U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Feingold KR, Anawalt B, Blackman MR, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.

Cover of Endotext

Endotext [Internet].

Show details

Use of Lipid Lowering Medications in Youth

, PharmD, , PharmD, and , PharmD.

Author Information and Affiliations

Last Update: November 26, 2023.

ABSTRACT

The first comprehensive pediatric dyslipidemia guidelines were published by the National Cholesterol Education Program’s Expert Panel on Blood Cholesterol Levels in Children and Adolescents in 1992 and were updated by the American Academy of Pediatrics (AAP) in 1998. In 2008 the AAP issued an updated clinical report detailing recommendations for screening and evaluation of cholesterol levels in children and adolescents as well as prevention and treatment strategies. Since the publication of the first guidelines, rates of pediatric obesity have significantly increased, resulting in a concomitant increase in dyslipidemia. Recently, options for pharmacotherapeutic interventions in pediatric patients have expanded with new FDA approved indications of several lipid lowering medications, as well as additional safety and efficacy data. In 2011, The National Heart Lung and Blood Institute (NHLBI) published its comprehensive report Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents. As with previous guidelines, lifestyle modifications with an emphasis on a heart-healthy diet and daily moderate to vigorous exercise remain an integral part of treatment for pediatric lipid disorders; however, the recommendations for patients requiring management with pharmacotherapy have changed, and will be the focus of this discussion. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION

The diagnosis, treatment, and monitoring of dyslipidemia in youth has undergone significant transformations in recent years. As detailed by the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) and Bogalusa Heart studies, dyslipidemia plays a vital role in both the initiation, as well as the progression of atherosclerotic lesions in children and adolescents (1-3). Because of their role in premature cardiovascular disease, control of dyslipidemia provides clinicians with an opportunity for reducing morbidity and mortality. Observational data from individuals with genetic mutations that lower atherogenic cholesterol, low-density lipoprotein cholesterol (LDL-C) and non-high-density lipoprotein cholesterol (non-HDL-C), over a lifetime are associated with fewer events and longer life expectancy (4, 5). While these observations are very encouraging, it is not known if achieving the same level of lipid lowering with medications over decades will offer the same protective effects as observed in individuals with life-long lower cholesterol secondary to a genetic mutation (6). Table 1 provides a comparison across the evolution of guideline recommendations for the initiation of pharmacologic intervention with the goal of balancing risk and benefit (7-11). Table 2 details the risk factors and risk conditions described in the NHLBI guidelines (9).

Table 1.

Comparison of Recommendations for Treatment


Guidelines
NCEP, AAP – 1992 & 1998AAP – 2008NHLBI, AAP – 2011
Pharmacologic Treatment Initiation Parameters*Age > 10 years with LDL-CAge ≥ 8 years with LDL-CAges 10-21 years with LDL-C
o≥ 190 mg/dLo≥ 190 mg/dLo≥ 190 mg/dL
o> 160 mg/dL in addition to a positive family history of premature CVD or presence of at least 2 CVD risk factors in the child/adolescento≥ 160 mg/dL in addition to a positive family history of premature CVD or presence of risk factorso160-189 mg/dL in addition to a positive family history of premature CVD or presence of 1 high level risk factor/condition or presence of 2 moderate level risk factors/conditions
o≥ 130 mg/dL in addition to presence of diabetes mellitus
Age < 8 years with LDL-C:o130-159 mg/dL in addition to the presence of 2 high level risk factors/conditions or 1 high level and at least 2 moderate level risk factors/conditions
o≥ 500 mg/dL
Age < 10 years with severe hyperlipidemia or high-risk conditions associated with serious morbidity
*After an adequate trial of diet and lifestyle management.Ages 8-9 years with LDL-C levels consistently ≥ 190 mg/dL in addition to a positive family history OR presence of risk factors
Pharmacologic Medication RecommendationsBile acid sequestrantsBile acid sequestrantsStatins
Cholesterol absorption inhibitors
Statins

Table 2.

NHLBI Risk Factors and Risk Conditions (9)

NHLBI, AAP 2011 Guidelines: Risk Factors and Risk Conditions
High Level Risk FactorsHypertension requiring drug therapy
Tobacco use
BMI ≥ 97th percentile
High risk conditions
Moderate Level Risk FactorsHypertension not requiring drug therapy
BMI ≥ 95th percentile, < 97th percentile
HDL < 40 mg/dL
Moderate risk conditions
High Risk ConditionsT1DM and T2DM
CKD, ESRD, post-renal transplant
Post-orthotopic heart transplant
Kawasaki disease with current aneurysms
Moderate Risk ConditionsKawasaki disease with regressed aneurysms
Chronic inflammatory disease
HIV infection
Nephrotic syndrome

PHARMACOTHERAPEUTIC TREATMENT IN YOUTH

The treatment of youth with lipid lowering medications presents some unique challenges and consideration due to their developmental stage, the possibility of extended durations of treatment, and the potential use of concurrent medications that may be counter-productive by increasing lipid levels. As patients progress into adolescence it is particularly important for the patient to understand not only the need for their lipid lowering therapies, but also the consequences of non-compliance. Counseling regarding pharmacotherapy should begin at an early age with developmentally appropriate explanations and expand as patients mature. During adolescence when patients are developing their independence counseling that addresses how to integrate their therapy into their own social norms is important for achieving compliance to both pharmacotherapy as well as lifestyle modifications. Additionally, the cost of therapy significantly impacts compliance and should be factored into therapy decisions especially as youth transition into adulthood and may be faced with changes in insurance coverage. It is also critical to continually readdress the correct use of medications as patients are likely to be on multiple therapies and adolescents will begin some their own medication management as they mature. Regular monitoring for adverse events and side effects of therapy is essential as youth will have a greater lifetime exposure compared to adults and long-term data is generally limited.

As with all pharmacotherapy careful consideration should be given to potential drug interactions including those that may increase lipid levels. It is not uncommon for adolescent patients to be prescribed medications which have the potential to negatively impact lipid levels such as systemic steroids or oral contraceptive pills. Each patient case must be evaluated on an individual basis to determine the risk and benefit of prescribing medications which negatively alter lipid levels for patients also utilizing lipid lowering therapies. It should be noted there is significant risk for adolescent females should they become pregnant while taking lipid lowering medications as some have demonstrated a negative impact on fetal development. Adolescent females should be counselled regarding pregnancy and methods of contraception should be discussed. The US Medical Eligibility Criteria for Contraceptive Use compiled by the CDC details several contraceptive methods where the benefits generally outweigh any theoretical or proven risk for patients with hyperlipidemias (12).

HMG-CoA REDUCTASE INHIBITORS

HMG-CoA reductase inhibitors, or statins, are recommended as first line treatment of youth with severe dyslipidemia who fail non-pharmacologic interventions (i.e., diet and lifestyle modification) (8-11). Statins first debuted in clinical practice in 1987 with the FDA’s approval of lovastatin. At present, there are seven HMG-CoA reductase inhibitors with FDA approval, at varying dosages, for youth with heterozygous familial hypercholesterolemia. Lovastatin, simvastatin, atorvastatin and fluvastatin are approved for children 10 years of age and older. Pitavastatin and pravastatin are approved starting at 8 years of age and rosuvastatin is indicated in children as early as age 6 (13-21). Table 3 provides a summary of HMG-CoA reductase inhibitors, pediatric approval and indications, recommended dosing ranges, comments on dosing, and supporting clinical trials (13-21).

Table 3.

HMG-CoA Reductase Inhibitors

MedicationPediatric Approvals & IndicationsDosingCommentsSupporting
Clinical Trials
Atorvastatin Age 10-17
Heterozygous familial hypercholesterolemia
10-20 mg/dayMay be titrated at ≥ 4-week intervalsMcCrindle, et al (22)
Fluvastatin Age 10-16
Heterozygous familial hypercholesterolemia
20-80 mg/dayMay be titrated at ≥ 6-week intervalsvan der Graaf, et al (23)
Lovastatin Age 10-17
Heterozygous familial hypercholesterolemia
10-40 mg/dayInitiated at 20 mg/day for ≥20% LDL reduction, may be titrated at ≥ 4-week intervalsClauss, et al (24)
Lambert, et al (25)
Stein, et al (26)
Pravastatin Age 8 and older
Heterozygous familial hypercholesterolemia
20-40 mg/dayAge 8-13: 20 mg/day
Age 14-18: 40 mg/day
Knipscheer, et al (27)
Wiegman, et al (28)
Rodenburg, et al (29)
Rosuvastatin Age 6 and older
Heterozygous familial hypercholesterolemia
5-20 mg/dayMay be titrated at ≥ 4-week intervalsAvis, et al (30)
Simvastatin Age 10-17
Heterozygous familial hypercholesterolemia
10-40 mg/dayMay be titrated at ≥ 4-week intervalsde Jongh, et al (31)
de Jongh, et al (32)
Pitavastatin Age 8 and older
Heterozygous familial hypercholesterolemia
1-4 mg/dayMay be titrated at ≥ 4-week intervalsFerrari, et al (20)

The above are approved as an adjunct to a diet that is low in cholesterol and saturated fat. The above agents are approved for both males and females (females must be at least one-year post-menarche) if, despite an adequate diet and other non-pharmacologic measures, the following are present: LDL-C ≥ 190 mg/dL or LDL-C ≥ 160 mg/dL and the patient has a family history of premature cardiovascular disease or two or more cardiovascular disease risk factors. Abbreviations: mg=milligrams, LDL=low density lipoprotein.

