archived

Final Evidence Summary

Lipid Disorders in Children: Screening

July 15, 2007

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.

Elizabeth M. Haney, MD;1,2 Laurie Hoyt Huffman, MS;1 Christina Bougatsos, BS;1 Michele Freeman, MPH;1 Robert D. Steiner, MD;3 Heidi D. Nelson, MD, MPH.1,2,4

The authors of this article are responsible for its contents, including any clinical or treatment recommendations. No statement in this article should be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

This article was first published in the Pediatrics.

Return to Table of Contents

Objective: This was a systematic evidence review for the U.S. Preventive Services Task Force, intended to synthesize the published evidence regarding the effectiveness of selecting, testing, and managing children and adolescents with dyslipidemia in the course of routine primary care.

Methods: Literature searches were performed to identify published articles addressing 10 key questions. The review focused on screening relevant to primary care of children without previously identified dyslipidemias, but included treatment trials of children with dyslipidmia because some drugs have only been tested in that population.

Results: Normal values for lipids for children and adolescents are defined according to population levels (percentiles). Age, sex, and racial differences and temporal trends may alter these statistical cut points. Approximately 40-55% of children with elevated total cholesterol and low-density lipoprotein will continue to have elevated lipids on follow-up. Current screening recommendations based on family history will fail to detect substantial numbers (30-60%) of children with elevated lipids.

Drug treatment for dyslipidemia in children has been studied and shown to be effective only for suspected or proven familial monogenic dyslipidemias. Intensive dietary counseling and follow-up can result in improvements in lipids, but these results have not been sustained after the cessation of the intervention. The few trials of exercise are of fair-poor quality and show little or no improvements in lipids for children without monogenic dyslipidemias. Although reported adverse effects were not serious, studies were generally small and not of sufficient duration to determine long-term effects of either short or extended use.

Conclusions: Several key questions about screening and treatment of dyslipidemia in children and adolescents could not be addressed because of lack of studies, including effectiveness of screening on adult coronary heart disease (CHD) or lipid outcomes, optimal ages and intervals for screening children, or effects of treatment of childhood lipid levels on adult CHD outcomes.

Return to Table of Contents

Dyslipidemias are disorders of lipoprotein metabolism resulting in abnormal excesses of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), or triglycerides, or deficiency of high-density lipoprotein cholesterol (HDL-C).1,2 Dyslipidemia is an established risk factor for coronary heart disease (CHD)—the leading cause of death for adults in the US.3 Dyslipidemia rarely leads to adverse health outcomes in childhood, but its long-term effects may be considerable. While no long-term studies of the direct relationship between lipid levels measured in children and CHD later in life have been conducted, this relationship can be inferred. Large epidemiologic studies indicate that children's lipid levels correlate with those of adult family members.4 Children of parents with CHD have a higher prevalence of dyslipidemia in childhood,5 and identification of dyslipidemia in children can identify families at increased risk for CHD.4 Studies of children and young adults who died accidentally report correlations between lipid levels and arterial fat deposition,6,7 and note early lesions of atherosclerosis (fatty streaks) in the abdominal aorta at age three years, coronary arteries at age 10 years, and further progression with age.8-12 Increasing prevalence of risk factors for CHD among children, including metabolic syndrome and obesity, as well as continued emphasis on primary prevention of CHD has raised interest in screening children for dyslipidemia.13-15

Dyslipidemia is defined by laboratory testing and statistically determined criteria. Elevated LDL-C is the most common clinically significant marker of dyslipidemia in children. The majority of children with dyslipidemia will have idiopathic dyslipidemias (polygenic, risk factor associated, or multi-factorial), while a minority will have monogenic or secondary dyslipidemias. The more common genetic dyslipidemias include familial hypercholesterolemia (FH), familial combined hyperlipidemia (FCH), familial defective apoprotein-B, and familial hypertriglyceridemia.

Most treatment recommendations advise a low-fat, low-cholesterol diet, such as the American Heart Association (AHA) Step I diet, for children with dyslipidemia beginning at age two years and older.14 Children younger than two years should not be prescribed a low-fat, low-cholesterol diet because their rapid growth and development require adequate fat and cholesterol intake.16,17 Children and adolescents with FH or FCH are the only non-adults for whom trials of drug therapy are available and drugs are approved by the US Food and Drug Administration (FDA). Bile acid-binding resins are the only medications approved for treatment of dyslipidemia for children younger than eight years of age. HMG Co-A reductase inhibitors (statins) are approved for use in older children with heterozygous FH.18,19 Other medications used in adults for treatment of hyperlipidemia, such as niacin, are either not recommended for children or have not been adequately evaluated for safety and efficacy in children. Additional interventions for children include dietary supplements (fiber, sterol or stanol margarines, omega-3 fatty acids), exercise, weight loss for overweight children, and identification and treatment of diabetes mellitus or other causes of secondary dyslipidemia.

The relationship between childhood and adult dyslipidemia, increasing prevalence of related CHD risk factors in children (e.g., obesity and diabetes),13-15 and continued emphasis on a primary prevention approach for CHD has raised interest in screening children for dyslipidemia. Identifying children with dyslipidemia could lead to interventions or treatments that could prevent or delay adult dyslipidemia and CHD. This rationale lends support to consideration of screening for dyslipidemia as part of well-child care and at other opportunities. Clinic-based screening, neonatal screening, community-based screening, and other prevention strategies have been proposed, but most recommendations support selective strategies testing children who have family members with dyslipidemia or premature CHD and those with unknown family histories.16,20

This evidence review focuses on the strengths and limitations of evidence for identifying and managing children and adolescents with dyslipidemia determined by screening in the course of routine primary care. Its objective is to determine the balance of potential benefits and adverse effects of screening for development of guidelines by the US Preventive Services Task Force (USPSTF). The target population includes children and adolescents age 0 to 21 years without previously-known conditions associated with dyslipidemia. There is potential to identify children and adolescents with dyslipidemia in this population from among three groups: those with undiagnosed monogenic dyslipidemias, such as familial hypercholesterolemia; those with undiagnosed secondary causes of dyslipidemia (diabetes, nephrotic syndrome, hypothyroidism, others); and those with idiopathic dyslipidemia (polygenetic, risk factor associated, or multi-factorial) (Figure 1). Although children and adolescents with idiopathic dyslipidemia generally have less severe lipid abnormalities than children and adolescents with monogenic disorders, such abnormal levels could still potentially improve with intervention.

Return to Table of Contents

Evidence reviews for the USPSTF follow a specific methodology21 (Figure 2). Key questions examine a chain of evidence about the accuracy and feasibility of screening children and adolescents for dyslipidemia in primary care or community settings (Key Question 1), abnormal lipid values (Key Question 2a), appropriate tests (Key Question 2b), tracking of lipid levels through childhood to adulthood (Key Question 2c), accuracy of family history (Key Question 2d), role of risk factors in selecting children and adolescents for screening (Key Question 2e), effectiveness of interventions for children and adolescents identified with dyslipidemia (Key Questions 4-8, 10), and adverse effects of screening and interventions (Key Questions 3, 9).

Studies that addressed Key Question 1 (Figure 2) include all components in the continuum of the screening process: the screening evaluation, diagnostic evaluation for those identified by the screening results, interventions for those diagnosed with dyslipidemia, and outcome measures allowing determination of the effectiveness of the overall screening process.

Studies of children with previously diagnosed conditions known to cause dyslipidemia were not included because the scope of this review is screening children without known diagnoses. Specifically, studies of children with diabetes were not included because these children would already be under surveillance for dyslipidemia as a result of their primary disease. This review includes treatment trials of children and adolescents using dietary, exercise, and drug interventions. Trials of drug therapy in children with heterozygous FH or FCH are included because drug treatment trials have been conducted exclusively in this population.

Relevant studies were identified from multiple searches of MEDLINE® (1966 through September 2005).22 We obtained additional articles from recent systematic reviews, reference lists of related studies, reviews, editorials, and Web sites, and from consulting experts. Retrieved abstracts were entered into an electronic database (EndNote®).

Investigators reviewed all identified abstracts and determined eligibility by applying inclusion and exclusion criteria specific to each key question. Full-text articles of included abstracts were reviewed for relevance. Eligible studies were English-language, applicable to US clinical practice, and provided primary data relevant to key questions. Studies of risk factors were included only if they provided multivariate adjusted analyses.

For treatment studies, full text randomized controlled trials (RCTs), non-controlled clinical trials, and non-controlled prospective studies providing data on the treatment of children and adolescents with diet, drug therapy, exercise, or combinations of these were initially reviewed. Subsequently, only RCTs and meta-analyses of RCTs that reported serum lipid outcomes were included. Crossover trials were included if they reported data prior to crossover. For Key Question 10, outcomes included either adult lipid levels or adult CHD. Information about adverse effects of treatment was obtained from RCTs and additional sources, such as non-randomized controlled treatment trials and non-comparative studies of treatment.

Data were extracted from each study, entered directly into evidence tables, and summarized. Benefits and adverse effects of therapies were considered equally important and both types of outcomes were abstracted. Trials of therapy for children and adolescents with dyslipidemia were categorized by population and intervention. Two reviewers independently rated the RCTs' quality using US Preventive Services Task Force criteria (Appendix 1).21

Return to Table of Contents

Our literature search identified 2,507 unique citations, including 144 papers about screening and testing for dyslipidemia (Key Question 2); 43 about interventions and tracking of lipid values over time (Key Questions 4-8 and 10); 6 about the adverse effects of screening (Key Question 3) and 84 about adverse effects of treatment (Key Question 9).

Key Question 1. Is screening for dyslipidemia in children/adolescents effective in delaying the onset and reducing the incidence of CHD-related events?

No studies evaluated the effect of screening children and adolescents on adult lipid or disease outcomes.

Key Question 2. What is the accuracy of screening for dyslipidemia in identifying children/adolescents at increased risk of CHD-related events and other outcomes?

Key Question 2a. What are abnormal lipid values in children/adolescents?

While several studies conducted in the US during the 1970s obtained lipid levels from large samples of normal healthy children,23-25 current recommendations14,16,20,26 are based on distributions of lipid and lipoprotein levels obtained from the Lipid Research Clinics (LRC) Prevalence Study.27 This study included one Canadian and nine US sites and enrolled subjects primarily based on residency within census tracts, school enrollment, and employment in occupational and industrial groups. Fasting (≥12 hours) lipoprotein levels were obtained in 15,626 children age 0 to 19 years between 1972 and 1976. The selected populations included a broad range of geographic, socio-economic, occupational, sex, and ethnic groups, but were not selected to be a representative sample of the North American population.

In the LRC sample, TC levels increased from birth and stabilized at approximately 2 years of age. At puberty, TC levels declined slightly for both boys and girls, and HDL-C levels declined for boys. For all children, the mean serum level for TC was approximately 160 mg/dL and for LDL-C was 100 mg/dL. The 95th percentile level was 200 mg/dL for TC and 130 mg/dL for LDL-C. While results for African American children were similar, they were based on smaller numbers and provided only TC and triglyceride data.27

More recent data from the National Health and Nutrition Examination Survey (NHANES) III (1988 to 1994) were derived from 7,499 children and adolescents ages 4 to 19 years. These provided 95th percentile levels of 216 mg/dL for serum TC, and 152 mg/dL for LDL-C.28 Mean age-specific TC levels peaked at 171 mg/dL at 9 to 11 years and declined at older ages. Girls had significantly higher mean TC and LDL-C levels than boys (p<0.005). Non-Hispanic Black children and adolescents had significantly higher mean TC, LDL-C, and HDL-C levels compared to non-Hispanic White and Mexican-American children and adolescents. In linear regression models of these data, age, sex, and race have significant effects on lipid levels questioning the utility of fixed screening cut points.29

Key Question 2b. What are the appropriate tests? How well do screening tests (non-fasting total cholesterol, fasting total cholesterol, fasting lipoprotein analysis) identify children and adolescents with dyslipidemia?

In the American Academy of Pediatrics (AAP) and the National Cholesterol Education Program (NCEP) guidelines, TC is used as an initial laboratory measurement for children tested because of a family history of high cholesterol or vascular disease, and a lipoprotein profile is obtained if the patient has a TC over a certain defined target.16,20 In children LDL-C is the basis for initiating treatment and determining goals of therapy.

How well TC levels detect elevated LDL-C levels has been examined with LRC data (ages 6 to19, n=1325),30 and data from the biracial Bogalusa cohort (ages 5 to 17, n=2,857).31 Elevated levels were defined as >95th percentile. With LRC data, an elevated fasting TC detected children with elevated LDL-C and elevated triglycerides with 69% sensitivity and 98% specificity.30

In the Bogalusa cohort, elevated TC detected elevated LDL-C with 44% (white females) to 50% (white males, African American males and females) sensitivity and 90% specificity (African American and white males and females).31

In adults, both TC and HDL-C are recommended for screening. While this has not been recommended in guidelines for children and adolescents, it is common in practice.32 HDL-C may help distinguish false negatives from true negatives when used with TC.30 In 260 African American adolescents ages 12 to 20 years, fasting TC minus HDL-C above the 95th percentile was 88-96% sensitive and 98% specific for predicting LDL-C ≥130 mg/dL.33 Using a lower threshold of fasting TC ≥ the 75th percentile to detect LDL-C ≥ the 95th percentile is a sample of Hispanic children ages 4-5, sensitivities were 86% (using an LRC defined 75th percentile) and 96% (using the sample-defined 75th percentile), and specificities were 93% (LRC defined) and 87% (sample defined).34 A TC > 215 mg/dL is required, however, to accurately identify a child with elevated LDL-C with 95% confidence. No single TC value places a child in the borderline category (170-200 mg/dL) with 95% confidence.35 Direct measurement of LDL-C can be done using non-fasting serum samples and may be as precise as calculated LDL-C, but this remains controversial.36,37

Key Question 2c. How well do lipid levels track from childhood to adulthood?

