regulator of membrane fluidity and signaling pathways as well, via a wide range of protein modification procedures [2,3]. Cholesterol homeostasis, consisting of multiple distinct pathways (endogenous biosynthesis, intestinal absorption, biliary/intestinal reabsorption, and efflux), is of great importance for cellular and systemic functions, considering cholesterol’s interactions with a variety of adjacent molecules and its involvement in enzymatic and non-enzymatic processes [4,5,6]. Moreover, given the multiple vital roles of cholesterol and its metabolism, disruptions in cholesterol homeostasis may lead to severe pathologic adverse events (elevated oxidative stress, persistent inflammatory responses, reduced autophagy, accelerated apoptotic activity), and in some cases, to systemic diseases, including cardiovascular disease, tumorigenic activity, immune system abnormalities, and eye disorders [4,5]. Dysregulation of cholesterol metabolism, congenital or acquired, contributes to disease pathogenesis through several mechanisms related to the different cholesterol metabolic pathways, which can serve as potential therapeutic targets.
Familial hypercholesterolemia (FH) is an inherited autosomal dominant or codominant transmitted disorder of lipoprotein metabolism that results in persistent elevation of serum low-density lipoprotein cholesterol (LDL-C) concentrations, being a predisposing factor for premature cardiovascular diseases and, subsequently, premature death [7,8]. FH is the result of genetic alterations in genes encoding proteins involved in the clearance of LDL-C particles. More than 90% of the patients with genetically confirmed FH have mutations in the gene encoding the LDL-C receptor (LDLR), and a lower percentage of FH diagnoses are associated with alterations in genes encoding apolipoprotein B (ApoB), proprotein convertase subtilisin–kexin type 9 (PCSK9), or LDL receptor adaptor protein 1 (LDLRAP1) [9]. Heterozygous FH has a frequency of 1 in approximately 300 individuals, while homozygous FH has a frequency of 1 in approximately 250,000–360,000 of the population [10,11]. The diagnosis of FH is based on several sets of criteria (Figure 1), all of which rely on elevated LDL-C levels, usually including clinical features (e.g., arcus cornealis or tendinous xanthomas), family history, and, in some cases, being aided by genetic testing, which is estimated to reveal pathogenic FH mutations in approximately half of pediatric FH patients [12,13]. The European diagnostic criteria of pediatric FH include: (i) elevated persistent LDL-C over 140 mg/dL, (ii) family history (parents or siblings) of FH, (iii) parental history of LDL-C over 180 mg/dL or family history of premature coronary artery disease, after excluding other causes of primary and secondary LDL-C [14]. In cases characterized by difficulties in diagnosis and management, genetic testing should be offered [14]. FH remains a largely underdiagnosed and, therefore, undertreated entity, leading to severe complications in some affected individuals [8]. The main complication of FH is cardiovascular disease, while other serious adverse events, including aortic disease and valvular disease (aortic aneurysm, aortic stenosis, and supravalvular stenosis), have been associated with FH in some cohorts [15].
Considering the arterial functional and morphological deformities that have been described in pediatric patients with FH and the severe complications presented later in the course of the disease, early initiation of treatment is warranted to prevent cardiovascular events in adult life [16]. Current evidence suggests that the induction of lipid-lowering therapy at a young age may decrease the LDL-C burden, improving endothelial functionality, regressing the atherosclerotic events, and ruling out coronary complications in the majority of pediatric cohorts [17,18,19,20]. Established disease guidelines recommend that the treatment of children with FH include a healthy lifestyle with a heart-healthy diet, as well as statin and/or ezetimibe therapy in affected individuals between 8 and 10 years of age, and in younger children only when there is an extreme elevation of LDL-C and the presence of associated risk factors [21,22]; however, continuous research on novel therapeutic targets is currently being conducted. The use of statins in pediatric patients with FH has shown variable response, which differs depending on the specific statin used and, in some cases, the sex of the patient. In female pediatric patients, the use of lovastatin resulted in a 23–27% reduction in LDL-C levels [23], while in male patients the same dose resulted in a 17–24% reduction in LDL-C [24]. Pravastatin has been shown to reduce LDL-C levels to 24.1% from baseline [25], while simvastatin showed a statistically significant decrease (p < 0.001, compared to the control group) in LDL-C (41%), TC (31%), apolipoprotein B (APOB) (34%), very low-density lipoprotein (VLDL) cholesterol (21%), and triglycerides (TG) (9%) [26]. Another study comparing the use of rosuvastatin in three doses (5 mg, 10 mg, and 20 mg) versus placebo reported statistically significant changes in LDL-C, TC, and APOB levels for all three doses (p < 0.001) [27]. Moreover, a cohort study on statin efficacy in a large pediatric FH population of 214 patients showed a 32% decrease in LDL-C levels (from 6.13 to 4.16 mmol/L), with treatment goals achieved in 20% of the cohort (37 patients) [28]. Data on ezetimibe treatment in pediatric patients with FH are limited. A recent study reported a 22% reduction in LDL-C levels (from 7.3 ± 1.0 mmol/L to 5.7 ± 1.0 mmol/L) after a mean of 105 days of treatment, without significant changes in total cholesterol (TC) or HDL-C levels. The patients remained on ezetimibe for up to 3.5 years, while no medication-related adverse effects were reported [29].
Based on the results of the aforementioned clinical studies, the response of pediatric patients with FH to statins and ezetimibe (measured by reductions in LDL-C levels) does not always achieve treatment targets. Furthermore, evidence from long-term monitoring for adverse events remains limited, underscoring the need for further studies to evaluate the safety of current treatment options throughout the lifetime of FH patients. Future research should also include pediatric patients with secondary forms of dyslipidemia, comparing the outcomes of combination therapy in both inherited and secondary disease groups. As a basis for future research, this review explores the mechanisms of intestinal cholesterol absorption and their relevance to pediatric FH, examining key pathways such as dietary cholesterol uptake via NPC1L1, bile acid reabsorption, and cholesterol efflux mediated by ATP-binding cassette transporters. It also discusses clinical and experimental evidence on pharmacologic and dietary interventions that modulate these pathways. Furthermore, given the implications of the dysregulation of these mechanisms in pediatric patients, we analyze several emerging therapeutic options targeting the gut to reduce LDL-C levels. A deeper understanding of cholesterol metabolism, the emerging role of the gut microbiota, and innovative therapeutic agents can support the development of additional effective and personalized approaches to the treatment of children with FH.

2. Pathways Involved in Cholesterol Homeostasis and Basic Aspects of Pediatric FH

2.1. Cholesterol Metabolism

2.1.1. Endogenous Biosynthesis

Hepatocytes are the main site of cholesterol synthesis using Acetyl-CoA, although smaller amounts are produced by all nucleated cells [5,30]. The biosynthesis of cholesterol is strongly regulated by two major mechanisms of negative feedback, sterol regulatory element-binding protein-2 (SREBP-2) and HMG-CoA reductase (HMGCR) degradation, and consists of four distinct processes: (i) mevalonate (MVA) synthesis; (ii) isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP) production; (iii) squalene synthesis; and (iv) lanosterol formation via squalene cyclization and subsequent cholesterol synthesis [31,32] (Figure 2). MVA undergoes multiple cycles of phosphorylation, decarboxylation, dehydroxylation, and condensation reactions to generate squalene [5]. This squalene is then converted to lanosterol and 27-carbon cholesterol by the action of enzymes, including cyclase and oxygenase, located in the endoplasmic reticulum (ER) [5,30]. SREBP-2 is linked to two protein molecules within the ER membrane: SREBP cleavage-activating protein (SCAP) and insulin-induced gene protein-1 (Insig-1) (Figure 2) [33]. Cholesterol in the ER membrane inhibits the binding of coat proteins to SCAP, preventing the formation of COPII-coated vesicles [34]. A distinct loop region on the SCAP protein, termed methionine-glutamate-leucine-alanine-aspartate-leucine (MELADL), is suggested to regulate the aforementioned mechanism. It is suggested that this SCAP sequence contains multiple domains that are essential for the transport of the SCAP-SREBP complex from the ER to the Golgi apparatus [35]. Cholesterol elimination leads to the dissociation of the SCAP-SREBP protein complex from Insig-1 and to its transportation from the ER to the Golgi apparatus, while Insig-1 is epigenetically modified (mainly ubiquitinated) and subsequently degraded by the proteasome [36] (Figure 2). Elevated cholesterol levels lead to the formation of complexes between cholesterol-derived oxysterols and Insig1/2, which prevents SREBP2 transport to the ER [36].

2.1.2. Intestinal Absorption via NPC1L1

Cholesterol in the intestine is derived from biliary secretion or dietary intake [37]. The intestinal absorption of dietary cholesterol mainly depends on the polytopic transmembrane protein Niemann-Pick C1-Like 1 (NPC1L1), which is expressed both in the apical membrane of the small intestine and in hepatocytes, regulating extracellular sterol transport in the liver, where it inhibits excessive biliary cholesterol loss [38,39]. Intestinal cholesterol absorption, mainly regulated by NPC1L1, consists of three stages: (i) cholesterol solubilization in chylomicrons (CM), (ii) transportation across the apical membrane of enterocytes, and (iii) cholesterol secretion into the bloodstream. Specifically, NPC1L1-dependent cholesterol intestinal absorption is mechanistically regulated by clathrin-mediated endocytosis, and following this, cholesterol is transported to the ER, where it undergoes esterification reactions by acyl-CoA acyltransferase (ACAT), thus participating in the formation of CMs, which are finally secreted into the systemic circulation via the lymphatic system [40,41,42,43]. Lipoprotein lipase (LPL) catalyzes the hydrolysis of CM triglycerides in the peripheral blood, while the remaining CMs bind to the low-density lipoprotein receptor (LDLR) or the LDLR-related protein 1 (LRP) on hepatocyte membranes, enhancing their absorption by liver cells [44]. NPC1L1 is then transported back to the hepatic cell surface, a process that is regulated by several factors, including the microfilament-interacting motor myosin Vb, the small GTPase Rab11a, and the adaptor Rab11-FIP2 [45]. With this mechanism, free cholesterol is dissociated from the NPC1L1 protein during the early stages of endocytosis [45].
Recently, new evidence has emerged supporting the potential of NPC1L1 as a drug target. NPC1L1-mediated cholesterol absorption was fully described in 2020 by a study using cryo-electron microscopy, which suggested that after binding to the sterol-binding specific region, cholesterol triggers the NPC1L1 transporter, changing its formation to enhance the process of cholesterol absorption [46]. Moreover, NPC1L1 can bind different amounts of cholesterol, depending on the different concentrations of cholesterol levels; however, the ability to effectively bind cholesterol lies in its effective dimerization [47]. NPC1L1-mediated cholesterol absorption undergoes esterification by acyl-CoA to form chylomicrons, which are subsequently released into the circulation, where triglycerides are hydrolyzed in order to be used in peripheral tissues or absorbed by hepatocytes [48]. On the other hand, free cholesterol can be pumped back into the intestinal lumen via ATP-binding cassette transporters (ABCs): ABCG5 and ABCG8, or can be released directly into the circulation through ABCA1, in the form of HDL-C [38].
The effects of targeting intestinal cholesterol absorption via NPC1L1 are illustrated in Figure 3.

2.1.3. Bile Acid Cholesterol Re-Absorption

Hepatocytes play a crucial role in cholesterol downregulation by converting it into bile acids and secreting it into bile [49]. The primary mechanism for cholesterol catabolism is bile acid synthesis, which occurs exclusively in hepatocytes [50]. The enzyme cholesterol 7α-hydroxylase (CYP7A1) initiates this conversion, serving as a rate-limiting step [51]. Bile acids are secreted into bile through the bile salt export pump (BSEP), while cholesterol is transported by the ATP-binding cassette sub-family G member 5 and 8 (ABCG5 and ABCG8) transporters [52] (Figure 4). Most bile acids are reabsorbed by enterocytes through the apical sodium-dependent bile acid transporter (ASBT) and transported back to the liver through the portal circulation [52] (Figure 4). Conjugated bile acids are taken up in hepatocytes by basolateral Na(+)-taurocholate co-transporting polypeptide (NTCP) [52]. Bile acid homeostasis is primarily regulated by feedback mechanisms involving the farnesoid X receptor (FXR), which is found in hepatocytes and intestinal cells, where it reduces CYP7A1 gene transcription through various pathways by the induction of the small heterodimer partner (SHP) and the RNA-binding protein ZFP36L1, which destabilize CYP7A1 mRNA [52,53]. Furthermore, in the intestine, FXR activates fibroblast growth factor 15 (FGF15), which inhibits the expression of CYP7A1 in the liver [52] (Figure 4).

