Evidence Summary

Lipid Disorders in Children and Adolescents: Screening

July 18, 2023

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

By Janelle M. Guirguis-Blake, MD; Corinne V. Evans, MPP; Erin L. Coppola, MPH; Nadia Redmond, MSPH; Leslie A. Perdue

The information in this article is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This article is intended as a reference and not as a substitute for clinical judgment.

This article may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied.

This article was published online in JAMA on July 18, 2023 (JAMA. 2023;330(3):261-274. doi:10.1001/jama.2023.886).

Return to Table of Contents

Importance: Lipid screening in childhood and adolescence can lead to early dyslipidemia diagnosis. The long-term benefits of lipid screening and subsequent treatment in this population are uncertain.

Objective: To review benefits and harms of screening and treatment of pediatric dyslipidemia due to familial hypercholesterolemia (FH) and multifactorial dyslipidemia.

Data Sources: MEDLINE and the Cochrane Central Register of Controlled Trials through May 16, 2022; literature surveillance through March 24, 2023.

Study Selection: English-language randomized clinical trials (RCTs) of lipid screening; recent, large US cohort studies reporting diagnostic yield or screen positivity; and RCTs of lipid-lowering interventions.

Data Extraction and Synthesis: Single extraction, verified by a second reviewer. Quantitative synthesis using random-effects meta-analysis.

Main Outcomes and Measures: Health outcomes, diagnostic yield, intermediate outcomes, behavioral outcomes, and harms.

Results: Forty-three studies were included (n = 491,516). No RCTs directly addressed screening effectiveness and harms. Three US studies (n = 395,465) reported prevalence of phenotypically defined FH of 0.2% to 0.4% (1:250 to 1:500). Five studies (n = 142,257) reported multifactorial dyslipidemia prevalence; the prevalence of elevated total cholesterol level (≥200 mg/dL) was 7.1% to 9.4% and of any lipid abnormality was 19.2%. Ten RCTs in children and adolescents with FH (n = 1230) demonstrated that statins were associated with an 81- to 82-mg/dL greater mean reduction in levels of total cholesterol and LDL-C compared with placebo at up to 2 years. Nonstatin-drug trials showed statistically significant lowering of lipid levels in FH populations, but few studies were available for any single drug. Observational studies suggest that statin treatment for FH starting in childhood or adolescence reduces long-term cardiovascular disease risk. Two multifactorial dyslipidemia behavioral counseling trials (n = 934) demonstrated 3- to 6-mg/dL greater reductions in total cholesterol levels compared with the control group, but findings did not persist at longest follow-up. Harms reported in the short-term drug trials were similar in the intervention and control groups.

Conclusions and Relevance: No direct evidence on the benefits or harms of pediatric lipid screening was identified. While multifactorial dyslipidemia is common, no evidence was found that treatment is effective for this condition. In contrast, FH is relatively rare; evidence shows that statins reduce lipid levels in children with FH, and observational studies suggest that such treatment has long-term benefit for this condition.

Return to Table of Contents

Screening can identify abnormal lipid levels with genetic and nongenetic etiologies. Familial hypercholesterolemia (FH) is an autosomal codominant genetic disorder of cholesterol lipid metabolism associated with elevated levels of low-density lipoprotein cholesterol (LDL-C), which causes premature atherosclerosis and early cardiovascular morbidity and mortality.1 Multifactorial dyslipidemia refers to dyslipidemias involving abnormal lipid levels that are not attributable to FH. Multifactorial dyslipidemia may be associated with environmental factors, such as lifestyle behaviors, with or without an inherited component from single-nucleotide variants with smaller additive effects.2-4

Therapies to reduce lipid levels in adulthood are well established,5 and there is also a body of evidence for reducing lipid levels in children and adolescents with FH.6 Evidence is uncertain, however, about when in the life span to begin screening for abnormal lipid levels. In 2016, the US Preventive Services Task Force (USPSTF) found insufficient evidence to assess the balance of benefits and harms of routine screening for any lipid disorders, including FH, in children and adolescents.7 This systematic review updates the body of evidence on screening for dyslipidemia in children and adolescents and was used to update the prior USPSTF recommendation.

Return to Table of Contents

 

Scope of Review

Figure 1 shows the analytic framework, key questions (KQs) that guided the systematic review, and the contextual questions intended to provide additional background information. In addition to systematic review of the KQs, this review looked for evidence about the association between lipid-related outcomes in childhood and adolescence and adult health outcomes and the optimal timing of statin treatment initiation in FH. Additional methodological details, analyses, results for other lipid outcomes in treatment trials other than total cholesterol and LDL-C (high-density lipoprotein cholesterol [HDL-C], triglycerides, non–HDL-C), as well as treatment trials of fibrates and supplements, are available in the full evidence report.9

Data Sources and Searches

MEDLINE and the Cochrane Central Register of Controlled Trials were searched for relevant English-language articles published after the search dates for the prior reviews of lipid disorders in children and adolescents previously conducted for the USPSTF (January 1, 2016, to May 16, 2022) (eMethods in the JAMA Supplement).6,10 All studies in the prior reviews were evaluated,6,10 as well as reference lists of related systematic reviews. ClinicalTrials.gov was searched for relevant ongoing trials. Active surveillance was conducted through March 24, 2023, via article alerts and targeted journal searches to identify major studies that might affect the conclusions of the review or understanding of the evidence. One new study was identified;11 however, it did not substantively change the review’s interpretation of findings or conclusions and is not addressed further.

Study Selection

Two independent reviewers screened titles, abstracts, and full-text articles against a priori eligibility criteria (eTable 1 in the JAMA Supplement). Eligible studies included children and adolescents 20 years and younger. Populations with homozygous FH, those already being followed up for dyslipidemia, or those with diagnoses associated with secondary dyslipidemia were excluded, as were populations with an established family history of FH. For KQ1, randomized clinical trials (RCTs) and controlled clinical trials comparing universal or selective serum lipid screening with no screening were used to evaluate the effectiveness of screening with nonfasting or fasting serum lipid tests typically ordered in primary care. Cascade screening was excluded because this represents a case-finding approach as opposed to population screening.

For KQ2, large, recent US cohort studies were used for assessing diagnostic yield of screening. Studies reporting positive predictive value of a first elevated screening lipid result for a second confirmatory test were sought; however, no included studies used a confirmatory test, and thus studies reporting screen positivity based on a single lipid test were accepted. Author-defined thresholds for abnormal lipid levels were used. For KQ4, RCTs of treatments for dyslipidemia including drugs, behavioral counseling, and supplements were used to assess benefits. Outcomes for treatment benefits included health outcomes (myocardial infarction, ischemic stroke, cardiovascular disease [CVD] mortality, or all-cause mortality); the intermediate outcomes of serum lipid concentrations (total cholesterol, LDL-C, HDL-C, triglycerides, or non–HDL-C), atherosclerosis markers (carotid intima-media thickness, calcium score, or pathological findings), and body mass index (BMI); and behavioral outcomes (physical activity, sedentary behavior, or dietary intake). For KQ3 (screening harms) and KQ5 (treatment harms), RCTs, controlled clinical trials, and nonrandomized studies of interventions were accepted.

Data Extraction and Quality Assessment

Two reviewers independently applied USPSTF design-specific criteria to critically appraise each study (eTable 2 in the JAMA Supplement).8 Each study was assigned a rating of “good,” “fair,” or “poor.” Discordant ratings were resolved by consensus. Poor-quality studies were excluded. One reviewer extracted data into standardized evidence tables and a second reviewer checked the tables for accuracy.

Data Synthesis and Analysis

All results were synthesized separately for FH and multifactorial dyslipidemia. Evidence related to the prevalence of FH and multifactorial dyslipidemia (KQ2) were synthesized narratively and summarized in tables. For treatment studies (KQ4 and KQ5), results were synthesized by intervention. Only statins had a sufficient number of contributing studies for quantitative pooling; other interventions were summarized narratively and in tables. The random-effects restricted maximum likelihood method with the Knapp-Hartung correction was applied in meta-analyses for statins because of either high statistical heterogeneity (commonly I2 > 50%) or small number of trials to be pooled.12,13 For pooling statin studies with multiple randomized groups with differing statin intensity, we selected the group receiving the highest-intensity dose. Statin intensity categorizations were based on 2018 guidelines for the management of cholesterol levels in adults,14 because intensity categorizations are not established for pediatric populations.

Statistical heterogeneity among pooled studies was evaluated using standard χ2 tests and the magnitude of heterogeneity was estimated using the I2 statistic. Due to the limited number of trials (<10) for pooled analyses of statins, assessment of small-study effects and publication bias were not performed.15,16

All quantitative analyses were performed using Stata version 16.1 (StataCorp). All significance testing was 2-sided, and results were considered statistically significant at P < .05.

The aggregate strength of evidence was assessed for each KQ using the approach described in the Methods Guide for Effectiveness and Comparative Effectiveness Reviews,17 based on the number, quality, and size of studies and the consistency and precision of results.