Twenty-three prospective cohort studies contributed information on tracking lipid levels during childhood.38-60 These studies drew from seven US cohorts and eight non-US cohorts. Approximately 40% to 55% of children with elevated lipids, defined by percentile within a population distribution, will continue to have elevated lipids on follow-up (4-15 years later).22 None of these studies, however, evaluated the proportion of children and adolescents with lipid levels above the 95th percentile who remained in the top 5% at follow-up.

Key Question 2d. What is the accuracy of family history in determining risk?

Several good-quality studies of diagnostic accuracy evaluated the sensitivity and specificity of family history information in determining risk for dyslipidemia in children and adolescents (Table 1).33,34,61-74 Studies used different definitions of family history such as any parental history of heart attack, other parental risk factors, and varying age definitions of early CHD, and selected different levels of LDL-C or TC as the lipid detection threshold. For example, parental history of early CHD alone was 5% to 17% sensitive for TC >170 mg/dL or LDL-C >130 mg/dL,34,63 whereas parent or grandparent history of early CHD was 46% sensitive for LDL >the 95th percentile.65

Regardless of the precise definition, using positive family history information to trigger lipid testing misses substantial numbers of children with elevated lipids, ranging from 17-90% overall and 30-60% in most studies.33,64,65,68,70,72,75-77 The proportion of children and adolescents qualifying for screening based on family history is generally between 25% to 55%, depending on the sensitivity of the specific family history question.33,34,61-65,67,70,71,73,78

Key Question 2e. What are other important risk factors?

Forty-three cohort and cross-sectional studies of mixed quality with adjusted statistical analyses contributed information on additional risk factors for identifying children at increased risk for elevated lipids and/or CHD-related events.66,79-120 Thirty studies examined overweight or body fat composition measures as a risk factor for dyslipidemia.79-82,84-86,89-95,99,101-104,106-112,114,115,117,119 These measures were the most consistently effective in predicting risk of dyslipidemia, compared to other factors assessed.22 Childhood overweight, as measured by BMI, was the best independent predictor of adult dyslipidemia after LDL-C, specifically when considering BMI increases from childhood to adulthood.121 Five of six studies evaluating overweight as a risk found that overweight was associated with abnormal lipid levels.85,86,94,110,115,117

Key Question 2f. What are effective screening strategies for children/adolescents (including frequency of testing, optimal age for testing)?

Thirty-two studies evaluated screening strategies among children in various settings.33,34,61,63-66,68,70,72,76,77,122-141 The only RCT compared two regimens for screening college students.131 All others were non-comparative prospective studies tht described screening interventions and differed considerably in venue (school, pediatric clinic, hospital, or population-based cohort), methods (fasting or non-fasting samples, method for detecting of positive family history), and outcomes. Most reported low parental compliance with follow-up testing,76,136-139 even when follow-up was provided free of charge, as in pre-paid health plans.

Studies demonstrate low compliance among primary care physicians in following current guidelines for screening.140 In an ancillary study of the Child Adolescent Trial for Cardiovascular Health (CATCH), parents were given recommendations to consult their child's physician if TC exceeded 200 mg/dL on one or more occasions.141 After physicians examined the children, only 59% were further evaluated for possible elevated cholesterol. Of these, half of the physicians repeated cholesterol tests, 42% asked about family history, 38% made recommendations for dietary management, and only 12% referred children to dietitians.141

Neonatal screening for dyslipidemia has been examined in multiple studies of either cord blood testing,54,142-155 dried filter paper blood spots from cord blood,156 or heel sticks of three to seven day old infants.157-162 No studies screened a general population of infants and followed abnormal results with mutation analysis or LDL-C receptor activity assays making it difficult to determine the value of such screening.

Key Question 3. What are the adverse effects of screening (including false positives, false negatives, labeling)?

Potential adverse effects of screening for dyslipidemia among children were examined in one randomized controlled trial163 and five non-comparative studies.76,136-139 Although one small study showed increased parental reporting of behavior difficulties among children with dyslipidemia, these reports were not objectively confirmed.139 No studies reported increased anxiety or depression among screened children or their parents.137,138 139

Key Question 4. In children/adolescents, what is the effectiveness of drug, diet, exercise, and combination therapy in reducing the incidence of adult dyslipidemia, and delaying the onset and reducing the incidence of CHD-related events (including optimal age for initiation of treatment)?

No studies evaluated the effect of a childhood intervention on the incidence of adult dyslipidemia or CHD-related events and outcomes.

Key Questions 5-8. What is the effectiveness of drug, diet, exercise, and combination therapy for treating dyslipidemia in children/adolescents?

Forty RCTs meeting the inclusion criteria addressed the effectiveness of interventions for treatment of dyslipidemia in children and adolescents.18,19,164-201 Statins, bile-acid binding resins, and fibrates have been tested and reported only in children with FH and FCH. Applicability of results from these trials to children without these conditions may be limited. In addition, 18 studies used populations recruited from single lipid clinics.18,165-169,176,178,179,181,182,185,186,189,191,193,196,202 Major limitations of trials include fewer than 20 subjects in each study arm,168,175,178,181,182,185,193,195 high loss to follow-up,177,187,191 failure of blinding,174,191,192,196-198 lack of results presented for the period prior to crossover,166-168,176,178,180-182,185,189,190,192,195,198,199,201 lack of intention to treat analyses,164,166,177-180,182,184,187,189,191-194,196-198 and lack of data reported for the placebo group.179

Studies in children with probable or definite familial hypercholesterolemia

Drug treatment. Eleven trials evaluated drug therapies for treatment of children with probable or definite heterozygous familial hypercholesterolemia (Table 2).18,19,163,167,170,171,177,182,184,185,186 Most of these included children who were already compliant with a recommended low-saturated fat, low-cholesterol diet, and both treatment and control groups were maintained on the diet during the trials.

All the trials of statin drugs,18,19,165,169,172,173,179,184,188 demonstrated improvement in TC and LDL-C among children and adolescents with FH. The decrease in TC compared to baseline ranged from 17-32% for treatment groups vs. changes of +3.6% to -2.3% for placebo groups. The decreases in LDL-C ranged from 19-41% for treatment groups, vs. changes of +0.67% to -3% for placebo groups. Changes in HDL-C and triglycerides were mixed.165,169,172,173,179,184,188

Trials of cholestyramine187 and colestipol186 demonstrated decreased total cholesterol and LDL-C, but no change in HDL-C or triglycerides. Trials evaluating bezafibrate,193 vitamins C and E,182 DHA,199,201 p-aminosalicylic acid,185 combined colestipol and pravastatin vs. colestipol alone166 and powder vs. pill form of cholestyramine174 failed to report pre-crossover data.

Diet treatment. Five trials evaluating diet treatments in children with FH or FCH met inclusion criteria.167,168,178,180,200 Although trials of sterol margarines and psyllium were crossover trials without pre-crossover results presented, the wash-out periods between treatment phases were four to six weeks, suggesting that results may be valid.167,178,180 TC and LDL-C reductions were significant in these trials (reduction of 7.4-11% and 10-14% respectively). There was no significant improvement in lipid levels with eight weeks of garlic extract treatment.200

Exercise treatment. No studies evaluated exercise treatment for lipid lowering in children with FH.

Studies in children with elevated lipids but not meeting criteria for familial hypercholesterolemia

Drug treatment. No studies evaluated drug interventions in children without monogenic dyslipidemia.

Diet treatment. Dietary interventions in general populations of children and adolescents were addressed in seven studies (Table 3).170,171,174,190,191,194,196 A trial conducted by the DISC Collaborative Research Group showed that intensive dietary counseling over three years was effective (8% improvement in LDL-C compared to control),171 but not sustained at five and seven year follow-ups once the intervention ceased.170 A study of the Parent-Child AutoTutorial (PCAT) program174 reported 8% improvement in LDL-C compared to the at-risk control group (p<0.05). One trial of psyllium did not present pre-crossover data.81

Exercise treatment. Six studies 183,197,198,189,192,195 evaluated exercise in normal or obese children with elevated lipids (Table 3). Three studies were limited by differential or low completion rates, small numbers of participants, or other deficiencies (lack of blinding, lack of intention to treat analysis).189,192,195 Four trials comparing supervised, scheduled sessions of aerobic and fitness training to control groups showed minimal or no change in lipids compared to control groups.189,192,197,198 Two trials showed improvements in HDL-C for the exercise intervention group compared to controls.183,195

Combination diet and exercise treatment. Three trials175,177,164 evaluated combined regimens of diet and exercise (Table 3). While all interventions showed some improvement in lipid levels, a group undertaking exercise, diet, and behavior change had a 23% increase in HDL-C, compared to both the diet plus behavior change group and the control group.175

Key Question 9. What are the adverse effects of drug, diet, exercise, and combination therapy in children/adolescents?

Drug treatment. Information about adverse events was reported in 15 studies of statins,18,19,165,169,172,173,179,184,188,203-208 in 22 studies of bile-acid binding resins166,176,186,187,209-227 and in eight studies of various other drugs or drug combinations (Table 4).26,185,193,228-232 Studies used RCT, open-label trial, and observational designs.

Statins were associated with increased ALT and/or AST levels in some,169,188,204,207 but not all, studies.18,165,203,205 Reports of elevated CK levels were similarly conflicting.172,173,184,188,204,205,207,18,165,172,203

Bile-acid binding resins were associated with gastrointestinal complaints (8-26%), such as flatulence and constipation,166,176,185-187,211,214,216,218,223,224,229,230 and unpalatability (up to 50%).212,216-219,222,224 One study of cholestyramine reported transient increases in LDH and abnormalities in AST that persisted for six months,211 but others showed normal liver function tests.224,226,227 Growth was reported normal in nine studies.26,186,187,193,215,220,221,225,227 One study reported a child whose height for age dropped below -2 S.D. while on colestipol (1 S.D. = 2.4 cm),213 while growth was normal in all other children in the study. Sexual maturation was followed over 4.3 years of treatment and found to be normal.225

Two studies of niacin reported increased liver enzymes (6 of 21 children in one study), and multiple other symptoms such as flushing, abdominal pain, nausea, and headache.229,231 There are also case reports of hepatitis229 and hepatotoxicity231 with niacin.

Low-fat diet. Nineteen studies of dietary fat restriction reported effects on growth, nutrient intake, laboratory safety parameters, or other adverse effects.170,171,190,233-248

Twelve studies reported normal height growth,170,190,234-236,239,240,242,243,245-247 although weight loss occurred among three children in two of these studies.235,242 Growth failure in one study occurred among 8 of 40 (20%) children with dyslipidemia, three (7.5%) of whom had nutritional dwarfing and no progression of puberty.241 In this study, families were unsupervised in the implementation of low-fat, low-cholesterol diets for a period up to 4.5 years; those with nutritional dwarfing had longer periods of time between diagnosis and formal dietary assessment and counseling.241 Failure to thrive has been demonstrated in children under age two years eating fat-restricted diets.249 although these diets are not recommended for this age group.16

Dietary supplements. Fourteen studies provided information about adverse effects of various dietary supplements.168,178,181,200,250-259 Two children (4% of the treatment group) reported abdominal discomfort using fiber tablets (containing 50% wheat bran and 50% pectin) administered at 100-150 mg/kg/day.181,253,256 There were no adverse effects with psyllium fiber in two other studies.181,253 Other adverse effects of dietary supplements were mild or transient.22

Exercise. A school-based program examined the effect of supervised exercise training on the lipid profiles of normal prepubertal children and reported 100% adherence and no adverse effects.260 In another study, treadmill tests elicited an exaggerated blood pressure response in boys with dyslipidemia.261

Key Question 10. Does improving dyslipidemia in childhood reduce the risk of dyslipidemia in adulthood?

No studies were identified that directly evaluated whether treatment of idiopathic dyslipidemia in childhood reduces risk of dyslipidemia in adulthood.

Return to Table of Contents
Although many studies have addressed the various aspects of dyslipidemia in children, few key questions about screening have been resolved (Table 5). Studies are not available that address the overarching key question about efficacy of screening children and adolescents for dyslipidemia in delaying the onset and reducing the incidence of CHD-related events (Key Question 1), effectiveness of treatments (drug, diet, exercise and combination) on reducing incidence of adult dyslipidemia or delaying the onset and reducing the risk of CHD-related events (Key Question 4), or whether improving dyslipidemia in children and adolescents reduces the risk of adult dyslipidemia (Key Question 10).