2.1.4. Efflux Through ABC Transporters

Although cholesterol can be produced by almost all cells in the human body, only a few cell types can effectively metabolize it [4]. Reverse cholesterol transport, which refers to the transport of cholesterol to the liver through the systemic circulation to maintain excess cholesterol elimination, can be achieved only by cells that expel elevated cholesterol levels [54]. This complex process is regulated by liver X receptors (LXRs) and the ABC transporters ABCA1 and ABCG1 [55]. LXR represents a ligand-activated nuclear transcription factor that counteracts the effects of the SREBP2 protein, which is activated during cholesterol accumulation [56]. Beyond cholesterol metabolism, LXR represents a crucial regulator of glucose metabolism via increasing the expression levels of glucose transporter 4 (GLUT4) in type 2 diabetes mouse models and modulating inflammatory responses by inhibiting inflammatory factors, primarily NF-κB [57,58].
Human tissues contain two highly homologous LXR proteins that are distributed across various tissues, specifically: (i) LXRα, primarily found in the intestine, liver, kidney, fat tissue, and macrophages, and (ii) LXRβ, which is widely expressed in all human tissues [59]. LRX is heterodimerized in the cell nucleus with the retinoid X receptor (RXR) [4,59]. Binding of the LRX agonist leads to stimulation of target genes including ABCA1, ABCG1, SREBP-1c, and ApoE [60]. Regarding LXR downstream targets, ABCA1 and ABCG1, they both use ATP as an energy source to facilitate the transmembrane transport of lipids, sugars, and amino acids [4]. ABCA1 is a full transporter protein expressed throughout the body, characterized by two tandem repeats of transmembrane domains, each containing six segments and a large glycosylated extracellular domain [61]. During the cholesterol efflux process, ABCA1 transfers intracellular cholesterol to lipid-free ApoA1 when activated by LXR, leading to the formation of nascent high-density lipoprotein cholesterol (HDL-C) [4]. Subsequently, lecithin-cholesterol acyltransferase (LCAT) promotes HDL-C maturation. Recent evidence indicates that mutations or inactivation of ABCA1 significantly impede the efflux of cellular cholesterol to ApoA1, as seen in patients with Tangier disease and Abca1-negative mice, who are unable to form mature HDL-C and exhibit low total plasma cholesterol and abnormal lipid deposition in various tissues [62,63]

2.1.5. Cholesterol Balance

Cholesterol homeostasis is regulated by a tight relationship between absorption and synthesis, which is considered crucial to maintaining cholesterol balance in the human body (Figure 5) [5]. Cholesterol synthesis negatively regulates cholesterol absorption and vice versa, maintaining the balance between dietary cholesterol intake and excretion [64]. Studies on cholesterol balance suggest significant variability in individual responses to dietary cholesterol absorption, which is partially explained by the wide variety of protein regulatory mechanisms related to cholesterol handling [64,65]. Increased dietary cholesterol intake increases bile acid synthesis, improving cholesterol excretion through the feces [64]. Moreover, higher cholesterol intake generally reduces endogenous synthesis as a compensatory response, helping to prevent persistently elevated cholesterol levels [66]. Additionally, given that the liver converts cholesterol into bile acids for digestion, which are then reabsorbed in the ileum and returned to the liver, any loss of bile acids (around 5% in feces) is compensated by the ongoing synthesis in the liver, regulated by feedback mechanisms from the reabsorbed bile acids [67].

2.2. Basic Aspects of Pediatric FH

Approximately 450,000 children are born with FH each year; however, only about 2.1% of the adult FH population is diagnosed before the age of 18 using current diagnostic methods [68]. Although selective screening based on family history is recommended to detect FH, real-world evidence shows that many cases are missed, in part due to the absence of symptoms related to atherosclerosis during childhood. As a result, most FH cases are discovered incidentally through blood tests during routine pediatric monitoring, following national guidelines [69]. In 2011, the National Heart, Lung, and Blood Institute Expert Panel recommended universal screening for children between 9 and 11 years of age, with a second screening at ages 17 to 21, alongside selective screening, to improve detection of heterozygous FH in the pediatric population [70]. This diagnostic strategy was reaffirmed in the 2018 American College of Cardiology and American Heart Association cholesterol guidelines, which also suggest “cascade screening” as a promising approach [71]. The diagnostic criteria for pediatric FH established in 2017 differ from those in adults in terms of LDL-C thresholds and family history considerations and introduce the category of “probable FH” as previously described [14].
These approaches aim to enable early identification and monitoring of FH in children to reduce missed diagnoses. Following an FH diagnosis, early initiation of lifestyle modifications is recommended. When lifestyle modifications are not sufficient, statin-based pharmacological therapy is considered effective in lowering LDL-C levels and is safe in the short and medium term, reducing disease-related complications [72]. Additional lipid-lowering medications can be used in combination with statins in patients for whom statin monotherapy is inadequate, particularly in homozygous FH patients that often require aggressive multimodal treatment [73]. If left untreated, FH is associated with an increased risk of premature coronary artery disease. Several predictive markers have been proposed, including combinations of clinical and biochemical markers, genetic risk scoring systems, inflammatory markers, and imaging techniques (e.g., carotid intima-media thickness and coronary artery calcium scoring) [74]. However, these predictive markers have not yet demonstrated sufficient efficacy for widespread use in daily clinical practice. Therefore, more studies are needed to refine diagnostic and prognostic algorithms, determine the optimal timing and intensity of statin therapy, and establish the long-term safety and outcomes of lipid-lowering treatments in pediatric cohorts with FH.

3. Therapeutic Options for Pediatric FH

Recent studies indicate that the pathophysiological atherosclerotic changes in individuals with FH occur early in their course of the disease; therefore, therapeutic interventions in childhood aiming to reduce LDL-C levels may be important to prevent FH complications, mainly cardiovascular disease [18]. Statins are the first-line medications for FH in pediatric patients, while ezetimibe can be induced at the age of 8–10 years for heterozygous FH patients and from diagnosis for homozygous FH patients [75]. However, even in cases of early induction of established therapeutic choices, guideline-recommended LDL-C cholesterol levels cannot be achieved in some pediatric FH patients; thus, we should suggest the use of novel therapeutic agents, which mainly target the intestinal cholesterol absorption [14,76].

3.1. Established Therapeutic Options for Pediatric FH and Ezetimibe Mechanism of Action

Regarding the established therapeutic options of pediatric FH, the European Atherosclerosis Society and the American College of Cardiology (American Heart Association) have proposed several guidelines that indicate the importance of lifestyle changes in combination with the initiation of statin therapy, from 8 to 10 years of age for FH heterozygotes and from the time of diagnosis for FH homozygotes [75,77]. Balanced dietary intake, regular physical activity, medical interventions for female adolescents with dyslipidemia (birth control), and monitoring for common risk factors (smoking, obesity, and diabetes) are considered key components of successful long-term pediatric FH management, decreasing the risk of developing severe complications, especially in patients with homozygous FH [28]. The 2017 guidelines for dyslipidemia suggest a fat-energy ratio of 20–25% and a carbohydrate-energy ratio of 50–60% for pediatric patients with FH, according to the indications for the adult population [78]. In pediatric patients with LDL-C levels equal to or above 180 mg/dL, pharmacotherapy is suggested, including statins as the first-line treatment and ezetimibe as the second-line treatment [78].
Serum cholesterol levels are regulated through a dynamic relationship between cholesterol biosynthesis in the liver and intestinal absorption [79]. Statins are competitive inhibitors of the HMG-CoA reductase enzyme and decrease serum LDL-C by inhibiting cholesterol biosynthesis, leading to elevated expression of the LDL receptor, thus facilitating the removal of LDL-C from the systemic circulation. Inhibition of LDL-C biosynthesis decreases total cholesterol expression in the ER, resulting in the transportation of SREBPs from the endoplasmic reticulum to the Golgi apparatus and subsequent cleavage into active transcription factors [80,81]. SREBPs translocate to the nucleus, elevating the expression of several genes, including HMG-CoA reductase, restoring liver cholesterol biosynthesis and the number of LDL receptors on the plasma membrane of hepatocytes, further resulting in increased clearance of LDL, thus reducing LDL-C levels [82,83]. In homozygous FH, which is characterized by the total absence of normally functioning LDL receptors, statin monotherapy is not considered effective in decreasing LDL-C levels [79].
The efficacy of using statins in decreasing LDL-C levels in FH pediatric patients has not been established yet, while long-term evidence on the effect of statin therapy in protecting against disease complications is currently lacking, especially when compared to adults, for whom the importance of statin treatment in the prevention of cardiovascular disease is based on robust data [84,85,86]. Luirinik et al. reported a 20-year follow-up study in FH pediatric patients, who were followed up alongside their unaffected healthy siblings in terms of the therapeutic results of statin use and alongside their affected parents regarding the incidence of clinical cardiovascular disease [28]. The mean levels of LDL-C reduced by 32% from baseline, while the mean progression of carotid intima—median thickness, which was used to evaluate the effect of statins, was similar between the two groups (patients and healthy siblings) during the study period [28]. The incidence of cardiovascular adverse events and cardiovascular-related death was significantly lower in patients with FH at 39 years of age compared to their FH counterparts who started statin use later in the course of the disease [28]. This study, which is, to the best of our knowledge, the largest one to evaluate statin use in pediatric FH patients, suggests that early initiation of statins in this population is safe and effective and reduces the long-term risk of cardiovascular complications [28].
Ezetimibe is an NPC1L1 inhibitor, representing an effective treatment option to decrease LDL-C cholesterol levels [41,87]. Ezetimibe inhibits caveolin-1 and annexin-2, decreasing intestinal cholesterol absorption and reducing liver cholesterol levels [88]. Specifically, ezetimibe prevents cholesterol absorption by directly binding to a transmembrane loop for NPC1L1, inhibiting its ability to bind with clathrin/AP2 that is required for its endocytosis, by which cholesterol is released into the cytosol and is esterified by cholesterol acyltransferase (ACAT) to form cholesterol esters [89,90]. These cholesterol esters, combined with triacylglycerol particles by facilitation by microsomal triacylglycerol transfer protein (MTP), form ApoB-48 containing chylomicrons, which will be taken up in the systemic circulation [91,92]. Ezetimibe inhibits this mechanism, selectively affecting cholesterol without impacting the absorption of triglycerides, enhancing the expression of liver LDL receptor levels, and resulting in an 18–20% decrease in circulating LDL-C levels [93]. Ezetimibe is metabolized in the liver and intestine, has a long half-life of 22 h, and is predominantly excreted in the feces while minimally interacting with medications metabolized by cytochrome P450 [94]. In particular, ezetimibe, by decreasing LDL-C levels, inhibits the inflammatory and tumorigenic effects of excess LDL-C in the systemic circulation, presenting antiangiogenetic, antiproliferative, and immune regulatory properties (Figure 6) [95].
Although statin monotherapy has been shown to decrease LDL-C levels, it stimulates intestinal cholesterol absorption, which is considered a limiting factor in its efficacy [96]. Combined treatment with statins and ezetimibe shows a significant synergistic effect in decreasing LDL-C levels in pediatric patients, while recent evidence suggests ezetimibe monotherapy as a safe and effective therapeutic option [97,98,99]. A prospective study in pediatric patients with FH, who received ezetimibe monotherapy at 10 mg/day for a median time of 15.9 months, showed a significant decrease (315.3 +/− 41.8 to 233.3 +/− 36.8 mg/dL) in total cholesterol and LDL-C levels. Their triglyceride levels and biochemical profile remained stable, and no adverse events were observed in this cohort [100]. Regarding combinational treatment schemas, a recently published meta-analysis of 41 studies including 4667 children with heterozygous HF and a median age of 12 years reported a 33.44% median reduction in LDL-C levels with statin and ezetimibe co-administration [101]. Moreover, a registry analysis of 11,848 FH pediatric patients revealed that only 28.5% were on lipid-lowering treatment during the course of the disease and that median LDL-C levels were higher in children not receiving treatment as compared to their counterparts under lipid-lowering treatment [102].