Return to Table of Contents

 

Two reviewers evaluated 7058 abstracts and 272 full-text articles for KQ eligibility (Figure 2). Overall, 43 studies (65 publications) met inclusion criteria for this systematic review.18-83 Thirteen of these studies evaluated the benefits of supplement interventions and 10 reported on the harms of supplement interventions. These studies were small, of short duration, and had few contributing studies for any one supplement. Evidence was generally insufficient and these interventions are not addressed further; additional details are available in the full report.9

Screening Benefits and Harms

KQ1. Does screening for familial hypercholesterolemia (FH) or multifactorial dyslipidemia in asymptomatic children and adolescents delay or reduce the incidence of health outcomes (eg, CVD events or mortality) or improve intermediate outcomes (eg, serum lipid levels and atherosclerotic markers) in children, adolescents, or adults?

No studies met inclusion criteria for this KQ.

Diagnostic Yield

KQ2. What is the diagnostic yield of serum lipid screening for FH or multifactorial dyslipidemia in children and adolescents?

No studies performed a confirmatory lipid or genetic test; thus, the evidence on lipid screening for identifying FH or multifactorial dyslipidemia is limited to the prevalence of single positive screening test results rather than the diagnostic yield as defined by confirmatory testing.

Familial Hypercholesterolemia

A summary of the evidence related to familial hypercholesterolemiais provided in Table 1.Three fair-quality US studies (n = 395,465) including the National Health and Nutrition Examination Survey (NHANES),82 a Texas blood donor program,81 and the West Virginia Coronary Artery Risk Detection in Appalachian Communities (CARDIAC) study,72 reported the prevalence of FH (eTables 3 and 4 in the JAMA Supplement). Using diagnostic criteria exclusively based on lipid levels (LDL-C 190 mg/dL or total cholesterol 270 mg/dL [to convert LDL-C and total cholesterol values to mmol/L, multiply by 0.0259]), prevalence ranged from 0.2% to 0.4% (1:250 to 1:500). One study showed that targeted screening in persons with a family history of hypercholesterolemia would miss many cases of children with LDL-C levels of 160 mg/dL or greater (prevalence in those with family history, 1.2%; prevalence in those without family history, 1.7%).70

Multifactorial Dyslipidemia

A summary of the evidence related to multifactorial dyslipidemia is provided in Table 2. Five fair-quality studies (n = 142,257), including NHANES,23 HEALTHY,25 the Study of Latino Youth,22 Poudre Valley Health System Healthy Hearts Club,24 and CARDIAC,72 reported the prevalence of multifactorial dyslipidemia (eTables 5 and 6 in the JAMA Supplement). Lipid abnormalities were common, being generally more common for the parameters of HDL-C and triglycerides.

Prevalence ranged from 7.1% to 9.4% for elevated total cholesterol level (≥200 mg/dL), 6.4% to 7.4% for elevated LDL-C (≥130 mg/dL), 12.1% to 22.2% for low HDL-C (<40 mg/dL), 8.0% to 17.3% for elevated triglycerides (using various thresholds), and 6.4% to 13.0% for elevated non–HDL-C (≥145 mg/dL) (to convert HDL-C and non–HDL-C values to mmol/L, multiply by 0.0259). Prevalence of any lipid abnormality in 6- to 19-year-olds was 19.2% based on NHANES data (2013-2016, n = 4381). Prevalence of abnormal lipid levels by population characteristics are shown in eFigures 1-3 in the JAMA Supplement.

Screening Harms

KQ3. What are the harms of screening for FH or multifactorial dyslipidemia in children and adolescents?

No studies met inclusion criteria for this KQ.

Treatment Benefit

KQ4. Does treatment of FH or multifactorial dyslipidemia with behavioral interventions, lipid-lowering medications, or both in children and adolescents delay or reduce the incidence of health outcomes (eg, CVD events or mortality) or improve intermediate outcomes (eg, serum lipid levels and atherosclerotic markers) in children, adults, or both?

Familial Hypercholesterolemia

A summary of the evidence related to familial hypercholesterolemia is provided in Table 1. No treatment trials reported long-term health outcomes. Twenty-two fair- to good-quality trials (n = 2257) examined the effectiveness of various lipid-lowering treatments for FH including pharmacotherapy, behavioral counseling, and dietary supplements. Trials were generally small and short-term. Overall, this body of evidence demonstrated that pharmacotherapy appears beneficial for total cholesterol and LDL-C outcomes, with the largest evidence available for statins; behavioral counseling was not effective.

Ten fair- to good-quality RCTs (n = 1230) of statins with follow-up for up to 2 years comprised the largest body of evidence addressing FH treatment, but only 1 trial is new in this update.56-65 Pooled analyses demonstrated that statins were associated with an 81- to 82-mg/dL greater mean reduction in total cholesterol and LDL-C levels compared with placebo at up to 2 years’ follow-up (total cholesterol: 7 studies [n = 706]; mean difference [MD] in change, −82.1mg/dL [95% CI, −101.1 to −63.2]; I2 = 83.0%; LDL-C: 8 studies [n = 742]; MD in change, −81.3 mg/dL [95% CI, −97.6 to −65.0]; I2 = 81.6%) (Figure 3; eFigures 4 and 5 in the JAMA Supplement). One good-quality and 2 fair-quality bile acid sequestrant trials (n = 332) demonstrated that treatment was associated with a significantly greater reduction in total cholesterol level compared with placebo.78-80 Total cholesterol reductions ranged from −22.1 mg/dL to −40.6 mg/dL and LDL-C reductions from −13.2 mg/dL to −45.9 mg/dL at 8 weeks (eFigures 6 and 7 in the JAMA Supplement). One good-quality ezetimibe trial (n = 138) showed a statistically significant reduction in total cholesterol (MD in change, −64.0 mg/dL [95% CI, −81.1 to −46.9]) and LDL-C (MD in change, −63.0 mg/dL [95% CI, −79.5 to −46.5]) (eFigures 6 and 7 in the JAMA Supplement).75

One new good-quality trial of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors (n = 158) demonstrated that evolocumab was associated with a statistically significant 68.6-mg/dL reduction in LDL-C level (95% CI, −83.1 to −54.1) (eFigure 7 in the JAMA Supplement).69 One trial of combination drug therapy of a statin plus ezetimibe compared with a statin alone (n = 248) showed that the 2-drug intervention was associated with a greater reduction in total cholesterol level (MD in change, −40.1 mg/dL [95% CI, −51.1 to −29.2]) and LDL-C (MD in change, −37.5 mg/dL [95% CI, −48.0 to −27.0]) compared with the single-drug intervention control group at 33 weeks (eFigures 6 and 7 in the JAMA Supplement).76

One very small, fair-quality behavioral counseling trial in an FH population (n = 21) tested a low-intensity diet and exercise counseling intervention of a single in-person 60-minute individual session with a dietitian and 4 follow-up sessions via email or telephone over a 12-week period.73 The trial reported no statistically significant improvement in lipid levels (MD in LDL-C, −13.9 mg/dL [95% CI, −32.0 mg/dL to 4.2 mg/dL]) (eFigure 7 in the JAMA Supplement), overlapping confidence intervals for physical activity outcomes, and mixed results for dietary outcomes (eTable 7 in the JAMA Supplement).73

Multifactorial Dyslipidemia

A summary of the evidence related to multifactorial dyslipidemia is provided in Table 2. There were no included trials of drug interventions in child and adolescent populations with multifactorial dyslipidemia. There were 2 fair- to good-quality behavioral counseling trials (n = 934); both focused on dietary changes.18,83 Overall, this body of evidence showed that behavioral counseling interventions were associated with nonsustained, short-term reductions in levels of total cholesterol and LDL-C, with some improvements in dietary intake. The first trial, in which intervention continued throughout a mean follow-up of 7.4 years, evaluated an intensive intervention of 19 individual sessions with a case manager and 31 group sessions led by dietitians, behaviorists, and health educators.83 The second trial was a 10-week, low-intensity intervention RCT with 1 year of follow-up and included 2 intervention groups.The first group received a home-based, social cognitive theory–based intervention with 10 audiotape story books with accompanying picture books, child activity books, and a parent manual to be reviewed over 10 weeks; the second intervention group received a child-parent in-person 45- to 60- minute counseling session with a pediatric registered dietitian and homeprint materials with access to the dietitian by phone with any questions after the session.18 These 2 trials demonstrated statistically significant 3- to 6-mg/dL greater reductions in levels of total cholesterol and LDL-C and improvements in dietary intake outcomes in the intervention group compared with the control group during the first follow-up for each trial, but findings did not persist at the second follow-up (eTables 8 and 9 in the JAMA Supplement). No treatment trials reported long-term health outcomes.

Treatment Harms

KQ5. What are the harms of treatment of FH or multifactorial dyslipidemia in children and adolescents?

Familial Hypercholesterolemia

A summary of the evidence related to familial hypercholesterolemia is provided in Table 1. Overall, harms reported in pharmacotherapy trials were similar in the intervention and control groups; however, most studies were relatively short-term and small with few events, leading to imprecise estimates. Further, the clinical importance of transient elevations in laboratory values was unknown.