As Table 4 outlines, statin therapies have demonstrated variable efficacy involving clinical trials of youth (21-32). With the longest half-life, rosuvastatin is the most potent statin, followed by atorvastatin (33). Given this knowledge, if pediatric patients who are initiated on statin therapy are having trouble meeting LDL-C goals, consideration should be given to switching to rosuvastatin or atorvastatin given potency prior to exploring second-line therapy options. Simvastatin is a moderately potent statin at clinically tolerable maximum doses of 40 mg/day (33-35). Lovastatin, pravastatin, and fluvastatin, respectively, are the least potent statins (34, 35). As many studies have demonstrated, reduced potency can be compensated by an increase in the amount of statin given; however, dose escalation is often associated with an increased occurrence of adverse events (21-35). As a result, selection of a specific statin therapy should be individualized and capable of reaching treatment goals. Equally important, consideration should be given to the prevalence and severity of reported side effects (36).

Table 4.

Statin Therapy Results

StudyMedicationDoseResults
LDL-CHDL-CTCTG
McCrindle, et al (22)Atorvastatin10-20 mg/day-40%+6%-30%-13%
van der Graaf, et al (23)Fluvastatin80 mg/day-34%+5%-27%-5%
Clauss, et al (24)Lovastatin40 mg/day-27%+3%-22%-23%
Lambert, et al (25)Lovastatin10 mg/day-21%+9%-17%-18%
Lambert, et al (25)Lovastatin20 mg/day-24%+2%-19%+9%
Lambert, et al (25)Lovastatin30 mg/day-27%+11%-21%+3%
Lambert, et al (25)Lovastatin40 mg/day-36%+3%-29%-9%
Stein, et al (26)Lovastatin10 mg/day-17%+4%-13%+4%
Stein, et al (26)Lovastatin20 mg/day-24%+4%-19%+8%
Stein, et al (26)Lovastatin40 mg/day-27%+5%-21%+6%
Knipscheer, et al (27)Pravastatin5 mg/day-23%+4%-18%+2%
Knipscheer, et al (27)Pravastatin10 mg/day-24%+6%-17%+7%
Knipscheer, et al (27)Pravastatin20 mg/day-33%+11%-25%+3%
Rodenburg, et al (29)Pravastatin20 mg/day or 40 mg/day-29%+3%-23%-2%
Wiegman, et al (28)Pravastatin20-40 mg/day-24%+6%-19%-17%
Avis, et al (30)Rosuvastatin5 mg/day-38%+4%-30%-13%
Avis, et al (30)Rosuvastatin10 mg/day-45%+10%-34%-15%
Avis, et al (30)Rosuvastatin20 mg/day-50%+9%-39%-16%
de Jongh, et al (31)Simvastatin10-40 mg/day-41%+3%-31%-9%
de Jongh, et al (32)Simvastatin40 mg/day-40%+5%-30%-17%

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report. Pediatrics. 2011;128(S5):S213-S256: Table 9-11. Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

Although long term studies evaluating the safety and efficacy of lipid-lowering medications in youth are lacking, results of short term observational and randomized controlled trials are encouraging. For example, atorvastatin was found to be well tolerated with no statistically significant differences in adverse events reported for either the treatment or placebo groups (22). Additionally, the percentage of patients with abnormal laboratory results was similar for both groups; the only noted difference was an increased percentage of patients with elevated triglycerides in the placebo group. Treatment with atorvastatin resulted in no significant difference in sexual development as assessed by Tanner staging. Van der Graaf and colleagues found that in youth treated with fluvastatin, 58 (68.2%) reported non-serious adverse events. Only four were believed to be drug related. Treatment with fluvastatin resulted in no abnormalities in hormone levels or sexual maturation (23).

In their study of lovastatin, Clauss and colleagues reported no clinically significant alterations in vital signs; growth; hormone levels including luteinizing hormone, follicle-stimulating hormone, dehydroepiandosterone sulfate, estradiol, and cortisol; menstrual cycle length; liver function tests; or muscle function tests (24). Lambert and colleagues also found lovastatin generally well tolerated with no serious clinical adverse effects noted (25). While increased, levels of aspartate aminotransferase did not exceed two times the upper limit of normal and alanine aminotransferase did not display significant changes in study participants. Creatine kinase was elevated, greater than three times the upper limit of normal, in three patients. All subjects remained asymptomatic and the elevated creatine kinase levels resolved spontaneously while no adjustment was need in their medication. Assessment of growth and sexual maturation, by Tanner staging and estimation of testicular volumes, in youth treated with lovastatin found no significant differences between the treatment groups and placebo at either 24 or 48 weeks (26). While the authors reported no significant change in serum hormone levels or biochemical parameters of nutrition, they noted that the study was under powered to detect statistically significant changes in these safety parameters.

Use of pravastatin has been shown to have minimal adverse events dispersed evenly between the active drug recipients and those who received placebo (27). Plasma thyroid-stimulating hormone, adrenocorticotropic hormone, cortisol, creatine phosphokinase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, and alkaline phosphatase levels failed to show significant changes from baseline in all treatment groups. Rodenburg and colleagues also evaluated the safety of pravastatin based on annual or biannual evaluation of plasma creatine phosphokinase levels, liver enzymes, sex steroids, gonadotropins, and hormones of the pituitary-adrenal axis (29). Height, weight, age at menarche, Tanner staging, and testicular volume were recorded at baseline and either annually or biannually. Two subjects demonstrated elevated creatine phosphokinase levels which returned to normal without adjustments in therapy, and were presumed to be due to extreme physical exercise. No occurrence of myalgia was associated with elevation in levels of creatinine phosphokinase. None of the subjects discontinued therapy due to adverse events or laboratory abnormalities. Similarly, Wiegman and colleagues examined the safety of pravastatin evaluated at baseline, one year, and two years via multiple variables including: sex steroids, endocrine function parameters, height, weight, body surface area, Tanner staging, menarche or testicular volume, alanine aminotransferase, aspartate aminotransferase, and creatine phosphokinase (28). All safety parameters demonstrated no statistically significant differences between active drug recipients verses placebo in changes from baseline.

The safety of rosuvastatin was assessed by Avis and colleagues. Laboratory monitoring including liver enzymes and creatine kinase levels, and markers of growth and development, such as Tanner staging (30). Two serious adverse events were reported including blurred vision in one patient in the placebo group and a vesicular rash progressing to cellulitis in one patient taking rosuvastatin 20 mg. Transaminase levels either remained normal or normalized without permanent discontinuation of treatment. While elevations in creatine kinase and reports of myalgia did occur, symptoms and creatine kinase levels normalized without permanent discontinuation of therapy. Normal progression of height, weight, and sexual development were observed.

The safety of simvastatin during short term therapy has also been reported. Levels of alanine aminotransferase, aspartate aminotransferase, creatine kinase, and physical examination all demonstrated no significant differences between simvastatin treated and placebo participants (31). de Jongh and colleagues evaluated the safety of simvastatin in a second trial by monitoring adverse events as well as changes in alanine aminotransferase, aspartate aminotransferase, and creatine kinase levels (32). Of note, none of the differences reported in events or laboratory values between simvastatin recipients and placebo reached statistical significance. Additionally, there were no statistically significant differences documented for height, body mass index, cortisol levels, testicular size and testosterone levels, menstrual cycle and estradiol levels, and Tanner staging. Dehydroepiandrosterone sulfate levels demonstrated a statistically significant decrease in the simvastatin group compared to placebo.

Braamskamp and colleagues investigated the efficacy and safety of pitavastatin in pediatric patients diagnosed with hyperlipidemia. 106 patients were enrolled in the study, ages 6-17, for a 12-week period (37). Patients were randomly assigned to 4 different groups categorized by dose 1 mg, 2 mg, 4 mg, or placebo (37). The results showed a reduction in all 3 dose groups in comparison to placebo, the 1 mg group showed a 23.5% reduction in LDL-C, the 2 mg group showed a 30.1% reduction, and the 4 mg group showed a 39.3% reduction (37). There was also a 52-week extension period where patients assigned to the 1 mg group were up-titrated to a maximum dose of 4 mg to try and achieve an LDL-C level of >110 mg/dL (37). There were no safety issues of concern throughout the study. The results indicated that pitavastatin is safe and efficacious for use in pediatric patients, 6-17 years of age, and it was well-tolerated (37).

Long-term data regarding the impact of statin therapy on growth, development, and reduction of cardiac risk are limited, particularly for high intensity statin therapy. Recently, Kusters and colleagues evaluated the safety of statin therapy in children and adolescents with familial hypercholesterolemia after 10 years of treatment comparing laboratory safety markers as well as growth and maturation in untreated siblings (36). Only three patients discontinued therapy due to adverse events. Safety parameters such as aspartate aminotransferase, alanine aminotransferase, creatine kinase, estimated glomerular filtration rate, c-reactive protein, and age of menarche did not differ between treated patients and siblings, demonstrating safety over the 10-year treatment period. The authors do note; however, that the study was underpowered to detect the occurrence of rare events (36).