Studies evaluating risk factors are also limited. Risk factors that might contribute to a risk assessment tool have not been adequately studied. Family history questions are not standardized and have limited diagnostic accuracy. Evidence for risk factors other than family history for predicting dyslipidemia in children is strongest for overweight, but the magnitude of the effect of overweight on lipid levels, and the potential impact of incorporating overweight into a screening strategy for dyslipidemia, have not been addressed. Multiple other risk factors such as diet, physical inactivity, and aerobic capacity/fitness have not been evaluated adequately to assess their contribution to dyslipidemia or their usefulness as screening tools either alone or in combination.

Currently recommended screening strategies have low adherence by providers and limited compliance by parents and children. No trials compared strategies by location, venue, age, or provider. No studies addressed the frequency and optimal age for testing. Adverse effects of screening for dyslipidemia have not been adequately studied.

Drug treatments for dyslipidemia in children have been studied only in children with FH or FCH, the population for whom these drugs are FDA-approved and recommended by the NCEP. Statins are effective for reducing TC and LDL-C in children with FH. It is not clear how this efficacy translates to children with milder and/or non-monogenic dyslipidemia, and it is not known how frequently these medications are used in children without FH in practice. There are no trials with long-term follow-up for adult lipid outcomes or CHD-related events. Adverse effects of treatment are reported in controlled and non-controlled studies of drug, diet, exercise, and combination therapy in children and adolescents. Studies were generally not of sufficient duration to determine long-term effects of either short or extended use.

Directions for future research should include identification of the impact of risk factors other than family history, such as overweight and physical inactivity, on lipids in order to develop risk assessment strategies. Such tools may provide a better indication of actual risk, and could facilitate screening by narrowing the number of children requiring serum lipid testing. New vascular markers such as apolipoprotein B and apolipoprotein A-I may prove to be useful for screening in children.263,264 There is a growing literature on non-invasive vascular outcomes such as carotid intima-media thickness (IMT), nitrate dilation, and brachial IMT. Carotid IMT is significantly higher in overweight children, and adult IMT measurements appear to correlate with lipid measurements taken in childhood.83,265-268 Further evaluation of arterial IMT as a risk factor identifiable in children and its usefulness as a screening tool may be warranted.

Randomized controlled clinical trials of screening strategies to determine which are more effective than current practice both in terms of parental compliance and provider adherence to guidelines are important. Screening strategies for ensuring adequate assessment of minorities and those with unknown family history deserve attention. Continued follow-up of currently established cohorts to assess the impact of screening for dyslipidemia in childhood on adult CHD outcomes is important.

More rigorous study designs, enrollment of larger population-based samples, and systematic reporting of adverse effects could improve studies of dyslipidemia treatments. Long-term follow-up of children treated with statins to determine the impact of sustained improvement of lipid levels in childhood on adult lipid levels, adult CHD-outcomes, and long-term safety will help further asses the efficacy and safety of treatment options. Effect of exercise on lipid levels should be evaluated further, particularly in children with lipid levels above the 95th percentile. Standardized methods for collecting and reporting adverse effects in treatment trials would facilitate combining data across trials, and lead to more thorough understanding of the risks of treatment among children and adolescents.

Return to Table of Contents

Author Affiliation

1 Oregon Evidence-based Practice Center, Department of Medical Informatics and Clinical Epidemiology.
2 Oregon Evidence-based Practice Center, Department of Medicine.
3 Departments of Pediatrics and Molecular & Medical Genetics, Oregon Health & Science University, Portland, Oregon.
4 Women and Children's Health Research Center, Providence Health System, Portland, Oregon.

Corresponding Author: Elizabeth Haney, MD, Oregon Health and Science University, Mail Code L-475, 3181 SW Sam Jackson Park Road, Portland Oregon 97239, E-mail: haneye@ohsu.edu.

Copyright and Source Information

This document is in the public domain within the United States.

Requests for linking or to incorporate content in electronic resources should be sent via the USPSTF contact form.

Support: This study was conducted by the Oregon Evidence-based Practice Center under contract to the Agency for Healthcare Research and Quality Contract Number 290-02-0024, Task Order Number #2, Rockville MD.

Source: Haney E, Huffman L, Bougatsos C, Freeman M, Steiner R, Nelson H. Screening and treatment for lipid disorders in children and adolescents: Systematic Evidence Review for the U.S. Preventive Services Task Force. Pediatrics 2007;120(1):e180-e214.