3.2. Novel FH Therapeutic Strategies Targeting Intestinal Cholesterol Metabolism

The therapeutic options previously analyzed, namely statins and ezetimibe, are dependent on LDL receptors, demonstrating limited efficacy in patients with homozygous FH variants, who do not reach the LDL-C levels recommended by the guidelines [103,104,105,106]. Lipoprotein apheresis is an alternative therapeutic option indicated for patients with early-onset FH, which can effectively decrease LDL-C levels by 50–70%. However, this decrease is temporary, and each treatment, which is necessary every 1–2 weeks, is time-consuming and affects the quality of life of patients [107,108]. Considering that there is a percentage of pediatric FH patients who do not reach target LDL-C levels despite treatment, the need to establish new therapeutic goals remains undeniable, especially in individuals who respond poorly to conventional therapies, even when receiving the maximum tolerated doses of first-line and second-line medications. Furthermore, given the efficacy of ezetimibe in children with FH, current research has focused on novel drugs that target intestinal cholesterol absorption. In this context, recent advances in FH drugs acting on the intestinal tract have surfaced, and several new treatment options are currently being investigated in randomized clinical trials [109]. It is also worth noting that other treatment alternatives targeting angiopoietin-like 3 (ANGPTL3) have emerged as candidates for FH treatment, especially for individuals with refractory FH [110]. ANGPTL3 inhibitors, namely evinacumab, promote the excretion of the very low-density lipoprotein before it can be converted to LDL-C [111]. In the following paragraphs, we focus on drugs that target intestinal cholesterol absorption in pediatric patients with FH.
Novel medications for pediatric FH mainly include PCSK9 inhibitors. PCSK9 primarily regulates LDL-C levels by targeting and degrading LDL receptors (LDLR) [112]. This process includes the interaction of PCSK9 with LDLR’s EGF-A domain, directing the LDL-C/LDLR complex to lysosomes for degradation, thus reducing LDLR on cell surfaces and leading to increased circulating LDL-C [112]. PCSK9 is expressed in the liver and small intestine, with hepatic PCSK9 playing a dominant role in cholesterol regulation [113]. Current research indicates that intestine-specific PCSK9 has less impact on lipidemia. Additional roles of PCSK9 include involvement in hepatic inflammation; inflammatory stimuli increase PCSK9 expression, decreasing LSLR in the liver and promoting inflammation through macrophage activation and cytokine production. This creates a cycle that worsens liver inflammation through the NLRP3 inflammasome and TLR4/NFκB signaling pathways [114]. Evolocumab is a monoclonal antibody targeting intestinal cholesterol absorption and specifically pro-protein convertase subtilisin-kexin type 9 (PCSK9) expression in the small intestine, preventing PCSK9 from degrading the LDL-C receptor, thus increasing LDL-C receptor availability and subsequently decreasing LDL-C levels up to 60%, according to current real-world data [115]. It is worth noting that PCSK9 interacts with CD36, a platelet glycoprotein that enhances LDL-C uptake, leading to decreased lipid uptake, increased inflammation, and accelerated progression of atherosclerosis [116]. Therefore, PCSK9 inhibitors can decrease LDL-C as well as thrombosis markers in patients with FH, preventing arterial clotting events, including heart attacks and ischemic strokes [116].
PCSK9 inhibition was initially approved in 2015 for adult patients with hyperlipidemia and in 2017 for people with cardiovascular disease and increased risk for myocardial infarction, stroke, and revascularization in atherosclerotic cardiovascular disease [117,118,119,120]. PCSK9 inhibitors, namely evolocumab and alirocumab, can reduce LDL-C levels by approximately 20–25% and 55–60% in homozygous FH patients and people with severe heterozygous FH, respectively [110]. Considering that the intestine influences the overall lipid levels via lipid absorption and that the direct effects of PCSK9 inhibition are centered on hepatic pathways (as hepatic PCSK9 plays the major role in cholesterol regulation), targeting PCSK with novel medication, including evolocumab, alirocumab, and inclisiran, is a promising option for non-responders to 1st and 2nd line FH treatment [116]. In pediatric cohorts, evolocumab and alirocumab have been shown to be effective and well-tolerated [110] and are recommended for FH pediatric patients ≥12 years of age who cannot tolerate statins as monotherapy or in combination with other agents or fail to respond to conventional treatment (ezetimibe) [101]. Furthermore, the liver production of PCSK9 has not yet been targeted by inclisiran, a gene-suppressing drug that is effective in adults but is still being investigated for pediatric use [121]. A summary of FH therapeutic options for the pediatric population, including intestinal absorption-targeting medication, is illustrated in Figure 7.
Only a limited number of clinical studies have evaluated the use of PCSK9 inhibitors in pediatric patients with FH. In more detail, six studies have investigated the efficacy of evolocumab or alirocumab on LDL-C levels in pediatric cohorts of FH, and the results on therapeutic efficacy demonstrated great variability, which may reflect the variability of LDL receptor activity as a result of the heterogenous genetic profile of the patients. Homozygous FH was subcategorized into three types, namely defective/defective, defective/null, and null/null, according to whether the activity of the LDL receptor in each allele was null or defective [118,122,123,124,125,126,127]. Real-world evidence on alirocumab [128] and evolocumab, obtained from the RAMAN and HAUSER-OLE studies, suggests using PCSK9 inhibitors to achieve a reduction equal to or above 15%, translating into an absolute reduction of >60 mg/dL, for the treatment of pediatric FH [129]. Two other clinical trials, namely TESLA and TAUSSIG, which evaluated 15 homozygous children with FH in total, indicate the use of evolocumab in these cohorts [125,128]. Evolocumab had a great safety profile, with no adverse events reported, while LDL-C levels were significantly reduced, as well as other variables of the lipid profile, including apolipoprotein B and lipoprotein-a [125,128]. Regarding heterozygous pediatric FH, the phase 2 ODYSSEY KIDS study, involving 42 pediatric patients, reported a significant decrease in LDL-C levels within 8 weeks of alirocumab induction [130]. Along the same line, the HAUSER-RCT phase 3 clinical trial evaluated the efficacy and safety of evolocumab in combination with statins with or without ezetimibe in 157 pediatric heterozygous FH patients in whom LDL-C guideline-approved levels had not been achieved, reporting a median decrease of 77.5 mg/dL in LDL-C levels [131], while no clinically relevant effects of evolocumab on steroid hormone biosynthesis and vitamin (A, D, E, and K) levels were observed [131]. (Table 1.)

4. Discussion and Future Perspectives

The purpose of the FH therapeutic intervention is to reduce the cumulative burden of persistently elevated LDL-C levels and, at the same time, to prevent the development of complications, especially cardiovascular adverse events [130]. Effective monitoring and control of LDL-C has been associated with a significant reduction in morbidity and mortality in FH patients, highlighting the importance of lipid-lowering treatment, which is suggested to be administered early in the course of the disease [132]. Statins represent the mainstay of FH treatment, especially when combined with a low-saturated fat diet, which lowers cholesterol and improves cardiovascular outcomes [133]. The recommended statin dose in the pediatric population is lower compared to adults; however, the majority of children diagnosed with FH receive statin monotherapy (first-line treatment), with or without ezetimibe (second-line treatment), without the need for additional medication. Unfortunately, some pediatric patients do not reach the LDL-C levels recommended by the guidelines, mainly due to limited drug response or poor treatment adherence [133]. To overcome the non-response in these cases, a slew of novel medications that aim to reduce LDL-C levels in severe cases, mostly with combinational lipid-lowering regimens, are being tested in ongoing clinical trials.
The cholesterol pool in the human body is derived from both biosynthesis and dietary uptake. Although statin monotherapy has been proven to be effective in reducing cholesterol biosynthesis, it lacks the ability to inhibit the absorption of cholesterol; this point is where current research is directed, providing novel treatment options targeting intestinal cholesterol absorption. Evolocumab and alirocumab are the new intestine-targeting drugs evaluated in pediatric FH cohorts, presenting promising effects in reducing LDL-C levels, with most studies suggesting a median decrease of 15% in half of patients [129]. It is worth noting that the response depends on the genetic profile of pediatric patients with FH, since homozygotes respond poorly to PCSK9 inhibition compared to their heterozygote peers. However, the effects of evolocumab and alirocumab can be safely evaluated after their implementation in large cohorts, with appropriate adaptation to the genetic heterogeneity of pediatric patients and/or differences in existing treatment [132]. It is, however, suggested that PCSK9 inhibitors represent a viable therapeutic option for homozygous FH children who are not responding adequately to high-intensity statin and ezetimibe therapy; if at least a 15% additional decrease in LDL-C levels is observed, PCSK9 inhibitor therapy may be continued, otherwise, this therapy is discontinued [107]. Familial hypercholesterolemia in pediatric cohorts is associated with a significant risk of cardiovascular complications; therefore, early recognition and timely therapeutic evaluation are considered necessary to achieve an optimal prognosis. Many pediatric patients with FH do not benefit from first-line statin treatment alone, nor from second-line ezetimibe-targeted treatment.
Recently developed monoclonal antibodies targeting the gut can be considered additional lipid-lowering therapeutic options, as recommended by ongoing clinical trials. Evolocumab and alirocumab, targeting PCSK9, have been shown to be effective in significantly reducing LDL-C levels in the pediatric population with FH. However, more research is needed on their long-term safety and efficacy, as evidence on long-term monitoring, especially for FH homozygotes, currently remains limited. The most promising future research directions for pediatric and, in particular, homozygous FH patients include inhibitors of cholesterylester transfer protein (CETP) (obicetrapib), RNAi agents (ARO-ANG3, olpasiran), antisense oligonucleotides, and emerging therapies such as ASGR1 inhibitors and HMGCR degraders, all of which aim to reduce serum LDL-C, with subsequent improvement in cardiovascular health of children.
The mainstay of treatment continues to be statins, often in combination with ezetimibe, PCSK9 inhibitors, or newer agents such as inclisiran and bempedoic acid. For homozygous FH, therapies such as lomitapide and evinacumab offer hope to lower lipids in refractory cases. Despite the availability of these options, the real-world adherence to therapy and the achievement of LDL-C targets remain suboptimal. Public health strategies must prioritize education, early detection, and access to advanced therapies to mitigate the long-term cardiovascular burden of FH. Additionally, emerging therapeutic approaches, including gene-based treatments, hold promise for a more definitive correction of the underlying genetic defect in the future. Continued research is essential to refine diagnostic algorithms, personalize therapy based on genotype-phenotype correlations, and develop cost-effective screening models. Multidisciplinary collaboration between clinicians, researchers, and policymakers will be crucial to overcoming current challenges and improving the prognosis of individuals with FH. Ultimately, a combination of early identification, aggressive lipid management, and innovations in therapy will be the key to reducing the global impact of this inherited lipid disorder.
Based on our daily clinical experience, pediatric patients with homozygous familial hypercholesterolemia are typically closely monitored to prevent and effectively manage cardiovascular complications related to the disease. The diagnosis of homozygous FH is most often prompted by the presence of xanthomas and total cholesterol levels exceeding 240 mg/dL. Until about two decades ago, homozygous FH was associated with high early mortality rates due to cardiovascular complications. The absence of effective, established medications made survival particularly difficult for these patients, often resulting in psychological distress and unnecessary adoption of extremely restrictive diets, which alone could not sufficiently improve their health. Current clinical practice in our country recommends a healthy diet in combination with pharmacologic treatment, primarily statins, for patients with homozygous FH in order to control lipid levels and reduce cardiovascular risk. Statin therapy has significantly improved clinical outcomes and quality of life for our patients; however, the introduction of novel agents could further enhance health outcomes and help eliminate disease-related complications. In general, the use of appropriate diagnostic algorithms to effectively detect and prevent severe forms of FH, together with the implementation of effective strategies to manage cardiovascular complications, are key components of our routine clinical care for patients with homozygous FH.

5. Conclusions

Pediatric FH represents a critical and often overlooked challenge in cardiovascular prevention. Early onset of elevated low-density lipoprotein cholesterol levels in children with FH substantially increases the lifetime risk of premature atherosclerosis and cardiovascular disease. Despite advances in lipid-lowering therapies and increasing awareness, the condition continues to be underdiagnosed and undertreated, especially in its heterozygous and homozygous forms. The complex genetic underpinnings of FH-ranging from monogenic to polygenic causes-highlight the need for comprehensive diagnostic approaches that combine clinical criteria with genetic testing. Early detection, ideally through cascade screening of family members, is crucial to initiating timely and effective interventions, and current guidelines recommend the initiation of lifestyle modifications and statin therapy in childhood. However, many children do not reach the LDL-C target levels with conventional therapies alone, which underscores the need for complementary and alternative strategies. A central focus of current research is the role of the gastrointestinal tract in cholesterol homeostasis and its growing therapeutic relevance. Intestinal cholesterol absorption, largely mediated by the NPC1L1 transporter, has emerged as a promising target to reduce LDL-C in pediatric FH. Ezetimibe, a selective inhibitor of NPC1L1, has been shown to be safe and effective in reducing LDL-C levels in children, both as monotherapy and in combination with statins. In addition, emerging treatments such as PCSK9 inhibitors and other agents targeting intestinal mechanisms present additional options for children who are statin-intolerant or are not adequately controlled with existing therapies. Understanding and modulating intestinal cholesterol absorption not only complements hepatic-targeted therapies but also opens new avenues for personalized care in pediatric FH. Future research should continue to refine these strategies, exploring age-specific pharmacodynamics, long-term safety, and the role of the gut microbiome. Early, targeted intervention-especially through intestinal pathways-offers a compelling opportunity to alter the natural course of FH and significantly improve cardiovascular outcomes for affected children.