When initiating HMG-CoA reductase inhibitor therapy, as with any new medication therapy, it is imperative for clinicians to establish an accurate baseline, monitor for new symptoms, and counsel both patients and family members regarding potential adverse events. Females should be informed about the need to avoid pregnancy and breastfeeding while using statins. Statins may be taken with or without meals, but are commonly given with the evening meal or at bedtime as this has the potential to improve LDL-C reduction (38). As a major substrate of P450, such as CYP3A4, there are multiple drug interactions associated with statin therapy. Grapefruit juice has gained considerable notoriety as a potential food interaction; however, it should be noted that more than a quart of grapefruit juice would have to be consumed to increase serum statin levels. Of more concern is the possible interaction of gemfibrozil and statins, which should either be avoided as it can increase the toxicity of HMG-CoA reductase inhibitors. Macrolides and antifungal azoles are classes of drugs commonly prescribed to children, and they should be avoided as often as possible as they increase serum statin levels leading to potentially enhanced myopathic effects. Additionally, patients should avoid herbal products and nutraceuticals, such as red yeast rice, which may further enhance adverse effects.

BILE ACID SEQUESTRANTS

Bile acid sequestrants, or bile acid binding resins, present an additional treatment option for youth with severe dyslipidemia. Bile acid sequestrants represent one of the oldest classes of medications available to treat dyslipidemia and were the only medication recommended in the 1992 NCEP Pediatric Panel Report, at a time when no data were available for statin use in youth (4). While no longer a recommended first-line therapy, bile acid sequestrants do have potential as a treatment option either alone or in combination with a statin (9, 10). At present, colesevelam is the only bile acid sequestrant with FDA approval for youth age 10 years and older with heterozygous familial hypercholesterolemia (39, 40). Despite the lack of FDA approval, both colestipol and cholestyramine have been studied in pediatric patients (41-49).

A number of clinical trials have evaluated the bile acid sequestrants in pediatric patients with heterozygous familial hypercholesterolemia and other forms of severe dyslipidemia. While palatability and tolerance remain potential barriers to effective therapy, in general, bile acid sequestrants have demonstrate significant reductions in both total cholesterol and LDL-cholesterol in study subjects (40, 43, 44, 48, 49). Table 5 provides a summary of bile acid sequestrants, pediatric approval and indications, recommended dosing ranges, comments on therapy, and supporting clinical trials. As outlined in Table 6, studies demonstrated some variability in efficacy for the available bile acid sequestrants (40, 43, 44, 48, 49).

Table 5.

Bile Acid Sequestrants

MedicationPediatric Approvals & IndicationsDosingCommentsClinical Trials
Colesevelam Age 10-17
Heterozygous familial hypercholesterolemia
1.875 g twice daily or 3.75 g dailyMay be used as monotherapy or in combination with a statinStein, et al (40)
Colestipol (Note: Not FDA Approved)
Age 7-12
Primary hypercholesterolemia
5 g twice daily, 10 g daily, or
125-500 mg/kg/day
N/AMcCrindle, et al (43)
Tonstad, et al (44)
(Note: Not FDA Approved)
Age ≥12
Primary hypercholesterolemia
10-15 g/dayN/A
Cholestyramine (Note: Not FDA Approved)
Age 6-12
Hypercholesterolemia adjunct
240 mg/kg/day divided three times daily before mealsInitiate at 2-4 g twice dailyMcCrindle, et al (48)
Tonstad, et al (49)
(Note: Not FDA Approved)
Age ≥12
8 g/day divided twice daily before mealsN/A

Colesevelam is approved as an adjunct to a diet that is low in cholesterol and saturated fat. Colesevelam is approved for both males and females (females must be at least one-year post-menarche) if, despite an adequate diet and other non-pharmacologic measures, the following are present: LDL-C ≥ 190 mg/dL or LDL-C ≥ 160 mg/dL and the patient has a family history of premature cardiovascular disease or two or more cardiovascular disease risk factors. Abbreviations: g=grams, mg=milligrams, kg=kilograms, N/A=not applicable.

Table 6.

Bile Acid Sequestrant Results

StudyMedicationDoseResults
LDL-CHDL-CTCTG
Stein, et al (40)Colesevelam1.875 g/day-6%+5%-3%+6%
Stein, et al (40)Colesevelam3.75 g/day-13%+8%-7%+5%
McCrindle, et al (43)Colestipol10 g/day-10%+2%-7%+12%
McCrindle, et al (43)Colestipol & PravastatinColestipol: 5 g/day
Pravastatin: 10 mg/day
-17%+4%-13%+8%
Tonstad, et al (44)Colestipol2-12 g/day-20%-7%-17%-13%
McCrindle, et al (48)Cholestyramine8 g/day-10% to
-15%
+2% to +4%-7% to
-11%
+6% to +9%
Tonstad, et al (49)Cholestyramine8 g/day-17%+8%-12%N/A

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report. Pediatrics. 2011;128(S5):S213-S256: Table 9-11. Abbreviations: g=grams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

Stein and colleagues assessed the safety of colesevelam at weeks 8-26 during an open-label study from. All subjects received colesevelam 3.75 grams per day in addition to a statin (40). Safety was measured via adverse events, vital signs and physical exam, laboratory monitoring, and Tanner staging. The most common adverse events related to use of colesevelam were gastrointestinal, including diarrhea, nausea, vomiting, and abdominal pain. It is important to note that no choking or difficulty swallowing were reported with the use of colesevelam. Vital signs, physical exams, laboratory monitoring, and Tanner staging remained the same or progressed as expected throughout the study period.

McCrindle and colleagues evaluated conventional high-dose colestipol versus a combination of low-dose colestipol plus pravastatin, but did not cite safety as an endpoint for their study (40). The researchers did conduct safety monitoring in the form of laboratory tests, physical evaluation, and adverse event reporting. Significant deviations from baseline were noted for alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase at various time intervals with different medication regimens; however, the authors noted that when compared to reference values, none of the laboratory results were considered abnormal. Study participants in the two medication regimens did not significantly vary in weight gain, height changes, or body mass index. While the majority of patients experienced no adverse events, gastrointestinal symptoms such as constipation, gas or bloating, or stomach ache were more commonly reported by the patients taking the high-dose colestipol. The authors found similar suboptimal compliance with both regimens as determined by medication counts at the end of each study period. Tonstad and colleagues also assessed the tolerability of colestipol granules by monitoring side effects as well as by having subjects’ complete subjective evaluations (44). Side effects associated with colestipol included constipation, dyspepsia, flatulence, nausea, reduction in appetite, and abdominal pain. The subjective evaluations indicated that only 21% of patients liked the taste of the colestipol; however, of those who had previously taken bile acid binding resin, 86% preferred the taste of the newer orange flavored granules. Thirty seven percent of subjects also reported that they frequently forgot to take the medication, while 44% intentionally eliminated the medication from their routine on special occasions or during trips.

The acceptability and compliance of cholestyramine has been studied (48). Eighty two percent (82%) of children preferred the pill formulation of cholestyramine compared to 16% who preferred the powder. Two percent of children in the study preferred neither form of the medication. Compliance was significantly impacted by medication formulation with patients taking the pill form reporting 61% compliance while those on the powder formulation were only 50% compliant. Compliance increased by at least 25% for 42% of patients when they switched to the pill formulation. Tonstad and colleagues assessed the safety of cholestyramine by measuring height velocity, erythrocyte folate, total plasma homocysteine, serum fat-soluble vitamins, and side effects (49). Weight and mean height velocity standard deviation scores were not statistically significant between treatment and placebo groups during the study. The cholestyramine active treatment group demonstrated decreased vitamin D levels and increased homocysteine levels. Differences in erythrocyte folate were not significant between the active treatment and placebo groups. Reported adverse events included intestinal obstruction, abdominal pain, nausea, and loose stools. Unpalatability was a common reason participants withdrew from the study.

As demonstrated by the previous studies, while bile acid sequestrants do present an effective therapy option, their side effect profile, issues of tolerability and drug interactions with statins make their use clinically challenging. It is generally recommended that all concurrent medications be given either one hour before or four hours after bile acid sequestrants to prevent decreased absorption of the additional therapies (41, 46). Use of bile acid sequestrants is generally limited to patients optimized on statin therapy who require additional therapy to achieve goal or those that cannot tolerate statins. Data on long-term safety, however, are generally lacking. It should also be noted that bile acid sequestrants can increase triglyceride levels and should not be used in patients with increased triglyceride levels.

FIBRIC ACID DERIVATIVES

Experience with fibric acid derivatives in youth is limited. Currently there are no fibric acid derivatives with FDA approval for use in pediatric patients. Both fenofibrate and gemfibrozil are available in the United Sates, but lack pediatric data on safety, efficacy, and dosing (50, 51). While there is very limited information on the use of bezafibrate in youth, the product is not available in the United States (52). It should be noted that fibric acid derivatives have the potential to increase the incidence in adverse events, such as rhabdomyolysis, when used with statins (45, 50, 51). However, the use of fibrates should be considered and can be beneficial in pediatric patients who also have triglyceride abnormalities (TG levels > 500 mg/dL) (9, 53).

NIACIN

Niacin provides a potential adjunct therapeutic option for youth with severe dyslipidemia who have not achieved their lipid goal. Extended-release niacin is the only formulation that has FDA-approval for use in children ≥ 16 years of age (54). Despite a lack of FDA approval for ages younger than 16, limited efficacy and safety data are published for the use of niacin in children 10 years of age as older as adjunct therapy (54). Table 7 summarizes data on recommended dosing ranges, comments on dose adjustments, and references supporting clinical trials.