Return to Table of Contents
  1. Katzmarzyk PT, Perusse L, Malina RM, Bergeron J, Despres JP, Bouchard C. Stability of indicators of the metabolic syndrome from childhood and adolescence to young adulthood: the Quebec Family Study. J Clin Epidemiol 2001;54(2):190-195.
  2. Kallio MJ, Salmenpera L, Siimes MA, Perheentupa J, Miettinen TA. Tracking of serum cholesterol and lipoprotein levels from the first year of life. Pediatrics 1993;91(5):949-954.
  3. Bastida S, Sanchez-Muniz FJ, Cuena R, Perea S, Aragones A. High density lipoprotein-cholesterol changes in children with high cholesterol levels at birth. Eur J Pediatr 2002;161(2):94-98.
  4. Porkka KV, Viikari JS, Akerblom HK. Tracking of serum HDL-cholesterol and other lipids in children and adolescents: the Cardiovascular Risk in Young Finns Study. Prev Med 1991;20(6):713-724.
  5. Porkka KV, Viikari JS, Taimela S, Dahl M, Akerblom HK. Tracking and predictiveness of serum lipid and lipoprotein measurements in childhood: a 12-year follow-up. The Cardiovascular Risk in Young Finns study. Am J Epidemiol 1994;140(12):1096-1110.
  6. Fuentes RM, Notkola IL, Shemeikka S, Tuomilehto J, Nissinen A. Tracking of serum total cholesterol during childhood: an 8-year follow-up population-based family study in eastern Finland. Acta Paediatr 2003;92(4):420-424.
  7. Eisenmann JC, Welk GJ, Wickel EE, Blair SN, Aerobics Center Longitudinal S. Stability of variables associated with the metabolic syndrome from adolescence to adulthood: the Aerobics Center Longitudinal Study. Am J Hum Biol 2004;16(6):690-696.
  8. Nicklas TA, von Duvillard SP, Berenson GS. Tracking of serum lipids and lipoproteins from childhood to dyslipidemia in adults: the Bogalusa Heart Study. Int J Sports Med 2002;23(Suppl 1):S39-43.
  9. Orchard TJ, Donahue RP, Kuller LH, Hodge PN, Drash AL. Cholesterol screening in childhood: does it predict adult hypercholesterolemia? The Beaver County experience. J Pediatr 1983;103(5):687-691.
  10. Bell MM, Joseph S. Screening 1140 fifth graders for hypercholesterolemia: family history inadequate to predict results. J Am Board Fam Pract 1990;3(4):259-263.
  11. Davidson DM, Van Camp J, Iftner CA, Landry SM, Bradley BJ, Wong ND. Family history fails to detect the majority of children with high capillary blood total cholesterol. J Sch Health 1991;61(2):75-80.
  12. Diller PM, Huster GA, Leach AD, Laskarzewski PM, Sprecher DL. Definition and application of the discretionary screening indicators according to the National Cholesterol Education Program for Children and Adolescents. J Pediatr 1995;126(3):345-352.
  13. Gagliano NJ, Emans SJ, Woods ER. Cholesterol screening in the adolescent. J Adolesc Health 1993;14(2):104-108.
  14. Griffin TC, Christoffel KK, Binns HJ, McGuire PA. Family history evaluation as a predictive screen for childhood hypercholesterolemia. Pediatric Practice Research Group. Pediatrics 1989;84(2):365-373.
  15. Muhonen LE, Burns TL, Nelson RP, Lauer RM. Coronary risk factors in adolescents related to their knowledge of familial coronary heart disease and hypercholesterolemia: the Muscatine Study. Pediatrics 1994;93(3):444-451.
  16. Primrose ED, Savage JM, Boreham CA, Cran GW, Strain JJ. Cholesterol screening and family history of vascular disease. Arch Dis Child 1994;71(3):239-242.
  17. Resnicow K, Cross D, Lacosse J, Nichols P. Evaluation of a school-site cardiovascular risk factor screening intervention. Prev Med 1993;22(6):838-856.
  18. Sanchez Bayle M, Gonzalez Vergaz A, Garcia Cuartero B, Santos Tapia M, Gonzalez Requejo A. Is a parental history of coronary arterial disease in children as discriminating as their lipoprotein profile? Nino Jesus Group. Int J Cardiol 1992;36(3):267-271.
  19. Steiner NJ, Neinstein LS, Pennbridge J. Hypercholesterolemia in adolescents: effectiveness of screening strategies based on selected risk factors. Pediatrics 1991;88(2):269-275.
  20. Troxler RG, Park MK, Miller MA, Karnavas BA, Lee DH. Predictive value of family history in detecting hypercholesterolemia in predominantly Hispanic adolescents. Tex Med 1991;87(11):75-79.
  21. Dennison BA, Kikuchi DA, Srinivasan SR, Webber LS, Berenson GS. Parental history of cardiovascular disease as an indication for screening for lipoprotein abnormalities in children. J Pediatr 1989;115(2):186-194.
  22. O'Loughlin J, Lauzon B, Paradis G, et al. Usefulness of the American Academy of Pediatrics recommendations for identifying youths with hypercholesterolemia. Pediatrics 2004;113(6):1723-1727.
  23. Wadowski SJ, Karp RJ, Murray-Bachmann R, Senft C. Family history of coronary artery disease and cholesterol: screening children in a disadvantaged inner-city population. Pediatrics 1994;93(1):109-113.
  24. Washington RL, Harrigan L, Day M. Early experience in treating hyperlipidemia in children. Ann N Y Acad Sci 1991;623:434.
  25. Lannon CM, Earp J. Parents' behavior and attitudes toward screening children for high serum cholesterol levels. Pediatrics 1992;89(6 Pt 2):1159-1163.
  26. Boulton TJ. The validity of screening for hypercholesterolaemia at different ages from 2 to 17 years. Aust N Z J Med 1979;9(5):542-546.
  27. Williams RR, Hunt SC, Barlow GK, et al. Prevention of familial cardiovascular disease by screening for family history and lipids in youths. Clin Chem 1992;38(8B Pt 2):1555-1560.
  28. Andersen LB, Wedderkopp N, Hansen HS, Cooper AR, Froberg K. Biological cardiovascular risk factors cluster in Danish children and adolescents: the European Youth Heart Study. Prev Med 2003;37(4):363-367.
  29. Bergstrom E, Hernell O, Persson LA. Endurance running performance in relation to cardiovascular risk indicators in adolescents. Int J Sports Med 1997;18(4):300-307.
  30. Bonora E, Zenere M, Branzi P, et al. Influence of body fat and its regional localization on risk factors for atherosclerosis in young men. Am J Epidemiol 1992;135(11):1271-1278.
  31. Bonora E, Targher G, Branzi P, et al. Cardiovascular risk profile in 38-year and 18-year-old men. Contribution of body fat content and regional fat distribution. Int J Obes Relat Metab Disord 1996;20(1):28-36.
  32. Davis PH, Dawson JD, Riley WA, Lauer RM. Carotid intimal-medial thickness is related to cardiovascular risk factors measured from childhood through middle age: the Muscatine Study. Circulation 2001;104(23):2815-2819.
  33. Demerath E, Muratova V, Spangler E, Li J, Minor VE, Neal WA. School-based obesity screening in rural Appalachia. Prev Med 2003;37(6 Pt 1):553-560.
  34. DeStefano F, Berg RL, Griese GG, Jr. Determinants of serum lipid and lipoprotein concentrations in children. Epidemiology 1995;6(4):446-449.
  35. Douglas MB, Birrer RB, Medidi S, Schlussel YR. Obese children should be screened for hypercholesterolemia. J Health Care Poor Underserved 1996;7(1):24-35.
  36. Durant RH, Linder CW, Jay S, Harkness JW, Gray RG. The influence of a family history of CHD risk factors on serum lipoprotein levels in black children and adolescents. J Adolesc Health Care 1982;3(2):75-81.
  37. Durant RH, Linder CW, Mahoney OM. Relationship between habitual physical activity and serum lipoprotein levels in white male adolescents. J Adolesc Health Care 1983;4(4):235-240.
  38. Durant RH, Baranowski T, Rhodes T, et al. Association among serum lipid and lipoprotein concentrations and physical activity, physical fitness, and body composition in young children. J Pediatr 1993;123(2):185-192.
  39. Dwyer T, Gibbons LE. The Australian Schools Health and Fitness Survey. Physical fitness related to blood pressure but not lipoproteins. Circulation 1994;89(4):1539-1544.
  40. Eisenmann JC, Malina RM. Age-related changes in subcutaneous adipose tissue of adolescent distance runners and association with blood lipoproteins. Ann Hum Biol 2002;29(4):389-397.
  41. Freedman DS, Dietz WH, Srinivasan SR, Berenson GS. The relation of overweight to cardiovascular risk factors among children and adolescents: the Bogalusa Heart Study. Pediatrics 1999;103(6 Pt 1):1175-1182.
  42. Fripp RR, Hodgson JL, Kwiterovich PO, Werner JC, Schuler HG, Whitman V. Aerobic capacity, obesity, and atherosclerotic risk factors in male adolescents. Pediatrics 1985;75(5):813-818.
  43. Giovannini M, Bellu R, Ortisi MT, Incerti P, Riva E. Cholesterol and lipoprotein levels in Milanese children: relation to nutritional and familial factors. J Am Coll Nutr 1992;11(Suppl):28S-31S.
  44. Glassman MS, Schwarz SM. Cholesterol screening in children: should obesity be a risk factor? J Am Coll Nutr 1993;12(3):270-273.
  45. Howard JK, Bindler RM, Dimico GS, et al. Cardiovascular risk factors in children: a Bloomsday research report. J Pediatr Nurs 1991;6(4):222-229.
  46. Jarvisalo MJ, Jartti L, Nanto-Salonen K, et al. Increased Aortic Intima-Media Thickness: A Marker of Preclinical Atherosclerosis in High-Risk Children. Circulation 2001;104(24):2943-2947.
  47. Larsson B, Vaara I. Cholesterol screening of seven-year-old children. How to identify children at risk. Acta Paediatr 1992;81(4):315-318.
  48. 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.
  49. Macek M, Bell D, Rutenfranz J, et al. A comparison of coronary risk factors in groups of trained and untrained adolescents. Eur J Appl Physiol Occup Physiol 1989;58(6):577-582.
  50. Marti B, Vartiainen E. Relation between leisure time exercise and cardiovascular risk factors among 15-year-olds in eastern Finland. J Epidemiol Community Health 1989;43(3):228-233.
  51. Raitakari OT, Taimela S, Porkka KV, Viikari JS. Effect of leisure-time physical activity change on high-density lipoprotein cholesterol in adolescents and young adults. Ann Med 1996;28(3):259-263.
  52. Resnicow K, Futterman R, Vaughan RD. Body mass index as a predictor of systolic blood pressure in a multiracial sample of US schoolchildren. Ethn Dis 1993;3(4):351-361.
  53. Ribeiro JC, Guerra S, Oliveira J, et al. Physical activity and biological risk factors clustering in pediatric population. Prev Med 2004;39(3):596-601.
  54. Shear CL. The relationship between parental history of vascular disease and cardiovascular disease risk factors in children: the Bogalusa Heart Study. Am J Epidemiol 1985;122(5):762-771.
  55. Simon JA, Morrison JA, Similo SL, McMahon RP, Schreiber GB. Correlates of high-density lipoprotein cholesterol in Black girls and White girls: the NHLBI Growth and Health Study. Am J Pub Health 1995;85(12):1698-1702.
  56. Suter E, Hawes MR. Relationship of physical activity, body fat, diet, and blood lipid profile in youths 10-15 yr. Med Sci Sports Exerc 1993;25(6):748-754.
  57. Thorland WG, Gilliam TB. Comparison of serum lipids between habitually high and low active pre-adolescent males. Med Sci Sports Exerc 1981;13(5):316-321.
  58. Tolfrey K, Campbell IG, Jones AM. Selected predictor variables and the lipid-lipoprotein profile of prepubertal girls and boys. Med Sci Sports Exerc 1999;31(11):1550-1557.
  59. Tonstad S, Leren TP, Sivertsen M, Ose L. Determinants of lipid levels among children with heterozygous familial hypercholesterolemia in Norway. Arterioscler Thromb Vasc Biol 1995;15(8):1009-1014.
  60. Twisk JW, Kemper HC, Van Mechelen W, Post GB. Clustering of risk factors for coronary heart disease. the longitudinal relationship with lifestyle. Ann Epidemiol 2001;11(3):157-165.
  61. Twisk JW, Kemper HC, van Mechelen W, Post GB, van Lenthe FJ. Body fatness: longitudinal relationship of body mass index and the sum of skinfolds with other risk factors for coronary heart disease. Int J Obes Relat Metab Disord 1998;22(9):915-922.
  62. Twisk JW, Kemper HC, Mellenbergh GJ, van Mechelen W, Post GB. Relation between the longitudinal development of lipoprotein levels and lifestyle parameters during adolescence and young adulthood. Ann Epidemiol 1996;6(3):246-256.
  63. van Lenthe FJ, van Mechelen W, Kemper HC, Twisk JW. Association of a central pattern of body fat with blood pressure and lipoproteins from adolescence into adulthood. The Amsterdam Growth and Health Study. Am J Epidemiol 1998;147(7):686-693.
  64. van Stiphout WA, Hofman A, de Bruijn AM, Valkenburg HA. Distributions and determinants of total and high-density lipoprotein cholesterol in Dutch children and young adults. Prev Med 1985;14(2):169-180.
  65. Ward SD, Melin JR, Lloyd FP, Norton JA, Jr., Christian JC. Determinants of plasma cholesterol in children—a family study. Am J Clin Nutr 1980;33(1):63-70.
  66. Wong ND, Hei TK, Qaqundah PY, Davidson DM, Bassin SL, Gold KV. Television viewing and pediatric hypercholesterolemia. Pediatrics 1992;90(1 Pt 1):75-79.
  67. Jiang X, Srinivasan SR, Webber LS, Wattigney WA, Berenson GS. Association of fasting insulin level with serum lipid and lipoprotein levels in children, adolescents, and young adults: the Bogalusa Heart Study. Arch Intern Med 1995;155(2):190-196.
  68. Gliksman MD, Lazarus R, Wilson A. Differences in serum lipids in Australian children: is diet responsible? Int J Epidemiol 1993;22(2):247-254.
  69. Kunz F, Pechlaner C, Hortnagl H, Pfister R. The smoker's paradox and the real risk of smoking. Eur J Epidemiol 2005;20(2):161-167.
  70. Berenson GS, Srinivasan S. Cholesterol as a risk factor for early atherosclerosis: the Bogalusa Heart Study. Prog Pediatr Cardiol 2003;17:113-122.
  71. Bachman RP, Schoen EJ, Stembridge A, Jurecki ER, Imagire RS. Compliance with childhood cholesterol screening among members of a prepaid health plan. Am J Dis Child 1993;147(4):382-385.
  72. Bistritzer T, Rosenzweig L, Barr J, et al. Lipid profile with paternal history of coronary heart disease before age 40. Arch Dis Child 1995;73(1):62-65.
  73. Davidson DM, Bradley BJ, Landry SM, Iftner CA, Bramblett SN. School-based blood cholesterol screening. J Pediatr Health Care 1989;3(1):3-8.
  74. Dennison BA, Jenkins PL, Pearson TA. Challenges to implementing the current pediatric cholesterol screening guidelines into practice. Pediatrics 1994;94(3):296-302.
  75. Faigel HC. Screening college students for hypercholesterolemia. J Am Coll Health 1992;40(6):272-275.
  76. Garcia RE, Moodie DS. Routine cholesterol surveillance in childhood. Pediatrics 1989;84(5):751-755.
  77. Hammond J, Chinn S, Richardson H, Rona R. Response to venepuncture for monitoring in primary schools. Arch Dis Child 1994;70(5):367-369.
  78. Heyden S, Schneider KA, Roberts KF. Effectiveness of education-screening on cholesterol levels of students. Ann Nutr Metab 1991;35(2):71-76.
  79. Lansing AM, Barbie RN, Shaheen KA. Regional cholesterol screening: problems and potentials. J Ky Med Assoc 1990;88(4):170-174.
  80. Manchester RA, McDuffie C, Diamond E. Screening for hypercholesterolemia in college students. J Am Coll Health 1989;37(4):149-153.
  81. Polonsky SM, Bellet PS, Sprecher DL. Primary hyperlipidemia in a pediatric population: classification and effect of dietary treatment. Pediatrics 1993;91(1):92-96.
  82. Skovby F, Micic S, Jepsen B, et al. Screening for familial hypercholesterolaemia by measurement of apolipoproteins in capillary blood. Arch Dis Child 1991;66(7):844-847.
  83. Sveger T, Flodmark CE, Nordborg K, Nilsson-Ehle P, Borgfors N. Hereditary dyslipidemias and combined risk factors in children with a family history of premature coronary artery disease. Arch Dis Child 2000;82(4):292-296.
  84. Goff DC, Jr., Donker GA, Ragan JD, Jr., et al. Cholesterol screening in pediatric practice. Pediatrics 1991;88(2):250-258.
  85. 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.
  86. Rosenberg E, Lamping DL, Joseph L, Pless IB, Franco ED. Cholesterol screening of children at high risk: behavioural and psychological effects. CMAJ 1997;156(4):489-496.
  87. Tonstad S. Familial hypercholesterolaemia: a pilot study of parents' and children's concerns. Acta Paediatr 1996;85(11):1307-1313.
  88. Rosenthal SL, Knauer-Black S, Stahl MP, Catalanotto TJ, Sprecher DL. The psychological functioning of children with hypercholesterolemia and their families. A preliminary investigation. Clinical Pediatrics 1993;32(3):135-141.
  89. Sinaiko AR, Prineas RJ. Reduction of cardiovascular disease: what is the role of the pediatrician? Pediatrics 1998;102(5):E61.
  90. Nader PR, Yang M, Luepker RV, et al. Parent and physician response to children's cholesterol values of 200 mg/dL or greater: the Child and Adolescent Trial for Cardiovascular Health Experiment. Pediatrics 1997;99(5):E5.
  91. Bastida S, Perea S, Sanchez-Muniz FJ. Do neonates with high serum cholesterol levels have a different high density lipoprotein composition? Eur J Pediatr 1998;157(1):66-70.
  92. Andersen GE, Lous P, Friis-Hansen B. Screening for hyperlipoproteinemia in 10,000 Danish newborns. Follow-up studies in 522 children with elevated cord serum VLDL-LDL-cholesterol. Acta Paediatr Scand 1979;68(4):541-545.
  93. Kenny D, Ward OC, Mulhern B, Rashied AA. Failure of cord serum cholesterol and betalipoprotein as screening tests for familial hyperlipoproteinaemia. Ir J Med Sci 1984;153(3):100-105.
  94. Cress HR, Shaher RM, Greenberg MH, Laffin R. Lipoproteins in neonates. J Pediatr 1974;84(4):585-587.
  95. Barnes K, Nestel PJ, Pryke ES, Whyte HM. Neonatal plasma lipids. Med J Austr 1972;2(18):1002-1005.
  96. Andersen GE, Brokhattingen K, Lous P. Familial hypobetalipoproteinaemia in 9 children diagnosed as the result of cord blood screening for hypolipoproteinaemia in 10,000 Danish newborns. Arch Dis Child 1979;54(9):691-694.
  97. Goldstein JL, Albers JJ, Schrott HG, Hazzard WR, Bierman EL, Motulsky AG. Plasma lipid levels and coronary heart disease in adult relatives of newborns with normal and elevated cord blood lipids. Am J Hum Genet 1974;26(6):727-735.
  98. Darmady JM, Fosbrooke AS, Lloyd JK. Prospective study of serum cholesterol levels during first year of life. Br Med J 1972;2(815):685-688.
  99. Ahmed SM, Clasen ME, Donnelly JE. Management of dyslipidemia in adults. Am Fam Physician 1998;57(9):2192-2204.
  100. Dorlands Illustrated Medical Dictionary, 29th ed. Philadelphia, PA: W.B. Saunders Company; 2000.
  101. Mortality From Coronary Heart Disease and Acute Myocardial Infarction—United States, 1998. MMWR 2001;50(6):90-93.
  102. Schrott HG, Bucher KA, Clarke WR, Lauer RM. The Muscatine hyperlipidemia family study program. Prog Clin Biol Res 1979;32:619-646.
  103. Bao W, Srinivasan SR, Valdez R, Greenlund KJ, Wattigney WA, Berenson GS. Longitudinal changes in cardiovascular risk from childhood to young adulthood in offspring of parents with coronary artery disease: the Bogalusa Heart Study. JAMA 1997;278(21):1749-1754.
  104. Kwiterovich PO, Jr. Prevention of coronary disease starting in childhood: what risk factors should be identified and treated? Coron Artery Dis 1993;4(7):611-630.
  105. Pathological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Relationship of atherosclerosis [in young men] to serum lipoprotein cholesterol concentrations and smoking. JAMA 1990;264:3018-3024.
  106. Newman WPI, Freedman DS, Voors AW, Gard PD, al. e. Relation of serum lipoprotein levels and systolic bolld pressure to early atherosclerosis. N Engl J Med 1986;315:138-144.
  107. Freedman DS, Newman WP, 3rd, Tracy RE, et al. Black-white differences in aortic fatty streaks in adolescence and early adulthood: the Bogalusa Heart Study. Circulation 1988;77(4):856-864.
  108. Berenson GS, Wattigney WA, Tracy RE, et al. Atherosclerosis of the aorta and coronary arteries and cardiovascular risk factors in persons aged 6 to 30 years and studied at necropsy (The Bogalusa Heart Study). Am J Cardiol 1992;70(9):851-858.
  109. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, 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.
  110. Strong JP, Zieske AW, Malcom GT. Lipoproteins and atherosclerosis in children: an early marriage? Nutr Metab Cardiovasc Dis 2001;11(Suppl 5):16-22.
  111. Eyre H, Kahn R, Robertson RM, et al. Preventing cancer, cardiovascular disease, and diabetes: a common agenda for the American Cancer Society, the American Diabetes Association, and the American Heart Association. Circulation 2004;109(25):3244-3255.
  112. Kavey R-EW, Daniels SR, Lauer RM, Atkins DL, Hayman LL, Taubert K. American heart association guidelines for primary prevention of atherosclerotic cardiovascular disease beginning in childhood. J Pediatr 2003;142(4):368-372.
  113. Williams CL, Hayman LL, Daniels SR, et al. Cardiovascular health in childhood: A statement for health professionals from the Committee on Atherosclerosis, Hypertension, and Obesity in the Young (AHOY) of the Council on Cardiovascular Disease in the Young, American Heart Association. Circulation 2002;106(1):143-160.
  114. American Academy of Pediatrics. Committee on Nutrition. Cholesterol in childhood. Pediatrics 1998;101(1 Pt 1):141-147.
  115. American Heart Association. Step I, Step II, and TLC Diets. Available at: http://www.americanheart.org/presenter.jhtml?identifier=4764. Accessed April 29, 2005.
  116. 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.
  117. 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.
  118. National Cholesterol Education Program (NCEP). Report of the expert panel on blood cholesterol levels in children and adolescents. Pediatrics 1992;89(3 Pt 2):525-584.
  119. Harris RP, Helfand M, Woolf SH, et al. Current methods of the U.S. Preventive Services Task Force. A review of the process. Am J Prev Med 2001;30(3S):21-35.
  120. Haney EM, Huffman LH, Bougatsos CM, et al. Screening for dyslipidemia in children and adolescents: Systematic evidence synthesis. Rockville, MD: Agency for Healthcare Research and Quality; 2006 (in press).
  121. National Heart Lung and Blood Institute. Cardiovascular profile of 15,000 children of school age in three communitites 1971-1975. Bethesda, MD: U.S. Department of Health, Education, and Welfare, Public Health Service, National Institutes of Health; 1978.
  122. Berenson GS. Cardiovascular risk factors in children. New York: Oxford University Press; 1980.
  123. National Center for Health Statistics. Serum cholesterol levels of persons 4-74 years of age by socioeconomic characteristics: United States, 1971-74. Hyattsville, MD: U.S. Department of Health Education and Welfare, Public Health Service, National Center for Health Statistics; 1980.
  124. Baker SG, Joffe BI, Mendelsohn D, Seftel HC. Treatment of homozygous familial hypercholesterolaemia with probucol. S Afr Med J 1982;62(1):7-11.
  125. The Lipid Research Clinics Population Studies Data Book. Vol 1. Bethesda, MD: National Institutes of Health; Lipid Metabolism Branch; Division of Heart and Vascular Diseases; National Heart, Lung, and Blood Institute; 1980.
  126. Hickman TB, Briefel RR, Carrol MD, et al. Distributions and trends of serum lipid levels among United States children and adolescents ages 4-19 years: data from the third National Health and Nutrition Examination Survey. Prev Med 1998;27:879-890.
  127. Labarthe DR, Dai S, Fulton JE. Cholesterol screening in children: insights from Project HeartBeat! and NHANES III. Prog Pediatr Cardiol 2003;17:169-178.
  128. Kwiterovich PO, Jr., Heiss G, Johnson N, Chase GA, Tamir I, Rifkind B. Assessment of plasma total cholesterol as a test to detect elevated low density (beta) lipoprotein cholesterol levels (type IIa hyperlipoproteinemia) in young subjects from a population-based sample. Am J Epidemiol 1982;115(2):192-204.
  129. Dennison BA, Kikuchi DA, Srinivasan SR, Webber LS, Berenson GS. Serum total cholesterol screening for the detection of elevated low-density lipoprotein in children and adolescents: the Bogalusa Heart Study. Pediatrics 1990;85(4):472-479.
  130. E. Neufeld, M.D., Ph.D., Personal communication regarding screening tests for children, 2005.
  131. Rifai N, Neufeld E, Ahlstrom P, Rimm E, D'Angelo L, Hicks JM. Failure of current guidelines for cholesterol screening in urban African-American adolescents. Pediatrics 1996;98(3 Pt 1):383-388.
  132. Shea S, Basch CE, Irigoyen M, et al. Failure of family history to predict high blood cholesterol among hispanic preschool children. Prev Med 1990;19(4):443-455.
  133. Gillman MW, Cupples LA, Moore LL, Ellison RC. Impact of within-person variability on identifying children with hypercholesterolemia: Framingham Children's Study. J Pediatr 1992;121(3):342-347.
  134. Harris N, Neufeld EJ, Newburger JW, et al. Analytical performance and clinical utility of a direct LDL-cholesterol assay in a hyperlipidemic pediatric population. Clin Chem 1996;42(8 Pt 1):1182-1188.
  135. Ticho BS, Neufeld EJ, Newburger JW, Harris N, Baker A, Rifai N. Utility of direct measurement of low-density lipoprotein cholesterol in dyslipidemic pediatric patients. Arch Pediatr Adol Med 1998;152(8):787-791.
  136. Bao W, Srinivasan SR, Wattigney WA, Berenson GS. Usefulness of childhood low-density lipoprotein cholesterol level in predicting adult dyslipidemia and other cardiovascular risks. The Bogalusa Heart Study. Arch Intern Med 1996;156(12):1315-1320.
  137. Freedman DS, Shear CL, Srinivasan SR, Webber LS, Berenson GS. Tracking of serum lipids and lipoproteins in children over an 8-year period: the Bogalusa Heart Study. Prev Med 1985;14(2):203-216.
  138. Berenson GS, Srinivasan SR, Frerichs RR, Webber LS. Serum high density lipoprotein and its relationship to cardiovascular disease risk factor variables in children—the Bogalusa heart study. Lipids 1979;14(1):91-98.
  139. 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.
  140. Clarke WR, H.G. S, Leaverton PE, Conner WE, Lauer RM. Tracking of blood lipids and blood pressures in school age children: the Muscatine study. Circulation 1978;58(4):626-634.
  141. Lauer RM, Lee J, Clarke WR. Predicting adult cholesterol levels from measurements in childhood and adolescence: the Muscatine Study. Bull N Y Acad Med 1989;65(10):1127-1142.
  142. Stuhldreher WL, Orchard TJ, Donahue RP, Kuller LH, Gloninger MF, Drash AL. Cholesterol screening in childhood: sixteen-year Beaver County Lipid Study experience. J Pediatr 1991;119(4):551-556.
  143. Namboodiri KK, Green PP, Walden C, et al. The Collaborative Lipid Research Clinics Program Family Study. II. Response rates, representativeness of the sample, and stability of lipid and lipoprotein levels. Am J Epidemiol 1984;119(6):944-958.
  144. Laskarzewski P, Morrison JA, de Groot I, et al. Lipid and lipoprotein tracking in 108 children over a four year period. Pediatrics 1979;64(5):584-591.
  145. Guo S, Beckett L, Chumlea WC, Roche AF, Siervogel RM. Serial analysis of plasma lipids and lipoproteins from individuals 9-21 y of age. Am J Clin Nutr 1993;58(1):61-67.
  146. Baumgartner RN, Guo S, Roche AF. Tracking of lipids and lipoproteins in adolescents from 12-22 years of age. The Fels longitudinal study. Ann N Y Acad Sci 1991;623:406-409.
  147. Twisk JW, Kemper HC, van Mechelen W, Post GB. Tracking of risk factors for coronary heart disease over a 14-year period: a comparison between lifestyle and biologic risk factors with data from the Amsterdam Growth and Health Study. Am J Epidemiol 1997;145(10):888-898.
  148. Mohler B, Ackermann-Liebrich U, Steffen T, Staehelin HB. Cholesterol screening in childhood: results of a 9-year follow-up study in Swiss and Italian children in Switzerland. Soz Praventivmed 1996;41(6):333-340.
  149. Vobecky JS, David P, Vobecky J. Dietary habits in relation to tracking of cholesterol level in young adolescents: a nine-year follow-up. Ann Nutr Metab 1988;32(5-6):312-323.
Return to Table of Contents