In the 9 statin studies reporting transaminitis of 3 times or more the upper limit of normal, this outcome occurred in 0% to 4.5% in intervention groups and 0% to 1.9% in control groups (eFigure 8 in the JAMA Supplement).47,56-58,60,63-65,75 The largest trial (n = 214) with 2-year follow-up reported no cases in the statin group and only 2 cases of aspartate aminotransferase levels more than 3 times the upper limit of normal in the control group.56 In the 10-year observational follow-up of this trial, transaminitis at this threshold was similarly rare (alanine aminotransferase: 1 case of >3 times elevation in the statin group; aspartate aminotransferase: 1 case of >3 times elevation each in the statin and control group).55 Abnormal creatine kinase level of 10 times or greater the upper limit of normal was reported as zero in 2 trials57,63 and up to 4.5%in the statin groups and up to 1.7% in the control groups (eFigure9 in the JAMA Supplement).60,65 One trial’s 10-year observational follow-up reported no instances of elevated creatine kinase level in participants taking statins and in 2 siblings without FH not taking statins.55

Two observational studies evaluated statin harms in populations with dyslipidemia, without specification of the type of dyslipidemia. One fair-quality observational study evaluated the association of statins and new-onset diabetes (n = 9393), showing no difference in new diabetes diagnoses over up to 9 years’ follow-up in individuals taking statins compared with controls. One fair-quality observational study (n = 943) reported alanine aminotransferase levels more than 3 times the upper limit of normal, with a frequency of 4.4% in the statin group and 1.5% in the control group over 3.5 years of observation.

In the statin trials, no significant differences between Tanner stages56-58,67 or other hormonal adverse events like abnormal levels of adrenocorticotropic hormone,59 cortisol,59 dehydroepiandrosterone sulfate,55 follicle-stimulating hormone,55 or thyrotropin59 were reported in the RCTs or in longer observational follow-up (eTables 10 and 11 in the JAMA Supplement). Harms in the 3 bile acid sequestrant trials (n = 332) were similar in the intervention and control groups; however, the trials were generally small with few events, and significance testing was not reported.78-80 Harms in the ezetimibe trial (n = 138),75 PCSK9 inhibitor trial (n = 158),69 and combination statin plus ezetimibe vs statin trial (n = 248)76 showed similar rates of total adverse events in the intervention and control groups. The diet and physical activity counseling intervention did not mention harms.73

Multifactorial Dyslipidemia

A summary of the evidence related to multifactorial dyslipidemia is provided in Table 2. Overall, behavioral counseling interventions do not appear to be associated with important harms (eTables 12 and 13 in the JAMA Supplement).18,83 The 2 behavioral counseling trials in children with multifactorial dyslipidemia (n = 934) reported no adverse effects in terms of growth and Tanner staging;83 nutrient adequacy in ferritin, retinol, zinc, or albumin;83 and psychosocial outcomes18,83 in the dietary intervention group compared with the control group.

Contextual Questions

Contextual question details are reported in the eDiscussion and eFigure 10 in the JAMA Supplement. Contextual question 1 focuses on the indirect evidence linking childhood lipid levels to adult health outcomes. Robust evidence suggests that abnormal lipid levels in childhood and young adulthood are highly associated with adult CVD events. For example, the 35-year follow-up from the i3C Consortium (n = 38,589) reported hazard ratios for a fatal CVD event in adulthood of 1.30 (95% CI, 1.14-1.47) per unit increase in the z score for total cholesterol in childhood.84

Meta-analysis of 6 US-based cohort studies demonstrated the independent association between exposure to high lipid levels in young adulthood (age 18-39 years) and later CVD events, taking into account exposure to elevated lipid levels in later adulthood (≥40 years). In this study, exposure to LDL-C levels 100 mg/dL or higher in young adulthood was associated with an adjusted hazard ratio of 1.64 (95% CI, 1.27-2.11) for coronary heart disease, compared with LDL-C levels lower than 100 mg/dL in young adulthood.85 Similarly, a mendelian randomization study of 9 single-nucleotide variants in an LDL-C gene suggested that lower LDL-C levels throughout the life span are associated with substantially lower incidence of coronary heart disease in adulthood.86

Studies of lipid values over the life course show that it is common but not inevitable for high lipid levels in childhood to persist into adulthood.84,87-90 There is robust evidence supporting the association between adult lipid levels and adult health outcomes from observational evidence and statin treatment trials.5,86,91-93

Contextual question 2 addresses the optimal timing of statin initiation in FH. In summary, there is no direct comparative effectiveness evidence to determine the exact age to start statin treatment for heterozygous FH, but earlier initiation is supported by indirect observational evidence. Markers of atherosclerosis are evident as early as age 8 years in children with FH compared with unaffected siblings or healthy controls; these subclinical atherosclerotic markers include higher carotid intima-media thickness, endothelial dysfunction, and arterial stiffness.94-97

Observational evidence further supports early treatment improvements in intermediate and health outcomes. At 10- to 20-year follow-up, carotid intima-media thickness progression rates converged in children with pathogenic variant–confirmed FH treated with statins and their unaffected siblings.42,55 One compelling observation from a 20- year follow-up study of 214 treated patients from the statin trial by Wiegman et al42,56 was that initiation of statins in adolescence was associated with an improved cumulative CVD-free survival at age 39 years. Participants with pathogenic variant–confirmed FH who started statins in youth (mean statin initiation age, 14.0 [SD, 3.1] years) had higher rates of CVD-free survival compared with their parents, for whom statins were not available until adulthood (99% v 74% CVD-free survival; hazard ratio, 11.8 [95% CI, 3.0-107.0] adjusted for sex, smoking status).42

Return to Table of Contents

Summary

This review, performed since the previous systematic reviews for the USPSTF,6,10 included the following new data: 7 studies of prevalence, 16 treatment trials, and 2 nonrandomized studies of interventions. Despite the inclusion of new evidence, the conclusions are similar to those of the prior reviews (Table 1 and Table 2). There is no direct evidence from population-based screening trials addressing the benefits and harms of pediatric lipid screening for intermediate, behavioral, or health outcomes.

Dyslipidemia is common in contemporary pediatric populations in the US, with a prevalence of 19.2% for any lipid abnormality and heterozygous FH prevalence (as defined by phenotype) estimated at 0.2% to 0.4% (1:250 to 1:500). The body of evidence on treatment benefit is strongest for statins in children and adolescents with FH, with pooled analysis showing beneficial effects on total cholesterol and LDL-C levels; these results were based on mostly small, short-term studies, with the longest trial lasting 2 years.

Most of the evidence for statin harms is from small, short-term studies. Limited longer-term evidence shows few withdrawals due to adverse events, slightly higher rates of liver and musculoskeletal laboratory elevations, and no significant differences in Tanner staging or hormonal adverse events between statin and placebo groups. These safety and efficacy findings are consistent with those from another recent systematic review98 and from 1- to 20-year observational follow-up studies of children and adolescents taking statins.99-107 Additional observational long-term reporting of health outcomes and statin safety (including diabetes, transaminitis) in those with FH for whom statins were initiated at various time points in childhood and adolescence would provide additional data for long-term benefits and harms. The nonstatin-drug trials show reductions in 1 or more lipid parameters and are generally associated with low withdrawals due to adverse events. There is scant evidence on behavioral counseling interventions in FH.

The body of evidence on treatment of multifactorial dyslipidemia is sparse, being limited to 2 behavioral counseling interventions showing modest short-term benefits in lipid levels that did not persist with longer follow-up. These results are consistent with short-term quality improvement projects in specialty settings that have shown that clinician advice targeting lifestyle modifications has shown promising results, especially for reductions in LDL-C levels.107

Single Screening Test Identifies Distinct Conditions

The natural history of FH and multifactorial dyslipidemia are quite different. While a single screening lipid panel identifies both conditions, FH is far less common and more prognostically severe. Further, the strength of the bodies of treatment literature are quite distinct for different dyslipidemias. Some observers have argued that the rationale for universal lipid screening in childhood is solely or primarily to identify those with FH because identifying FH has more potential benefit in reducing premature CVD events and death.42 While the treatment evidence for multifactorial dyslipidemia is scant, some observers have suggested that early identification of any dyslipidemia could lead to earlier nonpharmacologic interventions or pharmacologic management for significantly elevated LDL-C levels and potentially improve health outcomes.108 However, there is no direct evidence to suggest an effective lipid-lowering intervention for the nearly 20% of children and adolescents in whom screening would identify abnormal lipid levels.

Lipid screening may lead to additional benefits beyond identifying children with dyslipidemia, including discovery and treatment of secondary comorbid conditions (eg, diabetes, hypothyroidism) and identification and treatment of this condition in other family members via cascade testing. However, there is limited direct evidence about additional benefits of screening beyond the child.70,109-112

Other observers have surmised that screening and identification of dyslipidemia in children and adolescents with elevated BMI may make weight management interventions more effective; however, limited existing evidence does not support this hypothesis.113,114 Behavioral counseling intervention trials in children with multifactorial dyslipidemia with and without elevated BMI are needed in addition to trials of behavioral counseling as an adjunct to pharmacotherapy in children with FH.

Limitations of the Literature and Future Research Needs

Familial hypercholesterolemia diagnostic criteria in yield studies were limited to lipid levels alone; this is inconsistent with treatment trial criteria, which also included genetic, family, or clinical history components in addition to lipid levels. Consistency in the use of FH criteria between screening studies and treatment studies would facilitate more direct interpretation of evidence to clinical practice.

Outcomes for treatment trials were limited to intermediate outcomes with insufficient follow-up periods to assess long-term health effects or harms. Obtaining such health outcome data may be quite difficult. To report on health outcomes for CVD events occurring in adulthood, these large cohort studies would need to be conducted over a period of decades while maintaining adequate follow-up.