Colletti and colleagues conducted a retrospective review to evaluate the efficacy and adverse effect profile of niacin for children with severe hypercholesterolemia (54). The effects on serum lipid profiles are detailed in Table 8. Adverse effects were common, affecting 76% of children, and similar to those reported for adults including: flushing, abdominal pain, vomiting, headache, and elevated liver enzymes. Due to the high prevalence of adverse effects, use of niacin should be limited to patients not achieving goal with other therapies or those who cannot tolerate alternative adjunctive options. As with fibrates, niacin can also be considered for the purposes of treating pediatric patients who are concurrently diagnosed with hypertriglyceridemia (9).

Table 7.

Niacin

MedicationPediatric Approvals & IndicationsDosingCommentsClinical Trials
Niacin Extended release: >16 years of age
Note: Immediate release is not FDA Approved:
Age ≥ 10
Adjunct therapy
Initial: 100-250 mg/day
(Max: 10 mg/kg/day)
divided three times daily with meals
May titrate weekly by 100 mg/day or every 2-3 weeks by 250 mg/dayColletti, et al (54)

Abbreviations: mg=milligrams, kg=kilograms.

Table 8.

Niacin

StudyMedicationDoseResults
Colletti, et al (54)Niacin500-2,250 mg/dayLDL-CHDL-CTCTG
-17%+4%-13%+13%

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report. Pediatrics. 2011;128(S5):S213-S256: Table 9-11. Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

EZETIMIBE

Ezetimibe is FDA approved for adolescents 10 years of age and older with FH (53, 55), and it presents a potential therapy option either as monotherapy or when synergistically paired with an HMG-CoA reductase inhibitor (56-58). Due to its favorable tolerability, it has become the most frequently used second-line agent (59). Table 9 summarizes data on recommended dosing ranges and references supporting clinical trials while Table 10 details efficacy of therapy.

Tolerability of ezetimibe was prospectively evaluated by Yeste and colleagues via a combination of biochemical markers and adverse event reports (56). No change was seen in hemogram, transaminases, creatinine, calcium, phosphorus, and vitamins A and E for any of the 17 patients. Additionally, there were no reports of adverse events during the study period. Clauss and colleagues retrospectively evaluated ezetimibe; therefore, safety parameters were less defined, but included intermittent measurement of liver enzymes, occasional CK levels, and adverse event reports (57). There were no reported abnormalities in liver enzymes for study participants. Ultimately, one patient was discontinued from ezetimibe therapy for asymptomatic elevated CK levels, later determined to be likely unrelated to therapy.

Table 9.

Ezetimibe

MedicationPediatric Approvals & IndicationsDosingCommentsClinical Trials
Ezetimibe Age ≥10
Homozygous familial hypercholesterolemia
10 mg/dayN/AYeste, et al (56)
Clauss, et al (57)
van der Graaf, et al (58)

Abbreviations: mg=milligrams, N/A=not applicable.

Table 10.

Ezetimibe

StudyMedicationDoseResults
Yeste, et al (56)Ezetimibe10 mg/dayLDL-CHDL-CTCTG
PH-42%N/A-31%N/A
FH-30%-15%-26%N/A
Clauss, et al (57)Ezetimibe10 mg/dayLDL-CHDL-CTCTG
FH-28%N/A-22%N/A
FCHLN/A-13%N/A
van der Graaf, et al (58)Ezetimibe
&
Simvastatin
Ezetimibe: 10 mg/day
Simvastatin: 10-40 mg/day
LDL-CHDL-CTCTG
-49%+7%-38%-17%

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report. Pediatrics. 2011;128(S5):S213-S256: Table 9-11. Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides, PH=polygenic hypercholesterolemia, FH=familial hypercholesterolemia, FCHL=familial combined hyperlipidemia.

Van der Graaf and colleagues assessed the safety of combination therapy with ezetimibe and simvastatin based on reported adverse events as well as laboratory monitoring and clinical examination (58). After 53 weeks, 71% of study participants reported some types of treatment-emergent adverse events. Of those events reported, only influenza, nasopharyngitis, and headache occurred in greater than 5% of participants. Consecutive transaminase elevations of at least three times the upper limit of normal were reported in 6 participants; however, all values resolved with interruption or discontinuation of therapy. Elevations in creatine phosphokinase occurred infrequently and were not associated with myalgia at levels greater than three times the upper limit of normal. Height, weight, and sexual maturation were not significantly impacted by therapy. Ezetimibe affords flexibility in administration time with the ability to administer it without regard to meals or time of day (45). HMG-CoA reductase inhibitors have the risk of increasing myopathy and elevation in hepatic transaminases, but are generally considered a safe combination with ezetimibe.

OMEGA-3 FISH OILS

Omega-3 fish oils are a class of therapy for which there is significantly limited data in youth. To date the FDA approved formulations of omega-3 fatty acid lack a pediatric indication. High dose omega-3 fatty acid supplementation was evaluated by de Ferranti and colleagues, but ultimately the authors found no statistically significant improvement when subjects were compared to placebo (60). Chahal and colleagues similarly found no significant impact on hypertriglyceridemia when treating pediatric patients with fish oil (61).

Khorshidi and associates performed a systematic review and meta-analysis of the effect of omega-3 supplementation on lipid profiles in children and adolescents. They found that omega-3 supplementation improved triglyceride levels in patients diagnosed with hypertriglyceridemia that were less than or equal to 13 years of age; however, there was no significant effect seen in HDL, LDL, or TC values (62). Omega-3 fish oils can be considered in those who have elevated triglyceride levels.

FAMILIAL HYPERCHOLESTEROLEMIA AND THERAPEUTIC ADVANCES

Familial hypercholesterolemia (FH) is a common, but often misdiagnosed inherited gene disorder (63). The most common gene mutation seen in FH is in the low-density lipoprotein receptor gene (LDLR) accounting for 85-90% of cases, followed by the apolipoprotein (ApoB) gene (5-15% of cases) and the proprotein convertase subtilisin kexin 9 (PCSK9) genes (1% of cases) (63, 64). Patients who are diagnosed with FH have abnormally elevated LDL levels from birth. FH is associated with a twenty-fold increased risk in premature cardiovascular disease and cardiovascular events (65). There are two different types of FH, heterozygous (HeFH) and homozygous (HoFH). Heterozygous patients have one mutated allele and are more commonly seen in practice, while homozygous patients have two mutated alleles and are very rare (66). Distinction between the two types is imperative because HoFH patients tend to be treatment resistant and carry a worse prognosis, if left untreated, these patients rarely make it past age 20 (66).

Lipid lowering in these patients can be quite challenging, especially HoFH patients. Most typical therapies for lipid lowering require functional LDL receptors, therefore given the gene mutations which often render the receptors inactive, modest reductions of 10-25% in HoFH patients are usually all that will be gained (67). HeFH patients tend to see higher rates of reduction (25-40%) (68). However, first line treatment for both forms of FH is still high-intensity statin therapy at moderate to high doses to be initiated as early as age 8 (69). All seven statins are FDA-approved for the treatment of FH and have been proven to slow down the progression of carotid intima-media thickness (63). Statins also reduce the incidence of cardiovascular events and cardiac death (63). Until recently, long term data on the use of statins in this patient population was not available, but in 2019, a 20-year follow-up study of statins in pediatric patients with FH was published (70). The results found that the incidence of cardiovascular events and death was much lower in patients treated with statins (70). If LDL-C goals are not being met with the use of statins alone, the next recommend agent is ezetimibe; however, this should not be used in patients younger than 10 years of age (59).

Ezetimibe is the second-line option to statins in these patients. It’s use in combination with statins has demonstrated a reduction of LDL-C levels below 135 in more than 90% of children with FH (71). In one of the only studies that assessed the coadministration of simvastatin and ezetimibe in children with HeFH, it was found to be safe, well tolerated and provided a higher LDL reduction (15%) compared with simvastatin alone in HeFH patients (72). However, when it was investigated as a monotherapy option for children with HeFH, it only produced LDL-C lowering of 27% (71, 73). It is more appropriate to use as an adjunct therapy in this population of patients.

Alternative medications that can be considered other second-line options are bile acid sequestrants (71). Colesevelam is safe to use, but limited to children >10 years of age (71). These drugs however have minimal LDL-C lowering effects, usually only seeing a 10-20% reduction, and more importantly are very poorly tolerated due to gastrointestinal side effects (71).

PCSK9 Inhibitors

Given the difficulty and importance of treating these patients, especially those with HoFH, there is a need for stronger lipid lowering options, which is where PCSK9 inhibitors come into play. These are a more recent class addition to the therapy options for managing FH, which help to reduce the degradation of LDL receptors and the removal of LDL-cholesterol (74). In a recent study assessing the efficacy/safety of lipid-lowering agents in patients with familial hypercholesterolemia, it was concluded that PCSK9 inhibitors were the most effective in lowering lipid levels (75). They have none of the same side effects as statins and produced similar CV benefits. Therefore, based on these conclusions, PCSK9 inhibitors are recommended as first-line agents in patients with hypercholesteremia that have intolerances or resistance to statins (75). There is currently one PCSK9 inhibitor approved for pediatric use. Repatha (evolocumab) was originally approved for use in patients 13 years of age and older (76), but the HAUSER-RCT study assessed the use of Repatha in patients ages 10-17 years of age for 24 weeks. It showed that the drug improved lipid levels (by approximately 38% in HeFH patients and 21-24% in HoFH patients) and was safe for use as the incidence of adverse events was similar in both the drug and placebo groups (71, 77). So now it is considered safe to use in pediatric patients 10 years of age and older. The HAUSER-RCT trial was then continued for another 80 weeks to further assess the safety and efficacy of Repatha (78). This new trial, HAUSER-OLE, further confirmed that the drug was safe and well-tolerated (78).