Diagnostic Accuracy Studies1

Criteria:

  • Screening test relevant, available for primary care, adequately described.
  • Study uses a credible reference standard, performed regardless of test results.
  • Reference standard interpreted independently of screening test.
  • Handles indeterminate results in a reasonable manner.
  • Spectrum of patients included in study.
  • Sample size.
  • Administration of reliable screening test.

Definition of ratings based on above criteria:

Good: Evaluates relevant available screening test; uses a credible reference standard; interprets reference standard independently of screening test; reliability of test assessed; has few or handles indeterminate results in a reasonable manner; includes large number (more than 100) broad-spectrum patients with and without disease.
Fair: Evaluates relevant available screening test; uses reasonable although not best standard; interprets reference standard independent of screening test; moderate sample size (50 to 100 subjects) and a “medium” spectrum of patients.
Poor: Has important limitation such as: uses inappropriate reference standard; screening test improperly administered; biased ascertainment of reference standard; very small sample size of very narrow selected spectrum of patients.

Randomized Controlled Trials (RCTs) and Cohort Studies

Criteria:

  • Initial assembly of comparable groups: RCTs—adequate randomization, including concealment and whether potential confounders were distributed equally among groups; cohort studies—consideration of potential confounders with either restriction or measurement for adjustment in the analysis; consideration of inception cohorts.
  • Maintenance of comparable groups (includes attrition, cross-overs, adherence, contamination).
  • Important differential loss to followup or overall high loss to followup.
  • Measurements: equal, reliable, and valid (includes masking of outcome assessment).
  • Clear definition of interventions.
  • Important outcomes considered.
  • Analysis: adjustment for potential confounders for cohort studies, or intention-to-treat analysis for RCTs.

Definition of ratings based on above criteria:

Good: Meets all criteria: Comparable groups are assembled initially and maintained throughout the study (followup at least 80 percent); reliable and valid measurement instruments are used and applied equally to the groups; interventions are spelled out clearly; important outcomes are considered; and appropriate attention to confounders in analysis.
Fair: Studies will be graded “fair” if any or all of the following problems occur, without the important limitations noted in the “poor” category below: Generally comparable groups are assembled initially but some question remains whether some (although not major) differences occurred in follow-up; measurement instruments are acceptable (although not the best) and generally applied equally; some but not all important outcomes are considered; and some but not all potential confounders are accounted for.
Poor: Studies will be graded “poor” if any of the following major limitations exists: Groups assembled initially are not close to being comparable or maintained throughout the study; unreliable or invalid measurement instruments are used or not applied at all equally among groups (including not masking outcome assessment); and key confounders are given little or no attention.

Case Control Studies

Criteria:

  • Accurate ascertainment of cases.
  • Nonbiased selection of cases/controls with exclusion criteria applied equally to both.
  • Response rate.
  • Diagnostic testing procedures applied equally to each group.
  • Measurement of exposure accurate and applied equally to each group.
  • Appropriate attention to potential confounding variable.

Definition of ratings based on criteria above:

Good: Appropriate ascertainment of cases and nonbiased selection of case and control participants; exclusion criteria applied equally to cases and controls; response rate equal to or greater than 80 percent; diagnostic procedures and measurements accurate and applied equally to cases and controls; and appropriate attention to confounding variables.
Fair: Recent, relevant, without major apparent selection or diagnostic work-up bias but with response rate less than 80 percent or attention to some but not all important confounding variables.
Poor: Major selection or diagnostic work-up biases, response rates less than 50 percent, or inattention to confounding variables.

Reference

1. Harris RP, Helfand M, Woolf SH, Lohr KN, Mulrow CD, Teutsch SM et al. Current methods of the US Preventive Services Task Force: a review of the process. Am J Prev Med 2001;20(3 Suppl):21-35.

Return to Table of Contents

A large oval labeled 'Phenotypic Dyslipidemia' contains three slightly overlapping circles.  The central circle is shaded gray and is labeled 'Screening Population. Idiopathic Dyslipidemia: Polygenic, risk factor associated, multifactorial.' The circle to the left is labeled 'Secondary Dyslipidemia: Secondary to other diagnosed conditions (e.g., diabetes, nephrotic syndrome, organ transplant).' The section where these two circles overlap is labeled 'Undiagnosed secondary dyslipidemia.' The circle to the right is labeled 'Monogenic Dyslipidemia: Diagnosed monogenic syndromes (e.g., familial hypercholesterolemia, familial defective ApoB).' The section where this circle overlaps the central circle is labeled 'Undiagnosed monogenic dyslipidemia.

Children and adolescents identified by screening include those with undiagnosed monogenic dyslipidemia, undiagnosed secondary dyslipidemia, and idiopathic (polygenic or risk factor driven) dyslipidemia. Children and adolescents with previously known monogenic or secondary dyslipidemia would be specifically evaluated for these indications and are not included in the screening pool for the general population.