Limitations

The accuracy of FH diagnostic criteria was not systematically reviewed; instead, this review accepted studies of FH as defined by study authors. Familial hypercholesterolemia is genetically heterogeneous, and the relationship between the FH genotype and FH phenotype as expressed by elevated LDL-C level is not straightforward.115-118 Further, diagnosis of FH by genetic testing is rare in the US, further limiting the direct applicability of trials that use genetically confirmed FH to real-life practice, where FH is generally phenotypically defined. Furthermore, this review did not include other less common monogenic or polygenic dyslipidemias, so estimates of the positivity rates for screening may be an underestimate of familial dyslipidemias.

Return to Table of Contents

No direct evidence on the benefits or harms of pediatric lipid screening was identified. While multifactorial dyslipidemia is common, no evidence was found that treatment is effective for this condition. In contrast, FH is relatively rare; evidence shows that statins reduce lipid levels in children with FH, and observational studies suggest that such treatment has long-term benefit for this condition.

Return to Table of Contents

Source: This article was published online in JAMA on July 18, 2023 (JAMA. 2023;330(3):261-274. doi:10.1001/jama.2023.886).

Conflict of Interest Disclosures: None reported.

Funding/Support: This research was funded under contract 75Q80120D00004, Task Order 75Q80120F32001, from the Agency for Healthcare Research and Quality (AHRQ), US Department of Health and Human Services.

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ had no role in study selection, quality assessment, or synthesis. AHRQ staff provided project oversight; reviewed the report to ensure that the analysis met methodological standards; and distributed the draft for peer review. Otherwise, AHRQ had no role in the conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript findings. The opinions expressed in this document are those of the authors and do not reflect the official position of AHRQ or the US Department of Health and Human Services.

Additional Information: A draft version of this evidence report underwent external peer review from 3 content experts (Stephen R. Daniels, MD, PhD, University of Colorado School of Medicine and Children’s Hospital Colorado; Juanita Redfield, MD, Kaiser Permanente Colorado [retired]; and Justin P. V. Zachariah, MD, MPH, Baylor College of Medicine and Texas Children’s Hospital) and 5 individuals at USPSTF Federal Partner agencies (Office of Genomics and Precision Public Health and National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention; National Heart, Lung, and Blood Institute, National Institutes of Health). Comments were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Return to Table of Contents