Author Contributions

Conceptualization, K.A., T.K. and M.D.; writing—original draft preparation, K.A., C.A., C.T. and E.C.; writing—review and editing, K.A., C.A., G.K., E.S. and T.K.; visualization, C.T., V.K., G.K. and E.S.; supervision, V.K., G.G. and M.D. 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

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CADcoronary arterial disease
EASEuropean Atherosclerosis Society
FHfamilial hypercholesterolemia
LDL-Clow-density lipoprotein cholesterol
ANGPTL3angiopoietin protein like 3
PCSK9proprotein convertase subtilisin kexin-9
ACATacyl-coenzyme A: cholesterol acyltransferase
CEcholesterol esters
LDLlow-density lipoprotein
MTPmicrosomal triacylglycerol transfer protein
NPL1L1Niemann-Pick C1-Like1
ERendoplasmic reticulum
HDLhigh-density lipoprotein
LXRαliver X receptor α
SREBP2sterol regulatory element-binding protein 2
VLDLvery low-density lipoprotein
LDLRAP1LDL receptor adaptor protein 1
HMGCRHMG-CoA reductase
MVAmevalonate synthesis
IPPisopentenyl pyrophosphate dimethylallyl pyrophosphate
DMAPPdimethylallyl pyrophosphate
CETPcholesterylester transfer protein
LCATlecithin-cholesterol acyltransferase
RXRretinoid X receptor
LPLlipoprotein lipase
MELADLmethionine-glutamate-leucine-alanine-aspartate-leucine
GLUT4glucose transporter 4
SCAPSREBP cleavage-activating protein
Insig-1insulin-induced gene protein-1