Praluent (Alirocumab) is another PCSK9 inhibitor that is available for treatment of FH, however it is not currently approved for use in pediatric patients (79). Nevertheless, there are studies currently assessing the safety and efficacy within this population. Bruckert and associates utilized Praluent and conducted an open-label phase 3 study specifically in pediatric patients (8-17 years of age) diagnosed with HoFH that were inadequately controlled (80). Patients received 75 or 150 mg of the drug based on weight (<50 or >50 kg, respectively) every 2 weeks for 12 weeks (80). The primary endpoint was percent change in LDL-C levels from week 0 to 12 (80). Interestingly, the results showed only a 4.1% decrease in LDL-C levels by week 12 (80). The secondary endpoints (assessing percent change LDL-C levels from baseline to weeks 24 and 48, changes in other lipid parameters from baseline to weeks 12, 24, 48, patients with a reduction of more than 15% in LDL-C levels at weeks 12, 24 and 48, and absolute change in LDL-C from baseline to weeks 12, 24, and 48) produced incredibly variable results (80). Overall, there were quite small changes in LDL-C levels observed in this study with mean reductions of LDL-C levels noted to range anywhere from ~33 to 52 mg/dL (80). More importantly, previous studies have shown that PCSK9 inhibitors are linked to a decrease in major coronary/vascular events and all-cause mortality, so although the results produced small values, the changes seen are still clinically significant based on these added benefits (80). It was also noted that this study produced similar results when compared to the ODYSSEY study which assessed the use of Praluent in adult patients with HoFH (80). The drug was deemed safe and there were no issues with tolerability (80). The study supports the use of Praluent as an adjunct therapy in HoFH patients already on first- and second-line therapies and not reaching their goal LDL-C levels (80).

Another study assessed the use of Praluent in pediatric patients diagnosed with HeFH. The ODYSSEY KIDS study was a phase 2 dose-finding study that enrolled pediatric patients anywhere from 8-17 years of age (81). Patients were split into 4 cohorts and dosed every 2 weeks. Dosing was determined by weight and the primary endpoint assessed percent change in LDL-C from baseline to week 8 (81). Praluent demonstrated the best reduction in LDL-C levels in the highest dosed cohorts and was well-tolerated. This study also supported the use of the drug (with further analysis) in patients who require adjunct therapy, there is a phase 3 trial planned to assess the doses from this study that resulted in the greatest reduction in LDL-C levels. Overall, it is important to note that HoFH patients are more likely to fail PCKS9 inhibitors (82). This is attributable to their mechanism of action. This class of medication requires functional LDL receptors, and this is impaired or completely absent in HOFH patients (82). Therefore, effectiveness of PCSK9 inhibitors tends to be much higher in HeFH patients (82).

Leqvio (inclisiran) is also another PCSK9 inhibitor currently not approved for pediatric use (83). The mechanism of action of this drug differs from Repatha and Praluent. Leqvio is a small interfering RNA (siRNA) that utilizes the RNA interference mechanism to cause the catalytic breakdown of mRNA for PCSK9, thus stopping the translation of the protein (84). It also only requires administration twice yearly as opposed to biweekly (84). There are currently ongoing studies investigating the possibility of using Leqvio in pediatric patients. ORION-13 and ORION-16 are studies assessing the efficacy, safety and tolerability of Leqvio in pediatric patients diagnosed with HoFH and HeFH, respectively (84). They are two-part (1-year double blind, the other year open-label) phase 3 trials consisting of patients aged 12 to <18 years with FH (84). The primary endpoint is the percentage change in LDL-C from baseline to day 330 (84). Based on the results, this could be another drug option as adjunct therapy to consider for use.

Angiopoietin-Like Protein 3 (ANGPTL3)

Angiopoietin-like protein 3 (ANGPTL3) also presents a novel target of adjunctive therapy for patients with homozygous familial hypercholesterolemia that are not meeting LDL-C goals with first-line agents (85, 86). Evkeeva (evinacumab-dgnb) is a monoclonal ANGPTL3 inhibitor that is FDA-approved specifically for the adjunctive treatment of homozygous familial hypercholesterolemia in patients 5 years of age and older (87).

Lomitapide

Lomitapide is another potential treatment option for patients with HoFH. This medication works differently from more conventional options. It binds to microsomal triglyceride transfer protein (MTP) and prevents the production of lipoproteins that contain apo-B (88). This causes a decrease in the production of very-low-density lipoprotein (VLDL) and chylomicrons. Since VLDL is converted into LDL, this mechanism ultimately causing a decrease in LDL-C levels (89). It is administered once daily at doses ranging from 5 to 60 mg (88). The side effect profile of lomitapide can be difficult for patients as it can cause severe gastrointestinal side effects (due to the decrease in absorption of fats in the intestines), most often diarrhea (89). But it is also associated with raised hepatic fats and enzymes (82). It is currently approved for adult use only, but it has become an option for use in pediatric patients through an expanded access program or a named patient basis (82). There was a case series done exploring the effect of lomitapide in 11 pediatric patients diagnosed with HoFH. It demonstrated that the drug was effective in reducing LDL-C with all 11 HoFH patients and showed a similar side effect profile to that seen in adult patients (82). GI complaints were moderated and did not cause any discontinuation of use (82). It also showed greater reduction in LDL-C levels at lower doses (82). The greatest benefit of lomitapide was associated with its ability to reduce or stop the need for lipoprotein apheresis in the patients incorporated in this case study (82). An interesting mention about lomitapide from the case series is that adult patient data shows that early intervention utilizing the drug showed a potential for increased life expectancy and a delay in the time to first major adverse cardiovascular event (82). There is also currently an ongoing phase 3, open label trial investigating the efficacy and safety of lomitapide in pediatric patients with HoFH, estimated completion date is April of 2024 (82).

Bempedoic Acid

Bempedoic acid is a new medication that exerts its effects very similarly to that of statins. It works in the same pathway as statins and targets cholesterol biosynthesis (90-92). It is administered however as a prodrug and converted to active drug only in the liver and not in the muscles (90-92). The other difference between the two classes is that bempedoic acid inhibits ATP-citrate lyase (ACL), while statins inhibit HMG CoA reductase (90-92). Due to the lack of activation in skeletal muscles, this drug is a promising alternative to patients unable to take statins due to muscle related symptoms (90-92). The medication is FDA approved for use in patients with HeFH and those with established cardiovascular disease (93). It has shown promising results in adult trials, but there are currently no published pediatric trials to date assessing the safety or efficacy of use of the drug (93). There does however appear to be a trial in development: “An Open-Label Study to Evaluate the Pharmacokinetics, Pharmacodynamics, and Safety of Bempedoic acid in Pediatric Patients with Heterozygous Familial Hypercholesterolemia.” The results are highly anticipated so that this can offer another promising drug class for use in patients intolerant or unable to meet their LDL-C goals.

FAMILIAL CHYLOMICRONEMIA SYNDROME

Familial chylomicronemia syndrome (FCS) is an incredibly rare autosomal recessive gene disorder (94). There is reduced or absent lipoprotein lipase activity causing disruption in chylomicron metabolism leading to severely elevated triglyceride levels resulting in acute recurrent pancreatitis (94). There is not however an increased risk of ASCVD with an FCS diagnosis (94). The best way to treat FCS is also often referred to as the most difficult as it requires patients to restrict dietary intake to <10-15% of daily calories (94). Other treatment options utilized are fibrates, omega-3 fatty acids and statins with variable responses, but the use of these medications is most commonly seen in patients who have multifactorial chylomicronemia syndrome (94). Given the difficulty of ensuring these patients maintain low levels of triglycerides, medications like volanesorsen are being examined (94, 95).

Volanesorsen

Volanesorsen is a second-generation 2’-O-methoxyethyl (2’-MOE) antisense therapeutic oligonucleotide. It works by inhibiting apoC3 thus lowering triglyceride plasma levels (94). When it binds to apoC3, this interrupts mRNA translation which consequently promotes triglyceride clearance/lowering of triglyceride plasma levels (94). The efficacy and safety of volanesorsen was assessed in the APPROACH study (96). It included 67 patients that were randomized to either weekly volanesorsen or placebo for 3 months (97). The results showed a 77% reduction in triglyceride plasma levels at the end of the study period and there was only 1 event of pancreatitis in the study group (97). The largest trial performed assessing the use of volanesorsen was the COMPASS trial (97). It included 114 patients who were randomized to either weekly injections of volanesorsen or placebo for a total of 26 weeks (97). The results showed that patients in the treatment group saw a reduction in triglyceride levels, chylomicron triglycerides, VLDL levels and apoC3 levels by more than 70% (97). There were also no occurrences reported of pancreatitis in any of the patients randomized to the volanesorsen group (93). In both trials, volanesorsen proved itself as a promising agent for treatment of hypertriglyceridemia in FCS patients (95-98). This drug is not approved for use in the US but is approved in other countries.