Return to Table of Contents

Text Description

This figure displays graphically the logical progression of the systematic evidence review on Screening for Lipid Disorders in Children and Adolescents. A series of arrows represents key questions which, if answered with evidence from systematic reviews, may form an indirect chain of evidence linking screening for lipid disorders with improved health outcomes.

The figure proceeds from left to right, with two downward side-tracks for key questions 3 and 9, which relate to the adverse effects of screening and detection of the disease, respectively.

The key questions are listed at the bottom of the figure. Key question 2 includes 6 individual questions, numbered 2a through 2f.

Key question 1 is indicated by the topmost arrow. It relates the over-arching question: "Is screening for dyslipidemia in children/adolescents effective in delaying the onset and reducing the incidence of coronary heart disease (CHD) events. "Risk assessment and testing," indicated by a bold heading on the upper left of the figure, is linked to the ultimate desired outcome, "delayed onset and reduced incidence of CHD-related events," indicated in the rectangle on the extreme right side of the figure.

Key question 2 is indicated by an arrow connecting the population under investigation, "children and adolescents in the general population," to the information to be gained by answering the question, indicated in the rectangular lozenge-shaped box labeled "detection of children and adolescents with dyslipidemia." The sub-questions 2a through 2f are not displayed graphically. (Dyslipidemia is a technical term for "lipid disorders.")

Key question 3 is indicated by the downward-curving arrow linked to the oval labeled "adverse effects."

Key question 4 is indicated by an arrow just under the arrow for key question 1, in the right-hand half of the figure. It connects the section of the figure labeled by the bold header "treatment," consisting of a list of treatments: diet, exercise, drug [treatments], and combination [treatments], to a lozenge-shaped box labeled "reduced incidence of adult dyslipidemia" and to the rectangle at the extreme right side of the figure indicating the ultimate desired outcome of treatment, "delayed onset and reduced incidence of CHD-related events."

An arrow connects the lozenge "detection of children and adolescents with dyslipidemia" to a series of branching arrows for key questions 5, 6, 7, and 8. These key questions attempt to relate each specific treatment (diet, exercise, drug, and combination) to an intermediate outcome in children—reduced total cholesterol. This intermediate outcome is displayed in a lozenge-shaped box containing total cholesterol and its 4 sub-elements, with short arrows to the left of each element and sub-element to indicate the direction of desirable change. Only one arrow, the bottom one, points upward; this is because an increase in HDL-C levels is protective against CHD events.

Key question 9 is indicated by a downward-curving arrow beginning before the arrow on treatment branches off into key questions 5 through 8, and connecting with a second oval labeled "adverse effects." It refers to the adverse effects of the treatments, taken together, which are investigated individually key questions 5 through 8.

Key question 10 is indicated by an arrow connecting the lozenge containing the intermediate outcomes of treatment in children with a lozenge containing a desired outcome in adults, "reduced incidence of adult dyslipidemia." This lozenge includes the 5 elements of reduced total cholesterol and its sub-elements, with arrows indicating the direction of desired change, as in the lozenge to the left describing reduced total cholesterol in children.

A broken arrow connects this last lozenge with the rectangle on the extreme right side of the figure. This indicates either that there is an already-established evidence-based link between the intermediate outcomes of reduced cholesterol in adults and the ultimate desired outcome, "delayed onset and reduced incidence of CHD-related events," or that no key question has been framed which can establish this connection.

Key Questions

  1. Is screening for dyslipidemia in children/adolescents effective in delaying the onset and reducing the incidence of CHD-related events?
  2. What is the accuracy of screening for dyslipidemia in identifying children/adolescents at increased risk of CHD-related events?
    2a. What are abnormal lipid values in children/adolescents?
    2b. What are appropriate tests? How well do screening tests (non-fasting total cholesterol, fasting total cholesterol, fasting lipoprotein analysis) identify individuals with dyslipidemia?
    2c. How well do lipid levels track from childhood to adulthood?
    2d. What is the accuracy of family history in determining risk?
    2e. What are other important risk factors?
    2f. What are effective screening strategies for children/adolescents (including frequency of testing, optimal age for testing)?
  3. What are the adverse effects of screening (including false positives, false negatives, labeling)?
  4. In children/adolescents, what is the effectiveness of drug, diet, exercise, and combination therapy in reducing the incidence of adult dyslipidemia and delaying the onset and reducing the incidence of CHD-related events (including optimal age for initiation of treatment)?
  5. 6, 7, 8. What is the effectiveness of drug, diet, exercise, and combination therapy for treating dyslipidemia in children/adolescents?
  6. What are the adverse effects of drug, diet, exercise, and combination therapy in children/adolescents?
  7. Does improving dyslipidemia in childhood reduce the risk of dyslipidemia in adulthood?

The analytic framework represents an outline of the systematic evidence review and includes patient populations, risk assessment and testing, treatment, and outcomes. The key questions examine a chain of evidence about the accuracy, effectiveness, feasibility of screening asymptomatic children for dyslipidemia in primary care settings, adverse effects of screening, risk factors, effectiveness of interventions, and adverse effects of interventions.

* Includes those without previously known conditions that cause dyslipidemia such as genetic dyslipidemia, diabetes, nephrotic syndrome, organ transplant, and others.

Return to Table of Contents
Study, year Population -
N, age
Method Thresholda Sensitivity Specificity Number eligible for screening (based on population of 1,000)b Number missed (based on population of 1,000)b Comments
Bell, 199061 1,140 5th graders Family history of high cholesterol or MI < age 60 in parent or grandparent Non-fasting TC > 200 mg/dL 64% 47% 540 46  
1,140 5th graders As above, plus family history of stroke, angina or hypertension Non-fasting TC > 200 mg/dL 77% 24% 760 31  
Davidson, 199162 1,118 4th graders Family history from parents (regarding parents, siblings, grandparents, aunts, uncles); early MI defined as < age 56 for men and women TC > 200 mg/dL 41% 68% 330 83 RR of 48%
1,118 4th graders Parental questionnaire, definition using AAP criteria for early CHD (< age 50 for men, < age 60 for women) TC > 200 mg/dL 31% 66% 330 96  
Dennison, 198972 1,214, ages 4-10, Bogalusa Heart Study Parental questionnaire asking parental history of any vascular disease (CHD, HTN, diabetes, stroke) Fasting TC ≥ 95th percentile 38% for W; 27% for AA 73% for W; 65% for AA N/A N/A  
2,099, ages 11-17, Bogalusa Heart Study Parental questionnaire asking parental history of any vascular disease (CHD, HTN, diabetes, stroke) Fasting TC ≥ 95th percentile 59% for W; 25% for AA 67% for W; 56% for AA N/A N/A  
1,214,ages 4-10, Bogalusa Heart Study Parental questionnaire asking parental history of any vascular disease (CHD, HTN, diabetes, stroke) Fasting LDL ≥ 95th percentile 41% for W; 20% for AA 73% for W; 63% for AA N/A N/A  
2,099, ages 11-17, Bogalusa Heart Study Parental questionnaire asking parental history of any vascular disease (CHD, HTN, diabetes, stroke) Fasting LDL > 95th percentile 37% for W; 22% for AA 67% for W; 56% for AA N/A N/A  
Diller, 199563 232, ages 2-19, Cincinnati MI Hormone Study Parental questionnaire using NCEP definition of family history of premature CVD LDL ≥ 130 mg/dL 17% 75% 246 207  
232, ages 2-19, Cincinnati MI Hormone Study Parental questionnaire asking family history of cholesterol > 240 LDL ≥ 130 mg/dL 61% 74% 293 99  
232, ages 2-19, Cincinnati MI Hormone Study Both family history of elevated cholesterol and premature CVD LDL ≥ 130 mg/dL 74% 55% 478 65  
232, ages 2-19, Cincinnati MI Hormone Study Other indicators: obesity, smoking, use of lipid raising medications, high fat diet, HTN LDL ≥ 130 mg/dL 17.4% for obesity, 9-48% for others 86% for obesity, 69-95% for others 547 86  
232, ages 2-19, Cincinnati MI Hormone Study Family history of premature CHD (NCEP definition), TC > 240 mg/dL, or any other risk factor (obesity, smoking, lipid raising medication, high fat diet or HTN). LDL ≥ 130 mg/dL 96% 28% 746 13  
Gagliano, 199364 224, ages 11-20 Family history of early MI (< age 50 for men, < age 60 for women) or elevated lipids (TC > 200 mg/dL), history obtained from adolescent TC > 85th percentile for gender 36% 69% 320 94  
224, ages 11-20 Family history as above, history obtained from parent TC above the 85th percentile for gender 65% 46% 589 54  
224, ages 11-20 Use of combined family history from adolescent and parent TC above the 85th percentile for gender 45% 69% 361 80  
Griffin, 198965 1,005, ages 2-13, 8 office practices Parent and grandparent history of hypercholesterolemia or CHD < age 55 Fasting LDL > 95th percentile 46% NR N/A 147  
1,005, ages 2-13, 8 office practices Parent and grandparent history of any risk factor or complication (hypercholesterolemia, diabetes, HTN, gout, obesity and atherosclerosis prior to age 55) Fasting LDL > 95th percentile 78%
 
NR N/A 59  
1,005, ages 2-13, 8 office practices Parent and grandparent history of hypercholesterolemia or CHD < age 55 Fasting LDL > 90th percentile 51% 63% 385 48 51% of children with elevated LDL would have been detected by AAP screening criteria.; specificity calculated.
1,005, ages 2-13, 8 office practices Any history of parent or grandparent with a risk factor or complication (hypercholesterolemia, diabetes, HTN, gout, obesity and atherosclerosis prior to age 55) Fasting LDL > 90th percentile 80%
38% for high cholesterol alone
31% for obesity
18% for sudden death
17% for gout
13% for PVD
37% 650 20 specificity calculated
1,005, ages 2-13, 8 office practices Overweight (weight for height >> 95th percentile) plus family history of early CHD or hypercholesterolemia Fasting LDL > 90th percentile 57% NR N/A 42  
1,005, ages 2-13, 8 office practices Overweight (weight for height > 95th percentile) plus family history any risk factor or complication Fasting LDL > 90th percentile 84% 31.0% 704 16 specificity calculated
Muhonen, 199466 599, ages 14-20, Muscatine, IA Parental history of high cholesterol Highest decile of fasting TC 34% 76% N/A N/A "unsure" responses were not included in calculations of sensitivity/specificity
599, ages 14-20, Muscatine, IA Parental history of high cholesterol Highest decile of fasting LDL 34% 76% N/A N/A  
599, ages 14-20, Muscatine, IA Parental history of high cholesterol Lowest decile of fasting HDL 26% 75% N/A N/A  
O'Loughlin, 200473 2,217, ages 9, 13, and 16, Quebec Parental questionnaire asking personal history of 1) high cholesterol 2) medications for cholesterol 3) heart attack, angina, 4) stroke, CVD or PVD or 5) medications for the heart; unknown family history coded as negative Fasting LDL ≥ 109 mg/dL ("borderline") 33% 76% 256 44  
2,217, ages 9, 13, and 16, Quebec Parental questionnaire asking personal history of 1) high cholesterol 2) medications for cholesterol 3) heart attack, angina, 4) stroke, CVD or PVD or 5) medications for the heart; unknown family history coded as negative Fasting LDL ≥ 131.5 mg/dL, ("high") 41% 75% 256 12  
2,217, ages 9, 13, and 16, Quebec Parental questionnaire asking personal history of 1) high cholesterol 2) medications for cholesterol 3) heart attack, angina, 4) stroke, CVD or PVD or 5) medications for the heart; unknown family history excluded Fasting LDL ≥ 109 mg/dL, ("borderline") 42% 70% N/A 85  
2,217, ages 9, 13, and 16, Quebec Parental questionnaire asking personal history of 1) high cholesterol 2) medications for cholesterol 3) heart attack, angina, 4) stroke, CVD or PVD or 5) medications for the heart; unknown family history excluded Fasting LDL ≥ 131.5 mg/dL, ("high") 51% 69% N/A 19  
Primrose, 199467 1,012, ages 12-15, Ireland History of stroke, angina or heart attack in either parent at any age or in 1st degree grandparents, uncles or aunts < age 55. Questionnaires completed by parents Non-fasting TC > 95th percentile according to LRC 33% 72% 293 125  
Resnicow, 199368 574, elementary school age Parental cholesterol ≥ 240 in 1 parent only with known and reported value by that parent Non-fasting TC > 200 mg/dL 10% 91% 90 106  
Rifai, 199633 260, ages 12-20, AA Family history of early CHD or hyperlipidemia Fasting LDL > 110 mg/dL 10% NR 365 184 37% targeted for screening based on AAP/NCEP recommendations
Sanchez Bayle, 199269 2,224, ages 2-18, Spain Parental history of MI Fasting TC > 200 mg/dL 7% 96% 49 140  
2,224, ages 2-18, Spain Parental history of MI Fasting LDL > 135 mg/dL 9% 96% 49 101  
2,224, ages 2-18, Spain Parental history of stroke, HTN, diabetes, or hypercholesterolemia (but not MI) Fasting TC > 200 mg/dL 14% 90% 98 129  
2,224, ages 2-18, Spain Parental history of stroke, HTN, diabetes, or hypercholesterolemia (but not MI) Fasting LDL > 135 mg/dL 14% 91% 98 95  
Shea, 199034 108, ages 4-5, Hispanic, Study of Childhood Activity & Nutrition AAP definition (maternal hypertension, diabetes, obesity, hyperlipidemia or family history of premature CHD or hyperlipidemia) Fasting TC > 170 mg/dL 57% 59% 493 148 accuracy 58%
108, ages 4-5, Hispanic, Study of Childhood Activity & Nutrition AHA and NIH Consensus Conference definition (history of hyperlipidemia or premature CHD in the child's parent, aunt, uncle or grandparent) Fasting TC > 170 mg/dL 46% 70% 352 185 accuracy 62%
108, ages 4-5, Hispanic, Study of Childhood Activity & Nutrition NCEP guidelines (history of MI or sudden death in the child's parent, aunt, uncle, or grandparent; CHD prior to age 55). Fasting TC > 170 mg/dL 5% 92% 74 324 accuracy 62%
Steiner, 199170 1,001, ages 12-21 (38% Hispanic, 33.5% W, 15% AA, 11% Asian), Kaiser population AAP 1998 criteria (known hyperlipidemia in parent or sibling, known MI/angina, current corticosteroid use, juvenile diabetes, hypothyroidism, renal/endocrine/hepatic disease in teenager) Non-fasting TC ≥ 200 mg/dL,
repeated fasting TC if initial test ≥ 200 mg/dL, repeated a 3rd time if more than 30 mg/dL variability between the 1st two measurements
63% 60% 400 24 1/3 of 1/2 of teenagers with elevated cholesterol levels would have been missed by family history target screening alone
Troxler, 199171 110 mostly Hispanic senior high school students Questionnaires completed with parental assistance; family history in parents or grandparents of high cholesterol or CHD age < 55 (AAP) Fasting TC > 75th percentile (175 mg/dL) 38% 79% 218 245  
Wadowski, 199474 300 AA, ages 2-14 Family history of CHD in parent or grandparent at age < 55 Fasting TC > 215 mg/dL 59% 72% 327 23  