1. de Ferranti SD, Steinberger J, Ameduri R, et al. Cardiovascular risk reduction in high-risk pediatric patients: a scientific statement from the American Heart Association. Circulation. 2019;139(13):e603-e634. doi:10.1161/CIR.0000000000000618
2. Parks EJ, Hellerstein MK. Carbohydrate-induced hypertriacylglycerolemia: historical perspective and review of biological mechanisms. Am J Clin Nutr. 2000;71(2):412-433. doi:10.1093/ajcn/71.2.412
3. Martinez-Gomez D, Rey-López JP, Chillón P, et al; AVENA Study Group. Excessive TV viewing and cardiovascular disease risk factors in adolescents: the AVENA cross-sectional study. BMC Public Health. 2010;10:274. doi:10.1186/1471-2458-10-274
4. Jarauta E, Bea-Sanz AM, Marco-Benedi V, Lamiquiz-Moneo I. Genetics of hypercholesterolemia: comparison between familial hypercholesterolemia and hypercholesterolemia nonrelated to LDL receptor. Front Genet. 2020;11:554931. doi:10.3389/fgene.2020.554931
5. Mihaylova B, Emberson J, Blackwell L, et al; Cholesterol Treatment Trialists’ (CTT) Collaborators. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet. 2012;380(9841):581-590. doi:10.1016/S0140-6736(12)60367-5
6. Lozano P, Henrikson NB, Dunn J, et al. Lipid Screening in Childhood and Adolescence for Detection of Familial Hypercholesterolemia: A Systematic Evidence Review for the US Preventive Services Task Force. Evidence Synthesis No. 141. Agency for Healthcare Research and Quality; 2016. AHRQ publication 14-05204-EF-2.
7. US Preventive Services Task Force. Screening for lipid disorders in children and adolescents: US Preventive Services Task Force recommendation statement. JAMA. 2016;316(6):625-633. doi:10.1001/jama.2016.9852
8. US Preventive Services Task Force Procedure Manual. US Preventive Services Task Force. Published May 2021. Accessed June 5, 2023. https://uspreventiveservicestaskforce.org/uspstf/about-uspstf/methods-and-processes/procedure-manual
9. Guirguis-Blake JM, Evans CV, Coppola EL, Redmond N, Perdue LA. Screening for Lipid Disorders in Children and Adolescents: An Evidence Update for the US Preventive Services Task Force. Evidence Synthesis No. 229. Agency for Healthcare Research and Quality; 2023. AHRQ publication 22-05301-EF-1.
10. Lozano P, Henrikson NB, Morrison CC, et al. Lipid Screening in Childhood for Detection of Multifactorial Dyslipidemia: A Systematic Evidence Review for the US Preventive Services Task Force. Evidence Synthesis No. 140. Agency for Healthcare Research and Quality; 2016. AHRQ publication 14- 05204-EF-1.
11. Liu J, Ma J, Orekoya O, Vangeepuram N, Liu J. Trends in metabolic syndrome among US youth, from 1999 to 2018. JAMA Pediatr. 2022;176(10):1043-1045. doi:10.1001/jamapediatrics.2022.1850
12. Knapp G, Hartung J. Improved tests for a random effects meta-regression with a single covariate. Stat Med. 2003;22(17):2693-2710. doi:10.1002/sim.1482
13. Veroniki AA, Jackson D, Bender R, et al. Methods to calculate uncertainty in the estimated overall effect size from a random-effects meta-analysis. Res Synth Methods. 2019;10(1):23-43. doi:10.1002/jrsm.1319
14. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1082-e1143. doi:10.1161/CIR.0000000000000625
15. Terrin N, Schmid CH, Lau J. In an empirical evaluation of the funnel plot, researchers could not visually identify publication bias. J Clin Epidemiol. 2005;58(9):894-901. doi:10.1016/j.jclinepi.2005.01.006
16. Sterne JA, Sutton AJ, Ioannidis JP, et al. Recommendations for examining and interpreting funnel plot asymmetry inmeta-analyses of randomised controlled trials. BMJ. 2011;343:d4002. doi:10.1136/bmj.d4002
17. Berkman ND, Lohr KN, Ansari M, et al. Grading the strength of a body of evidence when assessing health care interventions for the Effective Health Care Program of the Agency for Healthcare Research and Quality: an update. In: Methods Guide for Effectiveness and Comparative Effectiveness Reviews. AHRQ publication 10(14)-EHC063-EF. Agency for Healthcare Research and Quality. Published 2013. Accessed June 21, 2023. https://effectivehealthcare.ahrq.gov/products/methods-guidance- grading-evidence/methods
18. Shannon BM, Tershakovec AM, Martel JK, et al. Reduction of elevated LDL-cholesterol levels of 4- to 10-year-old children through home-based dietary education. Pediatrics. 1994;94(6, pt 1):923- 927. doi:10.1542/peds.94.6.923
19. Nguyen D, Kit B, Carroll M. Abnormal cholesterol among children and adolescents in the United States, 2011-2014. NCHS Data Brief. 2015; (228):1-8.
20. Wong H, Chahal N, Manlhiot C, Niedra E, McCrindle BW. Flaxseed in pediatric hyperlipidemia: a placebo-controlled, blinded, randomized clinical trial of dietary flaxseed supplementation for children and adolescents with hypercholesterolemia. JAMA Pediatr. 2013;167(8):708-713. doi:10.1001/jamapediatrics.2013.1442
21. Gidding SS, Prospero C, Hossain J, et al. A double-blind randomized trial of fish oil to lower triglycerides and improve cardiometabolic risk in adolescents. J Pediatr. 2014;165(3):497-503.e2. doi:10.1016/j.jpeds.2014.05.039
22. Reina SA, Llabre MM, Vidot DC, et al. Metabolic syndrome in Hispanic youth: results from the Hispanic Community Children’s Health Study/Study of Latino Youth. Metab Syndr Relat Disord. 2017;15(8):400-406. doi:10.1089/met.2017.0054
23. Perak AM, Ning H, Kit BK, et al. Trends in levels of lipids and apolipoprotein B in US youths aged 6 to 19 years, 1999-2016. JAMA. 2019;321(19):1895-1905. doi:10.1001/jama.2019.4984
24. Nelson TL, Puccetti N, Luckasen GJ. Healthy hearts: a cross-sectional study of clinical cardiovascular disease risk factors in Northern Colorado school children (1992-2013). BMC Obes. 2015;2:48. doi:10.1186/s40608-015-0078-9
25. Bauer KW, Marcus MD, El ghormli L, Ogden CL, Foster GD. Cardio-metabolic risk screening among adolescents: understanding the utility of body mass index, waist circumference and waist to height ratio. Pediatr Obes. 2015;10(5):329-337. doi:10.1111/ijpo.267
26. Hirst K, Baranowski T, DeBar L, et al; HEALTHY Study Group. HEALTHY study rationale, design and methods: moderating risk of type 2 diabetes in multi-ethnic middle school students. Int J Obes (Lond). 2009;33(suppl 4):S4-S20. doi:10.1038/ijo.2009.112
27. Kwiterovich PO Jr, Barton BA, McMahon RP, et al. Effects of diet and sexual maturation on low-density lipoprotein cholesterol during puberty: the Dietary Intervention Study in Children (DISC). Circulation. 1997;96(8):2526-2533. doi:10.1161/01.CIR.96.8.2526
28. Dorgan JF, Liu L, Barton BA, et al. Adolescent diet and metabolic syndrome in young women: results of the Dietary Intervention Study in Children (DISC) follow-up study. J Clin Endocrinol Metab. 2011;96(12):E1999-E2008. doi:10.1210/jc.2010-2726
29. Obarzanek E, Kimm SY, Barton BA, et al; DISC Collaborative Research Group. Long-term safety and efficacy of a cholesterol-lowering diet in children with elevated low-density lipoprotein cholesterol: seven-year results of the Dietary Intervention Study in Children (DISC). Pediatrics. 2001;107(2):256-264. doi:10.1542/peds.107.2.256
30. Obarzanek E, Hunsberger SA, Van Horn L, et al. Safety of a fat-reduced diet: the Dietary Intervention Study in Children (DISC). Pediatrics. 1997;100(1):51-59. doi:10.1542/peds.100.1.51
31. Lavigne JV, Brown KM, Gidding S, et al. A cholesterol-lowering diet does not produce adverse psychological effects in children: three-year results from the dietary intervention study in children. Health Psychol. 1999;18(6):604-613. doi:10.1037/0278-6133.18.6.604
32. DISC Collaborative Research Group. Dietary intervention study in children (DISC) with elevated low-density-lipoprotein cholesterol: design and baseline characteristics. Ann Epidemiol. 1993;3(4): 393-402. doi:10.1016/1047-2797(93)90067-E
33. Engler MM, Engler MB, Arterburn LM, et al. Docosahexaenoic acid supplementation alters plasma phospholipid fatty acid composition in hyperlipidemic children: results from the Endothelial Assessment of Risk from Lipids in Youth (EARLY) study. Nutr Res. 2004;24(9):721-729. doi:10.1016/j.nutres.2004.06.004
34. Amundsen AL, Ntanios F, Put Nv, Ose L. Long-term compliance and changes in plasma lipids, plant sterols and carotenoids in children and parents with FH consuming plant sterol ester-enriched spread. Eur J Clin Nutr. 2004;58(12):1612-1620. doi:10.1038/sj.ejcn.1602015
35. Tershakovec AM, Shannon BM, Achterberg CL, et al. One-year follow-up of nutrition education for hypercholesterolemic children. Am J Public Health. 1998;88(2):258-261. doi:10.2105/AJPH.88.2.258
36. Tershakovec AM, Jawad AF, Stallings VA, et al. Growth of hypercholesterolemic children completing physician-initiated low-fat dietary intervention. J Pediatr. 1998;133(1):28-34. doi:10.1016/S0022-3476(98)70173-8
37. McHale SM, Tershakovec AM, Corneal DA, Tournier BA, Shannon BM. Psychosocial factors in nutrition education for hypercholesterolemic children. Ann Behav Med. 1998;20(3):233-240. doi:10.1007/BF02884966
38. Guaraldi F, Deon V, Del Bo’ C, et al. Effect of short-term hazelnut consumption on DNA damage and oxidized LDL in children and adolescents with primary hyperlipidemia: a randomized controlled trial. J Nutr Biochem. 2018;57:206-211. doi:10.1016/j.jnutbio.2018.03.012
39. Ramaswami U, Cooper J, Humphries SE; FH Paediatric Register Steering Group. The UK Paediatric Familial Hypercholesterolaemia Register: preliminary data. Arch Dis Child. 2017;102(3):255-260. doi:10.1136/archdischild-2015-308570
40. Avis HJ, Hargreaves IP, Ruiter JP, Land JM, Wanders RJ, Wijburg FA. Rosuvastatin lowers coenzyme Q10 levels, but not mitochondrial adenosine triphosphate synthesis, in children with familial hypercholesterolemia. J Pediatr. 2011;158 (3):458-462. doi:10.1016/j.jpeds.2010.08.015
41. Braamskamp MJ, Kusters DM, Wiegman A, et al. Gonadal steroids, gonadotropins and DHEAS in young adults with familial hypercholesterolemia who had initiated statin therapy in childhood. Atherosclerosis. 2015;241(2):427-432. doi:10.1016/j. atherosclerosis.2015.05.034
42. Luirink IK, Wiegman A, Kusters DM, et al. 20-Year follow-up of statins in children with familial hypercholesterolemia. N Engl J Med. 2019;381(16): 1547-1556. doi:10.1056/NEJMoa1816454
43. Braamskamp MJAM, Kastelein JJP, Kusters DM, Hutten BA, Wiegman A. Statin initiation during childhood in patients with familial hypercholesterolemia: consequences for cardiovascular risk. J Am Coll Cardiol. 2016;67(4): 455-456. doi:10.1016/j.jacc.2015.11.021
44. Braamskamp MJ, Kusters DM, Avis HJ, et al. Long-term statin treatment in children with familial hypercholesterolemia: more insight into tolerability and adherence. Paediatr Drugs. 2015;17(2):159-166. doi:10.1007/s40272-014-0116-y
45. Rodenburg J, Vissers MN, Wiegman A, et al. Statin treatment in children with familial hypercholesterolemia: the younger, the better. Circulation. 2007;116(6):664-668. doi:10.1161/CIRCULATIONAHA.106.671016
46. Joyce NR, Zachariah JP, Eaton CB, Trivedi AN, Wellenius GA. Statin use and the risk of type 2 diabetes mellitus in children and adolescents. Acad Pediatr. 2017;17(5):515-522. doi:10.1016/j.acap.2017.02.006
47. Desai NK, Mendelson MM, Baker A, et al. Hepatotoxicity of statins as determined by serum alanine aminotransferase in a pediatric cohort with dyslipidemia. J Pediatr Gastroenterol Nutr. 2019;68(2):175-181. doi:10.1097/MPG.0000000000002174
48. Dennison BA, Levine DM. Randomized, double-blind, placebo-controlled, two-period crossover clinical trial of psyllium fiber in children with hypercholesterolemia. J Pediatr. 1993;123(1):24-29. doi:10.1016/S0022-3476(05)81532-X
49. Guardamagna O, Amaretti A, Puddu PE, et al. Bifidobacteria supplementation: effects on plasma lipid profiles in dyslipidemic children. Nutrition. 2014;30(7-8):831-836. doi:10.1016/j.nut.2014.01.014
50. Del Bo’ C, Deon V, Abello F, et al. Eight-week hempseed oil intervention improves the fatty acid composition of erythrocyte phospholipids and the omega-3 index, but does not affect the lipid profile in children and adolescents with primary hyperlipidemia. Food Res Int. 2019;119:469-476. doi:10.1016/j.foodres.2018.12.045
51. Deon V, Del Bo’ C, Guaraldi F, et al. Effect of hazelnut on serum lipid profile and fatty acid composition of erythrocyte phospholipids in children and adolescents with primary hyperlipidemia: a randomized controlled trial. Clin Nutr. 2018;37(4):1193-1201. doi:10.1016/j.clnu.2017.05.022
52. Martino F, Martino E, Morrone F, Carnevali E, Forcone R, Niglio T. Effect of dietary supplementation with glucomannan on plasma total cholesterol and low density lipoprotein cholesterol in hypercholesterolemic children. Nutr Metab Cardiovasc Dis. 2005;15(3):174-180. doi:10.1016/j.numecd.2004.04.004
53. Guardamagna O, Abello F, Cagliero P, Visioli F. Could dyslipidemic children benefit from glucomannan intake? Nutrition. 2013;29(7-8):1060-1065. doi:10.1016/j.nut.2013.02.010
54. Verduci E, Agostoni C, Radaelli G, Banderali G, Riva E, Giovannini M. Blood lipids profile in hyperlipidemic children undergoing different dietary long chain polyunsaturated supplementations: a preliminary clinical trial. Int J Food Sci Nutr. 2014;65(3):375-379. doi:10.3109/09637486.2013.858239
55. Kusters 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. doi:10.1001/jama.2014.8892
56. 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. doi:10.1001/jama.292.3.331
57. Stein EA, Illingworth DR, Kwiterovich PO Jr, et al. Efficacy and safety of lovastatin in adolescent males with heterozygous familial hypercholesterolemia: a randomized controlled trial. JAMA. 1999; 281(2):137-144. doi:10.1001/jama.281.2.137