CONCLUSION

As noted in the 2011 NHLBI’s guidelines, available information regarding the treatment of youth with lipid disorders has greatly expanded. HMG-CoA reductase inhibitors, or statins, are now considered first-line pharmacologic treatment of children and adolescents with severe hypercholesterolemia who fail treatment with diet and exercise alone, although statins are only FDA approved for youth with familial hypercholesterolemia. Despite their ability to effectively reduced cholesterol levels, use of bile acid sequestrants continue to pose challenges for pediatric patients due to their unpalatability and are typically utilized as adjunctive therapy or for patients not able to tolerate statins. Fibric acid derivatives, as a class of medications, not only lack an FDA approved agent, but also continue to lack significant pediatric safety and efficacy data. Niacin, a potential adjunct therapy, lacks FDA approval for pediatric patients and is plagued by significant adverse effects, making it an unlikely therapeutic option for youth. Ezetimibe provides clinicians with an alternative adjunct therapy option when synergistically paired with an HMG-CoA reductase inhibitor or used as monotherapy for patients intolerant to statins and bile acid sequestrants. Despite their inherit appeal and popularity amongst the lay public, omega-3 fish oils have failed to demonstrate statistically significant cholesterol lowering in pediatric and adolescent patients, but can be used to lower triglyceride levels. PCSK9 and ANGPTL3 inhibiting agents are promising novel treatment options in pediatric patients diagnosed with FH. While recent years have witnessed a dramatic increase in studies of lipid lowering medications in youth, the long-term safety and efficacy data continue to present an active focus of research.