Notes:
a If not explicitly stated, values are mixed non-fasting/fasting or not reported.
b Number eligible for screening and number missed were calculated from available data. In some cases, reported data did not allow for these calculations (these indicated with N/A).

Abbreviations:
AA = African American, AAP = American Academy of Pediatrics, AHA = American Heart Association, CHD = Coronary heart disease, CVD = Cardiovascular disease, HTN = Hypertension, LDL = Low-density lipoprotein, LRC = Lipid Research Clinic, MI = Myocardial infarction, N/A = Not applicable, NCEP = National Cholesterol Education Program, NIH = National Institutes of Health, PVD = Peripheral vascular disease, TC = Total cholesterol, W = White.

Return to Table of Contents
Author, year Drug Population -
N, age
Duration of trial Significant changes vs. control Quality rating
TC HDL LDL TG
Statins
Clauss, 200519 Lovastatin 20 mg/d vs. 40 mg/d vs. placebo 54 girls, 11-18 y 24 wk a O;c atd> O;c Good
Couture, 1998179 Simvastatin 20 mg/d vs. placebo 63, 8-17 y 6 wk a b a a Fair
de Jongh, 2002165 Simvastatin 10 mg/d, doubled every 8 wk up to 40 mg/d vs. placebo 50, 9-18 y 28 wk a NR a a Poor
de Jongh, 2002188 Simvastatin 10 mg/d titrating up to 40 mg/d vs. placebo 173, 10-17 y 48 wk a NR a NR Good
Knipscheer, 1996173 Pravastatin in 3 active drug groups: 5, 10, or 20 mg/d vs. placebo 72, 11-17 y 12 wk a O;c a O;c Good
Lambert, 1996184 Lovastatin at 10, 20, 30, or 40 mg/d. 4 active drug groups, no placebo) 69 boys, ≤ 17 y 8 wk a b a NR Fair
McCrindle, 2003169 Atorvastatin 10 mg/d vs. placebo 187, 10-17 y 26 wk a b a a Good
Stein, 1999172 Lovastatin starting at 10mg/d, titrating to 40 mg/d vs. placebo 132 boys, 10-17 y 48 wk a O;c a O;c Good
Wiegman, 200418 Pravastatin 40 mg/d vs. placebo 214, 8-18 y 2 y a O;c a NR Good
Bile-acid Resins
Tonstad, 1996186 Colestipol 10 gm/d or 5 gm twice daily vs. placebo 66 adolescents, NR 8 wk a O;c a O;c Poor
Tonstad, 1996187 Cholestyramine titrating up from 4 gm/d to 8 gm/d vs. placebo 96 boys, 6-11 y 1 y a O;c a O;c Fair

Notes:
a ↓ = significant decrease.
b ↑ = significant increase.
c 0 = no significant change.

Abbreviations:
D = Day(s), HDL = High-density lipoprotein, LDL = Low-density lipoprotein, NR = not reported, TC = Total cholesterol, TG = Triglycerides, Wk = Week(s), Y= Year(s).

Return to Table of Contents
Author, year Intervention(s) Population - N, age/description Duration of trial Significant changes vs. control Quality rating
TC HDL LDL TG
Diet
DISC Collaborative Research Group, 1995]]171]] Family oriented behavioral intervention to promote dietary adherence vs. usual care 663, 8-10 y 3 y a a
y 1 only
a Oc Good
Gold, 1991196 Oat bran supplemented cereal within AHA Step 1 diet vs. cereal within Step 1 diet and no oat bran 49, 10 y (mean) with TC > 185 mg/dL 4 wk NR Oc Oc Oc Poor
Kuehl, 1993190 4 90-minute family-oriented nutrition sessions vs. 1 90-minute session 295, 2-15 y with TC > 185 16 wk Oc Oc Oc Oc Poor
Obarzanek, 2001170 Counseling intervention (same as DISC above) vs. usual care 663, 8-10 y 4 y (7 y total follow-up) Oc
5 & 7 y
Oc
5 & 7 y
Oc
5 & 7 y
Oc
5 & 7 y
Good
Shannon, 1994174 Parent-Child Auto Tutorial Program (PCAT): 10 talking book lessons and follow-up paper and pencil games for children with a manual for parents, vs. 45-60 minute counseling session with parent, child and registered dietitian, and take home print materials for both 261, 4-10 y with elevated LDL 3 mo follow-up NR NR a NR Good
Stallings, 1993191 Parent-Child Auto Tutorial Program (PCAT): 10 sessions total, 1 per week completed in home by child and parents vs. usual care 44, 4-10 y with LDL 90-99th percentile 6 mo NR NR Oc NR Poor
Williams, 1995194 Fiber cereal with 3.2 grams soluble fiber per serving. Dose=1 box of cereal/d for 3 wk, then 2 boxes/d. Children ages 2-5 consumed only 1 box/d throughout study. Compared to placebo cereal with 0.5 grams fiber 58, 2-11 y with TC > 170 mg/dL and LDL > 110mg/dL 12 wk a Oc a Oc Poor
Exercise
Boreham, 2000195 7 wk stair climbing program vs. no change in activity 25 sedentary females, 18-22 y 7 wk Oc bd NR NR Poor
Ferguson, 1999183 Exercise program 5 d/wk, 40 minutes/d; children were paid $1/session and given prizes for maintaining a heart rate > 150 beats per minute vs. no exercise program 81 obese children mean 9.5 y 4 mo Oc b Oc a Fair
Kang, 2002189 Physical activity training with lifestyle intervention 5 d/wk vs. lifestyle intervention alone 80 obese children, 13-16 y 8 mo Oc Oc Oc a Poor
Linder, 1983197 Physical conditioning program (PA) vs. usual activities 50 healthy boys, 11-17 y 8 wk Oc Oc Oc Oc Fair
Savage, 1986198 Walking/jogging/running 3 d/wk (1.6 km/session) high intensity (HR=75% of VO2max) vs. low intensity (HR=40% of VO2max). 663 boys, mean 8-9 y 11 wk NR NR Oc Oc Fair
Stergioulas, 1998192 Four 60 minute sessions/wk vs. no specific training program 58 sedentary boys, 10-14 y 2 mo NR Oc NR NR Poor
Diet and Exercise
Becque, 1988175 1. Diet and behavior change: met with dietician and behavior therapist 1 d/wk
2. Exercise plus diet and behavior change: same as above, with exercise program 50 minutes for 3 d/wk
3. No change in activity or diet
36 overweight adolescents, mean 13 y 20 wk Oc b Oc Oc Fair
Epstein, 1989164 Diet of 3800-5000 kJ/d monitored by a nutritionist.
Information on diet, exercise, stimulus control, reinforcement, modeling and contingency contracting presented to parents and their children in 8 weekly sessions followed by 4 monthly sessions
56 obese (> 20% of ideal weight) children,
8-12 y
6 mo a b NR a Poor
Walter, 1985177 "Know Your Body" curriculum yearly, taught 2 hours/wk by usual classroom teacher vs. standard curriculum 1,115 4th graders 1 y Oc Oc NR NR Fair

Notes:
a ↓ = significant decrease.
b ↑ = significant increase.
c 0 = no significant change.
d This trial reported significant pre-experimental differences between groups in HDL (p < 0.05).

Abbreviations:
AHA = American Heart Association, D = Day(s), DISC = Dietary Intervention Study in Children, HDL = High-density lipoprotein, LDL = Low-density lipoprotein, Mo = Month(s), NR= not reported, TC = Total cholesterol, TG = Triglycerides, Wk = Week(s), Y = Year(s).