58. McCrindle BW, Ose L, Marais AD. Efficacy and safety of atorvastatin in children and adolescents with familial hypercholesterolemia or severe hyperlipidemia: a multicenter, randomized, placebo-controlled trial. J Pediatr. 2003;143(1):74-80. doi:10.1016/S0022-3476(03)00186-0
59. Knipscheer HC, Boelen CC, Kastelein JJ, et al. Short-term efficacy and safety of pravastatin in 72 children with familial hypercholesterolemia. Pediatr Res. 1996;39(5):867-871. doi:10.1203/00006450-199605000-00021
60. de Jongh S, Ose L, Szamosi T, et al; Simvastatin in Children Study Group. 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. doi:10.1161/01.CIR.0000035247.42888.82
61. 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. doi:10.1016/S0735-1097(02)02593-7
62. Couture P, Brun LD, Szots F, et al. Association of specific LDL receptor gene mutations with differential plasma lipoprotein response to simvastatin in young French Canadians with heterozygous familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 1998;18(6):1007-1012. doi:10.1161/01.ATV.18.6.1007
63. Clauss SB, Holmes KW, Hopkins P, et al. Efficacy and safety of lovastatin therapy in adolescent girls with heterozygous familial hypercholesterolemia. Pediatrics. 2005;116(3):682-688. doi:10.1542/peds.2004-2090
64. Braamskamp MJ, Stefanutti C, Langslet G, et al; PASCAL Study Group. Efficacy and safety of pitavastatin in children and adolescents at high future cardiovascular risk. J Pediatr. 2015;167(2):338-43.e5. doi:10.1016/j.jpeds.2015.05.006
65. 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. doi:10.1016/j.jacc.2009.10.042
66. Gylling H, Siimes MA, Miettinen TA. Sitostanol ester margarine in dietary treatment of children with familial hypercholesterolemia. J Lipid Res. 1995;36(8):1807-1812. doi:10.1016/S0022-2275(20) 41499-3
67. de Jongh S, Vissers MN, Rol P, Bakker HD, Kastelein JJ, Stroes ES. Plant sterols lower LDL cholesterol without improving endothelial function in prepubertal children with familial hypercholesterolaemia. J Inherit Metab Dis. 2003;26(4):343-351. doi:10.1023/A:1025155002348
68. Amundsen AL, Ose L, Nenseter MS, Ntanios FY. Plant sterol ester-enriched spread lowers plasma total and LDL cholesterol in children with familial hypercholesterolemia. Am J Clin Nutr. 2002;76(2):338-344. doi:10.1093/ajcn/76.2.338
69. Santos RD, Ruzza A, Hovingh GK, et al; HAUSER-RCT Investigators. Evolocumab in pediatric heterozygous familial hypercholesterolemia. N Engl J Med. 2020;383(14):1317-1327. doi:10.1056/NEJMoa2019910
70. Ritchie SK, Murphy EC, Ice C, et al. Universal versus targeted blood cholesterol screening among youth: the CARDIAC project. Pediatrics. 2010;126(2):260-265. doi:10.1542/peds.2009-2546
71. Summary of results from 1998 to present: coronary artery risk detection in Appalachian communities 21 year summary—fifth grade 1998-2020. CARDIAC Project. Published 2020. Accessed June 20, 2022. https://www.cardiacwv.org/?pid=10
72. Elliott E, Lilly C, Murphy E, Pyles LA, Cottrell L, NealWA. The Coronary Artery Risk Detection in Appalachian Communities (CARDIAC) project: an 18 year review. Curr Pediatr Rev. 2017;13(4):265-276. doi:10.2174/1573400514666180117093652
73. Kinnear FJ, Lithander FE, Searle A, et al. Reducing cardiovascular disease risk among families with familial hypercholesterolaemia by improving diet and physical activity: a randomised controlled feasibility trial. BMJ Open. 2020;10(12):e044200. doi:10.1136/bmjopen-2020-044200
74. Wheeler KA, West RJ, Lloyd JK, Barley J. Double blind trial of bezafibrate in familial hypercholesterolaemia. Arch Dis Child. 1985;60(1):34-37. doi:10.1136/adc.60.1.34
75. Kusters DM, Caceres M, Coll M, et al. Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia. J Pediatr. 2015;166(6):1377-1384. doi:10.1016/j.jpeds.2015.02.043
76. van der Graaf A, Cuffie-Jackson C, Vissers MN, et al. Efficacy and safety of coadministration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008;52(17):1421-1429. doi:10.1016/j.jacc.2008.09.002
77. Engler MM, Engler MB, Malloy MJ, Paul SM, Kulkarni KR, Mietus-Snyder ML. Effect of docosahexaenoic acid on lipoprotein subclasses in hyperlipidemic children (the EARLY study). Am J Cardiol. 2005;95(7):869-871. doi:10.1016/j.amjcard.2004.12.014
78. Tonstad S, Sivertsen M, Aksnes L, Ose L. Low dose colestipol in adolescents with familial hypercholesterolaemia. Arch Dis Child. 1996;74(2):157-160. doi:10.1136/adc.74.2.157
79. 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. doi:10.1016/S0022-3476(96)70188-9
80. Stein EA, Marais AD, Szamosi T, et al. Colesevelam hydrochloride: efficacy and safety in pediatric subjects with heterozygous familial hypercholesterolemia. J Pediatr. 2010;156(2):231-6.e1,3. doi:10.1016/j.jpeds.2009.08.037
81. Jackson CL, Keeton JZ, Eason SJ, et al. Identifying familial hypercholesterolemia using a blood donor screening program with more than 1 million volunteer donors. JAMA Cardiol. 2019;4(7):685-689. doi:10.1001/jamacardio.2019.1518
82. de Ferranti SD, Rodday AM, Mendelson MM, Wong JB, Leslie LK, Sheldrick RC. Prevalence of familial hypercholesterolemia in the 1999 to 2012 United States National Health and Nutrition Examination Surveys (NHANES). Circulation. 2016;133(11):1067-1072. doi:10.1161/CIRCULATIONAHA.115.018791
83. Writing Group for the DISC Collaborative Research Group. Efficacy and safety of lowering dietary intake of fat and cholesterol in children with elevated low-density lipoprotein cholesterol: the Dietary Intervention Study in Children (DISC). JAMA. 1995;273(18):1429-1435. doi:10.1001/jama.1995.03520420045036
84. Jacobs DR Jr, Woo JG, Sinaiko AR, et al. Childhood cardiovascular risk factors and adult cardiovascular events. N Engl J Med. 2022;386(20):1877-1888. doi:10.1056/NEJMoa2109191
85. Zhang Y, Vittinghoff E, Pletcher MJ, et al. Associations of blood pressure and cholesterol levels during young adulthood with later cardiovascular events. J Am Coll Cardiol. 2019;74(3):330-341. doi:10.1016/j.jacc.2019.03.529
86. Ference BA, Yoo W, Alesh I, et al. Effect of long-term exposure to lower low-density lipoprotein cholesterol beginning early in life on the risk of coronary heart disease: a mendelian randomization analysis. J Am Coll Cardiol. 2012;60(25):2631-2639. doi:10.1016/j.jacc.2012.09.017
87. Adams C, Burke V, Beilin LJ. Cholesterol tracking from childhood to adult mid-life in children from the Busselton study. Acta Paediatr. 2005;94(3):275-280. doi:10.1111/j.1651-2227.2005.tb03069.x
88. Clarke WR, Schrott HG, Leaverton PE, Connor WE, Lauer RM. Tracking of blood lipids and blood pressures in school age children: the Muscatine study. Circulation. 1978;58(4):626-634. doi:10.1161/01.CIR.58.4.626
89. Kelder SH, Osganian SK, Feldman HA, et al. Tracking of physical and physiological risk variables among ethnic subgroups from third to eighth grade: the Child and Adolescent Trial for Cardiovascular Health cohort study. Prev Med. 2002;34(3):324-333. doi:10.1006/pmed.2001.0990
90. Magnussen CG, Raitakari OT, Thomson R, et al. Utility of currently recommended pediatric dyslipidemia classifications in predicting dyslipidemia in adulthood: evidence from the Childhood Determinants of Adult Health (CDAH) study, Cardiovascular Risk in Young Finns Study, and Bogalusa Heart Study. Circulation. 2008;117(1):32-42. doi:10.1161/CIRCULATIONAHA.107.718981
91. Lewington S, Whitlock G, Clarke R, et al; Prospective Studies Collaboration. Blood cholesterol and vascular mortality by age, sex, and blood pressure: ameta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths. Lancet. 2007;370(9602):1829-1839. doi:10.1016/S0140-6736(07)61778-4
92. Zhang Y, Pletcher MJ, Vittinghoff E, et al. Association between cumulative low-density lipoprotein cholesterol exposure during young adulthood and middle age and risk of cardiovascular events. JAMA Cardiol. 2021;6(12):1406-1413. doi:10.1001/jamacardio.2021.3508
93. Bahls M, Lorenz MW, Dörr M, et al; PROG-IMT Study Group. Progression of conventional cardiovascular risk factors and vascular disease risk in individuals: insights from the PROG-IMT consortium. Eur J Prev Cardiol. 2020;27(3):234-243. doi:10.1177/2047487319877078
94. Narverud I, Retterstøl K, Iversen PO, et al. Markers of atherosclerotic development in children with familial hypercholesterolemia: a literature review. Atherosclerosis. 2014;235(2):299-309. doi: 10.1016/j.atherosclerosis.2014.05.917
95. Braamskamp MJ, Hutten BA, Wiegman A. Early initiation of statin treatment in children with familial hypercholesterolaemia. Curr Opin Lipidol. 2015;26 (3):236-239. doi:10.1097/MOL.0000000000000177
96. Kusters DM, Wiegman A, Kastelein JJ, Hutten BA. Carotid intima-media thickness in children with familial hypercholesterolemia. Circ Res. 2014;114(2):307-310. doi:10.1161/CIRCRESAHA.114.301430
97. Wiegman A, de Groot E, Hutten BA, et al. Arterial intima-media thickness in children heterozygous for familial hypercholesterolaemia. Lancet. 2004;363(9406):369-370. doi:10.1016/S0140-6736(04)15467-6
98. Vuorio A, Kuoppala J, Kovanen PT, et al. Statins for children with familial hypercholesterolemia. Cochrane Database Syst Rev. 2019;2019(11):CD006401. doi:10.1002/14651858.CD006401.pub5
99. Cottrell L, John C, Murphy E, et al. Individual-, family-, community-, and policy-level impact of a school-based cardiovascular risk detection screening program for children in underserved, rural areas: the CARDIAC Project. J Obes. 2013;2013:732579. doi:10.1155/2013/732579
100. Karapostolakis G, Vakaki M, Attilakos A, et al. The effect of long-term atorvastatin therapy on carotid intima-media thickness of children with dyslipidemia. Angiology. 2021;72(4):322-331. doi: 10.1177/0003319720975635
101. Mamann N, Lemale J, Karsenty A, Dubern B, Girardet JP, Tounian P. Intermediate-term efficacy and tolerance of statins in children. J Pediatr. 2019; 210:161-165. doi:10.1016/j.jpeds.2019.03.032
102. Langslet G, Breazna A, Drogari E. A 3-year study of atorvastatin in children and adolescents with heterozygous familial hypercholesterolemia. J Clin Lipidol. 2016;10(5):1153-1162. doi:10.1016/j.jacl.2016.05.010
103. Carreau V, Girardet JP, Bruckert E. Long-term follow-up of statin treatment in a cohort of children with familial hypercholesterolemia: efficacy and tolerability. Paediatr Drugs. 2011;13(4):267-275. doi:10.2165/11591650-000000000-00000
104. Kavey RW, Manlhiot C, Runeckles K, et al. Effectiveness and safety of statin therapy in children: a real-world clinical practice experience. CJC Open. 2020;2(6):473-482. doi:10.1016/j.cjco.2020.06.002
105. Benekos T, Kosmeri C, Vlahos A, Milionis H. Nine-year overview of dyslipidemia management in children with heterozygous familial hypercholesterolemia: a university hospital outpatient lipid clinic project in Northwestern Greece. J Pediatr Endocrinol Metab. 2020;33(4):533-538. doi:10.1515/jpem-2019-0250
106. Humphries SE, Cooper J, Dale P, Ramaswami U, Group FHPRS. The UK Paediatric Familial Hypercholesterolaemia Register: statin-related safety and 1-year growth data. J Clin Lipidol. 2018;12(1):25-32. doi:10.1016/j.jacl.2017.11.005
107. Zachariah JP, Chan J, Mendelson MM, et al. Adolescent dyslipidemia and standardized lifestyle modification: benchmarking real-world practice. J Am Coll Cardiol. 2016;68(19):2122-2123. doi:10.1016/j.jacc.2016.08.041
108. Benuck I. Point: the rationale for universal lipid screening and treatment in children. J Clin Lipidol. 2015;9(5 suppl):S93-S100. doi:10.1016/j. jacl.2015.03.104
109. Pyles LA, Lilly CL, Joseph A, Mullett CJ, Neal WA. Cardiometabolic risk factors in siblings from a statewide screening program. J Clin Lipidol. 2020;14(6):762-771. doi:10.1016/j.jacl.2020.09.003
110. Wald DS, Bestwick JP, Morris JK, Whyte K, Jenkins L,Wald NJ. Child-parent familial hypercholesterolemia screening in primary care. N Engl J Med. 2016;375(17):1628-1637. doi:10.1056/NEJMoa1602777
111. Wald DS, Bestwick JP. Reaching detection targets in familial hypercholesterolaemia: comparison of identification strategies. Atherosclerosis. 2020;293:57-61. doi:10.1016/j.atherosclerosis.2019.11.028
112. Wald DS, Wald NJ. Integration of child-parent screening and cascade testing for familial hypercholesterolaemia. J Med Screen. 2019;26(2):71-75. doi:10.1177/0969141318796856
113. Doshi N, Perrin EM, Lazorick S, Esserman D, Steiner MJ. Short-term change in body mass index in overweight adolescents following cholesterol screening. Arch Pediatr Adolesc Med. 2009;163(9):812-817. doi:10.1001/archpediatrics.2009.152
114. Gregory EF, Miller JM, Wasserman RC, Seshadri R, Rubin DM, Fiks AG. Routine cholesterol tests and subsequent change in BMI among overweight and obese children. Acad Pediatr. 2019;19(7):773-779. doi:10.1016/j.acap.2019.05.131
115. Futema M, Cooper JA, Charakida M, et al; UK10K Consortium. Screening for familial hypercholesterolaemia in childhood: Avon Longitudinal Study of Parents and Children (ALSPAC). Atherosclerosis. 2017;260:47-55. doi:10.1016/j.atherosclerosis.2017.03.007
116. Damgaard D, Larsen ML, Nissen PH, et al. The relationship of molecular genetic to clinical diagnosis of familial hypercholesterolemia in a Danish population. Atherosclerosis. 2005;180(1):155-160. doi:10.1016/j.atherosclerosis.2004.12.001
117. Williams RR, Hunt SC, Schumacher MC, et al. Diagnosing heterozygous familial hypercholesterolemia using new practical criteria validated by molecular genetics. Am J Cardiol. 1993;72(2):171-176. doi:10.1016/0002-9149(93)90155-6
118. Austin MA, Hutter CM, Zimmern RL, Humphries SE. Genetic causes of monogenic heterozygous familial hypercholesterolemia: a HuGE prevalence review. Am J Epidemiol. 2004;160(5):407-420. doi:10.1093/aje/kwh236