REFERENCES

1.
McMahan CA, Gidding SS, Malcom GT, Tracy RE, Strong JP, McGill Jr HC. Pathobiological determinants of atherosclerosis in youth risk scores are associated with early and advanced atherosclerosis. (PDAY) Pediatrics. 2006;118(4): 1447-1455. [PubMed: 17015535]
2.
Newman III WP, Freedman DS, Voors AW, et al. Relation of serum lipoprotein levels and systolic blood pressure to early atherosclerosis. (The Bogalusa Heart Study). N Engl J Med. 1986;314(3):138-44. [PubMed: 3455748]
3.
Berenson GS, Srinivasan SR, Bao W, Newman III WP, Tracy RE, Wattigney WA. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. (The Bogalusa Heart Study). N Engl J Med. 1998;338(23):1650-1656. [PubMed: 9614255]
4.
National Cholesterol Education Program (NCEP): Highlights of the Report of the Expert Panel on Blood Cholesterol Levels in Children and Adolescents. Pediatrics. 1992;89:495-501. [PubMed: 1741227]
5.
American Academy of Pediatrics. Committee on Nutrition. Cholesterol in childhood. Pediatrics. 1998;101 (1 Pt 1): 141-7. [PubMed: 11345978]
6.
Daniels SR, Greer FR. Lipid Screening and cardiovascular health in childhood. Pediatrics. 2008;122:198-208. [PubMed: 18596007]
7.
National Center for Chronic Disease Prevention and Health Promotion, Division of Adolescent and School Health. Youth Risk Behavior Sheet(YRBS): National Trends, 1991-2009: Obesity, Dietary Behaviors, and Weight Control Practices. http://www​.cdc.gov/healthyyouth​/yrbs/pdf​/us_obesity_trend_yrbs.pdf. Updated October 5, 2011. Accessed December 2011.
8.
McCrindle BW, Urbina EM, Dennison BA, Jacobson MS, Steinberger J, Rocchini AP, Hayman LL, Daniels SR. American Heart Association scientific statement: drug therapy of high-risk lipid abnormalities in children and adolescents. Circulation. 2007;115:1948-67. [PubMed: 17377073]
9.
National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report. Pediatrics. 2011;128(S5):S213-S256. [PMC free article: PMC4536582] [PubMed: 22084329]
10.
Kavey RW, Allada V, Daniels SR, Hayman LL, McCrindle BW, Newburger JW, Parekh RS, Steinberger J. American Heart Association scientific statement: cardiovascular risk reduction in high-risk pediatric patients. Circulation. 2006;114:2710-38. [PubMed: 17130340]
11.
Sibley C, Stone NJ. Familial hypercholesterolemia: a challenge of diagnosis and therapy. Cleve Clin J Med. 2006;73:57-64. [PubMed: 16444917]
12.
Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion. US Medical Eligibility Criteria for Contraceptive Use, 2016 (US MEC). https://www​.cdc.gov/reproductivehealth​/contraception​/mmwr/mec/summary.html. Updated March 27, 2023. Accessed November 2023.
13.
Atorvastatin product monograph. Pfizer. New York, NY. 2007.
14.
Fluvastatin product monograph. Novartis. East Hanover, NJ. 2006.
15.
Lovastatin product monograph. Merck & Co. Whitehouse Station, NJ. 2007.
16.
Pravastatin product monograph. Bristol-Myers Squibb Company. Princeton, NJ. 2007.
17.
Rosuvastatin product monograph. AstraZeneca. Wilmington, DE. 2011.
18.
Simvastatin product monograph. Merck & Co. Whitehouse Station, NJ. 2006.
19.
Pitavastatin product monograph. Kowa. Montgomery, AL. 2022.
20.
Ferrari F., Martins V.M., Rocha V.Z., Santos R.D. Advances with lipid-lowering drugs for pediatric patients with familial hypercholesterolemia. Expert Opin. Pharmacother. 2021;22:483–495. doi: .10.1080/14656566.2020.1832991 [PubMed: 33016816] [CrossRef]
21.
Haney EM, Huffman LH, Bougatsos C, Freeman M, Steiner RD, Nelson HD. Screening and treatment for lipid disorders in children and adolescents: systematic evidence review for the US preventive services task force. Pediatrics. 2007;120:e189-214. [PubMed: 17606543]
22.
McCrindle BW, Ose L, Marais AD. Efficacy and safety of atorvastatin in children and adolescents with familial hypercholesterolemia or severe hyperlipidemia: a multi-center, randomized, placebo-controlled trial. J Pediatr. 2003;142:74-80. [PubMed: 12915827]
23.
van der Graaf A, Nierman MC, Firth JC, Wolmarans KH, Marais AD, de Groot E. Efficacy and safety of fluvastatin in children and adolescents with heterozygous familial hypercholesterolemia. Acta Paediatr. 2006;95(11):1461-1466. [PubMed: 17062478]
24.
Clauss SB, Holmes KW, Hopkins P, Stein E, Cho M, Tate A, Johnson-Levonas AO, Kwiterovich PO. Efficacy and safety of lovastatin therapy in adolescent girls with heterozygous familial hypercholesterolemia. Pediatrics. 2005;116(3):682-8. [PubMed: 16140708]
25.
Lambert M, Lupien PJ, Gagné C, et al. Treatment of familial hypercholesterolemia in children and adolescents: effect of lovastatin. Pediatrics. 1996;97(5):619-628. [PubMed: 8628597]
26.
Stein EA, Illingworth DR, Kwiterovich Jr PO, et al. Efficacy and safety of lovastatin in adolescent males with heterozygous familial hypercholesterolemia. JAMA. 1999;281(2):137-144. [PubMed: 9917116]
27.
Knipscheer HC, Boelen CCA, Kastelein JJP, et al. Short-term efficacy and safety of pravastatin in 72 children with familial hypercholesterolemia. Pediatr Res. 1996;39(5):867-871. [PubMed: 8726243]
28.
Wiegman A, Hutten BA, de Groot E, et al. Efficacy and safety of statin therapy in children with familial hypercholesterolemia: a randomized controlled trial. JAMA. 2004;292(3):331-337. [PubMed: 15265847]
29.
Rodenburg J, Vissers MN, Wiegman A, et al. Statin treatment in children with familial hypercholesterolemia: The younger, the better. Circulation. 2007;116:664-668. [PubMed: 17664376]
30.
Avis HJ, Hutten BA, Gagné C, et al. Efficacy and safety of rosuvastatin therapy for children with familial hypercholesterolemia. J Am Coll Cardiol. 2010;55(11):1121-1126. [PubMed: 20223367]
31.
de Jongh S, Ose L, Szamosi T, et al. Efficacy and safety of statin therapy in children with familial hypercholesterolemia: a randomized, double-blind, placebo-controlled trial with simvastatin. Circulation. 2002;106(17):2231-2237. [PubMed: 12390953]
32.
de Jongh S, Lilien MR, op’t Roodt J, Stroes ES, Bakker HD, Kastelein JJ. Early statin therapy restores endothelial function in children with familial hypercholesterolemia. J Am Coll Cardiol 2002;40(12):2117-2121. [PubMed: 12505222]
33.
Rosenson RS. Rosuvastatin: a new inhibitor of HMG-CoA reductase for the treatment of dyslipidemia. Expert Rev Cardiovasc Ther. 2003;1(4):495-505. [PubMed: 15030249]
34.
Jones PH, Davidson MH, Stein EA, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* trial). Am J Cardiol. 2003;92(2):152-160. [PubMed: 12860216]
35.
Brown AS, Bakker-Arkema RG, Yellen L, et al. Treating patients with documented atherosclerosis to National Cholesterol Education Program- recommended low-density-lipoprotein cholesterol goals with atorvastatin, fluvastatin, lovastatin, and simvastatin. J Am Coll Cariol. 1998;32(3):665-672. [PubMed: 9741509]
36.
Kuster DM, Avis HJ, de Groot E, et al. Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia. JAMA. 2014;312(10):1055-1057. [PubMed: 25203086]
37.
Braamskamp MJ, Stefanutti C, Langslet G, Drogari E, Wiegman A, Hounslow N, Kastelein JJ; PASCAL Study Group. Efficacy and Safety of Pitavastatin in Children and Adolescents at High Future Cardiovascular Risk. J Pediatr. 2015 Aug;167(2):338-43.e5.doi: . Epub 2015 Jun 6. PMID: .10.1016/j.jpeds.2015.05.006 [PubMed: 26059337] [CrossRef]
38.
Simvastatin: pediatric drug information. Lexicomp Online, Pediatric and Neonatal Lexi-Drugs Online, Hudson, Ohio: Lexi-Comp Inc.; 2015.
39.
Colesevelam product monograph. Daiichi Sankyo, Inc. Parsippany, NJ. 2009.
40.
Stein EA, Marais D, Szamosi T, et al. Colesevelam hydrochloride: efficacy and safety in pediatric subjects with heterozygous familial hypercholesterolemia. J Pediatr. 2010; 156(2):231-236.e1-3. [PubMed: 19879596]
41.
Gold Standard, Inc. Colestipol. Clinical Pharmacology [database online]. Available at: http://www​.clinicalpharmacology.com. Accessed 1/4/2008.
42.
Colestipol. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
43.
McCrindle BW, Helden E, Cullen-Dean G, Conner WT. A randomized crossover trial of combination pharmacologic therapy in children with familial hyperlipidemia. Pediatr Res. 2002:51(6):715-21. [PubMed: 12032266]
44.
Tonstad S, Sivertsen M, Aksnes L, Ose L. Low dose colestipol in adolescents with familial hypercholesterolaemia. Arch Dis Child. 1996;74(2):157-160. [PMC free article: PMC1511517] [PubMed: 8660081]
45.
Taketomo CK, Hodding JH, Kraus DM, eds. Lexi-Comp’s Pediatric Dosing Handbook. 15th edition. Hudson, OH: Lexi-Comp; 2008.
46.
Gold Standard, Inc. Cholestyramine. Clinical Pharmacology [database online]. Available at: http://www​.clinicalpharmacology.com. Accessed 1/4/2008.
47.
Cholesytramine. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
48.
McCrindle BW, O’Neill MB, Cullen-Dean G, Helden E. Acceptability and compliance with two forms of cholestyramine in the treatment of hypercholesterolemia in children: A randomized, crossover trial. J Pediatr. 1997;130(2):266-273. [PubMed: 9042130]
49.
Tonstad S, Knudtzon J, Sivertsen M, Refsum H, Ose L. Efficacy and safety of cholestyramine therapy in peripubertal and prepubertal children with familial hypercholesterolemia. J Pediatr. 1996;129(1):42-49. [PubMed: 8757561]
50.
Fenofibrate. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
51.
Gemfibrozil. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
52.
Wheeler KA, West RJ, Lloyd JK, Barley J. Double blind trial of bezafibrate in familial hypercholesterolaemia. Arch Dis Child. 1985;60(1):34-37. [PMC free article: PMC1777065] [PubMed: 3882058]
53.
Fiorentino R, Chiarelli F. Treatment of Dyslipidaemia in Children. Biomedicines. 2021 Aug 24;9(9):1078. doi: . PMID: ; PMCID: PMC8470054.10.3390/biomedicines9091078 [PMC free article: PMC8470054] [PubMed: 34572264] [CrossRef]
54.
Colletti RB, Roff NK, Neufeld EJ, Baker AL, Newburger JW, McAuliffe TL. Niacin treatment of hypercholesterolemia in children. Pediatrics. 1993;92(1):78-82. [PubMed: 8516088]
55.
Ezetimibe product monograph. Merck & CO., Inc. Whitehouse Station, NJ. 2012.
56.
Yeste D, Chacon P, Clemente M, et al. Ezetimibe as monotherapy in the treatment of hypercholesterolemia in children and adolescents. J Pediatr Endocrinol Metab. 2009;22:487-492. [PubMed: 19694195]
57.
Clauss S, Wai KM, Kavey REW, et al. Ezetimibe treatment of pediatric patients with hypercholesterolemia. J Pediatr. 2009;154:869-872. [PubMed: 19230898]
58.
van der Graaf A, Cuffie-Jackson C, Vissers MN, et al. Efficacy and safety of co-administration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008;52(17):1421-1429. [PubMed: 18940534]
59.
Hammersley D., Signy M. Ezetimibe: An update on its clinical usefulness in specific patient groups. Ther. Adv. Chronic Dis. 2017;8:4–11. doi: .10.1177/2040622316672544 [PMC free article: PMC5298356] [PubMed: 28203346] [CrossRef]
60.
de Ferranti SD, Milliren CE, Denhoff ER, et al. Using high-dose omega-3 fatty acid supplements to lower triglyceride levels in 10- to 19-year-olds. Clin Pediatr (Phila). 2014;53(5):428-438. [PMC free article: PMC4353654] [PubMed: 24707021]
61.