References

  1. Maxfield, F.R.; van Meer, G. Cholesterol, the Central Lipid of Mammalian Cells. Curr. Opin. Cell Biol. 2010, 22, 422–429. [Google Scholar] [CrossRef] [PubMed]
  2. Ali, O.; Szabó, A. Review of Eukaryote Cellular Membrane Lipid Composition, with Special Attention to the Fatty Acids. Int. J. Mol. Sci. 2023, 24, 15693. [Google Scholar] [CrossRef]
  3. Paukner, K.; Králová Lesná, I.; Poledne, R. Cholesterol in the Cell Membrane—An Emerging Player in Atherogenesis. Int. J. Mol. Sci. 2022, 23, 533. [Google Scholar] [CrossRef]
  4. Guo, J.; Chen, S.; Zhang, Y.; Liu, J.; Jiang, L.; Hu, L.; Yao, K.; Yu, Y.; Chen, X. Cholesterol Metabolism: Physiological Regulation and Diseases. MedComm 2024, 5, e476. [Google Scholar] [CrossRef]
  5. Duan, Y.; Gong, K.; Xu, S.; Zhang, F.; Meng, X.; Han, J. Regulation of Cholesterol Homeostasis in Health and Diseases: From Mechanisms to Targeted Therapeutics. Signal Transduct. Target. Ther. 2022, 7, 265. [Google Scholar] [CrossRef]
  6. Luu, W.; Sharpe, L.J.; Capell-Hattam, I.; Gelissen, I.C.; Brown, A.J. Oxysterols: Old Tale, New Twists. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 447–467. [Google Scholar] [CrossRef]
  7. Benito-Vicente, A.; Belloso-Uribe, K.; Jebari Benslaiman, S.; Galicia Garcia, U.; Ostolaza, H.; Martin, C. Familial Hypercholesterolemia: The Most Frequent Cholesterol Metabolism Disorder Caused Disease. Int. J. Mol. Sci. 2018, 19, 3426. [Google Scholar] [CrossRef]
  8. Abifadel, M.; Boileau, C. Genetic and Molecular Architecture of Familial Hypercholesterolemia. J. Intern. Med. 2023, 293, 144–165. [Google Scholar] [CrossRef]
  9. Vallejo-Vaz, A.J.; Seshasai, S.R.K.; Cole, D.; Hovingh, G.K.; Kastelein, J.J.P.; Mata, P.; Raal, F.J.; Santos, R.D.; Soran, H.; Watts, G.F. Familial Hypercholesterolaemia: A Global Call to Arms. Atherosclerosis 2015, 243, 257–259. [Google Scholar] [CrossRef]
  10. Hu, P.; Dharmayat, K.I.; Stevens, C.A.T.; Sharabiani, M.T.A.; Jones, R.S.; Watts, G.F.; Genest, J.; Ray, K.K.; Vallejo-Vaz, A.J. Prevalence of Familial Hypercholesterolemia among the General Population and Patients with Atherosclerotic Cardiovascular Disease: A Systematic Review and Meta-Analysis. Circulation 2020, 141, 1742–1759. [Google Scholar] [CrossRef]
  11. Defesche, J.C.; Gidding, S.S.; Harada-Shiba, M.; Hegele, R.A.; Santos, R.D.; Wierzbicki, A.S. Familial Hypercholesterolaemia. Nat. Rev. Dis. Prim. 2017, 3, 17093. [Google Scholar] [CrossRef]
  12. Futema, M.; Taylor-Beadling, A.; Williams, M.; Humphries, S.E. Genetic Testing for Familial Hypercholesterolemia–Past, Present, and Future. J. Lipid Res. 2021, 62, 100139. [Google Scholar] [CrossRef]
  13. Okada, K.; Warabi, E.; Sugimoto, H.; Horie, M.; Tokushige, K.; Ueda, T.; Harada, N.; Taguchi, K.; Hashimoto, E.; Itoh, K. Nrf2 Inhibits Hepatic Iron Accumulation and Counteracts Oxidative Stress-Induced Liver Injury in Nutritional Steatohepatitis. J. Gastroenterol. 2012, 47, 924–935. [Google Scholar] [CrossRef] [PubMed]
  14. Harada-Shiba, M.; Ohtake, A.; Sugiyama, D.; Tada, H.; Dobashi, K.; Matsuki, K.; Minamino, T.; Yamashita, S.; Yamamoto, Y. Guidelines for the Diagnosis and Treatment of Pediatric Familial Hypercholesterolemia 2022. J. Atheroscler. Thromb. 2023, 30, 531–557. [Google Scholar] [CrossRef]
  15. Prajapati, R.; Agrawal, V. Familial Hypercholesterolemia Supravalvular Aortic Stenosis and Extensive Atherosclerosis. Indian Heart J. 2018, 70, 575–577. [Google Scholar] [CrossRef]
  16. Fularski, P.; Hajdys, J.; Majchrowicz, G.; Stabrawa, M.; Młynarska, E.; Rysz, J.; Franczyk, B. Unveiling Familial Hypercholesterolemia—Review, Cardiovascular Complications, Lipid-Lowering Treatment and Its Efficacy. Int. J. Mol. Sci. 2024, 25, 1637. [Google Scholar] [CrossRef]
  17. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A. ESC Scientific Document Group. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Hear. J. 2020, 41, 111. [Google Scholar] [CrossRef]
  18. Wiegman, A.; Gidding, S.S.; Watts, G.F.; Chapman, M.J.; Ginsberg, H.N.; Cuchel, M.; Ose, L.; Averna, M.; Boileau, C.; Borén, J. Familial Hypercholesterolaemia in Children and Adolescents: Gaining Decades of Life by Optimizing Detection and Treatment. Eur. Heart J. 2015, 36, 2425–2437. [Google Scholar] [CrossRef]
  19. Braamskamp, M.J.A.M.; Langslet, G.; McCrindle, B.W.; Cassiman, D.; Francis, G.A.; Gagne, C.; Gaudet, D.; Morrison, K.M.; Wiegman, A.; Turner, T. Effect of Rosuvastatin on Carotid Intima-Media Thickness in Children with Heterozygous Familial Hypercholesterolemia: The CHARON Study (Hypercholesterolemia in Children and Adolescents Taking Rosuvastatin Open Label). Circulation 2017, 136, 359–366. [Google Scholar] [CrossRef]
  20. Vuorio, A.; Kuoppala, J.; Kovanen, P.T.; Humphries, S.E.; Tonstad, S.; Wiegman, A.; Drogari, E.; Ramaswami, U. Statins for Children with Familial Hypercholesterolemia. Cochrane Database Syst. Rev. 2017, 7, 7. [Google Scholar] [CrossRef]
  21. Law, M.R.; Wald, N.J.; Rudnicka, A.R. Quantifying Effect of Statins on Low Density Lipoprotein Cholesterol, Ischaemic Heart Disease, and Stroke: Systematic Review and Meta-Analysis. BMJ 2003, 326, 1423. [Google Scholar] [CrossRef]
  22. Watts, G.F.; Gidding, S.; Wierzbicki, A.S.; Toth, P.P.; Alonso, R.; Brown, W.V.; Bruckert, E.; Defesche, J.; Lin, K.K.; Livingston, M. Integrated Guidance on the Care of Familial Hypercholesterolemia from the International FH Foundation. J. Clin. Lipidol. 2014, 8, 148–172. [Google Scholar] [CrossRef]
  23. O’Gorman, C.S.M.; O’Neill, M.B.; Conwell, L.S. Considering Statins for Cholesterol-Reduction in Children If Lifestyle and Diet Changes Do Not Improve Their Health: A Review of the Risks and Benefits. Vasc. Health Risk Manag. 2010, 7, 1–14. [Google Scholar] [CrossRef] [PubMed]
  24. Avis, H.J.; Hutten, B.A.; Gagné, C.; Langslet, G.; McCrindle, B.W.; Wiegman, A.; Hsia, J.; Kastelein, J.J.P.; Stein, E.A. Efficacy and Safety of Rosuvastatin Therapy for Children with Familial Hypercholesterolemia. J. Am. Coll. Cardiol. 2010, 55, 1121–1126. [Google Scholar] [CrossRef] [PubMed]
  25. Radaelli, G.; Sausen, G.; Cesa, C.C.; Santos, F.d.S.; Portal, V.L.; Neyeloff, J.L.; Pellanda, L.C. Statin Treatments and Dosages in Children with Familial Hypercholesterolemia: Meta-Analysis. Arq. Bras. Cardiol. 2018, 111, 810–821. [Google Scholar] [CrossRef]
  26. Clauss, S.B.; Holmes, K.W.; Hopkins, P.; Stein, E.; Cho, M.; Tate, A.; Johnson-Levonas, A.O.; Kwiterovich, P.O. Efficacy and Safety of Lovastatin Therapy in Adolescent Girls with Heterozygous Familial Hypercholesterolemia. Pediatrics 2005, 116, 682–688. [Google Scholar] [CrossRef]
  27. Wiegman, A.; Hutten, B.A.; De Groot, E.; Rodenburg, J.; Bakker, H.D.; Büller, H.R.; Sijbrands, E.J.G.; Kastelein, J.J.P. Efficacy and Safety of Statin Therapy in Children with Familial Hypercholesterolemia: A Randomized Controlled Trial. Jama 2004, 292, 331–337. [Google Scholar] [CrossRef]
  28. Luirink, I.K.; Wiegman, A.; Kusters, D.M.; Hof, M.H.; Groothoff, J.W.; de Groot, E.; Kastelein, J.J.P.; Hutten, B.A. 20-Year Follow-up of Statins in Children with Familial Hypercholesterolemia. N. Engl. J. Med. 2019, 381, 1547–1556. [Google Scholar] [CrossRef]
  29. Clauss, S.; Wai, K.-M.; Kavey, R.-E.W.; Kuehl, K. Ezetimibe Treatment of Pediatric Patients with Hypercholesterolemia. J. Pediatr. 2009, 154, 869–872. [Google Scholar] [CrossRef]
  30. Rudney, H.; Sexton, R.C. Regulation of Cholesterol Biosynthesis. Annu. Rev. Nutr. 1986, 6, 245–272. [Google Scholar] [CrossRef]
  31. Guo, W.; Zhao, Y.; Zhang, Z.; Tan, N.; Zhao, F.; Ge, C.; Liang, L.; Jia, D.; Chen, T.; Yao, M.; et al. Disruption of XCT Inhibits Cell Growth via the ROS/Autophagy Pathway in Hepatocellular Carcinoma. Cancer Lett. 2011, 312, 55–61. [Google Scholar] [CrossRef] [PubMed]
  32. Cerqueira, N.; Oliveira, E.; Gesto, D.; Santos-Martins, D.; Moreira, C.; Moorthy, N.S.H.; Ramos, M.; Fernandes, P. Cholesterol Biosynthesis: A Mechanistic Overview. Biochemistry 2016, 55, 5483–5506. [Google Scholar] [CrossRef] [PubMed]
  33. Song, B.-L.; Javitt, N.B.; DeBose-Boyd, R.A. Insig-Mediated Degradation of HMG CoA Reductase Stimulated by Lanosterol, an Intermediate in the Synthesis of Cholesterol. Cell Metab. 2005, 1, 179–189. [Google Scholar] [CrossRef]
  34. Sun, L.-P.; Seemann, J.; Goldstein, J.L.; Brown, M.S. Sterol-Regulated Transport of SREBPs from Endoplasmic Reticulum to Golgi: Insig Renders Sorting Signal in Scap Inaccessible to COPII Proteins. Proc. Natl. Acad. Sci. USA 2007, 104, 6519–6526. [Google Scholar] [CrossRef]
  35. Brown, A.J.; Sun, L.; Feramisco, J.D.; Brown, M.S.; Goldstein, J.L. Cholesterol Addition to ER Membranes Alters Conformation of SCAP, the SREBP Escort Protein That Regulates Cholesterol Metabolism. Mol. Cell 2002, 10, 237–245. [Google Scholar] [CrossRef]
  36. Gong, Y.; Lee, J.N.; Lee, P.C.W.; Goldstein, J.L.; Brown, M.S.; Ye, J. Sterol-Regulated Ubiquitination and Degradation of Insig-1 Creates a Convergent Mechanism for Feedback Control of Cholesterol Synthesis and Uptake. Cell Metab. 2006, 3, 15–24. [Google Scholar] [CrossRef]
  37. Stieger, B. Biliary Cholesterol Secretion: More Lessons from Plants? J. Hepatol. 2003, 38, 843–846. [Google Scholar] [CrossRef]
  38. Jia, L.; Betters, J.; Yu, L. Niemann-Pick C1-Like 1 (NPC1L1) Protein in Intestinal and Hepatic Cholesterol Transport. Annu. Rev. Physiol. 2011, 73, 239–259. [Google Scholar] [CrossRef]
  39. Kobayashi, S. The Effect of Polyphenols on Hypercholesterolemia through Inhibiting the Transport and Expression of Niemann–Pick C1-Like 1. Int. J. Mol. Sci. 2019, 20, 4939. [Google Scholar] [CrossRef]
  40. Yu, L.; Bharadwaj, S.; Brown, J.M.; Ma, Y.; Du, W.; Davis, M.A.; Michaely, P.; Liu, P.; Willingham, M.C.; Rudel, L.L. Cholesterol-Regulated Translocation of NPC1L1 to the Cell Surface Facilitates Free Cholesterol Uptake. J. Biol. Chem. 2006, 281, 6616–6624. [Google Scholar] [CrossRef]
  41. Ge, L.; Wang, J.; Qi, W.; Miao, H.-H.; Cao, J.; Qu, Y.-X.; Li, B.-L.; Song, B.-L. The Cholesterol Absorption Inhibitor Ezetimibe Acts by Blocking the Sterol-Induced Internalization of NPC1L1. Cell Metab. 2008, 7, 508–519. [Google Scholar] [CrossRef]
  42. Brown, J.M.; Rudel, L.L.; Yu, L. NPC1L1 (Niemann–Pick C1-like 1) Mediates Sterol-Specific Unidirectional Transport of Non-Esterified Cholesterol in McArdle-RH7777 Hepatoma Cells. Biochem. J. 2007, 406, 273–283. [Google Scholar] [CrossRef] [PubMed]
  43. Luo, G.; Xiang, L.; Xiao, L. Acetyl-CoA Deficiency Is Involved in the Regulation of Iron Overload on Lipid Metabolism in Apolipoprotein E Knockout Mice. Molecules 2022, 27, 4966. [Google Scholar] [CrossRef] [PubMed]
  44. Hultin, M.; Olivecrona, T. Conversion of Chylomicrons into Remnants. Atherosclerosis 1998, 141, S25–S29. [Google Scholar] [CrossRef]
  45. Chu, B.-B.; Ge, L.; Xie, C.; Zhao, Y.; Miao, H.-H.; Wang, J.; Li, B.-L.; Song, B.-L. Requirement of Myosin Vb·Rab11a·Rab11-FIP2 Complex in Cholesterol-Regulated Translocation of NPC1L1 to the Cell Surface. J. Biol. Chem. 2009, 284, 22481–22490. [Google Scholar] [CrossRef]
  46. Johnson, T.A.; Pfeffer, S.R. Ezetimibe-Sensitive Cholesterol Uptake by NPC1L1 Protein Does Not Require Endocytosis. Mol. Biol. Cell 2016, 27, 1845–1852. [Google Scholar] [CrossRef]
  47. Hu, M.; Yang, F.; Huang, Y.; You, X.; Liu, D.; Sun, S.; Sui, S.-F. Structural Insights into the Mechanism of Human NPC1L1-Mediated Cholesterol Uptake. Sci. Adv. 2021, 7, eabg3188. [Google Scholar] [CrossRef]
  48. Long, T.; Liu, Y.; Qin, Y.; DeBose-Boyd, R.A.; Li, X. Structures of Dimeric Human NPC1L1 Provide Insight into Mechanisms for Cholesterol Absorption. Sci. Adv. 2021, 7, eabh3997. [Google Scholar] [CrossRef]
  49. Li, T.; Chiang, J.Y.L. Regulation of Bile Acid and Cholesterol Metabolism by PPARs. PPAR Res. 2009, 2009, 501739. [Google Scholar] [CrossRef]
  50. Zhang, B.; Kuipers, F.; de Boer, J.F.; Kuivenhoven, J.A. Modulation of Bile Acid Metabolism to Improve Plasma Lipid and Lipoprotein Profiles. J. Clin. Med. 2022, 11, 4. [Google Scholar] [CrossRef]
  51. Chiang, J.Y.L.; Ferrell, J.M. Up to Date on Cholesterol 7 Alpha-Hydroxylase (CYP7A1) in Bile Acid Synthesis. Liver Res. 2020, 4, 47–63. [Google Scholar] [CrossRef]
  52. Fleishman, J.S.; Kumar, S. Bile Acid Metabolism and Signaling in Health and Disease: Molecular Mechanisms and Therapeutic Targets. Signal Transduct. Target. Ther. 2024, 9, 97. [Google Scholar] [CrossRef] [PubMed]
  53. Coy, D.J.; Wooton-Kee, C.R.; Yan, B.; Sabeva, N.; Su, K.; Graf, G.; Vore, M. ABCG5/ABCG8-Independent Biliary Cholesterol Excretion in Lactating Rats. Am. J. Physiol. Liver Physiol. 2010, 299, G228–G235. [Google Scholar] [CrossRef]
  54. Cohen, D.E. Balancing Cholesterol Synthesis and Absorption in the Gastrointestinal Tract. J. Clin. Lipidol. 2008, 2, S1–S3. [Google Scholar] [CrossRef] [PubMed]
  55. Apfel, R.; Benbrook, D.; Lernhardt, E.; Ortiz, M.A.; Salbert, G.; Pfahl, M. A Novel Orphan Receptor Specific for a Subset of Thyroid Hormone-Responsive Elements and Its Interaction with the Retinoid/Thyroid Hormone Receptor Subfamily. Mol. Cell. Biol. 1994, 14, 7025–7035. [Google Scholar]
  56. Laffitte, B.A.; Chao, L.C.; Li, J.; Walczak, R.; Hummasti, S.; Joseph, S.B.; Castrillo, A.; Wilpitz, D.C.; Mangelsdorf, D.J.; Collins, J.L. Activation of Liver X Receptor Improves Glucose Tolerance through Coordinate Regulation of Glucose Metabolism in Liver and Adipose Tissue. Proc. Natl. Acad. Sci. USA 2003, 100, 5419–5424. [Google Scholar] [CrossRef]
  57. Hazra, S.; Rasheed, A.; Bhatwadekar, A.; Wang, X.; Shaw, L.C.; Patel, M.; Caballero, S.; Magomedova, L.; Solis, N.; Yan, Y. Liver X Receptor Modulates Diabetic Retinopathy Outcome in a Mouse Model of Streptozotocin-Induced Diabetes. Diabetes 2012, 61, 3270–3279. [Google Scholar] [CrossRef]
  58. El-Darzi, N.; Mast, N.; Buchner, D.A.; Saadane, A.; Dailey, B.; Trichonas, G.; Pikuleva, I.A. Low-Dose Anti-HIV Drug Efavirenz Mitigates Retinal Vascular Lesions in a Mouse Model of Alzheimer’s Disease. Front. Pharmacol. 2022, 13, 902254. [Google Scholar] [CrossRef]
  59. Calkin, A.C.; Tontonoz, P. Transcriptional Integration of Metabolism by the Nuclear Sterol-Activated Receptors LXR and FXR. Nat. Rev. Mol. Cell Biol. 2012, 13, 213–224. [Google Scholar] [CrossRef]
  60. Edwards, P.A.; Kennedy, M.A.; Mak, P.A. LXRs;: Oxysterol-Activated Nuclear Receptors That Regulate Genes Controlling Lipid Homeostasis. Vascul. Pharmacol. 2002, 38, 249–256. [Google Scholar] [CrossRef]
  61. Oram, J.F.; Vaughan, A.M. ATP-Binding Cassette Cholesterol Transporters and Cardiovascular Disease. Circ. Res. 2006, 99, 1031–1043. [Google Scholar] [CrossRef]
  62. Orsó, E.; Broccardo, C.; Kaminski, W.E.; Böttcher, A.; Liebisch, G.; Drobnik, W.; Götz, A.; Chambenoit, O.; Diederich, W.; Langmann, T. Transport of Lipids from Golgi to Plasma Membrane Is Defective in Tangier Disease Patients and Abc1-Deficient Mice. Nat. Genet. 2000, 24, 192–196. [Google Scholar] [CrossRef] [PubMed]
  63. Kennedy, M.A.; Barrera, G.C.; Nakamura, K.; Baldán, Á.; Tarr, P.; Fishbein, M.C.; Frank, J.; Francone, O.L.; Edwards, P.A. ABCG1 Has a Critical Role in Mediating Cholesterol Efflux to HDL and Preventing Cellular Lipid Accumulation. Cell Metab. 2005, 1, 121–131. [Google Scholar] [CrossRef]
  64. Afonso, M.S.; Machado, R.M.; Lavrador, M.S.; Quintao, E.C.R.; Moore, K.J.; Lottenberg, A.M. Molecular Pathways Underlying Cholesterol Homeostasis. Nutrients 2018, 10, 760. [Google Scholar] [CrossRef]
  65. Huang, C.-H.; Hsu, H.-S.; Chiang, M.-T. Influence of Varied Dietary Cholesterol Levels on Lipid Metabolism in Hamsters. Nutrients 2024, 16, 2472. [Google Scholar] [CrossRef]
  66. Soliman, G.A. Dietary Cholesterol and the Lack of Evidence in Cardiovascular Disease. Nutrients 2018, 10, 780. [Google Scholar] [CrossRef]