Return to Table of Contents

Figure 1 is the analytic framework that depicts the five Key Questions to be addressed in the systematic review. The figure illustrates how screening for familial hypercholesterolemia (FH) or multifactorial dyslipidemia in asymptomatic children and adolescents may result in delayed or reduced incidence of health outcomes (CVD events,*CVD mortality, or all-cause mortality) or improve intermediate outcomes (serum lipid levels, atherosclerotic markers, BMI, physical activity, sedentary behavior, dietary intake) in children, adults, or both (KQ1). There is also a question related to the diagnostic yield of serum lipid screening for FH or multifactorial dyslipidemia (KQ2) and the harms of screening for these conditions in children and adolescents (KQ3). Additionally, the figure illustrates how treatment of FH or multifactorial dyslipidemia with lifestyle modifications, lipid-lowering medications, or both in children and adolescents may delay or reduce the incidence of health outcomes (CVD events,*CVD mortality, or all-cause mortality) or improve intermediate outcomes (serum lipid levels, atherosclerotic markers, BMI, physical activity, sedentary behavior, dietary intake) in children, adults, or both (KQ4) and what harms are associated with these treatments (KQ5).

Evidence reviews for the US Preventive Services Task Force (USPSTF) use an analytic framework to visually display the key questions that the review will address to allow the USPSTF to evaluate the effectiveness and safety of a preventive service. The questions are depicted by linkages that relate interventions and outcomes. A dashed line indicates a health outcome that immediately follows an intermediate outcome. For more details see the USPSTF Procedure Manual.8 BMI indicates body mass index; CVD, cardiovascular disease.
a Defined as dyslipidemia not due to familial hypercholesterolemia.
b Defined as myocardial infarction or ischemic stroke.

Return to Table of Contents

Two reviewers evaluated 7058 abstracts and 272 full-text articles for KQ eligibility. Overall, 43 studies (65 publications) met inclusion criteria for this systematic review. Thirteen of these studies evaluated the benefits of supplement interventions and 10 reported on the harms of supplement interventions.

Reasons for exclusion: Study design: Study did not use an included design.
Setting: Study was not conducted in a country relevant to US practice.
Outcomes: Study did not have relevant outcomes or had incomplete outcomes.
Comparator: Study included a comparator group that was not included.
Intervention: Study used an excluded intervention or screening approach.
Population: Study was not conducted in an average-risk population. Quality: Study did not meet criteria for fair or good quality. Yield study superseded: Publication evaluated for KQ2 (yield) was superseded by another publication that was more contemporary, comprehensive, or more relevant. FH indicates familial hypercholesterolemia; KQ, key question; MFD, multifactorial dyslipidemia.
a Studies may appear in more than 1 KQ.
b One study reports both FH and MFD populations.