Chahal N, Manlhoit C, Wong H, McCrindle BW. Effectiveness of omega-3 polysaturated fatty acids (fish oil) supplementation for treating hypertriglyceridemia in children and adolescents. Clin Pediatr (Phila). 2014;53(7):645-657. [PubMed: 24647701]
62.
Khorshidi, M., Hazaveh, Z.S., Alimohammadi-kamalabadi, M. et al. Effect of omega-3 supplementation on lipid profile in children and adolescents: a systematic review and meta-analysis of randomized clinical trials. Nutr J 22, 9 (2023). https://doi.org/10.1186/s12937-022-00826-5 [PMC free article: PMC9912483] [PubMed: 36765362] [CrossRef]
63.
McGowan MP, Hosseini Dehkordi SH, Moriarty PM, Duell PB. Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia. J Am Heart Assoc. 2019 Dec 17;8(24):e013225. doi: . Epub 2019 Dec 16. PMID: ; PMCID: PMC6951065.10.1161/JAHA.119.013225 [PMC free article: PMC6951065] [PubMed: 31838973] [CrossRef]
64.
Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia Mary P. McGowan MD mary.p.mcgowan@hitchcock.org, Seyed Hamed Hosseini Dehkordi MD, Patrick M. Moriarty MD, and P. Barton Duell MD [PMC free article: PMC6951065] [PubMed: 31838973]
65.
Defesche JC, Gidding SS, Harada-Shiba M, Hegele RA, Santos RD, Wierzbicki AS. Familial hypercholesterolaemia. Nat Rev Dis Primers. 2017 Dec 7;3:17093. doi: . PMID: .10.1038/nrdp.2017.93 [PubMed: 29219151] [CrossRef]
66.
Alenizi MM, Almushir S, Sulimanaa I. Surgical Management and Outcomes of Homozygous Familial Hypercholesterolemia in Two Cousins: A Rare Case Report. Cureus. 2020 Nov 25;12(11):e11692. doi: . PMID: ; PMCID: PMC7769830.10.7759/cureus.11692 [PMC free article: PMC7769830] [PubMed: 33391926] [CrossRef]
67.
German, C. Homozygous Familial Hypercholesteremia: Diagnosis and Emerging Therapies. 2022 Feb 18. American College of Cardiology.
68.
Avis HJ, Vissers MN, Stein EA, Wijburg FA, Trip MD, Kastelein JJ, Hutten BA. A systematic review and meta-analysis of statin therapy in children with familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2007 Aug;27(8):1803-10. doi: . Epub 2007 Jun 14. PMID: .10.1161/ATVBAHA.107.145151 [PubMed: 17569881] [CrossRef]
69.
Mirzaee S, Cameron JD. Familial hypercholesterolemia and cardiovascular risk stratification. J Clin Lipidol. 2018 Sep-Oct;12(5):1328-1329. doi: . Epub 2018 Jul 10. PMID: .10.1016/j.jacl.2018.07.002 [PubMed: 30061010] [CrossRef]
70.
Luirink IK, Wiegman A, Kusters DM, Hof MH, Groothoff JW, de Groot E, Kastelein JJP, Hutten BA. 20-Year Follow-up of Statins in Children with Familial Hypercholesterolemia. N Engl J Med. 2019 Oct 17;381(16):1547-1556. doi: . PMID: .10.1056/NEJMoa1816454 [PubMed: 31618540] [CrossRef]
71.
Mainieri F, Tagi VM, Chiarelli F. Recent Advances on Familial Hypercholesterolemia in Children and Adolescents. Biomedicines. 2022; 10(5):1043. https://doi.org/10.3390/biomedicines10051043 [PMC free article: PMC9139047] [PubMed: 35625781] [CrossRef]
72.
van der Graaf A, Cuffie-Jackson C, Vissers MN, Trip MD, Gagné C, Shi G, Veltri E, Avis HJ, Kastelein JJ. Efficacy and safety of coadministration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008 Oct 21;52(17):1421-9. doi: . PMID: 10.1016/j.jacc.2008.09.002 [PubMed: 18940534] [CrossRef]
73.
Kusters DM, Caceres M, Coll M, Cuffie C, Gagné C, Jacobson MS, Kwiterovich PO, Lee R, Lowe RS, Massaad R, McCrindle BW, Musliner TA, Triscari J, Kastelein JJ. Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia. J Pediatr. 2015 Jun;166(6):1377-84.e1-3. doi: 10.1016/j.jpeds.2015.02.043. Epub 2015 Apr 1. PMID: 25841542.10.1016/j.jpeds.2015.02.043 [PubMed: 25841542] [CrossRef] [CrossRef]
74.
Hajar R. PCSK 9 Inhibitors: A Short History and a New Era of Lipid-lowering Therapy. Heart Views. 2019 Apr-Jun;20(2):74-75. doi: . PMID: ; PMCID: PMC6686613.10.4103/HEARTVIEWS.HEARTVIEWS_59_19 [PMC free article: PMC6686613] [PubMed: 31462965] [CrossRef]
75.
Zhao Z, Du S, Shen S, Luo P, Ding S, Wang G, Wang L. Comparative efficacy and safety of lipid-lowering agents in patients with hypercholesterolemia: A frequentist network meta-analysis. Medicine (Baltimore). 2019 Feb;98(6):e14400. doi: . PMID: ; PMCID: PMC6380691.10.1097/MD.0000000000014400 [PMC free article: PMC6380691] [PubMed: 30732185] [CrossRef]
76.
Repatha product monograph. Amgen. Thousand Oaks, California. 2017.
77.
Santos RD, Ruzza A, Hovingh GK, Wiegman A, Mach F, Kurtz CE, Hamer A, Bridges I, Bartuli A, Bergeron J, Szamosi T, Santra S, Stefanutti C, Descamps OS, Greber-Platzer S, Luirink I, Kastelein JJP, Gaudet D; HAUSER-RCT Investigators. Evolocumab in Pediatric Heterozygous Familial Hypercholesterolemia. N Engl J Med. 2020 Oct 1;383(14):1317-1327. doi: . Epub 2020 Aug 29. PMID: .10.1056/NEJMoa2019910 [PubMed: 32865373] [CrossRef]
78.
Santos RD, Ruzza A, Hovingh GK, Stefanutti C, Mach F, Descamps OS, Bergeron J, Wang B, Bartuli A, Buonuomo PS, Greber-Platzer S, Luirink I, Bhatia AK, Raal FJ, Kastelein JJP, Wiegman A, Gaudet D. Paediatric patients with heterozygous familial hypercholesterolaemia treated with evolocumab for 80 weeks (HAUSER-OLE): a single-arm, multicentre, open-label extension of HAUSER-RCT. Lancet Diabetes Endocrinol. 2022 Oct;10(10):732-740. doi: . Epub 2022 Sep 5. PMID: .10.1016/S2213-8587(22)00221-2 [PubMed: 36075246] [CrossRef]
79.
Praluent product monograph. Regeneron. Tarrytown, NY. 2021.
80.
Bruckert E, Caprio S, Wiegman A, Charng MJ, Zárate-Morales CA, Baccara-Dinet MT, Manvelian G, Ourliac A, Scemama M, Daniels SR. Efficacy and Safety of Alirocumab in Children and Adolescents With Homozygous Familial Hypercholesterolemia: Phase 3, Multinational Open-Label Study. Arterioscler Thromb Vasc Biol. 2022 Dec;42(12):1447-1457. doi: . Epub 2022 Nov 3. PMID: ; PMCID: PMC9750107.10.1161/ATVBAHA.122.317793 [PMC free article: PMC9750107] [PubMed: 36325897] [CrossRef]
81.
Daniels S, Caprio S, Chaudhari U, Manvelian G, Baccara-Dinet MT, 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 May-Jun;14(3):322-330.e5. doi: . Epub 2020 Mar 28. Erratum in: J Clin Lipidol. 2020 Sep - Oct;14(5):741. PMID: 32331936.10.1016/j.jacl.2020.03.001 [PubMed: 32331936] [CrossRef]
82.
Ben-Omran T, Masana L, Kolovou G, Ariceta G, Nóvoa FJ, Lund AM, Bogsrud MP, Araujo M, Hussein O, Ibarretxe D, Sanchez-Hernández RM, Santos RD. Real-World Outcomes with Lomitapide Use in Paediatric Patients with Homozygous Familial Hypercholesterolaemia. Adv Ther. 2019 Jul;36(7):1786-1811. doi: . Epub 2019 May 17. PMID: ; PMCID: PMC6824397.10.1007/s12325-019-00985-8 [PMC free article: PMC6824397] [PubMed: 31102204] [CrossRef]
83.
Leqvio product monograph. Novartis. East Hanover, NJ. 2021.
84.
Reijman MD, Schweizer A, Peterson ALH, Bruckert E, Stratz C, Defesche JC, Hegele RA, Wiegman A. Rationale and design of two trials assessing the efficacy, safety, and tolerability of inclisiran in adolescents with homozygous and heterozygous familial hypercholesterolaemia. Eur J Prev Cardiol. 2022 Jul 20;29(9):1361-1368. doi: . PMID: .10.1093/eurjpc/zwac025 [PubMed: 35175352] [CrossRef]
85.
Kosmas CE, Bousvarou MD, Sourlas A, Papakonstantinou EJ, Genao EP, Uceta RE, Guzman E. Angiopoietin-like protein 3 (ANGPTL3) inhibitors in the management of refractory hypercholesterolemia. Clin Pharmacol. 2022;14:49-59. doi: 10.2147/CPAA.S345072. Epub 2022 Jul 16. PMID: 358l73366.10.2147/CPAA.S345072 [PMC free article: PMC9300746] [PubMed: 35873366] [CrossRef] [CrossRef]
86.
Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, Ali S, Banerjee P, Chan K-C, Gipe DA, Khilla N, Pordy R, Weinreich DM, Yancopoulos GD, Zhang Y, Guadet D, ELIPSE HoFH Investigators. Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med. 2020 Aug 20;383(8):711-720. doi: . PMID: .10.1056/NEJMoa2004215 [PubMed: 32813947] [CrossRef]
87.
Evkeeza (Evinacumab-dgnb) product monograph. Regeneron. Tarrytown, NY. 2023.
88.
Rayan RA, Sharma S. Lomitapide. [Updated 2022 Jul 18]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-. Available from: https://www​.ncbi.nlm​.nih.gov/books/NBK560849/
89.
Goulooze SC, Cohen AF, Rissmann R. Lomitapide. Br J Clin Pharmacol. 2015 Aug;80(2):179-81. doi: . Epub 2015 Jul 2. PMID: ; PMCID: PMC4541964.10.1111/bcp.12612 [PMC free article: PMC4541964] [PubMed: 25702706] [CrossRef]
90.
Ray KK, Bays HE, Catapano AL, Lalwani ND, Bloedon LT, Sterling LR, Robinson PL, Ballantyne CM; CLEAR Harmony Trial. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med. 2019 Mar 14;380(11):1022-1032. doi: . PMID: .10.1056/NEJMoa1803917 [PubMed: 30865796] [CrossRef]
91.
Ballantyne CM, Bays H, Catapano AL, Goldberg A, Ray KK, Saseen JJ. Role of Bempedoic Acid in Clinical Practice. Cardiovasc Drugs Ther. 2021 Aug;35(4):853-864. doi: . Epub 2021 Apr 5. Erratum in: Cardiovasc Drugs Ther. 2021 Apr 19;: PMID: 33818688; PMCID: PMC8266788.10.1007/s10557-021-07147-5 [PMC free article: PMC8266788] [PubMed: 33818688] [CrossRef]
92.
Bempedoic acid product monograph. Esperion. Ann Arbor, MI. 2022.
93.
Sunil B, Foster C, Wilson DP, Ashraf AP. Novel therapeutic targets and agents for pediatric dyslipidemia. Ther Adv Endocrinol Metab. 2021 Nov 24;12:20420188211058323. doi: . PMID: ; PMCID: PMC8637781.10.1177/20420188211058323 [PMC free article: PMC8637781] [PubMed: 34868544] [CrossRef]
94.
Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, Yang Q, Hughes SG, Geary RS, Arca M, Stroes ESG, Bergeron J, Soran H, Civeira F, Hemphill L, Tsimikas S, Blom DJ, O'Dea L, Bruckert E. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N Engl J Med. 2019 Aug 8;381(6):531-542. doi: . PMID: .10.1056/NEJMoa1715944 [PubMed: 31390500] [CrossRef]
95.
Kolovou G, Kolovou V, Katsiki N. Volanesorsen: A New Era in the Treatment of Severe Hypertriglyceridemia. J Clin Med. 2022 Feb 13;11(4):982. doi: . PMID: ; PMCID: PMC8880470.10.3390/jcm11040982 [PMC free article: PMC8880470] [PubMed: 35207255] [CrossRef]
96.
Esan O, Wierzbicki AS. Volanesorsen in the Treatment of Familial Chylomicronemia Syndrome or Hypertriglyceridaemia: Design, Development and Place in Therapy. Drug Des Devel Ther. 2020 Jul 6;14:2623-2636. doi: . PMID: ; PMCID: PMC7351689.10.2147/DDDT.S224771 [PMC free article: PMC7351689] [PubMed: 32753844] [CrossRef]
97.
Gelrud A, Digenio A, Alexander V, et al. Treatment with Volanesorsen (VLN) Reduced Triglycerides and Pancreatitis in Patients with FCS and sHTG vs Placebo: results of the APPROACH and COMPASS . J Clin Lipidol. 2018;12(2):537. doi: 10.1016/j.jacl.2018.03.032 [CrossRef]
98.
Kohan AB. Apolipoprotein C-III: a potent modulator of hypertriglyceridemia and cardiovascular disease. Curr Opin Endocrinol Diabetes Obes. 2015 Apr;22(2):119-25. doi: . PMID: ; PMCID: PMC4524519.10.1097/MED.0000000000000136 [PMC free article: PMC4524519] [PubMed: 25692924] [CrossRef]
Copyright © 2000-2024, MDText.com, Inc.

This electronic version has been made freely available under a Creative Commons (CC-BY-NC-ND) license. A copy of the license can be viewed at http://creativecommons.org/licenses/by-nc-nd/2.0/.

Bookshelf ID: NBK395575PMID: 27809436

Views

  • PubReader
  • Print View
  • Cite this Page

LINKS TO WWW.ENDOTEXT.ORG

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...