  67. Ticho, A.L.; Malhotra, P.; Dudeja, P.K.; Gill, R.K.; Alrefai, W.A. Intestinal Absorption of Bile Acids in Health and Disease. In Comprehensive Physiology; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2019; pp. 21–56. ISBN 9780470650714. [Google Scholar]
  68. Dharmayat, K.I.; Vallejo-Vaz, A.J.; Stevens, C.A.T.; Brandts, J.M.; Lyons, A.R.M.; Groselj, U.; Abifadel, M.; Aguilar-Salinas, C.A.; Alhabib, K.; Alkhnifsawi, M.; et al. Familial Hypercholesterolaemia in Children and Adolescents from 48 Countries: A Cross-Sectional Study. Lancet 2024, 403, 55–66. [Google Scholar] [CrossRef]
  69. Timoshchenko, O.; Ivanoshchuk, D.; Semaev, S.; Orlov, P.; Zorina, V.; Shakhtshneider, E. Diagnosis of Familial Hypercholesterolemia in Children and Young Adults. Int. J. Mol. Sci. 2024, 25, 314. [Google Scholar] [CrossRef]
  70. Allen-Tice, C.; Steinberger, J.; Murdy, K.; Zierhut, H. Pediatric Cholesterol Screening Practices in 9- to 11-Year-Olds in a Large Midwestern Primary Care Setting. J. Clin. Lipidol. 2020, 14, 224–230. [Google Scholar] [CrossRef]
  71. Reyes-Soffer, G.; Yeang, C.; Michos, E.D.; Boatwright, W.; Ballantyne, C.M. High Lipoprotein(a): Actionable Strategies for Risk Assessment and Mitigation. Am. J. Prev. Cardiol. 2024, 18, 100651. [Google Scholar] [CrossRef]
  72. Sawhney, J.P.S.; Madan, K. Familial Hypercholesterolemia. Indian Heart J. 2024, 76, S108–S112. [Google Scholar] [CrossRef] [PubMed]
  73. Krzemińska, J.; Młynarska, E.; Radzioch, E.; Wronka, M.; Rysz, J.; Franczyk, B. Management of Familial Hypercholesterolemia with Special Emphasis on Evinacumab. Biomedicines 2022, 10, 3273. [Google Scholar] [CrossRef] [PubMed]
  74. Martin, A.C.; Gidding, S.S.; Wiegman, A.; Watts, G.F. Knowns and Unknowns in the Care of Pediatric Familial Hypercholesterolemia. J. Lipid Res. 2017, 58, 1765–1776. [Google Scholar] [CrossRef]
  75. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; De Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2019, 73, 3168–3209. [Google Scholar] [CrossRef]
  76. Ramaswami, U.; Futema, M.; Bogsrud, M.P.; Holven, K.B.; van Lennep, J.R.; Wiegman, A.; Descamps, O.S.; Vrablik, M.; Freiberger, T.; Dieplinger, H. Comparison of the Characteristics at Diagnosis and Treatment of Children with Heterozygous Familial Hypercholesterolaemia (FH) from Eight European Countries. Atherosclerosis 2020, 292, 178–187. [Google Scholar] [CrossRef]
  77. Wald, D.S.; Bestwick, J.P.; Morris, J.K.; Whyte, K.; Jenkins, L.; Wald, N.J. Child–Parent Familial Hypercholesterolemia Screening in Primary Care. N. Engl. J. Med. 2016, 375, 1628–1637. [Google Scholar] [CrossRef]
  78. Cohen, H.; Stefanutti, C.; Di Giacomo, S.; Morozzi, C.; Widhalm, K.; Bjelakovic, B.B.; Berni, A.; Martino, F.; Bosco, G. The Mighty Medic Satellite Research Group for Pediatric Dyslipidemia. Current Approach to the Diagnosis and Treatment of Heterozygote and Homozygous FH Children and Adolescents. Curr. Atheroscler. Rep. 2021, 23, 30. [Google Scholar] [CrossRef]
  79. Xu, D.; Ma, R.; Ju, Y.; Song, X.; Niu, B.; Hong, W.; Wang, R.; Yang, Q.; Zhao, Z.; Zhang, Y.; et al. Cholesterol Sulfate Alleviates Ulcerative Colitis by Promoting Cholesterol Biosynthesis in Colonic Epithelial Cells. Nat. Commun. 2022, 13, 4428. [Google Scholar] [CrossRef]
  80. Brown, M.S.; Radhakrishnan, A.; Goldstein, J.L. Retrospective on Cholesterol Homeostasis: The Central Role of Scap. Annu. Rev. Biochem. 2018, 87, 783–807. [Google Scholar] [CrossRef]
  81. Fryer, L.G.D.; Jones, B.; Duncan, E.J.; Hutchison, C.E.; Ozkan, T.; Williams, P.A.; Alder, O.; Nieuwdorp, M.; Townley, A.K.; Mensenkamp, A.R.; et al. The Endoplasmic Reticulum Coat Protein II Transport Machinery Coordinates Cellular Lipid Secretion and Cholesterol Biosynthesis. J. Biol. Chem. 2014, 289, 4244–4261. [Google Scholar] [CrossRef]
  82. Sakakura, Y.; Shimano, H.; Sone, H.; Takahashi, A.; Inoue, N.; Toyoshima, H.; Suzuki, S.; Yamada, N.; Inoue, K. Sterol Regulatory Element-Binding Proteins Induce an Entire Pathway of Cholesterol Synthesis. Biochem. Biophys. Res. Commun. 2001, 286, 176–183. [Google Scholar] [CrossRef]
  83. Xiao, X.; Luo, Y.; Peng, D. Updated Understanding of the Crosstalk Between Glucose/Insulin and Cholesterol Metabolism. Front. Cardiovasc. Med. 2022, 9, 879355. [Google Scholar] [CrossRef]
  84. Versmissen, J.; Oosterveer, D.M.; Yazdanpanah, M.; Defesche, J.C.; Basart, D.C.G.; Liem, A.H.; Heeringa, J.; Witteman, J.C.; Lansberg, P.J.; Kastelein, J.J.P. Efficacy of Statins in Familial Hypercholesterolaemia: A Long Term Cohort Study. BMJ 2008, 337, a2423. [Google Scholar] [CrossRef]
  85. Neil, A.; Cooper, J.; Betteridge, J.; Capps, N.; McDowell, I.; Durrington, P.; Seed, M.; Humphries, S.E. Reductions in All-Cause, Cancer, and Coronary Mortality in Statin-Treated Patients with Heterozygous Familial Hypercholesterolaemia: A Prospective Registry Study. Eur. Heart J. 2008, 29, 2625–2633. [Google Scholar] [CrossRef] [PubMed]
  86. Jiang, S.-Y.; Li, H.; Tang, J.-J.; Wang, J.; Luo, J.; Liu, B.; Wang, J.-K.; Shi, X.-J.; Cui, H.-W.; Tang, J.; et al. Discovery of a Potent HMG-CoA Reductase Degrader That Eliminates Statin-Induced Reductase Accumulation and Lowers Cholesterol. Nat. Commun. 2018, 9, 5138. [Google Scholar] [CrossRef]
  87. Enjoji, M.; Machida, K.; Kohjima, M.; Kato, M.; Kotoh, K.; Matsunaga, K.; Nakashima, M.; Nakamuta, M. NPC1L1 Inhibitor Ezetimibe Is a Reliable Therapeutic Agent for Non-Obese Patients with Nonalcoholic Fatty Liver Disease. Lipids Health Dis. 2010, 9, 29. [Google Scholar] [CrossRef]
  88. Van Heek, M.; Farley, C.; Compton, D.S.; Hoos, L.; Davis, H.R. Ezetimibe Selectively Inhibits Intestinal Cholesterol Absorption in Rodents in the Presence and Absence of Exocrine Pancreatic Function. Br. J. Pharmacol. 2001, 134, 409–417. [Google Scholar] [CrossRef]
  89. Wüstner, D.; Solanko, K. How Cholesterol Interacts with Proteins and Lipids during Its Intracellular Transport. Biochim. Biophys. Acta-Biomembr. 2015, 1848, 1908–1926. [Google Scholar] [CrossRef]
  90. Calandra, S.; Tarugi, P.; Speedy, H.E.; Dean, A.F.; Bertolini, S.; Shoulders, C.C. Mechanisms and Genetic Determinants Regulating Sterol Absorption, Circulating LDL Levels, and Sterol Elimination: Implications for Classification and Disease Risk. J. Lipid Res. 2011, 52, 1885–1926. [Google Scholar] [CrossRef]
  91. Kounatidis, D.; Vallianou, N.G.; Poulaki, A.; Evangelopoulos, A.; Panagopoulos, F.; Stratigou, T.; Geladari, E.; Karampela, I.; Dalamaga, M. ApoB100 and Atherosclerosis: What’s New in the 21st Century? Metabolites 2024, 14, 123. [Google Scholar] [CrossRef]
  92. Gugliucci, A. Triglyceride-Rich Lipoprotein Metabolism: Key Regulators of Their Flux. J. Clin. Med. 2023, 12, 4399. [Google Scholar] [CrossRef] [PubMed]
  93. Fraunberger, P.; Gröne, E.; Gröne, H.-J.; Drexel, H.; Walli, A.K. Ezetimibe Reduces Cholesterol Content and NF-KappaB Activation in Liver but Not in Intestinal Tissue in Guinea Pigs. J. Inflamm. 2017, 14, 3. [Google Scholar] [CrossRef] [PubMed]
  94. Shi, Z.; Han, S. Personalized Statin Therapy: Targeting Metabolic Processes to Modulate the Therapeutic and Adverse Effects of Statins. Heliyon 2025, 11, e41629. [Google Scholar] [CrossRef] [PubMed]
  95. Descamps, O.; Tomassini, J.E.; Lin, J.; Polis, A.B.; Shah, A.; Brudi, P.; Hanson, M.E.; Tershakovec, A.M. Variability of the LDL-C Lowering Response to Ezetimibe and Ezetimibe + Statin Therapy in Hypercholesterolemic Patients. Atherosclerosis 2015, 240, 482–489. [Google Scholar] [CrossRef]
  96. Hammersley, D.; Signy, M. Ezetimibe: An Update on Its Clinical Usefulness in Specific Patient Groups. Ther. Adv. Chronic Dis. 2017, 8, 4–11. [Google Scholar] [CrossRef]
  97. Hamilton-Craig, I.; Kostner, K.; Colquhoun, D.; Woodhouse, S. Combination Therapy of Statin and Ezetimibe for the Treatment of Familial Hypercholesterolemia. Vasc. Health Risk Manag. 2010, 6, 1023–1037. [Google Scholar] [CrossRef]
  98. Llewellyn, A.; Simmonds, M.; Marshall, D.; Harden, M.; Woods, B.; Humphries, S.E.; Ramaswami, U.; Priestley-Barnham, L.; Fisher, M.; Tata, L.J.; et al. Efficacy and Safety of Statins, Ezetimibe and Statins-Ezetimibe Therapies for Children and Adolescents with Heterozygous Familial Hypercholesterolaemia: Systematic Review, Pairwise and Network Meta-Analyses of Randomised Controlled Trials. Atherosclerosis 2025, 401, 118598. [Google Scholar] [CrossRef]
  99. Ezhov, M.V.; Sergienko, I.V.; Kryzhanovskiy, S.M.; Manko, K.S.; Timoshina, E. V Comparative Efficacy and Safety of Statin Monotherapy and Statin plus Ezetimibe Combination in a Real-World Setting. Diseases 2023, 11, 168. [Google Scholar] [CrossRef]
  100. Yeste, D.; Chacón, P.; Clemente, M.; Albisu, M.A.; Gussinyé, M.; Carrascosa, A. Ezetimibe as Monotherapy in the Treatment of Hypercholesterolemia in Children and Adolescents. J. Pediatr. Endocrinol. Metab. 2009, 22, 487–492. [Google Scholar] [CrossRef]
  101. Banach, M.; Bytyçi, I.; Bytyqi, S.; Lewek, J.; Surma, S.; Bajraktari, G.; Henein, M.; Sahebkar, A.; Alkhnifsawi, M.; Gouni-Berthold, I.; et al. Management of Children with Heterozygous Familial Hypercholesterolemia Worldwide: A Meta-Analysis. Eur. Heart J. Open 2025, 5, oeaf001. [Google Scholar] [CrossRef]
  102. Xiao, Y.; Ba, Z.; Pang, S.; Liu, D.; Wang, H.; Liang, H.; Wang, Y.; Yuan, J. PCSK9 Inhibitor: Safe Alternative to Fill the Treatment Gap in Statin-Limited Conditions? RCM 2022, 23, 380. [Google Scholar] [CrossRef]
  103. Rosenson, R.S. Existing and Emerging Therapies for the Treatment of Familial Hypercholesterolemia. J. Lipid Res. 2021, 62, 100060. [Google Scholar] [CrossRef]
  104. Thedrez, A.; Blom, D.J.; Ramin-Mangata, S.; Blanchard, V.; Croyal, M.; Chemello, K.; Nativel, B.; Pichelin, M.; Cariou, B.; Bourane, S. Homozygous Familial Hypercholesterolemia Patients With Identical Mutations Variably Express the LDLR (Low-Density Lipoprotein Receptor) Implications for the Efficacy of Evolocumab. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 592–598. [Google Scholar] [CrossRef]
  105. Stein, E.A.; Dann, E.J.; Wiegman, A.; Skovby, F.; Gaudet, D.; Sokal, E.; Charng, M.-J.; Mohamed, M.; Luirink, I.; Raichlen, J.S. Efficacy of Rosuvastatin in Children with Homozygous Familial Hypercholesterolemia and Association with Underlying Genetic Mutations. J. Am. Coll. Cardiol. 2017, 70, 1162–1170. [Google Scholar] [CrossRef]
  106. Cuchel, M.; Lee, P.C.; Hudgins, L.C.; Duell, P.B.; Ahmad, Z.; Baum, S.J.; Linton, M.F.; de Ferranti, S.D.; Ballantyne, C.M.; Larry, J.A. Contemporary Homozygous Familial Hypercholesterolemia in the United States: Insights from the CASCADE FH Registry. J. Am. Heart Assoc. 2023, 12, e029175. [Google Scholar] [CrossRef] [PubMed]
  107. Cuchel, M.; Raal, F.J.; Hegele, R.A.; Al-Rasadi, K.; Arca, M.; Averna, M.; Bruckert, E.; Freiberger, T.; Gaudet, D.; Harada-Shiba, M. 2023 Update on European Atherosclerosis Society Consensus Statement on Homozygous Familial Hypercholesterolaemia: New Treatments and Clinical Guidance. Eur. Heart J. 2023, 44, 2277–2291. [Google Scholar] [CrossRef] [PubMed]
  108. Raal, F.J.; Hovingh, G.K.; Catapano, A.L. Familial Hypercholesterolemia Treatments: Guidelines and New Therapies. Atherosclerosis 2018, 277, 483–492. [Google Scholar] [CrossRef]
  109. Fiorentino, R.; Chiarelli, F. Treatment of Dyslipidaemia in Children. Biomedicines 2021, 9, 78. [Google Scholar] [CrossRef]
  110. Sosnowska, B.; Adach, W.; Surma, S.; Rosenson, R.S.; Banach, M. Evinacumab, an ANGPTL3 Inhibitor, in the Treatment of Dyslipidemia. J. Clin. Med. 2023, 12, 168. [Google Scholar] [CrossRef]
  111. Gusarova, V.; Alexa, C.A.; Wang, Y.; Rafique, A.; Kim, J.H.; Buckler, D.; Mintah, I.J.; Shihanian, L.M.; Cohen, J.C.; Hobbs, H.H. ANGPTL3 Blockade with a Human Monoclonal Antibody Reduces Plasma Lipids in Dyslipidemic Mice and Monkeys. J. Lipid Res. 2015, 56, 1308–1317. [Google Scholar] [CrossRef]
  112. Coppinger, C.; Movahed, M.; Azemawah, V.; Peyton, L.; Gregory, J.; Hashemzadeh, M. A Comprehensive Review of PCSK9 Inhibitors. J. Cardiovasc. Pharmacol. Ther. 2022, 27, 107424842211001. [Google Scholar] [CrossRef]
  113. Xia, X.D.; Peng, Z.S.; Gu, H.M.; Wang, M.; Wang, G.Q.; Zhang, D.W. Regulation of PCSK9 Expression and Function: Mechanisms and Therapeutic Implications. Front. Cardiovasc. Med. 2021, 8, 764038. [Google Scholar] [CrossRef]
  114. Feingold, K.R.; Moser, A.H.; Shigenaga, J.K.; Patzek, S.M.; Grunfeld, C. Inflammation Stimulates the Expression of PCSK9. Biochem. Biophys. Res. Commun. 2008, 374, 341–344. [Google Scholar] [CrossRef]
  115. Ito, M.K.; Santos, R.D. PCSK9 Inhibition with Monoclonal Antibodies: Modern Management of Hypercholesterolemia. J. Clin. Pharmacol. 2017, 57, 7–32. [Google Scholar] [CrossRef] [PubMed]
  116. Abduljabbar, M.H. PCSK9 Inhibitors: Focus on Evolocumab and Its Impact on Atherosclerosis Progression. Pharmaceuticals 2024, 17, 1581. [Google Scholar] [CrossRef]
  117. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef]
  118. Santos, R.D.; Stein, E.A.; Hovingh, G.K.; Blom, D.J.; Soran, H.; Watts, G.F.; López, J.A.G.; Bray, S.; Kurtz, C.E.; Hamer, A.W. Long-Term Evolocumab in Patients with Familial Hypercholesterolemia. J. Am. Coll. Cardiol. 2020, 75, 565–574. [Google Scholar] [CrossRef]
  119. Raal, F.J.; Stein, E.A.; Dufour, R.; Turner, T.; Civeira, F.; Burgess, L.; Langslet, G.; Scott, R.; Olsson, A.G.; Sullivan, D. PCSK9 Inhibition with Evolocumab (AMG 145) in Heterozygous Familial Hypercholesterolaemia (RUTHERFORD-2): A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2015, 385, 331–340. [Google Scholar] [CrossRef]
  120. Koren, M.J.; Sabatine, M.S.; Giugliano, R.P.; Langslet, G.; Wiviott, S.D.; Ruzza, A.; Ma, Y.; Hamer, A.W.; Wasserman, S.M.; Raal, F.J. Long-Term Efficacy and Safety of Evolocumab in Patients with Hypercholesterolemia. J. Am. Coll. Cardiol. 2019, 74, 2132–2146. [Google Scholar] [CrossRef]
  121. Wołowiec, Ł.; Osiak, J.; Wołowiec, A.; Wijata, A.; Grześk, E.; Kozakiewicz, M.; Banach, J.; Nowaczyk, A.; Nowaczyk, J.; Grześk, G. Inclisiran—Safety and Effectiveness of Small Interfering RNA in Inhibition of PCSK-9. Pharmaceutics 2023, 15, 323. [Google Scholar] [CrossRef]
  122. Lagace, T.A. PCSK9 and LDLR Degradation: Regulatory Mechanisms in Circulation and in Cells. Curr. Opin. Lipidol. 2014, 25, 387–393. [Google Scholar] [CrossRef]
  123. Raal, F.J.; Hegele, R.A.; Ruzza, A.; López, J.A.G.; Bhatia, A.K.; Wu, J.; Wang, H.; Gaudet, D.; Wiegman, A.; Wang, J. Evolocumab Treatment in Pediatric Patients with Homozygous Familial Hypercholesterolemia: Pooled Data from Three Open-Label Studies. Arterioscler. Thromb. Vasc. Biol. 2024, 44, 1156–1164. [Google Scholar] [CrossRef]
  124. Raal, F.J.; Honarpour, N.; Blom, D.J.; Hovingh, G.K.; Xu, F.; Scott, R.; Wasserman, S.M.; Stein, E.A. Inhibition of PCSK9 with Evolocumab in Homozygous Familial Hypercholesterolaemia (TESLA Part B): A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2015, 385, 341–350. [Google Scholar] [CrossRef] [PubMed]
  125. Bansal, S.; Ruzza, A.; Sawhney, J.P.S.; Kulkarni, G.; Iyengar, S.; Mehta, V.; Hamer, A.; Wu, Y.; Raal, F.J. Evolocumab in Patients with Homozygous Familial Hypercholesterolemia in India. J. Clin. Lipidol. 2021, 15, 814–821. [Google Scholar] [CrossRef] [PubMed]
  126. Raal, F.J.; Mehta, V.; Kayikcioglu, M.; Blom, D.J.; Gupta, P.; Durst, R.; Asprusten, E.A.; Turner, T.; Bahassi, E.M.; Vest, J. Response to PCSK9 Inhibition in Homozygous Familial Hypercholesterolemia Patients Is Variable in Different Regions of the World and According to Age. Circulation 2023, 148, A15073. [Google Scholar] [CrossRef]
  127. Raal, F.; Bruckert, E.; Blom, D.; Kurtz, C.; Coll, B.; Tang, L.; Somaratne, R.; Stein, E.A. Evolocumab Treatment in Paediatric Patients with Homozygous Familial Hypercholesterolaemia: The Trial Assessing Long-Term Use of PCSK9 Inhibition in Subjects with Genetic LDL Disorders (TAUSSIG): 3105. Eur. Heart J. 2017, 38, ehx5043105. [Google Scholar] [CrossRef]