Return to Table of Contents

Intervention No. of included studies (No. of participants) Summary of findings Consistency and precision Other limitations Strength of evidence Applicability
KQ1: Benefits of screening
Universal or selective screening 0 NA NA NA Insufficient NA
KQ2: Yield
Universal or selective screening 3 (n = 395,465)
New: 3
Diagnostic yield:
   No studies reported true diagnostic yield, as there were no screening studies with genetic testing

Prevalence:
   Using thresholds of LDL-C ≥190 mg/dL or total cholesterol ≥270 mg/dL, FH prevalence was 0.20% to 0.42% (1:250 to 1:500)

Targeted screening based on family history would miss a substantial proportion of cases

Diagnostic yield: NA

Prevalence: reasonably consistent; reasonably precise

No genetic or family
history criteria; lipid values are used as a proxy for FH
Insufficient for diagnostic yield

Low for prevalence

US children and adolescents with most evidence for ages 10 y or older; applicability to various recruitment settings and geographic locations
KQ3: Harms of screening
Universal or selective screening 0 NA NA NA Insufficient NA
KQ4: Benefits of treatment
Statin 10 (n = 1230)
New: 1
Total cholesterol: 7 studies (n = 706); MD in change, −82.1 mg/dL (95% CI, −101.1 to −63.2); I2 = 83.0%

LDL-C: 8 studies (n = 742); MD in change, −81.3 mg/dL (95% CI, −97.6 to −65.0); I2 = 81.6%

Total cholesterol and LDL-C effects appear dose-related

Consistent; reasonably precise Heterogeneity of statin drugs and intensity

Short-term follow-up (one 2-y trial but all other trials <6 mo)

No health outcomes

Small sample sizes (range, 50-214)

Moderate for benefit Children and adolescents aged 6-18 y with FH defined using various diagnostic criteria
Bile acid sequestrants 3 (n = 332)
New: 0
Total cholesterol: MD in change, −22.1 to −40.6 mg/dL

LDL-C: MD in change, −13.2 to −45.9 mg/dL

Variation in effect by dose

Reasonably consistent; reasonably precise Different formulations of bile acid sequestrants

Short duration (8-52 wk)

No health outcomes

Low for benefit Children and adolescents aged 6-17 y with FH
Ezetimibe 1 (n = 138)
New: 0
Total cholesterol: MD in change, −64.0 mg/dL (95% CI, −81.1 to −46.9)

LDL-C: MD in change, −63.0 mg/dL (95% CI, −79.5 to −46.5)

Consistency NA; reasonably precise Short duration (12 wk)

No health outcomes

Low for benefit Children aged 6-11 y with FH
PCSK9 inhibitor 1 (n = 158)
New: 1
LDL-C: MD in change, −68.6 mg/dL (95% CI, −83.1 to −54.0) Consistency NA; reasonably precise Short duration (24 wk)

No health outcomes

Low for benefit Children and adolescents aged 10-17 with FH
Drug combination (simvastatin + ezetimibe) 1 (n = 248)
New: 0
Compared with single drug:
   Total cholesterol: MD in change, −40.1 mg/dL (95% CI, −51.1 to −29.2)
   LDL-C: MD in change, −37.5 mg/dL (95% CI, −48.0 to −27.0)
Consistency NA; reasonably precise Short duration (33 wk)

No health outcomes

Low for benefit Children and adolescents aged 10-17 with FH
Behavioral counseling 1 (n = 21)
New: 1
Lipids: no difference

Physical activity outcomes: overlapping confidence intervals for intervention vs control

Dietary outcomes: mixed results

Consistency NA; imprecise Very small trial

Short duration (12 wk)

No health outcomes

Insufficient Low-intensity diet and physical activity intervention for patients aged 10-18 y with FH
KQ5: Harms of treatment
Statin 12 (n = 1476 in trials, 10,336 in NRSI harms-only studies)

New: 3 (1 RCT,
2 NRSI)

Transaminitis >3× ULN: 0%-4.5% (intervention) vs 0%-1.9% (control), but largest trial (n = 214) with 2-y follow-up reported no cases in the statin group and 2 cases of AST >3× ULN in the control group

In the 10-y observational follow-up of this trial, transaminitis at this threshold was similarly rare (ALT: 1 case of >3× ULN elevation in the statin group; AST: 1 case of >3× ULN each in the statin and control group)

CK ≥10× ULN: 0 in 2 trials and up to 4.5% (intervention) vs 1.7% (control) but 1 trial’s 10-y observational follow-up reported no instances of elevated CK

1 NRSI (n = 943) reported ALT elevations of >3× ULN, with a frequency of 4.4% in the statin group and 1.5% in the control group over 3.5 y of observation

1 NRSI (n = 9393) showed no difference in new diabetes diagnoses over 9 y

Six trials (n = 931) and 1 NRSI (n = 309) reported no significant differences between Tanner stages or other hormonal adverse events

Inconsistent; imprecise Most trials were short-term and small with few events, leading to imprecise estimates

Clinical importance of transient elevations in these laboratory values is unknown

Low for reversible liver and musculoskeletal laboratory abnormalities

Insufficient for new-onset diabetes

Low for no growth or hormonal harms

Short-term harms
Bile acid sequestrants 3 (n = 332)
New: 0
Similar rates of total adverse events in intervention and control groups Relatively consistent, imprecise Different formulations, few events

Short duration (8-52 wk)

Low for minimal harm Children and adolescents aged 6-17 y with FH
Ezetimibe 1 (n = 138)
New: 0
Similar rates of total adverse events in intervention and control groups Consistency NA, imprecise Single trial

Short duration (12 wk)

Few events

Insufficient Children aged 6-11 y with FH
PCSK9 inhibitor 1 (n = 158)
New: 1
Similar rates of total adverse events in intervention and control groups Consistency NA, imprecise Single trial

Short duration (24 wk)

Few events

Insufficient Children and adolescents aged 10-17 with FH
Drug combination (simvastatin + ezetimibe) 1 (n = 248)
New: 0
Similar rates of total adverse events in intervention and control groups Consistency NA, imprecise High total adverse events in both the intervention and the control group

Short duration (33 wk)

Insufficient Children and adolescents aged 10-17 with FH
Behavioral counseling 0 NA NA NA Insufficient NA

Abbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CK, creatine kinase; DHA, docosahexaenoic acid; FDA, US Food and Drug Administration; FH, familial hypercholesterolemia; KQ, key question; LDL-C, low-density lipoprotein cholesterol; MD, mean difference; NA, not applicable; NRSI, nonrandomized controlled study of intervention; PCSK9, proprotein convertase subtilisin/kexin type 9; RCT, randomized clinical trial; ULN, upper limit of normal.
SI conversion factors: To convert LDL-C and total cholesterol values to mmol/L, multiply by 0.0259.

Return to Table of Contents

Intervention No. of included studies Summary of findings Consistency and precision Other limitations Strength of evidence Applicability
KQ1: Benefits of screening
Universal or selective screening 0 NA NA NA Insufficient NA
KQ2: Yield
Universal or selective screening 5 (n = 142,257)
New: 5
Diagnostic yield:
   No studies reported true diagnostic yield, as there were no screening studies with genetic testing

Prevalence:
   ≥1 Abnormal lipid value: 19.2% (NHANES [n = 4381])
   Total cholesterol ≥200 mg/dL: 7.1% (NHANES) to 9.4% (PVHS) (3 studies [n = 75,551])
   LDL-C ≥130 mg/dL: 6.4% (NHANES) to 7.4% (CARDIAC) (2 studies [n = 56,824])
   HDL-C <40 mg/dL: 12.1% (NHANES) to 22.2% (PVHS) (4 studies [n = 72,320])
   Triglycerides ≥130 mg/dL: 10.2% (NHANES) (1 study [n = 2045])
   Non–HDL-C ≥145 mg/dL: 6.4% (NHANES) and 13.0% (PVHS) (2 studies [n = 16,150])

Diagnostic yield: NA

Prevalence: Consistent; reasonably precise for total cholesterol and LDL-C but imprecise for other measures

No confirmatory testing

NHANES represents only national sample and included most recent years of 2016; fasting and nonfasting samples

Prevalence varies by population characteristics

Insufficient for diagnostic yield of screening tests

Moderate that abnormal lipid values are common

US children and adolescents aged 6-19 y

Overall prevalence lower in national data set (NHANES) compared with other geographically focused recruitment settings

KQ3: Harms of screening
Universal or selective screening 0 NA NA NA Insufficient NA
KQ4: Benefits of treatment
Behavioral counseling 2 (n = 934)
New: 1
One 7-y trial (DISC) of a high-intensity dietary intervention showed statistically significant reductions in total cholesterol and LDL-C (MD in change, −3.3 mg/dL for total cholesterol and LDL-C) at 3 y that were not sustained at 7-y follow-up

One low-intensity dietary 10-wk intervention with up to 1 y of follow-up: statistically significant reduction in LDL-C (MD in change, −6.7 mg/dL) at 3 mo not sustained at 1-y follow-up

Both trials reported that interventions were associated with improved dietary intake outcomes, which were attenuated at longer follow-up

Consistent, reasonably precise

Heterogeneous dietary interventions with variable intensity, duration, and follow-up

Low for no long-term benefit Children aged 4-10 y
KQ5: Harms of treatment
Behavioral counseling 2 (n = 934)
New: 1
No harmful effects identified in growth (BMI, weight, height), development (Tanner stage), nutritional outcomes (serum ferritin, red cell folate, zinc, albumin), or psychological outcomes (anxiety, depression, behavior)

One trial (DISC) reported better depression outcomes in the intervention group

Consistent, reasonably precise Heterogeneous dietary interventions with variable intensity, duration, and follow-up Low for no harms Children aged 4-10 y

Abbreviations: BMI, body mass index; CARDIAC, Coronary Artery Risk Detection in Appalachian Communities; DISC, Dietary Intervention Study in Children; KQ, key question; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; MD, mean difference; NA, not applicable; NHANES, National Health and Nutrition Examination Survey; non–HDL-C, non–high-density lipoprotein cholesterol; PVHS, Poudre Valley Health System study; RCT, randomized clinical trial.
SI conversion factors: To convert LDL-C, total cholesterol, HDL-C, and non–HDL-C values to mmol/L, multiply by 0.0259; triglyceride values to mmol/L, multiply by 0.0113.

Return to Table of Contents