Return to Table of Contents
Author, year, title Drug Population -
N, age
Duration of trial Adverse effects of treatment
Clinical effects Laboratory effects
Statins
McCrindle, 2003169 Atorvastatin 187, 10-17 y 26 wk None observed; No effect on sexual development. Increased AST and ALT (1% of patients). None withdrew or stopped medication as a result of increased transaminases.
Clauss, 200519 Lovastatin 54 girls, 10-17 y 24 wk Abdominal pain (2), diarrhea (1), nausea (1), headache (1), amenorrhea (1). All resolved with patient continuing medication. Transient decreased HCT.
Lambert, 1996184 Lovastatin 69 boys, < 18 y 8 wk None observed. Asymptomatic elevations in CK (3).
Stein, 1999172 Lovastatin 132, 13 y (mean) 48 wk No effect on growth or sexual development. Transient CK elevations in response to exercise. No effect on AST; ALT increased in placebo and treatment groups. DHEAS increased. Tocopheral, CD3, CD4, and CD8 counts decreased.
Wiegman, 200418 Pravastatin 214, 8-18 y 2 y No effect on growth or sexual development. No effects on muscle or liver enzyme levels.
Hedman, 2003203 Pravastatin 20, 4-15 y 8 wk Abdominal pain (1), loose stools (1), headache (4), sleep disturbance (2), muscle tenderness or pain at rest (1), muscle tenderness or pain associated with physical training (1). No effects on serum ALT, CK, or creatinine.
Knipscheer, 1996173 Pravastatin 72, 12 y (mean) 12 wk Rash, nose bleeding, headache, nausea/vomiting, abdominal pain. CK abnormal in placebo (8), 5 mg/d (6), 10 mg/d (11) and 20 mg/d groups (8). Cortisol abnormal in placebo (2), 5 mg/d (2), 10 mg/d (5), and 20 mg/d (3) groups.
Couture, 1998179 Simvastatin 63, 8-17 y 6 wk None observed. NR
De Jongh, 2002165 Simvastatin 69, 9-18 y 28 wk None observed. No significant effects on ALT, AST, and CK.
De Jongh, 2002188 Simvastatin 173, 10-17 y 48 wk Abdominal pain (3), chest pain (1), flatulence (1), myalgia (2), headache (4), sleep disorder (1), weight gain (1), pruritus (1). Increased ALT (3), AST (3), and CK (1).
Dirisamer, 2003204 Simvastatin 20, 10-17 y 18 mo Transient headache (2). Myalgia (1) for 2 weeks. Transient gastrointestinal complaints (2). Slightly higher values of CK (2); Transiently elevated ALT and glucose challenge test (1).
Ducobu, 1992207 Simvastatin 32, < 17 y 24-36 mo No effect on growth. Transient increases in transaminase (1) and CK (2).
Stefanutti, 1999208 Simvastatin 16, 7-12 y 12 mo None observed. NR
Various or unspecified statins
Sinzinger, 2004205 Various statins 22 professional athletes, 15-27 y 8 y Muscle pain reported in 84% of periods of statin therapy (mean time of onset was 8.3 d). Elevated CK in 3 subjects. No increase in liver enzymes.
De Jongh, 2003206 Various statins 69, 10-18 y NR None observed. NR
Bile-acid Binding Resins
Curtis, 1991209 Cholestyramine 1, 7 y 2 y Loss of dental enamel noted (presumed due to low pH 2.4 of cholestyramine mixed with Kool-Aid® for administration). Serum calcium, phosphorus, folate, B12 were normal.
Farah, 1977210 and Farah, 1977211 Cholestyramine 20, 4-23 y 16 d Febrile gastroenteritis (1) after 7 days treatment resulting in discontinuation of therapy. Serum folate decreased significantly in females. AST increases (2) persisted 6 mo. Transient LDH increases (2). No fat-soluble vitamin malabsorption.
Glueck, 1973227 Cholestyramine 36, 7-21 6 mo None observed. Normal growth. None observed.
Glueck, 1974226 Cholestyramine 30 on diet + BABR, 5-21 y 6 mo
average follow-up
NR Plasma vitamins A and E remained within the normal range.
Glueck, 1977224 Cholestyramine 16, 9-17 y 18 mo (16); 24 mo (12); 30-36 mo (7) Persistent constipation (11). Gritty sensation and poor palatability (5). Chronic fatigue (1). Drop outs after 2 y due to palatability. No effect on CBC, liver function tests, vitamin A and E, calcium, phosphorus, blood urea nitrogen, fasting blood sugar levels.
Glueck, 1986225 Cholestyramine 33, 10.3 y (mean) 4.3 y No effect on growth or sexual development; 1 competitive cross-country runner had persistently irregular periods. NR
Koletzko, 1992215 Cholestyramine 35 on diet; 14 on diet + BABR, 2-17 y Diet: mean 17.5 mo Diet + BABR: mean 27.9 mo None observed. No effect on growth. NR
Liacouras, 1993216 Cholestyramine 87, 10.6 y (mean) Up to 62 mo Nausea (12), abdominal bloating (2), severe constipation (1). Poor palatability (73%). No elevated prothrombin times.
McCrindle, 1997176 Cholestyramine 40, 10-18 y 28 wk Minor gastrointestinal complaints were frequent but did not result in any drop-out. NR
Tonstad, 1996187 Cholestyramine 96, 6-11 y 1 y No effect on growth. One case of intestinal obstruction caused by adhesions. Unpalatability, headaches, and vomiting were reasons for withdrawals. Folate deficiency (most subjects taking cholestyramine). Vitamin D levels decreased significantly for those not taking a multi-vitamin.
Tonstad, 1998219 Cholestyramine 96, 6-11 y 1 y Unpalatability in both treatment and placebo groups. During cholestyramine treatment, plasma total homocysteine increased in subjects with the C677T mutation in 1 or both alleles, but not in subjects with the CC genotype.
West, 1973220 Cholestyramine 19, 1-14 y Up to 20 mo Some had impaired fat absorption without diarrhea. Growth was normal. Serum folate decreased in all patients.
West, 1975221 Cholestyramine 18, 1-14 y 1 to 2.5 y No child developed diarrhea. No effect on growth Decreased red cell folate and mean serum levels of vitamins A, vitamin E and inorganic phosphorus.
West, 1975222 Cholestyramine 45, 1-16 y 2-8 y Adherence was poor due to unpalatability. Folate deficiency, steatorrhoea, and reduction in serum levels of vitamins A and E and of inorganic phosphorus although not to abnormally low values.
West, 1980223 Cholestyramine 35, 1-17 y 1-8 y Nausea, dizziness and malaise in a female aged 18 y. 1 boy died of intercurrent infection 10 mo after starting meds, not stated whether related to treatment. Transient gastric fullness. NR
Groot, 1983212 Colestipol 33, NR 16 wk Withdrawals due to unpalatability (5). NR
Hansen, 1992213 Colestipol 30, 1-17 y 8.5 y (diet); 5.5 y (diet followed by diet + BABR) 1 child's height/age decreased below -2 SD. Growth was normal in other children. NR
Harvengt, 1976214 Colestipol 3, 6-18 y Up to 36 mo Mild gastrointestinal complaints (flatulence, constipation) during first 3 months, but disappeared despite continued treatment. Low iron without anemia (1). Serum uric acid level increased during treatment but did not reach abnormal values.
McCrindle, 2002166 Colestipol 40, 9-18 y 36 wk Constipation (18%), stomachache (21%), headache (11%), muscle aches (6%). NR
Schwarz, 1980217 Colestipol 23, 5-17 y Up to 24 mo Poor palatability (6). Reynauld's phenomenon occurred during therapy (1) but treatment continued without recurrence. Serum vitamins A and E decreased significantly after 18-24 mo of colestipol.
Tonstad, 1996186 Colestipol 66, 13.2 y (mean) 52 wk Gastrointestinal side effects (8), including constipation, dyspepsia, flatulence, nausea, decreased appetite, abdominal pain. Growth was normal. Reduced serum folate after 8 wk. Decreased serum vitamin E and carotenoids. Decreased vitamin D levels (not significant) in subjects who were more compliant after 1 y.
Tonstad, 1996218 Colestipol 27, 10-16 y 6 mo for colestipol; 6 y (mean) for diet No effect on growth. Difficulty swallowing the tablets (2); flatulence (1); abdominal discomfort (1). NR
Other drugs and combinations
Baker, 198226 Probucol 7, 6-21 y 15-21 mo Nausea in 1 patient; No effect on growth and development. None observed.
Becker, 1992228 Sitosterol and bezafibrate, in sequence and in combination 7, 8.4 y (mean) 3 mo sitosterol; 3 mo bezafibrate; 24 mo sitosterol + bezafibrate Decreased appetite for the first 2 wk on sitosterol (2). Sitosterol: slight, significant decrease in hemoglobin
(-5%) and ALP (-19%). Bezafibrate: ALP remained lower; iron increased by 26%. Combination: transferrin increased 20% and reached abnormal levels in 2; all other lab values normal.
Colletti, 1993229 Niacin 21, 4-14 y 1-19 mo, average 8.1 mo 18 of 21 patients reported some adverse effect. Flushing (71%), itching (19%), abdominal pain (14%), nausea (14%), headache (14%), constipation (5%), hepatitis (1). Dose related, reversible serum aminotransferase elevations (6: 4 with crystalline and 2 with sustained release form of niacin).
Malloy, 1978185 P-amnosali-cylic acid 20, 5-21 y 6 mo Mild gastric irritation that remitted with oral antacid treatment. Normal AST, ALT, ALP, bilirubin, and glucose levels in fasting serum; normal TSH and thyroxine.
McDuffie, 2002230 Orlistat 20, 14.6 y (mean) 3 mo Gastrointestinal effects related to increased fat excretion that resolved within the first 6 wk of treatment. 1 subject withdrew because of intolerance of adverse effects. Decreased 25-hydroxy vitamin D levels at 1 mo; 3 subjects required additional vitamin D supplementation despite the prescription of a daily multivitamin containing vitamin D.
Stein, 1989231 Diet + drug or combined drugs: BABR; BABR + niacin; lovastatin or simvastatin 30, 1-20 y 1-9 y None observed. Resin + niacin together produced elevated AST and ALT, decreased albumin and clinical symptoms of hepatotoxicity (1).
Steinmetz, 1981232 Fenofibrate 17,4-19 y 18 mo NR Increased ALT and AST (4); Decreased uric acid, bilirubin, inorganic phosphates, ALP, and GGT.
Wheeler, 1985193 Bezafibrate 14,4-15 y 3 mo None observed. No effect on growth. All subjects declared preference for this drug over cholestyramine. Increased alkaline phosphatase (1), transient rise in ALT (1).

Key:
(#) = Number of participants experiencing effect.

Abbreviations:
ALP = Alkaline phosphate, ALT = Alanine aminotransaminase, AST = Aspartate aminotransferase, BABR = Bile acid binding resin, CBC = Complete blood count, CK = Creatine kinase, D = Day(s), DHEAS = Dehydroepiandrosterones, GGT = Gamma-Glutamyl Transpeptidase, HCT = Hematocrit, LDH = Lactate dehydrogenase, Mo = Month(s), NR = Not reported, RCT = Randomized controlled trial, TSH = Thyroid stimulating hormone, Wk = Week(s), Y = Year(s).

Return to Table of Contents
Arrow Key question Quality of evidence Conclusions
1 Is screening for dyslipidemia in children effective in delaying the onset and reducing the incidence of CHD-related events? No evidence. No evidence.
2 What is the accuracy of screening for dyslipidemia in identifying children at increased risk of CHD-related events? See below (subquestions). See below (subquestions).
2a What are abnormal lipid values in children/adolescents? Fair to Poor Normal values for lipids in children are currently defined according to population levels (percentiles). NCEP recommendations are based on LRC data, which defines the 95th percentile for TC as 200 mg/dL and for LDL as 130 mg/dL. There are more recent studies suggesting that age, gender, racial differences and temporal trends shift these cut points. The NCEP has defined levels of LDL for which drug treatment (LDL ≥ 190mg/dL or LDL ≥ 160mg/dL with family history of early CHD), further evaluation, diet therapy and testing (LDL > 130mg/dL) and diet therapy with increased surveillance (LDL110-129mg/dL) are recommended.
2b What are appropriate tests? How well do screening tests (non-fasting total cholesterol, fasting total cholesterol, fasting lipoprotein analysis) identify individuals with dyslipidemia? Poor The most appropriate test is one that accurately predicts future risk and benefit from treatment. In the general population of children there have not been adequate studies to determine these characteristics. Data from few studies suggest that TC above the 95th percentile predicts LDL above the 95th percentile with 44-69% sensitivity. TC minus HDL might be a more sensitive test, but has not been extensively evaluated. A single TC measurement is inadequate to classify children and adolescents into NCEP risk categories with 95% confidence.
2c How well do lipid levels track from childhood to adulthood? Good Serial correlations measured in individual children over time are higher for TC (r=0.38-0.78) and LDL (r=0.4-0.7) than for HDL and TG. Approximately 40-55% of children with elevated lipids (by percentile) will continue to have elevated lipids on follow-up.
2d What is the accuracy of family history in determining risk? Good Multiple good quality studies evaluating the use of family history as a diagnostic test for dyslipidemia in children using varied and large populations demonstrate that family history is an imperfect screening tool for detecting dyslipidemia among children.
2e What are other important risk factors? Good for family history; Good for obesity; Poor for all other risk factors. Evidence from epidemiologic cross-sectional and cohort studies establishes statistical associations between elevations in lipids and family history and overweight. There is inadequate evidence to show the magnitude of the effect of overweight on lipids, or the impact that incorporating weight measures into a screening tool could have. Multiple other risk factors (diet, physical inactivity, aerobic capacity/fitness, puberty level and smoking) have not been evaluated adequately to assess their contribution to dyslipidemia in children or their usefulness as screening tools.
2f What are effective screening strategies for children/adolescents (including frequency of testing, optimal age for testing)? Poor Currently recommended screening strategies have limited diagnostic accuracy, low adherence to guidelines by providers, and limited compliance by parents and children. No trials compare strategies of screening in children. No studies address the frequency and optimal age for testing.
3 What are the adverse effects of screening including false positives, false negatives, labeling, etc? Fair Studies demonstrate lack of parental compliance with screening and follow-up recommendations. Reasons for non-compliance include concern about test accuracy, lack of proof that intervention makes a difference in children, concern about upsetting the child, refusal by the child, inconvenience, or parental decision to institute a diet themselves and have child rechecked subsequently.
4 In children and adolescents, what is the effectiveness of drug, diet, exercise, and combination therapy in reducing the incidence of adult dyslipidemia, and delaying the onset and reducing the incidence of CHD-related events and other outcomes (including optimal age for initiation of treatment)? No evidence. No evidence.
5-8 What is the effectiveness of drug, diet, exercise, and combination therapy for treating dyslipidemia in children/adolescents (including the incremental benefit of treating dyslipidemia in childhood)? Good quality studies with fair external validity for drug therapy. Fair to poor for diet and exercise treatments. Statins are effective for reducing TC and LDL in children with familial hypercholesterolemia. It is not clear how this efficacy translates to children with milder and/or non-familial forms of dyslipidemia. Diet supplements (psyllium, oat, sterol margarine) and counseling were marginally effective in both FH/FCH children and adolescents and those without identified monogenic dyslipidemia. Exercise treatments showed minimal to no improvements in children without monogenic dyslipidemia.
9 What are the adverse effects of drug, diet, exercise, and combination therapy in children/adolescents? Fair Controlled and non-controlled studies of treatment reported adverse effects of drug, diet, exercise, and combination therapy in children and adolescents. Statin drugs were associated primarily with elevations in LFTs and CK. Bile-acid binding resins were associated with GI side effects and decreased levels of serum vitamins and minerals. Low fat diet has been associated with growth retardation and nutritional dwarfing in 3 children who were placed on low-fat diets without formal advice and monitoring. Most studies show normal growth and development in children over 2 years old on monitored low-fat diets. Few side effects other than elevated blood pressure were noted with exercise. The duration of follow-up in these studies ranged from 10 days to 8 years. Studies were generally not of sufficient duration to determine long-term effects of either short or extended use.
10 Does improving dyslipidemia in childhood reduce the risk of dyslipidemia in adulthood? No evidence. No evidence.

Abbreviations:
CHD = Coronary heart disease, CK = Creatine kinase, FH = Familial hyperlipidemia, FCH = Familial Combined Hyperlipidemia, HDL = High-density Lipoprotein, LDL = Low-density Lipoprotein, LFT = Liver function test, NCEP = National Cholesterol Education Program, RCT = Randomized controlled trial, TC = Total cholesterol, TG = Triglycerides.

Return to Table of Contents