  128. Bruckert, E.; Caprio, S.; Wiegman, A.; Charng, M.-J.; Zárate-Morales, C.A.; Baccara-Dinet, M.T.; Manvelian, G.; Ourliac, A.; Scemama, M.; Daniels, S.R. Efficacy and Safety of Alirocumab in Children and Adolescents with Homozygous Familial Hypercholesterolemia: Phase 3, Multinational Open-Label Study. Arterioscler. Thromb. Vasc. Biol. 2022, 42, 1447–1457. [Google Scholar] [CrossRef]
  129. Xiao, G.; Gao, S.; Xie, Y.; Wang, Z.; Shu, M. Efficacy and Safety of Evolocumab and Alirocumab as PCSK9 Inhibitors in Pediatric Patients with Familial Hypercholesterolemia: A Systematic Review and Meta-Analysis. Medicina 2024, 60, 1646. [Google Scholar] [CrossRef]
  130. Daniels, S.; Caprio, S.; Chaudhari, U.; Manvelian, G.; Baccara-Dinet, M.T.; Brunet, A.; Scemama, M.; Loizeau, V.; Bruckert, E. PCSK9 Inhibition with Alirocumab in Pediatric Patients with Heterozygous Familial Hypercholesterolemia: The ODYSSEY KIDS Study. J. Clin. Lipidol. 2020, 14, 322–330. [Google Scholar] [CrossRef]
  131. Santos, R.D.; Ruzza, A.; Hovingh, G.K.; Wiegman, A.; Mach, F.; Kurtz, C.E.; Hamer, A.; Bridges, I.; Bartuli, A.; Bergeron, J.; et al. Evolocumab in Pediatric Heterozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 1317–1327. [Google Scholar] [CrossRef]
  132. Mormone, A.; Tortorella, G.; Esposito, F.; Caturano, A.; Marrone, A.; Cozzolino, D.; Galiero, R.; Marfella, R.; Sasso, F.C.; Rinaldi, L. Advances in Pharmacological Approaches for Managing Hypercholesterolemia: A Comprehensive Overview of Novel Treatments. Biomedicines 2024, 12, 432. [Google Scholar] [CrossRef]
  133. Nordestgaard, B.G.; Langsted, A. Lipoprotein (a) as a Cause of Cardiovascular Disease: Insights from Epidemiology, Genetics, and Biology. J. Lipid Res. 2016, 57, 1953–1975. [Google Scholar] [CrossRef]
Figure 1. Diagnostic criteria for pediatric FH were established by the European Atherosclerosis Society and the Simon Broome Register Group. The European Atherosclerosis Society criteria include: (i) persistently elevated LDL-C over 140 mg/dL, (ii) family history (parents or siblings) of FH, and (iii) parental history of LDL-C over 180 mg/dL or family history of premature coronary artery disease. Regarding the number and the combinations of these criteria, the diagnosis of FH in a pediatric patient is considered unlikely, possible, probable, or definite. Simon Broome criteria diagnose pediatric FH based on clinical (total cholesterol value, tendon xanthomas), genetic (pathogenic variants: LDLR, ApoB, PCSK9), and familial (myocardial infarction, hypercholesterolemia in a first- or second-degree relative) characteristics. [CAD: coronary arterial disease; EAS: European Atherosclerosis Society; FH: familial hypercholesterolemia; LDL-C: low-density lipoprotein cholesterol]. (The image was created using BioRender software version 04, License #VI28C5TP5H).
Figure 1. Diagnostic criteria for pediatric FH were established by the European Atherosclerosis Society and the Simon Broome Register Group. The European Atherosclerosis Society criteria include: (i) persistently elevated LDL-C over 140 mg/dL, (ii) family history (parents or siblings) of FH, and (iii) parental history of LDL-C over 180 mg/dL or family history of premature coronary artery disease. Regarding the number and the combinations of these criteria, the diagnosis of FH in a pediatric patient is considered unlikely, possible, probable, or definite. Simon Broome criteria diagnose pediatric FH based on clinical (total cholesterol value, tendon xanthomas), genetic (pathogenic variants: LDLR, ApoB, PCSK9), and familial (myocardial infarction, hypercholesterolemia in a first- or second-degree relative) characteristics. [CAD: coronary arterial disease; EAS: European Atherosclerosis Society; FH: familial hypercholesterolemia; LDL-C: low-density lipoprotein cholesterol]. (The image was created using BioRender software version 04, License #VI28C5TP5H).
Nutrients 17 02357 g001
Figure 2. Endogenous synthesis of cholesterol. The key process includes the multiple-step production of lanosterol and cholesterol in the liver. The SCAP protein plays a crucial role as well, enhancing SREBP translocation to the Golgi apparatus, finally leading to the synthesis of lipogenic proteins and sterols, including LDL-C. [FDFT1: farnesyl diphosphate farnesyltransferase 1; FDPS: farnesyl dimethylallyl pyrophosphate synthase; FPP: farnesyl pyrophosphate; HMG-CoA: hydroxy-3-methylglutaryl coenzyme A; HMGCR: HMG-CoA reductase; INSIG: insulin-induced gene protein; IPP: isopentenyl pyrophosphate isomerase; LDL: low-density lipoprotein; SCAP: SREBP cleavage-activating protein; SQLE: squalene epoxidase; SREBP-2: sterol regulatory element-binding protein-2; VLDL: very low-density lipoprotein] (The image was created using BioRender software version 04, License #SV28G95283).
Figure 2. Endogenous synthesis of cholesterol. The key process includes the multiple-step production of lanosterol and cholesterol in the liver. The SCAP protein plays a crucial role as well, enhancing SREBP translocation to the Golgi apparatus, finally leading to the synthesis of lipogenic proteins and sterols, including LDL-C. [FDFT1: farnesyl diphosphate farnesyltransferase 1; FDPS: farnesyl dimethylallyl pyrophosphate synthase; FPP: farnesyl pyrophosphate; HMG-CoA: hydroxy-3-methylglutaryl coenzyme A; HMGCR: HMG-CoA reductase; INSIG: insulin-induced gene protein; IPP: isopentenyl pyrophosphate isomerase; LDL: low-density lipoprotein; SCAP: SREBP cleavage-activating protein; SQLE: squalene epoxidase; SREBP-2: sterol regulatory element-binding protein-2; VLDL: very low-density lipoprotein] (The image was created using BioRender software version 04, License #SV28G95283).
Nutrients 17 02357 g002
Figure 3. Illustration of the main pathway of cholesterol absorption from the intestinal lumen. High dietary cholesterol, transported via the chylomicron pathway, may lead to excessive cholesterol deposition in the liver, increasing bile cholesterol. Reducing cholesterol absorption in the small intestine, by targeting the NPC1L1 pathway, decreases its availability for secretion into the bile. [ABCG5/8: ATP-binding cassette G 5/8; ACAT2: acetyl-Coenzyme A acetyltransferase; APOB48: apolipoprotein B48; MTP: microsomal triglyceride transfer protein; NPC1L1: Niemann-Pick C1-Like1] (The image was created using BioRender software version 04, License #SV28G95J5X).
Figure 3. Illustration of the main pathway of cholesterol absorption from the intestinal lumen. High dietary cholesterol, transported via the chylomicron pathway, may lead to excessive cholesterol deposition in the liver, increasing bile cholesterol. Reducing cholesterol absorption in the small intestine, by targeting the NPC1L1 pathway, decreases its availability for secretion into the bile. [ABCG5/8: ATP-binding cassette G 5/8; ACAT2: acetyl-Coenzyme A acetyltransferase; APOB48: apolipoprotein B48; MTP: microsomal triglyceride transfer protein; NPC1L1: Niemann-Pick C1-Like1] (The image was created using BioRender software version 04, License #SV28G95J5X).
Nutrients 17 02357 g003
Figure 4. Bile acid cholesterol recycling: the role of ABCG transporters in mature HDL synthesis, the initiation of cholesterol conversion to bile acids via CYP7A1 (inhibited by FGF15), and the fate of bile acids, either being excreted or recirculated via the hepatic portal vein. [ABCA1: ATP-binding cassette A1; ABCG: ATP-binding cassette G; CYP7A1: cholesterol 7α-hydroxylase; FGF15: fibroblast growth factor 15; HDL: high-density lipoprotein] (The image was created using BioRender software version 04, License #VI28G959BA).
Figure 4. Bile acid cholesterol recycling: the role of ABCG transporters in mature HDL synthesis, the initiation of cholesterol conversion to bile acids via CYP7A1 (inhibited by FGF15), and the fate of bile acids, either being excreted or recirculated via the hepatic portal vein. [ABCA1: ATP-binding cassette A1; ABCG: ATP-binding cassette G; CYP7A1: cholesterol 7α-hydroxylase; FGF15: fibroblast growth factor 15; HDL: high-density lipoprotein] (The image was created using BioRender software version 04, License #VI28G959BA).
Nutrients 17 02357 g004
Figure 5. A schematic representation of cholesterol metabolism. Dietary cholesterol is absorbed by the intestine, participating in the assembly of chylomicrons, which are secreted into the blood circulation. After being hydrolyzed, the chylomicron remnants are digested by a low-density lipoprotein receptor on the hepatocyte membrane in order to be absorbed by the hepatocytes. NPL1L1 regulates intestinal cholesterol absorption and further leads to chylomicron release in muscles and adipose tissue. In the liver, cholesterol de novo biosynthesis consists of several enzymatic reactions that convert acetyl-CoA into cholesterol. Sterol regulatory element-binding protein 2 and liver X receptor α are the major regulators of liver cholesterol homeostasis. Reverse cholesterol transport includes transport of free cholesterol to a pre–β high-density lipoprotein particle, generation of mature HDL and VLDL particles through esterification reactions, and cholesterol delivery to the liver through the LDL receptor. LDL-C is taken into hepatic cells, and excess cholesterol is delivered to HDL particles. [ER: endoplasmic reticulum; HDL: high-density lipoprotein; LDL: low-density lipoprotein; LXRα; liver X receptor α; NPC1L1: Niemann-Pick C1-Like1; SREBP2: sterol regulatory element-binding protein 2; VLDL: very low-density lipoprotein]. (The image was created using BioRender software version 04, License #YE28C5T6LJ).
Figure 5. A schematic representation of cholesterol metabolism. Dietary cholesterol is absorbed by the intestine, participating in the assembly of chylomicrons, which are secreted into the blood circulation. After being hydrolyzed, the chylomicron remnants are digested by a low-density lipoprotein receptor on the hepatocyte membrane in order to be absorbed by the hepatocytes. NPL1L1 regulates intestinal cholesterol absorption and further leads to chylomicron release in muscles and adipose tissue. In the liver, cholesterol de novo biosynthesis consists of several enzymatic reactions that convert acetyl-CoA into cholesterol. Sterol regulatory element-binding protein 2 and liver X receptor α are the major regulators of liver cholesterol homeostasis. Reverse cholesterol transport includes transport of free cholesterol to a pre–β high-density lipoprotein particle, generation of mature HDL and VLDL particles through esterification reactions, and cholesterol delivery to the liver through the LDL receptor. LDL-C is taken into hepatic cells, and excess cholesterol is delivered to HDL particles. [ER: endoplasmic reticulum; HDL: high-density lipoprotein; LDL: low-density lipoprotein; LXRα; liver X receptor α; NPC1L1: Niemann-Pick C1-Like1; SREBP2: sterol regulatory element-binding protein 2; VLDL: very low-density lipoprotein]. (The image was created using BioRender software version 04, License #YE28C5T6LJ).
Nutrients 17 02357 g005
Figure 6. Illustration of the mechanism of action and therapeutic properties of ezetimibe. Ezetimibe inhibits cholesterol absorption by directly attaching to a specific transmembrane domain of the NPC1L1 protein. Therefore, internalization of the NPC1L1/cholesterol complex is inhibited, preventing cholesterol from entering the enterocyte. This blocks the formation of initial Apo-B48 chylomicrons (through the ACAT/MTP pathway) that normally enter the bloodstream, and, as a result, LDL-cholesterol levels are decreased. Ezetimibe presents anti-inflammatory and anti-tumorigenic (antiangiogenetic, antiproliferative) properties, as well as reducing the risk of hypercholesterolemia complications. [ACAT: acyl-coenzyme A: cholesterol acyltransferase; CE: cholesterol esters; LDL: low-density lipoprotein; MTP: microsomal triacylglycerol transfer protein; NPL1L1: Niemann-Pick C1-Like1]. (The image was created using BioRender software version 04, License #VF28C5SX19).
Figure 6. Illustration of the mechanism of action and therapeutic properties of ezetimibe. Ezetimibe inhibits cholesterol absorption by directly attaching to a specific transmembrane domain of the NPC1L1 protein. Therefore, internalization of the NPC1L1/cholesterol complex is inhibited, preventing cholesterol from entering the enterocyte. This blocks the formation of initial Apo-B48 chylomicrons (through the ACAT/MTP pathway) that normally enter the bloodstream, and, as a result, LDL-cholesterol levels are decreased. Ezetimibe presents anti-inflammatory and anti-tumorigenic (antiangiogenetic, antiproliferative) properties, as well as reducing the risk of hypercholesterolemia complications. [ACAT: acyl-coenzyme A: cholesterol acyltransferase; CE: cholesterol esters; LDL: low-density lipoprotein; MTP: microsomal triacylglycerol transfer protein; NPL1L1: Niemann-Pick C1-Like1]. (The image was created using BioRender software version 04, License #VF28C5SX19).
Nutrients 17 02357 g006
Figure 7. Algorithm for the therapeutic evaluation of pediatric familial hypercholesterolemia in homozygous and heterozygous patients. Lifestyle changes, as well as 1st and 2nd line treatment (statins and ezetimibe, respectively), apply both to homozygotes (maximum statin dose) and heterozygotes (low to moderate statin dose). In cases of non-response to treatment, which are more common in homozygous FH patients due to the absent or significantly defective LDL receptor activity, LDL-C apheresis and the use of PCSK9 inhibitors should be considered. [ANGPTL3: angiopoietin protein-like 3; FH: familial hypercholesterolemia; LDL-C: low-density lipoprotein cholesterol; PCSK9: proprotein convertase subtilisin kexin9]. (The image was created using BioRender software version 04, License #MD28C5S0B7).
Figure 7. Algorithm for the therapeutic evaluation of pediatric familial hypercholesterolemia in homozygous and heterozygous patients. Lifestyle changes, as well as 1st and 2nd line treatment (statins and ezetimibe, respectively), apply both to homozygotes (maximum statin dose) and heterozygotes (low to moderate statin dose). In cases of non-response to treatment, which are more common in homozygous FH patients due to the absent or significantly defective LDL receptor activity, LDL-C apheresis and the use of PCSK9 inhibitors should be considered. [ANGPTL3: angiopoietin protein-like 3; FH: familial hypercholesterolemia; LDL-C: low-density lipoprotein cholesterol; PCSK9: proprotein convertase subtilisin kexin9]. (The image was created using BioRender software version 04, License #MD28C5S0B7).
Nutrients 17 02357 g007
Table 1. Novel therapeutic strategies targeting intestinal cholesterol metabolism for the treatment of pediatric FH.
Table 1. Novel therapeutic strategies targeting intestinal cholesterol metabolism for the treatment of pediatric FH.
Clinical Studies/TrialsMedicationTargetMechanism of Action% Reduction in LDL-CEfficacy in Pediatric FHSerious Adverse Events
Approved
[117,118,119,120]
EvolocumabPCSK9Prevents PCSK9 from degrading LDL-C receptor, increasing LDL-C receptor availability20–60%Efficient and approved for pediatric FH from 12 years of ageNot reported
ODYSSEY KIDS
[130]
AlirocumabPCSK9Prevents PCSK9 from degrading LDL-C receptor, increasing LDL-C receptor availability21–62%Efficient and off-label used for pediatric patients above 8 years of age with severe FHNot reported
ORION-16
[121]
InclisiranPCSK9PCSK9 gene-silencing medication35–43%Evaluated in pediatric FH in the ORION-16 studyN/A
[FH: familial hypercholesterolemia; LDL-C: low density lipoprotein cholesterol; PCSK9: proprotein convertase subtilisin kexin 9].
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

Arvanitakis, K.; Chatzikalil, E.; Antza, C.; Topalidis, C.; Kalopitas, G.; Solomou, E.; Kotsis, V.; Germanidis, G.; Koufakis, T.; Doumas, M. Pediatric Familial Hypercholesterolemia: Targeting Intestinal Absorption and Other Therapeutic Strategies. Nutrients 2025, 17, 2357. https://doi.org/10.3390/nu17142357

AMA Style

Arvanitakis K, Chatzikalil E, Antza C, Topalidis C, Kalopitas G, Solomou E, Kotsis V, Germanidis G, Koufakis T, Doumas M. Pediatric Familial Hypercholesterolemia: Targeting Intestinal Absorption and Other Therapeutic Strategies. Nutrients. 2025; 17(14):2357. https://doi.org/10.3390/nu17142357

Chicago/Turabian Style

Arvanitakis, Konstantinos, Elena Chatzikalil, Christina Antza, Christos Topalidis, Georgios Kalopitas, Elena Solomou, Vasilios Kotsis, Georgios Germanidis, Theocharis Koufakis, and Michael Doumas. 2025. "Pediatric Familial Hypercholesterolemia: Targeting Intestinal Absorption and Other Therapeutic Strategies" Nutrients 17, no. 14: 2357. https://doi.org/10.3390/nu17142357

APA Style

Arvanitakis, K., Chatzikalil, E., Antza, C., Topalidis, C., Kalopitas, G., Solomou, E., Kotsis, V., Germanidis, G., Koufakis, T., & Doumas, M. (2025). Pediatric Familial Hypercholesterolemia: Targeting Intestinal Absorption and Other Therapeutic Strategies. Nutrients, 17(14), 2357. https://doi.org/10.3390/nu17142357

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop