LayerRx Mapping ID
733
Slot System
Featured Buckets
Featured Buckets Admin

Severely frail elderly patients do not need lipid-lowering drugs

Article Type
Changed
Mon, 07/31/2017 - 09:30
Display Headline
Severely frail elderly patients do not need lipid-lowering drugs

Frail elderly patients are at high risk of adverse clinical outcomes, including those due to polypharmacy. Several groups tackle “deprescribing” by developing lists of medications that are potentially inappropriate for the elderly, such as the Beers or STOPP/START criteria.1–4

See related editorial

In contrast, our group (the Palliative and Therapeutic Harmonization [PATH] program and the Dalhousie Academic Detailing Service) has developed evidence-based, frailty-specific guidelines for treating hypertension5 and diabetes,6 in which we advocate less-stringent treatment targets and tapering or discontinuing medications, as needed.

The PATH program7 is a clinical approach that prioritizes the consideration of frailty when making treatment decisions. The Dalhousie Academic Detailing Service collaborates with the Nova Scotia Health Authority to research and develop evidence-informed educational messages about the treatment of common medical conditions.

Here, we address lipid-lowering therapy in this population.

CONSIDERING FRAILTY

Frailty is defined in several ways. The Fried model8,9 identifies frailty when 3 of the following characteristics are present: unintentional weight loss, exhaustion, muscle weakness, slow walking speed, or low levels of activity. The Clinical Frailty Scale10,11 and the Frailty Assessment for Care-planning Tool (FACT)5 use deficits in cognition, function, and mobility to define frailty. According to these scales, people are considered severely frail when they require assistance with basic activities of daily living (such as bathing or dressing), owing to cognitive or physical deficits from any cause.

In reviewing the evidence, we consider five questions:

  • What is the quality of the evidence? (Up to 48% of clinical practice guideline recommendations may be based on low-level evidence or expert opinion.12)
  • How did the study population compare with the frail?
  • Are study outcomes and potential benefits clinically relevant to those who are frail?
  • How long did it take for the clinical benefit of a treatment to become apparent, and are the frail elderly likely to live that long?
  • Have the harms of treatment been sufficiently considered?

WHAT IS THE QUALITY OF THE EVIDENCE?

We found no studies that specifically evaluated the benefit of lipid-lowering for severely frail older adults. Therefore, we examined randomized controlled trials that enrolled non-frail older adults,13–28 subgroup analyses of randomized controlled trials,29,30 meta-analyses that analyzed subgroups of elderly populations,31,32 and publications describing the study designs of randomized controlled trials.33–37

Most of the evidence comes from post hoc subgroup analyses of elderly populations. Although meta-analysis is commonly used to compare subgroups, the Cochrane handbook and others consider subgroup comparisons observational by nature.38,39 (See Table 1 for lipid-lowering studies discussed in this article.)

Studies of statins for primary prevention of cardiovascular disease

For evidence of benefit from lipid-lowering for primary prevention (ie, to reduce the risk of cardiovascular events in patients with no known cardiovascular disease at baseline but at increased risk), we reviewed the meta-analysis conducted by the Cholesterol Treatment Trialists’ (CTT) Collaborators.32 Since this meta-analysis included the major trials that enrolled elderly patients, individual publications of post hoc, elderly subgroups were, for the most part, not examined individually. The exception to this approach was a decision to report on the PROSPER13 and JUPITER28 trials separately, because PROSPER is the most representative of the elderly population and JUPITER reached the lowest LDL-C of primary prevention trials published to date and included a large elderly subgroup (n = 5,695).

Savarese et al40 evaluated the benefits of statins for older adults who did not have established cardiovascular disease. We did not report on this meta-analysis, as not all of the subjects that populated the meta-analysis were representative of a typical prevention population. For instance, in the Anglo-Scandinavian Cardiac Outcomes Trial lipid-lowering arm,41 14% of the subjects had had a previous stroke or transient ischemic attack. In the Antihypertensive and Lipid-Lowering Treatment Trial,42 16% of the population had a family history of premature coronary heart disease.

In addition, all the trials in the Savarese meta-analysis were also included in the CTT meta-analysis.32 The CTT reports on baseline risk using patient-level data stratified by age and risk, which may be more relevant to the question of primary prevention for older adults, as highlighted in our review.

PROSPER (Prospective Study of Pravastatin in the Elderly at Risk),13 a well-conducted, double-blind, randomized controlled trial with low probability of bias, compared pravastatin 40 mg and placebo. It was the only study that specifically enrolled older adults, with prespecified analysis of primary and secondary prevention subgroups. The primary prevention subgroup accounted for 56% of the 5,084 participants.

JUPITER (Justification for the Use of Statins in Prevention)28 compared rosuvastatin 20 mg and placebo in 17,802 participants. All had low-density lipoprotein cholesterol (LDL-C) levels below 3.4 mmol/L (130 mg/dL) and elevated levels of the inflammatory biomarker high-sensitivity C-reactive protein (hsCRP), ie, 2 mg/L or higher. Subsequently, Glynn et al performed a post hoc, exploratory subgroup analysis of elderly participants (N = 5,695).29

The JUPITER trial had several limitations.43,44 The planned follow-up period was 5 years, but the trial was stopped early at 1.9 years, after a statistically significant difference was detected in the primary composite outcome of reduction in all vascular events. Studies that are stopped early may exaggerate positive findings.45

Further, JUPITER’s patients were a select group, with normal LDL-C levels, elevated hsCRP values, and without diabetes. Of 90,000 patients screened, 72,000 (80%) did not meet the inclusion criteria and were not enrolled. This high rate of exclusion limits the generalizability of study findings beyond the shortcomings of post hoc subgroup analysis.

The meta-analysis performed by the CTT Collaborators32 used individual participant data from large-scale randomized trials of lipid-modifying treatment. This analysis was specific to people at low risk of vascular disease. In a supplementary appendix, the authors described the reduction in major vascular events for each 1.0 mmol/L decrease in LDL-C in three age categories: under age 60, ages 61 to 70, and over age 70.

The authors also stratified the results by risk category and provided information about those with a risk of major vascular events of less than 20%, which would be more representative of a purer primary prevention population.

For the elderly subgroup at low risk, the CTT Collaborators32 only reported a composite of major vascular events (coronary death, nonfatal myocardial infarction [MI], ischemic stroke, or revascularization) and did not describe individual outcomes, such as prevention of coronary heart disease.

Study results are based on postrandomization findings and therefore may be observational, not experimental.46

Studies of statins for secondary prevention of cardiovascular disease

The aim of secondary prevention is to reduce the risk of recurrent cardiovascular events in patients who already have cardiovascular disease.

To address the question of whether statins reduce cardiovascular risk, we reviewed:

PROSPER,13 which included a preplanned analysis of the secondary prevention population.

Afilalo et al,31,47 who performed a meta-analysis of the elderly subgroups of nine major secondary prevention studies (19,569 patients) using published and unpublished data.

To address the question of whether statins benefit individuals with heart failure, we found two relevant studies:

GISSI-HF (Gruppo Italiano per lo Studio della Sopravvivenza nell’Insufficienza Cardiaca Heart Failure)25 and CORONA (Controlled Rosuvastatin Multinational Trial in Heart Failure),26 which were large, international, well-conducted randomized controlled trials that examined statin use in heart failure.

To answer the question of whether statins benefit individuals after a stroke or transient ischemic attack, we found one relevant study:

SPARCL (Stroke Prevention by Aggressive Reduction in Cholesterol Levels),27 which evaluated the benefit of statins in older adults with a history of stroke or transient ischemic attack. It was a prospective, double-blind, placebo-controlled, international trial conducted at 205 centers. One to 6 months after their cerebrovascular event, patients were randomized to receive either atorvastatin 80 mg or placebo. Given the young age of patients in this trial (mean age 63), we also reviewed a post hoc subgroup analysis of the elderly patients in SPARCL (age > 65).30

 

 

HOW DID THE STUDY POPULATION COMPARE WITH THOSE WHO ARE FRAIL?

Frail older adults are almost always excluded from large-scale clinical trials,48 leading to uncertainty about whether the conclusions can be applied to those with advanced frailty.

Although age is an imperfect proxy measure of frailty,49 we consider the age of the study population as well as their comorbidities.

Participants in the studies we reviewed were generally younger and healthier than those who are frail, with mean ages of about 75 or less (Table 1).

PROSPER was the most representative study, as it specifically enrolled older adults, albeit without frailty,13 and excluded people with poor cognitive function as defined by a Mini Mental State Examination score less than 24.

JUPITER enrolled a select population, as described above. The median age in the elderly subgroup was 74 (interquartile range 72–78).29

The Afilalo et al31 meta-analysis primarily included studies of young-elderly patients, with a mean age of less than 70. PROSPER13 was an exception.

The GISSI-HF study,25 which examined the benefit of statins in heart failure, described their study population as frail, although the mean age was only 68. Compared with those in GISSI-HF, the CORONA patients26 with heart failure were older (mean age 73) and had more severe heart failure. Accordingly, it is possible that many of the CORONA participants were frail.

ARE STUDY OUTCOMES CLINICALLY RELEVANT TO THOSE WHO ARE FRAIL?

Because baseline cardiovascular risk increases with age, the elderly should, in theory, experience greater absolute benefit from lipid-lowering. However, there is uncertainty about whether this is true in practice.

Some, but not all, epidemiologic studies show a weaker relationship between cholesterol levels and cardiovascular morbidity and mortality rates in older compared to younger adults.50,51 This may be because those with high cholesterol levels die before they get old (time-related bias), or because those with life-threatening illness may have lower cholesterol levels.50 In addition, classic risk factors such as age, sex, systolic blood pressure, cholesterol values, diabetes, smoking, and left ventricular hypertrophy on electrocardiography may have less power to predict cardiovascular risk among older patients.52

The goal of treatment in frailty is to prevent further disability or improve quality of life. Therefore, meaningful outcomes for lipid-lowering therapy should include symptomatic nonfatal MI and its associated morbidity (eg, heart failure and persistent angina) or symptomatic nonfatal stroke leading to disability. Outcomes without sustained clinical impact, such as transient ischemic attack, nondisabling stroke, or silent MI, while potentially important in other populations, are less relevant in severe frailty. Notably, in many statin studies, outcomes include asymptomatic heart disease (eg, silent MI and “suspected events”) and nondisabling stroke (eg, mild stroke, transient ischemic attack). When symptomatic outcomes are not reported separately, the impact of the reported benefit on quality of life and function is uncertain.

The outcome of all-cause mortality is generally recognized as a gold standard for determining treatment benefit. However, since advanced frailty is characterized by multiple competing causes for mortality, a reduction in all-cause mortality that is achieved by addressing a single issue in nonfrail populations may not extend to the frail.

To more fully understand the impact of lipid-lowering therapy on quality of life and function, we examined the following questions:

Do statins as primary prevention reduce symptomatic heart disease?

Outcomes for coronary heart disease from PROSPER and JUPITER are summarized in Table 2.

PROSPER. In the PROSPER primary prevention group,13 statin therapy did not reduce the combined outcome of coronary heart disease death and nonfatal MI.

The JUPITER trial demonstrated a statistically significant benefit for preventing MI in the elderly subpopulation (ages 70–97),29 but the number needed to treat was high (211 for 2 years), with a wide confidence interval (CI) (95% CI 106–32,924). The trial did not adequately differentiate between symptomatic and asymptomatic events, making it difficult to determine outcome relevance. Also, due to the methodologic limitations of JUPITER as described above, its results should be interpreted with caution.43,44

The CTT Collaborators32 did not report individual outcomes (eg, coronary heart disease) for the elderly low-risk subgroup and, therefore, this meta-analysis does not answer the question of whether statins reduce symptomatic heart disease in primary prevention populations.

Taken together, these findings do not provide convincing evidence that statin therapy as primary prevention reduces the incidence of symptomatic heart disease for severely frail older adults.

Do statins as secondary prevention reduce symptomatic heart disease?

Most studies defined secondary prevention narrowly as treatment for patients with established coronary artery disease. For instance, in the Afilalo et al meta-analysis,31 the small number of studies that included individuals with other forms of vascular disease (such as peripheral vascular disease) enrolled few participants with noncardiac conditions (eg, 29% in PROSPER13 and 13% in the Heart Protection Study20).

Therefore, any evidence of benefit for secondary prevention demonstrated in these studies is most applicable to patients with coronary heart disease, with less certainty for those with other forms of cardiovascular disease.

In PROSPER,13 the secondary prevention group experienced benefit in the combined outcome of coronary heart disease death or nonfatal MI. In the treatment group, 12.7% experienced this outcome compared with 16.8% with placebo, an absolute risk reduction of 4.1% in 3 years (P = .004, number needed to treat 25, 95% CI 15–77). This measure includes coronary heart disease death, an outcome that may not be generalizable to those who are frail. In addition, the outcome of nonfatal MI includes both symptomatic and suspected events. As such, there is uncertainty whether the realized benefit is clinically relevant to frail older adults.

The Afilalo et al meta-analysis31 showed that the number needed to treat to prevent one nonfatal MI was 38 (95% CI 16–118) over 5 years (Table 2). However, this outcome included both symptomatic and asymptomatic (silent) events.

Based on the available data, we conclude that it is not possible to determine whether statins reduce symptomatic heart disease as secondary prevention for older adults who are frail.

Do statins reduce heart disease in combined populations?

In the combined primary and secondary population from PROSPER,13 pravastatin decreased the risk of nonfatal symptomatic MI from 4.3% in the placebo group to 3.4%, a relatively small reduction in absolute risk (0.9%) and not statistically significant by our chi-square calculation (P = .099).

Do statins prevent a first symptomatic stroke in people with or without preexisting cardiovascular disease?

Preventing strokes that cause functional decline is an important outcome for the frail elderly. Stroke outcomes from PROSPER,13 JUPITER,29 and the Afilalo et al meta-analysis31 are summarized in Table 3.

For primary prevention:

In PROSPER (primary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

JUPITER,29 in contrast, found that rosuvastatin 20 mg reduced strokes in primary prevention, but the absolute benefit was small. In 2 years, 0.8% of the treatment group had strokes, compared with 1.4% with placebo, an absolute risk reduction of 0.6% (P = .023, number needed to treat 161, 95% CI 86–1,192).

Neither PROSPER nor JUPITER differentiated between disabling and nondisabling strokes.

For secondary prevention:

In PROSPER (secondary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

The Afilalo et al secondary prevention meta-analysis demonstrated a 25% relative reduction in stroke (relative risk 0.75, 95% CI 0.56–0.94, number needed to treat 58, 95% CI 27–177).31

Notably, the stroke outcome in Afilalo included both disabling and nondisabling strokes. For example, in the Heart Protection Study,20 the largest study in the Afilalo et al meta-analysis, approximately 50% of nonfatal, classifiable strokes in the overall study population (ie, both younger and older patients) were not disabling. Including disabling and nondisabling strokes in a composite outcome confounds the clinical meaningfulness of these findings in frailty, as the number needed to treat to prevent one disabling stroke cannot be calculated from the data provided.

 

 

Do statins prevent a second (symptomatic) stroke in people with a previous stroke?

SPARCL27 (Table 3) examined the question of whether statins decrease the risk of recurrent ischemic stroke for patients with a prior history of stroke or transient ischemic attack. There was a statistically significant reduction in the primary composite outcome of fatal and nonfatal stroke, with 11.2% of the treatment group and 13.1% of the placebo group experiencing this outcome, an absolute risk reduction of 1.9% at 5 years (P = .03; number needed to treat 52, 95% CI 26–1,303). However, the difference in nonfatal stroke, which is the outcome of interest for frailty (since mortality has uncertain relevance), was not statistically significant (10.4% with treatment vs 11.8% with placebo, P =.11).

An exploratory subgroup analysis of SPARCL patients based on age30 showed a smaller, nonsignificant reduction in the primary end point of fatal and nonfatal stroke in the group over age 65 (relative risk 0.90, 95% confidence interval 0.73–1.11, P = .33) compared with the younger group (age < 65) (relative risk 0.74, 95% CI 0.57–0.96, P = .02).

The applicability of these results to the frail elderly is uncertain, since the subgroup analysis was not powered to determine outcomes based on age stratification and there were differences between groups in characteristics such as blood pressure and smoking status. In addition, the outcome of interest, nonfatal stroke, is not provided for the elderly subgroup.

In conclusion, in both primary and secondary prevention populations, the evidence that statins reduce nonfatal, symptomatic stroke rates for older adults is uncertain.

Do statins decrease all-cause mortality for primary or secondary prevention?

Due to competing risks for death, the outcome of mortality may not be relevant to those who are frail; however, studies showed the following:

For primary prevention, there was no decrease in mortality in PROSPER13 or in the elderly subgroup of JUPITER.29

For secondary prevention, an analysis of PROSPER trial data by Afilalo et al31 showed a significant 18% decrease in all-cause mortality (relative risk 0.82, 95% CI 0.69–0.98) using pravastatin 40 mg.

A decrease in all-cause mortality with statins was also reported in the pooled result of the Afilalo et al meta-analysis.31

What are the reported composite outcomes for primary and secondary prevention?

While we were most interested in the symptomatic outcomes described above, we recognize that the small numbers of events make it difficult to draw firm conclusions. Therefore, we also considered composite primary outcomes, even though most included multiple measures that have varying associations with disability and relevancy to frail older adults.

For primary prevention, in the PROSPER preplanned subgroup analysis,13 there was no statistical benefit for any outcome, including the primary composite measure. In contrast, the elderly subpopulation in the JUPITER trial28 showed a treatment benefit with rosuvastatin 20 mg compared with placebo for the primary composite outcome of MI, stroke, cardiovascular death, hospitalization for unstable angina, or revascularization. The number needed to treat for 2 years was 62 (95% CI 39–148).

In the CTT meta-analysis,32 patients at all levels of baseline risk showed benefit up to age 70. However, there was no statistically significant benefit in the composite primary outcome of coronary deaths, nonfatal myocardial infarction, ischemic stroke, or revascularization in the population most representative of elderly primary prevention—those who were more than 70 years old with a 5-year baseline risk of less than 20%.

For secondary prevention, in PROSPER,13 the subpopulation of patients treated for secondary prevention experienced benefit in the primary composite outcome of coronary heart disease death, nonfatal MI, or fatal or nonfatal stroke, achieving a 4% absolute risk reduction with a number needed to treat of 23 (95% CI 14–81) over 3 years.

Do statins decrease disability?

PROSPER was the only study that reported on disability. Compared with placebo, pravastatin did not decrease disability in the total population as measured by basic and instrumental activities of daily living scales.

Do statins help patients with heart failure?

Neither GISSI-HF25 nor CORONA26 found significant benefit from rosuvastatin 10 mg, despite LDL-C lowering of 27% in GISSI-HF and 45% in CORONA.

Do ezetimibe or other nonstatin lipid-lowering agents improve outcomes?

There is no definitive evidence that ezetimibe provides clinically meaningful benefit as a single agent.

For combination therapy, the IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial)53 showed that adding ezetimibe 10 mg to simvastatin 40 mg after an acute coronary syndrome reduced the risk of nonfatal myocardial infarction compared with simvastatin monotherapy (event rate 12.8% vs 14.4%; hazard ratio 0.87, 95% CI 0.80–0.95; P = .002) for a population with a mean age of 64. The risk of any stroke was also reduced; strokes occurred in 4.2% of those receiving combination therapy vs 4.8% with monotherapy (hazard ratio 0.86, 95% CI 0.73–1.00, P = .05). After a median of 6 years, 42% of patients in each group had discontinued treatment. Given the very specific clinical scenario of acute coronary syndrome and the young age of the patients in this trial, we do not think that this study justifies the use of ezetimibe for severely frail older adults.

There is no evidence that other combinations (ie, a statin plus another lipid-lowering drug) improve clinical outcomes for either primary or secondary prevention in any population.54

WILL FRAIL PATIENTS LIVE LONG ENOUGH TO BENEFIT?

It is often difficult to determine the number of years that are needed to achieve benefit, as most trials do not provide a statistical analysis of varying time frames.

The PROSPER trial13 lasted 3.2 years. From the Kaplan-Meier curves in PROSPER, we estimate that it took about 1.5 years to achieve a 1% absolute risk reduction and 2.5 years for a 2% absolute risk reduction in coronary heart disease death and nonfatal MI in the combined primary and secondary groups.

JUPITER28 was stopped early at 1.9 years. The Afilalo et al meta-analysis31 was based on follow-up over 4.9 years.

IMPROVE-IT53 reported event rates at 7 years. The authors note that benefit in the primary composite outcome appeared to emerge at 1 year, although no statistical support is given for this statement and divergence in the Kaplan-Meier curves is not visually apparent.

The duration of other studies ranged between 2.7 and 4.9 years (Table 1).26–28

It has been suggested that statins should be considered for elderly patients who have a life expectancy of at least 5 years.3 However, many older adults have already been taking statins for many years, which makes it difficult to interpret the available timeframe evidence.

In a multicenter, unblinded, randomized trial,55 statins were either stopped or continued in older adults who had a short life expectancy and a median survival of approximately 7 months. Causes of death were evenly divided between cancer and noncancer diagnoses, and 22% of the patients were cognitively impaired. Discontinuing statin therapy did not increase mortality or cardiovascular events within 60 days. Nevertheless, stopping statin therapy did not achieve noninferiority for the primary end point, the proportion of participants who died within 60 days. Statin discontinuation was associated with improved quality of life, although the study was not blinded, which could have influenced results.

HAVE THE HARMS BEEN SUFFICIENTLY CONSIDERED?

Frail older adults commonly take multiple medications and are more vulnerable to adverse events.56

Many statins require dose reduction with severe renal impairment (creatinine clearance < 30 mL/min/1.73 m2), which would be a common consideration in severely frail older adults.

Myopathy

Myopathy, which includes myalgias and muscle weakness, is a statin-related adverse event that can impair quality of life. Myopathy typically develops within the first 6 months but can occur at any time during statin treatment.57 When muscle-related adverse effects occur, they may affect the elderly more significantly, particularly their ability to perform activities of daily living, rise from a chair, or mobilize independently. Another concern is that older adults with dementia may not be able to accurately report muscle-related symptoms.

It is difficult to ascertain the true prevalence of myopathy, especially in advanced age and frailty. Randomized controlled trials report incidence rates of 1.5% to 5%, which is comparable to placebo.57,58 However, inconsistent definitions of myopathy and exclusion of subjects with previous statin intolerance or adverse effects during run-in periods limit interpretability.57 Clinical experience suggests that muscle complaints may be relatively common.59–61

Advanced age, female sex, low body mass index, and multisystem disease are all associated with frailty and have also been described as risk factors for statin-associated muscle syndromes.61 Physiologic changes associated with frailty, such as reduced muscle strength, decreased lean body mass, impaired functional mobility, decreased reserve capacity, and altered drug metabolism may increase the risk and severity of myopathy.62

Adverse cognitive events

Meta-analyses of randomized clinical trials and narrative reviews find no definitive relationship between statin therapy and adverse cognitive events.63–67 Nevertheless, there have been case reports of memory loss associated with the use of statins, and the US Food and Drug Administration has issued a warning that statins have been associated with memory loss and confusion.68

It may be difficult to determine whether a statin is causing or aggravating cognitive symptoms among individuals with dementia without a trial withdrawal of the drug.

OUR RECOMMENDATIONS

The recommendations below are intended for adults with severe or very severe frailty (ie, a score of 7 or 8 on the Clinical Frailty Scale11 or FACT5 and therefore apply to most older adults living in long-term care facilities.

Primary prevention

There is no reason to prescribe or continue statins for primary prevention, as it is unlikely that they would provide benefit for outcomes that are relevant in this population.

Secondary prevention

Statin treatment is probably not necessary for secondary prevention in those with severe frailty, although there may be extenuating circumstances that justify statin use.

Heart failure

There is no reason to start or continue statins for heart failure, as there is insufficient evidence that they are effective for this indication in any population.

Ezetimibe

There is no evidence that ezetimibe reduces cardiovascular events in any population when used as monotherapy. For a select population with acute coronary syndromes, ezetimibe has a modest effect. Given the very specific clinical scenario of acute coronary syndrome, we do not think that the available evidence justifies the use of ezetimibe for severely frail older adults.

Agents other than ezetimibe combined with statins

There is no reason to start or continue other lipid-lowering drugs in conjunction with statins.

Statin dosing

As statin adverse effects have the potential to increase with advancing age and frailty, lower doses may be appropriate.68

Adverse events

Consider stopping statins on a trial basis if there is concern regarding myopathy, drug interactions, or other adverse effects.

BOTTOM LINE: DO STATINS IMPROVE QUALITY OF LIFE OR FUNCTION?

In primary prevention for older adults, there is doubt that statins prevent cardiovascular disease and stroke-related events because the main study involving the elderly did not show a benefit in the primary prevention subgroup.13 Additionally, there is no conclusive evidence that statin treatment decreases mortality in primary prevention.13,29

There is insufficient information to determine whether the frail elderly should receive statins for secondary prevention. Although there is evidence that treatment decreases measures of coronary heart disease and stroke, it is unclear whether it improves quality of life or function for those who are frail. To answer this question, we need more information about whether reported outcomes (such as stroke and MI) are associated with disability, which is not provided in many of the studies we reviewed. When disability was specifically considered in the PROSPER trial for the combined population of primary and secondary prevention, treatment with statins had no impact on basic and instrumental activities of daily living.

Some experts may not agree with our interpretation of the complex evidence presented in this article. Others may ask, “What is the harm in using statins, even if there is no definitive benefit?” However, the harms associated with statin therapy for the frail are poorly defined. In the face of these uncertainties and in the absence of definitive improvement in quality of life, we believe that “less is more” in the context of severe frailty.69

The cost of medications should also be considered, especially in long-term care facilities, where there is an added expense of drug administration that diverts human resources away from interactions that are more congruent with respecting the lifestage of frailty.

Careful review of evidence before applying clinical practice guidelines to those who are frail should become the norm. When considering treatment of frail patients, the five questions described in this review shed light on the applicability of clinical trial evidence. Therapies that are highly effective in healthier populations may be less effective when individuals are severely frail. Accordingly, we propose that medications should only be used if they improve quality of life or function.

References
  1. Ontario Pharmacy Research Collaboration. Deprescribing guidelines for the elderly. www.open-pharmacy-research.ca/research-projects/emerging-services/deprescribing-guidelines. Accessed December 28, 2016.
  2. Scott IA, Hilmer SN, Reeve E, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med 2015; 175:827–834.
  3. O’Mahony D, O’Sullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing 2015; 44:213–218.
  4. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc 2012; 60:616–631.
  5. Mallery LH, Allen M, Fleming I, et al. Promoting higher blood pressure targets for frail older adults: a consensus guideline from Canada. Cleve Clin J Med 2014; 81:427–437.
  6. Mallery LH, Ransom T, Steeves B, Cook B, Dunbar P, Moorhouse P. Evidence-informed guidelines for treating frail older adults with type 2 diabetes: from the Diabetes Care Program of Nova Scotia (DCPNS) and the Palliative and Therapeutic Harmonization (PATH) program. J Am Med Dir Assoc 2013; 14:801–808.
  7. Moorhouse P, Mallery L. Palliative and therapeutic harmonization: a model for appropriate decision-making in frail older adults. J Am Geriatr Soc 2012; 60:2326–2332.
  8. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study Collaborative Research Group. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
  9. Morley JE, Malmstrom TK, Miller DK. A simple frailty questionnaire (FRAIL) predicts outcomes in middle aged African Americans. J Nutr Health Aging 2012; 16:601–608.
  10. Rockwood K, Song Z, MacKnight C, et al. A global clinical measure of fitness and frailty in elderly people. CMAJ 2005; 173:489–495.
  11. Morley JE, Vellas B, van Kan GA, et al. Frailty consensus: a call to action. J Am Med Dir Assoc 2013; 14:392–397.
  12. Tricoci P, Allen JM, Kramer JM, Califf RM, Smith SC Jr. Scientific evidence underlying the ACC/AHA clinical practice guidelines. JAMA 2009; 301:831–841.
  13. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  14. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet 1994; 344:1383–1389.
  15. Miettien TA, Pyorala K, Olsson AG, et al. Cholesterol-lowering therapy in women and elderly patients with myocardial infarction or angina pectoris: findings from the Scandinavian Simvastatin Study Group (4S). Circulation 1997; 96:4211–4218.
  16. Lewis SJ, Moye LA, Sacks FM, et al. Effect of pravastatin on cardiovascular events in older patients with myocardial infarction and cholesterol levels in the average range. Results of the Cholesterol and Recurrent Events (CARE) trial. Ann Intern Med 1998; 129:681–689.
  17. Hunt D, Young P, Simes J, et al. Benefits of pravastatin on cardiovascular events and mortality in older patients with coronary heart disease are equal to or exceed those seen in younger patients: results from the LIPID trial. Ann Intern Med 2001; 134:931–940.
  18. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. N Engl J Med 1998; 339:1349–1357.
  19. Heart Protection Study Collaborative Group. The effects of cholesterol lowering with simvastatin on cause-specific mortality and on cancer incidence in 20,536 high-risk people: a randomized placebo-controlled trial. BMC Med 2005; 3:6.
  20. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomized placebo-controlled trial. Lancet 2002; 360:7–22.
  21. Pitt B, Mancini GB, Ellis SG, Rosman HS, Park JS, McGovern ME. Pravastatin limitation of atherosclerosis in the coronary arteries (PLAC 1): reduction in atherosclerosis progression and clinical events. PLAC 1 investigation. J Am Coll Cardiol 1995; 26:1133–1139.
  22. Jukema JW, Bruschke AV, van Boven AJ, et al. Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS). Circulation 1995; 91:2528–2540.
  23. Serruys PW, Foley DP, Jackson G, et al. A randomized placebo-controlled trial of fluvastatin for prevention of restenosis after successful coronary balloon angioplasty; final results of the fluvastatin angiographic restenosis (FLARE) trial. Eur Heart J 1999; 20:58–69.
  24. Serruys PW, de Feyter P, Macaya C, et al; Lescol Intervention Prevention Study (LIPS) Investigators. Fluvastatin for prevention of cardiac events following successful first percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 287:3215–3222.
  25. Tavazzi L, Maggioni AP, Marchioli R, et al; Gissi-HF Investigators. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomized, double-blind, placebo-controlled trial. Lancet 2008; 372:1231–1239.
  26. Kjekshus J, Apatrei E, Barrios V, et al; CORONA Group. Rosuvastatin in older patients with systolic heart failure. N Engl J Med 2007; 357:2248–2261.
  27. Amarenco P, Bogousslavsky J, Callahan A, et al; Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) Investigators. High-dose atorvastatin after stroke or transient ischemic attack. N Engl J Med 2006; 355:549–559.
  28. Ridker PM, Danielson E, Fonseca FA, et al; JUPITER Study Group. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med 2008; 359:2195–2207.
  29. Glynn RJ, Koenig W, Nordestgaard BG, Shepherd J, Ridker PM. Rosuvastatin for primary prevention in older persons with elevated C-reactive protein and low to average low-density lipoprotein cholesterol levels: exploratory analysis of a randomized trial. Ann Intern Med 2010; 152:488–496, W174.
  30. Chaturvedi S, Zivin J, Breazna A, et al; SPARCL Investigators. Effect of atorvastatin in elderly patients with a recent stroke or transient ischemic attack. Neurology 2009; 72:688–694.
  31. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  32. Cholesterol Treatment Trialists’ (CTT) Collaborators; Mihaylova B, Emberson J, Blackwell L, et al. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012; 380:581– 590.
  33. Sacks FM, Pfeffer MA, Moye L, et al. Rationale and design of a secondary prevention trial of lowering normal plasma cholesterol levels after acute myocardial infarction: the Cholesterol and Recurrent Events (CARE). Am J Cardiol 1991; 68:1436–1446.
  34. Armitage J, Collins R. Need for large scale randomised evidence about lowering LDL cholesterol in people with diabetes mellitus: MRC/BHF Heart Protection Study and other major trials. Heart 2000; 84:357–360.
  35. Design features and baseline characteristics of the LIPID (Long-Term Intervention with Pravastatin in Ischemic Disease) study: a randomized trial in patients with previous acute myocardial infarction and/or unstable angina pectoris. Am J Cardiol 1995; 76:474–479.
  36. Shepherd J, Blauw GJ, Murphy MB, et al. The design of a prospective study of Pravastatin in the Elderly at Risk (PROSPER). Am J Cardiol 1999; 84:1192–1197.
  37. Amarenco P, Bogousslavsky J, Callahan AS, et al; SPARCL Investigators. Design and baseline characteristics of the stroke prevention by aggressive reduction in cholesterol levels (SPARCL) study. Cerebrovasc Dis 2003; 16:389–395.
  38. Interpretation of subgroup analyses and meta-regressions. In: Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration, 2011. http://handbook.cochrane.org/chapter_9/9_6_6_interpretation_of_subgroup_analyses_and_meta_regressions.htm. Accessed December 5, 2016.
  39. Borenstein M, Higgins JP. Meta-analysis and subgroups. Prev Sci 2013; 14:134–143.
  40. Savarese G, Gotto AM Jr, Paolillo S, et al. Benefits of statins in elderly subjects without established cardiovascular disease: a meta-analysis. J Am Coll Cardiol 2013; 62:2090–2099.
  41. Sever PS, Dahlof B, Poulter NR, et al. Prevention of coronary and stroke events with atorvastatin in hypertensive patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet 2003; 361:1149–1158.
  42. The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial. Major outcomes in moderately hypercholesterolemic, hypertensive patients randomized to pravastatin vs usual care: The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT-LLT). JAMA 2002; 288:2998–3007.
  43. de Longeril M, Salen P, Abramson J, et al. Cholesterol lowering, cardiovascular diseases, and the rosuvastatin-JUPITER controversy: a critical reappraisal. Arch Intern Med 2010; 170:1032–1036.
  44. Yusuf S, Lonn E, Bosch J. Lipid lowering for primary prevention. Lancet 2009: 373:1152–1155.
  45. Briel M, Bassler D, Wang AT, Guyatt GH, Montori VM. The dangers of stopping a trial too early. J Bone Joint Surg Am 2012; 94(suppl 1):56–60.
  46. Hayward RA, Krumholz HM. Three reasons to abandon low-density lipoprotein targets: an open letter to the Adult Treatment Panel IV of the National Institutes of Health. Circ Cardiovasc Qual Outcomes 2012; 5:2–5.
  47. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical Bayesian meta-analysis. www.ncbi.nlm.nih.gov/pubmedhealth/PMH0026417. Accessed December 5, 2016.
  48. Holmes HM, Hayley DC, Alexander GC, Sachs GA. Reconsidering medication appropriateness for patients late in life. Arch Intern Med 2006; 166:605–609.
  49. Rockwood K, Mitnitski A. Frailty defined by deficit accumulation and geriatric medicine defined by frailty. Clin Geriatr Med 2011; 27:17–26.
  50. Petersen LK, Christensen K, Kragstrup J. Lipid-lowering treatment to the end? A review of observational studies and RCTs on cholesterol and mortality in 80+-year olds. Age Ageing 2010; 39:674–680.
  51. Psaty BM, Anderson M, Kronmal RA, et al. The association between lipid levels and the risks of incident myocardial infarction, stroke, and total mortality: the Cardiovascular Health Study. J Am Geriatr Soc 2004; 52:1639–1647.
  52. de Ruijter W, Westendorp RG, Assendelft WJ, et al. Use of Framingham risk score and new biomarkers to predict cardiovascular mortality in older people: population based observational cohort study. BMJ 2009; 338:a3083.
  53. Canon CP, Blazing MA, Giugliano RP, et al; IMPROVE-IT Investigators. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med 2015; 372:2387–2397.
  54. Anderson TJ, Gregoire J, Hegele RA, et al. 2012 update of the Canadian Cardiovascular Society guidelines for the diagnosis and treatment of dyslipidemia for the prevention of cardiovascular disease in the adult. Can J Cardiol 2013; 29:151–167.
  55. Kutner JS, Blatchford PJ, Taylor DH, et al. Safety and benefit of discontinuing statin therapy in the setting of advanced, life-limiting illness: a randomized clinical trial. JAMA Intern Med 2015; 175:691–700.
  56. Tinetti ME, Bogardus ST Jr, Agostini JV. Potential pitfalls of disease-specific guidelines for patients with multiple conditions. N Engl J Med 2004; 351:2870–2874.
  57. Rosenson RS. Current overview of statin-induced myopathy. Am J Med 2004; 116:408–416.
  58. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: proceedings of a Canadian Working Group Consensus Conference. Can J Cardiol 2011; 27:635–662.
  59. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208–215.
  60. Joy TR, Hegele RA. Narrative review: statin-related myopathy. Ann Intern Med 2009; 150:858–868.
  61. Talbert RL. Safety issues with statin therapy. J Am Pharm Assoc (2003) 2006; 46:479–490.
  62. Sewright KA, Clarkson PM, Thompson PD. Statin myopathy: incidence, risk factors, and pathophysiology. Curr Atheroscler Rep 2007; 9:389–396.
  63. Ott BR, Daiello LA, Dahabreh IJ, et al. Do statins impair cognition? A systematic review and meta-analysis of randomized controlled trials. J Gen Intern Med 2015; 30:348–358.
  64. Mancini GB, Tashakkor AY, Baker S, et al. Diagnosis, prevention and management of statin adverse effects and intolerance: Canadian Working Group Consensus update. Can J Cardiol 2013: 29:1553–1568.
  65. Rojas-Fernandez CH, Cameron JC. Is statin-associated cognitive impairment clinically relevant? A narrative review and clinical recommendations. Ann Pharmacother 2012; 46:549–557.
  66. McGuinness B, O’Hare J, Craig D, Bullock R, Malouf R, Passmore P. Cochrane review on ‘Statins for the treatment of dementia’. Int J Geriatr Psychiatry 2013; 28:119–126.
  67. Pandey RD, Gupta PP, Jha D, Kumar S. Role of statins in Alzheimer’s disease: a retrospective meta-analysis for commonly investigated clinical parameters in RCTs. Int J Neurosci 2013; 123:521–525.
  68. Food and Drug Administration (FDA). FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. www.fda.gov/drugs/ drugsafety/ucm293101.htm. Accessed December 5, 2016.
  69. Garfinkel D, Mangin D. Feasibility study of a systematic approach for discontinuation of multiple medications in older adults: addressing polypharmacy. Arch Intern Med 2010; 170:1648–1654.
Article PDF
Author and Disclosure Information

Laurie Herzig Mallery, MD, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Paige Moorhouse, MD, MPH, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Pam McLean Veysey, BSc (Pharm)
Team Lead, Drug Evaluation Unit, Department of Pharmacy, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada

Michael Allen, MD, MSc
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Isobel Fleming, BScPharm, ACPR
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Address: Laurie H. Mallery, MD, FRCPC, Camp Hill Veterans’ Memorial Building, 5955 Veterans’ Memorial Lane, Suite 2650, Halifax, NS B3H 2E1 Canada; laurie.mallery@nshealth.ca

Dr. Mallery and Dr. Moorhouse have disclosed partnership in Palliative and Therapeutic Harmonization Ltd.

Issue
Cleveland Clinic Journal of Medicine - 84(2)
Publications
Topics
Page Number
131-142
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, deprescribing, PATH program, Canada, JUPITER trial, PROSPER trial, SPARCL trial, Laurie Mallery, Paige Moorhouse, Pam Veysey, Michael Allen, Isobel Fleming
Sections
Author and Disclosure Information

Laurie Herzig Mallery, MD, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Paige Moorhouse, MD, MPH, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Pam McLean Veysey, BSc (Pharm)
Team Lead, Drug Evaluation Unit, Department of Pharmacy, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada

Michael Allen, MD, MSc
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Isobel Fleming, BScPharm, ACPR
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Address: Laurie H. Mallery, MD, FRCPC, Camp Hill Veterans’ Memorial Building, 5955 Veterans’ Memorial Lane, Suite 2650, Halifax, NS B3H 2E1 Canada; laurie.mallery@nshealth.ca

Dr. Mallery and Dr. Moorhouse have disclosed partnership in Palliative and Therapeutic Harmonization Ltd.

Author and Disclosure Information

Laurie Herzig Mallery, MD, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Paige Moorhouse, MD, MPH, FRCPC, MSM
Department of Medicine, Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Pam McLean Veysey, BSc (Pharm)
Team Lead, Drug Evaluation Unit, Department of Pharmacy, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada

Michael Allen, MD, MSc
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Isobel Fleming, BScPharm, ACPR
Academic Detailing Service, Continuing Professional Development, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Address: Laurie H. Mallery, MD, FRCPC, Camp Hill Veterans’ Memorial Building, 5955 Veterans’ Memorial Lane, Suite 2650, Halifax, NS B3H 2E1 Canada; laurie.mallery@nshealth.ca

Dr. Mallery and Dr. Moorhouse have disclosed partnership in Palliative and Therapeutic Harmonization Ltd.

Article PDF
Article PDF
Related Articles

Frail elderly patients are at high risk of adverse clinical outcomes, including those due to polypharmacy. Several groups tackle “deprescribing” by developing lists of medications that are potentially inappropriate for the elderly, such as the Beers or STOPP/START criteria.1–4

See related editorial

In contrast, our group (the Palliative and Therapeutic Harmonization [PATH] program and the Dalhousie Academic Detailing Service) has developed evidence-based, frailty-specific guidelines for treating hypertension5 and diabetes,6 in which we advocate less-stringent treatment targets and tapering or discontinuing medications, as needed.

The PATH program7 is a clinical approach that prioritizes the consideration of frailty when making treatment decisions. The Dalhousie Academic Detailing Service collaborates with the Nova Scotia Health Authority to research and develop evidence-informed educational messages about the treatment of common medical conditions.

Here, we address lipid-lowering therapy in this population.

CONSIDERING FRAILTY

Frailty is defined in several ways. The Fried model8,9 identifies frailty when 3 of the following characteristics are present: unintentional weight loss, exhaustion, muscle weakness, slow walking speed, or low levels of activity. The Clinical Frailty Scale10,11 and the Frailty Assessment for Care-planning Tool (FACT)5 use deficits in cognition, function, and mobility to define frailty. According to these scales, people are considered severely frail when they require assistance with basic activities of daily living (such as bathing or dressing), owing to cognitive or physical deficits from any cause.

In reviewing the evidence, we consider five questions:

  • What is the quality of the evidence? (Up to 48% of clinical practice guideline recommendations may be based on low-level evidence or expert opinion.12)
  • How did the study population compare with the frail?
  • Are study outcomes and potential benefits clinically relevant to those who are frail?
  • How long did it take for the clinical benefit of a treatment to become apparent, and are the frail elderly likely to live that long?
  • Have the harms of treatment been sufficiently considered?

WHAT IS THE QUALITY OF THE EVIDENCE?

We found no studies that specifically evaluated the benefit of lipid-lowering for severely frail older adults. Therefore, we examined randomized controlled trials that enrolled non-frail older adults,13–28 subgroup analyses of randomized controlled trials,29,30 meta-analyses that analyzed subgroups of elderly populations,31,32 and publications describing the study designs of randomized controlled trials.33–37

Most of the evidence comes from post hoc subgroup analyses of elderly populations. Although meta-analysis is commonly used to compare subgroups, the Cochrane handbook and others consider subgroup comparisons observational by nature.38,39 (See Table 1 for lipid-lowering studies discussed in this article.)

Studies of statins for primary prevention of cardiovascular disease

For evidence of benefit from lipid-lowering for primary prevention (ie, to reduce the risk of cardiovascular events in patients with no known cardiovascular disease at baseline but at increased risk), we reviewed the meta-analysis conducted by the Cholesterol Treatment Trialists’ (CTT) Collaborators.32 Since this meta-analysis included the major trials that enrolled elderly patients, individual publications of post hoc, elderly subgroups were, for the most part, not examined individually. The exception to this approach was a decision to report on the PROSPER13 and JUPITER28 trials separately, because PROSPER is the most representative of the elderly population and JUPITER reached the lowest LDL-C of primary prevention trials published to date and included a large elderly subgroup (n = 5,695).

Savarese et al40 evaluated the benefits of statins for older adults who did not have established cardiovascular disease. We did not report on this meta-analysis, as not all of the subjects that populated the meta-analysis were representative of a typical prevention population. For instance, in the Anglo-Scandinavian Cardiac Outcomes Trial lipid-lowering arm,41 14% of the subjects had had a previous stroke or transient ischemic attack. In the Antihypertensive and Lipid-Lowering Treatment Trial,42 16% of the population had a family history of premature coronary heart disease.

In addition, all the trials in the Savarese meta-analysis were also included in the CTT meta-analysis.32 The CTT reports on baseline risk using patient-level data stratified by age and risk, which may be more relevant to the question of primary prevention for older adults, as highlighted in our review.

PROSPER (Prospective Study of Pravastatin in the Elderly at Risk),13 a well-conducted, double-blind, randomized controlled trial with low probability of bias, compared pravastatin 40 mg and placebo. It was the only study that specifically enrolled older adults, with prespecified analysis of primary and secondary prevention subgroups. The primary prevention subgroup accounted for 56% of the 5,084 participants.

JUPITER (Justification for the Use of Statins in Prevention)28 compared rosuvastatin 20 mg and placebo in 17,802 participants. All had low-density lipoprotein cholesterol (LDL-C) levels below 3.4 mmol/L (130 mg/dL) and elevated levels of the inflammatory biomarker high-sensitivity C-reactive protein (hsCRP), ie, 2 mg/L or higher. Subsequently, Glynn et al performed a post hoc, exploratory subgroup analysis of elderly participants (N = 5,695).29

The JUPITER trial had several limitations.43,44 The planned follow-up period was 5 years, but the trial was stopped early at 1.9 years, after a statistically significant difference was detected in the primary composite outcome of reduction in all vascular events. Studies that are stopped early may exaggerate positive findings.45

Further, JUPITER’s patients were a select group, with normal LDL-C levels, elevated hsCRP values, and without diabetes. Of 90,000 patients screened, 72,000 (80%) did not meet the inclusion criteria and were not enrolled. This high rate of exclusion limits the generalizability of study findings beyond the shortcomings of post hoc subgroup analysis.

The meta-analysis performed by the CTT Collaborators32 used individual participant data from large-scale randomized trials of lipid-modifying treatment. This analysis was specific to people at low risk of vascular disease. In a supplementary appendix, the authors described the reduction in major vascular events for each 1.0 mmol/L decrease in LDL-C in three age categories: under age 60, ages 61 to 70, and over age 70.

The authors also stratified the results by risk category and provided information about those with a risk of major vascular events of less than 20%, which would be more representative of a purer primary prevention population.

For the elderly subgroup at low risk, the CTT Collaborators32 only reported a composite of major vascular events (coronary death, nonfatal myocardial infarction [MI], ischemic stroke, or revascularization) and did not describe individual outcomes, such as prevention of coronary heart disease.

Study results are based on postrandomization findings and therefore may be observational, not experimental.46

Studies of statins for secondary prevention of cardiovascular disease

The aim of secondary prevention is to reduce the risk of recurrent cardiovascular events in patients who already have cardiovascular disease.

To address the question of whether statins reduce cardiovascular risk, we reviewed:

PROSPER,13 which included a preplanned analysis of the secondary prevention population.

Afilalo et al,31,47 who performed a meta-analysis of the elderly subgroups of nine major secondary prevention studies (19,569 patients) using published and unpublished data.

To address the question of whether statins benefit individuals with heart failure, we found two relevant studies:

GISSI-HF (Gruppo Italiano per lo Studio della Sopravvivenza nell’Insufficienza Cardiaca Heart Failure)25 and CORONA (Controlled Rosuvastatin Multinational Trial in Heart Failure),26 which were large, international, well-conducted randomized controlled trials that examined statin use in heart failure.

To answer the question of whether statins benefit individuals after a stroke or transient ischemic attack, we found one relevant study:

SPARCL (Stroke Prevention by Aggressive Reduction in Cholesterol Levels),27 which evaluated the benefit of statins in older adults with a history of stroke or transient ischemic attack. It was a prospective, double-blind, placebo-controlled, international trial conducted at 205 centers. One to 6 months after their cerebrovascular event, patients were randomized to receive either atorvastatin 80 mg or placebo. Given the young age of patients in this trial (mean age 63), we also reviewed a post hoc subgroup analysis of the elderly patients in SPARCL (age > 65).30

 

 

HOW DID THE STUDY POPULATION COMPARE WITH THOSE WHO ARE FRAIL?

Frail older adults are almost always excluded from large-scale clinical trials,48 leading to uncertainty about whether the conclusions can be applied to those with advanced frailty.

Although age is an imperfect proxy measure of frailty,49 we consider the age of the study population as well as their comorbidities.

Participants in the studies we reviewed were generally younger and healthier than those who are frail, with mean ages of about 75 or less (Table 1).

PROSPER was the most representative study, as it specifically enrolled older adults, albeit without frailty,13 and excluded people with poor cognitive function as defined by a Mini Mental State Examination score less than 24.

JUPITER enrolled a select population, as described above. The median age in the elderly subgroup was 74 (interquartile range 72–78).29

The Afilalo et al31 meta-analysis primarily included studies of young-elderly patients, with a mean age of less than 70. PROSPER13 was an exception.

The GISSI-HF study,25 which examined the benefit of statins in heart failure, described their study population as frail, although the mean age was only 68. Compared with those in GISSI-HF, the CORONA patients26 with heart failure were older (mean age 73) and had more severe heart failure. Accordingly, it is possible that many of the CORONA participants were frail.

ARE STUDY OUTCOMES CLINICALLY RELEVANT TO THOSE WHO ARE FRAIL?

Because baseline cardiovascular risk increases with age, the elderly should, in theory, experience greater absolute benefit from lipid-lowering. However, there is uncertainty about whether this is true in practice.

Some, but not all, epidemiologic studies show a weaker relationship between cholesterol levels and cardiovascular morbidity and mortality rates in older compared to younger adults.50,51 This may be because those with high cholesterol levels die before they get old (time-related bias), or because those with life-threatening illness may have lower cholesterol levels.50 In addition, classic risk factors such as age, sex, systolic blood pressure, cholesterol values, diabetes, smoking, and left ventricular hypertrophy on electrocardiography may have less power to predict cardiovascular risk among older patients.52

The goal of treatment in frailty is to prevent further disability or improve quality of life. Therefore, meaningful outcomes for lipid-lowering therapy should include symptomatic nonfatal MI and its associated morbidity (eg, heart failure and persistent angina) or symptomatic nonfatal stroke leading to disability. Outcomes without sustained clinical impact, such as transient ischemic attack, nondisabling stroke, or silent MI, while potentially important in other populations, are less relevant in severe frailty. Notably, in many statin studies, outcomes include asymptomatic heart disease (eg, silent MI and “suspected events”) and nondisabling stroke (eg, mild stroke, transient ischemic attack). When symptomatic outcomes are not reported separately, the impact of the reported benefit on quality of life and function is uncertain.

The outcome of all-cause mortality is generally recognized as a gold standard for determining treatment benefit. However, since advanced frailty is characterized by multiple competing causes for mortality, a reduction in all-cause mortality that is achieved by addressing a single issue in nonfrail populations may not extend to the frail.

To more fully understand the impact of lipid-lowering therapy on quality of life and function, we examined the following questions:

Do statins as primary prevention reduce symptomatic heart disease?

Outcomes for coronary heart disease from PROSPER and JUPITER are summarized in Table 2.

PROSPER. In the PROSPER primary prevention group,13 statin therapy did not reduce the combined outcome of coronary heart disease death and nonfatal MI.

The JUPITER trial demonstrated a statistically significant benefit for preventing MI in the elderly subpopulation (ages 70–97),29 but the number needed to treat was high (211 for 2 years), with a wide confidence interval (CI) (95% CI 106–32,924). The trial did not adequately differentiate between symptomatic and asymptomatic events, making it difficult to determine outcome relevance. Also, due to the methodologic limitations of JUPITER as described above, its results should be interpreted with caution.43,44

The CTT Collaborators32 did not report individual outcomes (eg, coronary heart disease) for the elderly low-risk subgroup and, therefore, this meta-analysis does not answer the question of whether statins reduce symptomatic heart disease in primary prevention populations.

Taken together, these findings do not provide convincing evidence that statin therapy as primary prevention reduces the incidence of symptomatic heart disease for severely frail older adults.

Do statins as secondary prevention reduce symptomatic heart disease?

Most studies defined secondary prevention narrowly as treatment for patients with established coronary artery disease. For instance, in the Afilalo et al meta-analysis,31 the small number of studies that included individuals with other forms of vascular disease (such as peripheral vascular disease) enrolled few participants with noncardiac conditions (eg, 29% in PROSPER13 and 13% in the Heart Protection Study20).

Therefore, any evidence of benefit for secondary prevention demonstrated in these studies is most applicable to patients with coronary heart disease, with less certainty for those with other forms of cardiovascular disease.

In PROSPER,13 the secondary prevention group experienced benefit in the combined outcome of coronary heart disease death or nonfatal MI. In the treatment group, 12.7% experienced this outcome compared with 16.8% with placebo, an absolute risk reduction of 4.1% in 3 years (P = .004, number needed to treat 25, 95% CI 15–77). This measure includes coronary heart disease death, an outcome that may not be generalizable to those who are frail. In addition, the outcome of nonfatal MI includes both symptomatic and suspected events. As such, there is uncertainty whether the realized benefit is clinically relevant to frail older adults.

The Afilalo et al meta-analysis31 showed that the number needed to treat to prevent one nonfatal MI was 38 (95% CI 16–118) over 5 years (Table 2). However, this outcome included both symptomatic and asymptomatic (silent) events.

Based on the available data, we conclude that it is not possible to determine whether statins reduce symptomatic heart disease as secondary prevention for older adults who are frail.

Do statins reduce heart disease in combined populations?

In the combined primary and secondary population from PROSPER,13 pravastatin decreased the risk of nonfatal symptomatic MI from 4.3% in the placebo group to 3.4%, a relatively small reduction in absolute risk (0.9%) and not statistically significant by our chi-square calculation (P = .099).

Do statins prevent a first symptomatic stroke in people with or without preexisting cardiovascular disease?

Preventing strokes that cause functional decline is an important outcome for the frail elderly. Stroke outcomes from PROSPER,13 JUPITER,29 and the Afilalo et al meta-analysis31 are summarized in Table 3.

For primary prevention:

In PROSPER (primary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

JUPITER,29 in contrast, found that rosuvastatin 20 mg reduced strokes in primary prevention, but the absolute benefit was small. In 2 years, 0.8% of the treatment group had strokes, compared with 1.4% with placebo, an absolute risk reduction of 0.6% (P = .023, number needed to treat 161, 95% CI 86–1,192).

Neither PROSPER nor JUPITER differentiated between disabling and nondisabling strokes.

For secondary prevention:

In PROSPER (secondary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

The Afilalo et al secondary prevention meta-analysis demonstrated a 25% relative reduction in stroke (relative risk 0.75, 95% CI 0.56–0.94, number needed to treat 58, 95% CI 27–177).31

Notably, the stroke outcome in Afilalo included both disabling and nondisabling strokes. For example, in the Heart Protection Study,20 the largest study in the Afilalo et al meta-analysis, approximately 50% of nonfatal, classifiable strokes in the overall study population (ie, both younger and older patients) were not disabling. Including disabling and nondisabling strokes in a composite outcome confounds the clinical meaningfulness of these findings in frailty, as the number needed to treat to prevent one disabling stroke cannot be calculated from the data provided.

 

 

Do statins prevent a second (symptomatic) stroke in people with a previous stroke?

SPARCL27 (Table 3) examined the question of whether statins decrease the risk of recurrent ischemic stroke for patients with a prior history of stroke or transient ischemic attack. There was a statistically significant reduction in the primary composite outcome of fatal and nonfatal stroke, with 11.2% of the treatment group and 13.1% of the placebo group experiencing this outcome, an absolute risk reduction of 1.9% at 5 years (P = .03; number needed to treat 52, 95% CI 26–1,303). However, the difference in nonfatal stroke, which is the outcome of interest for frailty (since mortality has uncertain relevance), was not statistically significant (10.4% with treatment vs 11.8% with placebo, P =.11).

An exploratory subgroup analysis of SPARCL patients based on age30 showed a smaller, nonsignificant reduction in the primary end point of fatal and nonfatal stroke in the group over age 65 (relative risk 0.90, 95% confidence interval 0.73–1.11, P = .33) compared with the younger group (age < 65) (relative risk 0.74, 95% CI 0.57–0.96, P = .02).

The applicability of these results to the frail elderly is uncertain, since the subgroup analysis was not powered to determine outcomes based on age stratification and there were differences between groups in characteristics such as blood pressure and smoking status. In addition, the outcome of interest, nonfatal stroke, is not provided for the elderly subgroup.

In conclusion, in both primary and secondary prevention populations, the evidence that statins reduce nonfatal, symptomatic stroke rates for older adults is uncertain.

Do statins decrease all-cause mortality for primary or secondary prevention?

Due to competing risks for death, the outcome of mortality may not be relevant to those who are frail; however, studies showed the following:

For primary prevention, there was no decrease in mortality in PROSPER13 or in the elderly subgroup of JUPITER.29

For secondary prevention, an analysis of PROSPER trial data by Afilalo et al31 showed a significant 18% decrease in all-cause mortality (relative risk 0.82, 95% CI 0.69–0.98) using pravastatin 40 mg.

A decrease in all-cause mortality with statins was also reported in the pooled result of the Afilalo et al meta-analysis.31

What are the reported composite outcomes for primary and secondary prevention?

While we were most interested in the symptomatic outcomes described above, we recognize that the small numbers of events make it difficult to draw firm conclusions. Therefore, we also considered composite primary outcomes, even though most included multiple measures that have varying associations with disability and relevancy to frail older adults.

For primary prevention, in the PROSPER preplanned subgroup analysis,13 there was no statistical benefit for any outcome, including the primary composite measure. In contrast, the elderly subpopulation in the JUPITER trial28 showed a treatment benefit with rosuvastatin 20 mg compared with placebo for the primary composite outcome of MI, stroke, cardiovascular death, hospitalization for unstable angina, or revascularization. The number needed to treat for 2 years was 62 (95% CI 39–148).

In the CTT meta-analysis,32 patients at all levels of baseline risk showed benefit up to age 70. However, there was no statistically significant benefit in the composite primary outcome of coronary deaths, nonfatal myocardial infarction, ischemic stroke, or revascularization in the population most representative of elderly primary prevention—those who were more than 70 years old with a 5-year baseline risk of less than 20%.

For secondary prevention, in PROSPER,13 the subpopulation of patients treated for secondary prevention experienced benefit in the primary composite outcome of coronary heart disease death, nonfatal MI, or fatal or nonfatal stroke, achieving a 4% absolute risk reduction with a number needed to treat of 23 (95% CI 14–81) over 3 years.

Do statins decrease disability?

PROSPER was the only study that reported on disability. Compared with placebo, pravastatin did not decrease disability in the total population as measured by basic and instrumental activities of daily living scales.

Do statins help patients with heart failure?

Neither GISSI-HF25 nor CORONA26 found significant benefit from rosuvastatin 10 mg, despite LDL-C lowering of 27% in GISSI-HF and 45% in CORONA.

Do ezetimibe or other nonstatin lipid-lowering agents improve outcomes?

There is no definitive evidence that ezetimibe provides clinically meaningful benefit as a single agent.

For combination therapy, the IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial)53 showed that adding ezetimibe 10 mg to simvastatin 40 mg after an acute coronary syndrome reduced the risk of nonfatal myocardial infarction compared with simvastatin monotherapy (event rate 12.8% vs 14.4%; hazard ratio 0.87, 95% CI 0.80–0.95; P = .002) for a population with a mean age of 64. The risk of any stroke was also reduced; strokes occurred in 4.2% of those receiving combination therapy vs 4.8% with monotherapy (hazard ratio 0.86, 95% CI 0.73–1.00, P = .05). After a median of 6 years, 42% of patients in each group had discontinued treatment. Given the very specific clinical scenario of acute coronary syndrome and the young age of the patients in this trial, we do not think that this study justifies the use of ezetimibe for severely frail older adults.

There is no evidence that other combinations (ie, a statin plus another lipid-lowering drug) improve clinical outcomes for either primary or secondary prevention in any population.54

WILL FRAIL PATIENTS LIVE LONG ENOUGH TO BENEFIT?

It is often difficult to determine the number of years that are needed to achieve benefit, as most trials do not provide a statistical analysis of varying time frames.

The PROSPER trial13 lasted 3.2 years. From the Kaplan-Meier curves in PROSPER, we estimate that it took about 1.5 years to achieve a 1% absolute risk reduction and 2.5 years for a 2% absolute risk reduction in coronary heart disease death and nonfatal MI in the combined primary and secondary groups.

JUPITER28 was stopped early at 1.9 years. The Afilalo et al meta-analysis31 was based on follow-up over 4.9 years.

IMPROVE-IT53 reported event rates at 7 years. The authors note that benefit in the primary composite outcome appeared to emerge at 1 year, although no statistical support is given for this statement and divergence in the Kaplan-Meier curves is not visually apparent.

The duration of other studies ranged between 2.7 and 4.9 years (Table 1).26–28

It has been suggested that statins should be considered for elderly patients who have a life expectancy of at least 5 years.3 However, many older adults have already been taking statins for many years, which makes it difficult to interpret the available timeframe evidence.

In a multicenter, unblinded, randomized trial,55 statins were either stopped or continued in older adults who had a short life expectancy and a median survival of approximately 7 months. Causes of death were evenly divided between cancer and noncancer diagnoses, and 22% of the patients were cognitively impaired. Discontinuing statin therapy did not increase mortality or cardiovascular events within 60 days. Nevertheless, stopping statin therapy did not achieve noninferiority for the primary end point, the proportion of participants who died within 60 days. Statin discontinuation was associated with improved quality of life, although the study was not blinded, which could have influenced results.

HAVE THE HARMS BEEN SUFFICIENTLY CONSIDERED?

Frail older adults commonly take multiple medications and are more vulnerable to adverse events.56

Many statins require dose reduction with severe renal impairment (creatinine clearance < 30 mL/min/1.73 m2), which would be a common consideration in severely frail older adults.

Myopathy

Myopathy, which includes myalgias and muscle weakness, is a statin-related adverse event that can impair quality of life. Myopathy typically develops within the first 6 months but can occur at any time during statin treatment.57 When muscle-related adverse effects occur, they may affect the elderly more significantly, particularly their ability to perform activities of daily living, rise from a chair, or mobilize independently. Another concern is that older adults with dementia may not be able to accurately report muscle-related symptoms.

It is difficult to ascertain the true prevalence of myopathy, especially in advanced age and frailty. Randomized controlled trials report incidence rates of 1.5% to 5%, which is comparable to placebo.57,58 However, inconsistent definitions of myopathy and exclusion of subjects with previous statin intolerance or adverse effects during run-in periods limit interpretability.57 Clinical experience suggests that muscle complaints may be relatively common.59–61

Advanced age, female sex, low body mass index, and multisystem disease are all associated with frailty and have also been described as risk factors for statin-associated muscle syndromes.61 Physiologic changes associated with frailty, such as reduced muscle strength, decreased lean body mass, impaired functional mobility, decreased reserve capacity, and altered drug metabolism may increase the risk and severity of myopathy.62

Adverse cognitive events

Meta-analyses of randomized clinical trials and narrative reviews find no definitive relationship between statin therapy and adverse cognitive events.63–67 Nevertheless, there have been case reports of memory loss associated with the use of statins, and the US Food and Drug Administration has issued a warning that statins have been associated with memory loss and confusion.68

It may be difficult to determine whether a statin is causing or aggravating cognitive symptoms among individuals with dementia without a trial withdrawal of the drug.

OUR RECOMMENDATIONS

The recommendations below are intended for adults with severe or very severe frailty (ie, a score of 7 or 8 on the Clinical Frailty Scale11 or FACT5 and therefore apply to most older adults living in long-term care facilities.

Primary prevention

There is no reason to prescribe or continue statins for primary prevention, as it is unlikely that they would provide benefit for outcomes that are relevant in this population.

Secondary prevention

Statin treatment is probably not necessary for secondary prevention in those with severe frailty, although there may be extenuating circumstances that justify statin use.

Heart failure

There is no reason to start or continue statins for heart failure, as there is insufficient evidence that they are effective for this indication in any population.

Ezetimibe

There is no evidence that ezetimibe reduces cardiovascular events in any population when used as monotherapy. For a select population with acute coronary syndromes, ezetimibe has a modest effect. Given the very specific clinical scenario of acute coronary syndrome, we do not think that the available evidence justifies the use of ezetimibe for severely frail older adults.

Agents other than ezetimibe combined with statins

There is no reason to start or continue other lipid-lowering drugs in conjunction with statins.

Statin dosing

As statin adverse effects have the potential to increase with advancing age and frailty, lower doses may be appropriate.68

Adverse events

Consider stopping statins on a trial basis if there is concern regarding myopathy, drug interactions, or other adverse effects.

BOTTOM LINE: DO STATINS IMPROVE QUALITY OF LIFE OR FUNCTION?

In primary prevention for older adults, there is doubt that statins prevent cardiovascular disease and stroke-related events because the main study involving the elderly did not show a benefit in the primary prevention subgroup.13 Additionally, there is no conclusive evidence that statin treatment decreases mortality in primary prevention.13,29

There is insufficient information to determine whether the frail elderly should receive statins for secondary prevention. Although there is evidence that treatment decreases measures of coronary heart disease and stroke, it is unclear whether it improves quality of life or function for those who are frail. To answer this question, we need more information about whether reported outcomes (such as stroke and MI) are associated with disability, which is not provided in many of the studies we reviewed. When disability was specifically considered in the PROSPER trial for the combined population of primary and secondary prevention, treatment with statins had no impact on basic and instrumental activities of daily living.

Some experts may not agree with our interpretation of the complex evidence presented in this article. Others may ask, “What is the harm in using statins, even if there is no definitive benefit?” However, the harms associated with statin therapy for the frail are poorly defined. In the face of these uncertainties and in the absence of definitive improvement in quality of life, we believe that “less is more” in the context of severe frailty.69

The cost of medications should also be considered, especially in long-term care facilities, where there is an added expense of drug administration that diverts human resources away from interactions that are more congruent with respecting the lifestage of frailty.

Careful review of evidence before applying clinical practice guidelines to those who are frail should become the norm. When considering treatment of frail patients, the five questions described in this review shed light on the applicability of clinical trial evidence. Therapies that are highly effective in healthier populations may be less effective when individuals are severely frail. Accordingly, we propose that medications should only be used if they improve quality of life or function.

Frail elderly patients are at high risk of adverse clinical outcomes, including those due to polypharmacy. Several groups tackle “deprescribing” by developing lists of medications that are potentially inappropriate for the elderly, such as the Beers or STOPP/START criteria.1–4

See related editorial

In contrast, our group (the Palliative and Therapeutic Harmonization [PATH] program and the Dalhousie Academic Detailing Service) has developed evidence-based, frailty-specific guidelines for treating hypertension5 and diabetes,6 in which we advocate less-stringent treatment targets and tapering or discontinuing medications, as needed.

The PATH program7 is a clinical approach that prioritizes the consideration of frailty when making treatment decisions. The Dalhousie Academic Detailing Service collaborates with the Nova Scotia Health Authority to research and develop evidence-informed educational messages about the treatment of common medical conditions.

Here, we address lipid-lowering therapy in this population.

CONSIDERING FRAILTY

Frailty is defined in several ways. The Fried model8,9 identifies frailty when 3 of the following characteristics are present: unintentional weight loss, exhaustion, muscle weakness, slow walking speed, or low levels of activity. The Clinical Frailty Scale10,11 and the Frailty Assessment for Care-planning Tool (FACT)5 use deficits in cognition, function, and mobility to define frailty. According to these scales, people are considered severely frail when they require assistance with basic activities of daily living (such as bathing or dressing), owing to cognitive or physical deficits from any cause.

In reviewing the evidence, we consider five questions:

  • What is the quality of the evidence? (Up to 48% of clinical practice guideline recommendations may be based on low-level evidence or expert opinion.12)
  • How did the study population compare with the frail?
  • Are study outcomes and potential benefits clinically relevant to those who are frail?
  • How long did it take for the clinical benefit of a treatment to become apparent, and are the frail elderly likely to live that long?
  • Have the harms of treatment been sufficiently considered?

WHAT IS THE QUALITY OF THE EVIDENCE?

We found no studies that specifically evaluated the benefit of lipid-lowering for severely frail older adults. Therefore, we examined randomized controlled trials that enrolled non-frail older adults,13–28 subgroup analyses of randomized controlled trials,29,30 meta-analyses that analyzed subgroups of elderly populations,31,32 and publications describing the study designs of randomized controlled trials.33–37

Most of the evidence comes from post hoc subgroup analyses of elderly populations. Although meta-analysis is commonly used to compare subgroups, the Cochrane handbook and others consider subgroup comparisons observational by nature.38,39 (See Table 1 for lipid-lowering studies discussed in this article.)

Studies of statins for primary prevention of cardiovascular disease

For evidence of benefit from lipid-lowering for primary prevention (ie, to reduce the risk of cardiovascular events in patients with no known cardiovascular disease at baseline but at increased risk), we reviewed the meta-analysis conducted by the Cholesterol Treatment Trialists’ (CTT) Collaborators.32 Since this meta-analysis included the major trials that enrolled elderly patients, individual publications of post hoc, elderly subgroups were, for the most part, not examined individually. The exception to this approach was a decision to report on the PROSPER13 and JUPITER28 trials separately, because PROSPER is the most representative of the elderly population and JUPITER reached the lowest LDL-C of primary prevention trials published to date and included a large elderly subgroup (n = 5,695).

Savarese et al40 evaluated the benefits of statins for older adults who did not have established cardiovascular disease. We did not report on this meta-analysis, as not all of the subjects that populated the meta-analysis were representative of a typical prevention population. For instance, in the Anglo-Scandinavian Cardiac Outcomes Trial lipid-lowering arm,41 14% of the subjects had had a previous stroke or transient ischemic attack. In the Antihypertensive and Lipid-Lowering Treatment Trial,42 16% of the population had a family history of premature coronary heart disease.

In addition, all the trials in the Savarese meta-analysis were also included in the CTT meta-analysis.32 The CTT reports on baseline risk using patient-level data stratified by age and risk, which may be more relevant to the question of primary prevention for older adults, as highlighted in our review.

PROSPER (Prospective Study of Pravastatin in the Elderly at Risk),13 a well-conducted, double-blind, randomized controlled trial with low probability of bias, compared pravastatin 40 mg and placebo. It was the only study that specifically enrolled older adults, with prespecified analysis of primary and secondary prevention subgroups. The primary prevention subgroup accounted for 56% of the 5,084 participants.

JUPITER (Justification for the Use of Statins in Prevention)28 compared rosuvastatin 20 mg and placebo in 17,802 participants. All had low-density lipoprotein cholesterol (LDL-C) levels below 3.4 mmol/L (130 mg/dL) and elevated levels of the inflammatory biomarker high-sensitivity C-reactive protein (hsCRP), ie, 2 mg/L or higher. Subsequently, Glynn et al performed a post hoc, exploratory subgroup analysis of elderly participants (N = 5,695).29

The JUPITER trial had several limitations.43,44 The planned follow-up period was 5 years, but the trial was stopped early at 1.9 years, after a statistically significant difference was detected in the primary composite outcome of reduction in all vascular events. Studies that are stopped early may exaggerate positive findings.45

Further, JUPITER’s patients were a select group, with normal LDL-C levels, elevated hsCRP values, and without diabetes. Of 90,000 patients screened, 72,000 (80%) did not meet the inclusion criteria and were not enrolled. This high rate of exclusion limits the generalizability of study findings beyond the shortcomings of post hoc subgroup analysis.

The meta-analysis performed by the CTT Collaborators32 used individual participant data from large-scale randomized trials of lipid-modifying treatment. This analysis was specific to people at low risk of vascular disease. In a supplementary appendix, the authors described the reduction in major vascular events for each 1.0 mmol/L decrease in LDL-C in three age categories: under age 60, ages 61 to 70, and over age 70.

The authors also stratified the results by risk category and provided information about those with a risk of major vascular events of less than 20%, which would be more representative of a purer primary prevention population.

For the elderly subgroup at low risk, the CTT Collaborators32 only reported a composite of major vascular events (coronary death, nonfatal myocardial infarction [MI], ischemic stroke, or revascularization) and did not describe individual outcomes, such as prevention of coronary heart disease.

Study results are based on postrandomization findings and therefore may be observational, not experimental.46

Studies of statins for secondary prevention of cardiovascular disease

The aim of secondary prevention is to reduce the risk of recurrent cardiovascular events in patients who already have cardiovascular disease.

To address the question of whether statins reduce cardiovascular risk, we reviewed:

PROSPER,13 which included a preplanned analysis of the secondary prevention population.

Afilalo et al,31,47 who performed a meta-analysis of the elderly subgroups of nine major secondary prevention studies (19,569 patients) using published and unpublished data.

To address the question of whether statins benefit individuals with heart failure, we found two relevant studies:

GISSI-HF (Gruppo Italiano per lo Studio della Sopravvivenza nell’Insufficienza Cardiaca Heart Failure)25 and CORONA (Controlled Rosuvastatin Multinational Trial in Heart Failure),26 which were large, international, well-conducted randomized controlled trials that examined statin use in heart failure.

To answer the question of whether statins benefit individuals after a stroke or transient ischemic attack, we found one relevant study:

SPARCL (Stroke Prevention by Aggressive Reduction in Cholesterol Levels),27 which evaluated the benefit of statins in older adults with a history of stroke or transient ischemic attack. It was a prospective, double-blind, placebo-controlled, international trial conducted at 205 centers. One to 6 months after their cerebrovascular event, patients were randomized to receive either atorvastatin 80 mg or placebo. Given the young age of patients in this trial (mean age 63), we also reviewed a post hoc subgroup analysis of the elderly patients in SPARCL (age > 65).30

 

 

HOW DID THE STUDY POPULATION COMPARE WITH THOSE WHO ARE FRAIL?

Frail older adults are almost always excluded from large-scale clinical trials,48 leading to uncertainty about whether the conclusions can be applied to those with advanced frailty.

Although age is an imperfect proxy measure of frailty,49 we consider the age of the study population as well as their comorbidities.

Participants in the studies we reviewed were generally younger and healthier than those who are frail, with mean ages of about 75 or less (Table 1).

PROSPER was the most representative study, as it specifically enrolled older adults, albeit without frailty,13 and excluded people with poor cognitive function as defined by a Mini Mental State Examination score less than 24.

JUPITER enrolled a select population, as described above. The median age in the elderly subgroup was 74 (interquartile range 72–78).29

The Afilalo et al31 meta-analysis primarily included studies of young-elderly patients, with a mean age of less than 70. PROSPER13 was an exception.

The GISSI-HF study,25 which examined the benefit of statins in heart failure, described their study population as frail, although the mean age was only 68. Compared with those in GISSI-HF, the CORONA patients26 with heart failure were older (mean age 73) and had more severe heart failure. Accordingly, it is possible that many of the CORONA participants were frail.

ARE STUDY OUTCOMES CLINICALLY RELEVANT TO THOSE WHO ARE FRAIL?

Because baseline cardiovascular risk increases with age, the elderly should, in theory, experience greater absolute benefit from lipid-lowering. However, there is uncertainty about whether this is true in practice.

Some, but not all, epidemiologic studies show a weaker relationship between cholesterol levels and cardiovascular morbidity and mortality rates in older compared to younger adults.50,51 This may be because those with high cholesterol levels die before they get old (time-related bias), or because those with life-threatening illness may have lower cholesterol levels.50 In addition, classic risk factors such as age, sex, systolic blood pressure, cholesterol values, diabetes, smoking, and left ventricular hypertrophy on electrocardiography may have less power to predict cardiovascular risk among older patients.52

The goal of treatment in frailty is to prevent further disability or improve quality of life. Therefore, meaningful outcomes for lipid-lowering therapy should include symptomatic nonfatal MI and its associated morbidity (eg, heart failure and persistent angina) or symptomatic nonfatal stroke leading to disability. Outcomes without sustained clinical impact, such as transient ischemic attack, nondisabling stroke, or silent MI, while potentially important in other populations, are less relevant in severe frailty. Notably, in many statin studies, outcomes include asymptomatic heart disease (eg, silent MI and “suspected events”) and nondisabling stroke (eg, mild stroke, transient ischemic attack). When symptomatic outcomes are not reported separately, the impact of the reported benefit on quality of life and function is uncertain.

The outcome of all-cause mortality is generally recognized as a gold standard for determining treatment benefit. However, since advanced frailty is characterized by multiple competing causes for mortality, a reduction in all-cause mortality that is achieved by addressing a single issue in nonfrail populations may not extend to the frail.

To more fully understand the impact of lipid-lowering therapy on quality of life and function, we examined the following questions:

Do statins as primary prevention reduce symptomatic heart disease?

Outcomes for coronary heart disease from PROSPER and JUPITER are summarized in Table 2.

PROSPER. In the PROSPER primary prevention group,13 statin therapy did not reduce the combined outcome of coronary heart disease death and nonfatal MI.

The JUPITER trial demonstrated a statistically significant benefit for preventing MI in the elderly subpopulation (ages 70–97),29 but the number needed to treat was high (211 for 2 years), with a wide confidence interval (CI) (95% CI 106–32,924). The trial did not adequately differentiate between symptomatic and asymptomatic events, making it difficult to determine outcome relevance. Also, due to the methodologic limitations of JUPITER as described above, its results should be interpreted with caution.43,44

The CTT Collaborators32 did not report individual outcomes (eg, coronary heart disease) for the elderly low-risk subgroup and, therefore, this meta-analysis does not answer the question of whether statins reduce symptomatic heart disease in primary prevention populations.

Taken together, these findings do not provide convincing evidence that statin therapy as primary prevention reduces the incidence of symptomatic heart disease for severely frail older adults.

Do statins as secondary prevention reduce symptomatic heart disease?

Most studies defined secondary prevention narrowly as treatment for patients with established coronary artery disease. For instance, in the Afilalo et al meta-analysis,31 the small number of studies that included individuals with other forms of vascular disease (such as peripheral vascular disease) enrolled few participants with noncardiac conditions (eg, 29% in PROSPER13 and 13% in the Heart Protection Study20).

Therefore, any evidence of benefit for secondary prevention demonstrated in these studies is most applicable to patients with coronary heart disease, with less certainty for those with other forms of cardiovascular disease.

In PROSPER,13 the secondary prevention group experienced benefit in the combined outcome of coronary heart disease death or nonfatal MI. In the treatment group, 12.7% experienced this outcome compared with 16.8% with placebo, an absolute risk reduction of 4.1% in 3 years (P = .004, number needed to treat 25, 95% CI 15–77). This measure includes coronary heart disease death, an outcome that may not be generalizable to those who are frail. In addition, the outcome of nonfatal MI includes both symptomatic and suspected events. As such, there is uncertainty whether the realized benefit is clinically relevant to frail older adults.

The Afilalo et al meta-analysis31 showed that the number needed to treat to prevent one nonfatal MI was 38 (95% CI 16–118) over 5 years (Table 2). However, this outcome included both symptomatic and asymptomatic (silent) events.

Based on the available data, we conclude that it is not possible to determine whether statins reduce symptomatic heart disease as secondary prevention for older adults who are frail.

Do statins reduce heart disease in combined populations?

In the combined primary and secondary population from PROSPER,13 pravastatin decreased the risk of nonfatal symptomatic MI from 4.3% in the placebo group to 3.4%, a relatively small reduction in absolute risk (0.9%) and not statistically significant by our chi-square calculation (P = .099).

Do statins prevent a first symptomatic stroke in people with or without preexisting cardiovascular disease?

Preventing strokes that cause functional decline is an important outcome for the frail elderly. Stroke outcomes from PROSPER,13 JUPITER,29 and the Afilalo et al meta-analysis31 are summarized in Table 3.

For primary prevention:

In PROSPER (primary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

JUPITER,29 in contrast, found that rosuvastatin 20 mg reduced strokes in primary prevention, but the absolute benefit was small. In 2 years, 0.8% of the treatment group had strokes, compared with 1.4% with placebo, an absolute risk reduction of 0.6% (P = .023, number needed to treat 161, 95% CI 86–1,192).

Neither PROSPER nor JUPITER differentiated between disabling and nondisabling strokes.

For secondary prevention:

In PROSPER (secondary prevention),13 there was no statistically significant benefit in the combined outcome of fatal and nonfatal stroke or the single outcome of transient ischemic attack after 3.2 years.

The Afilalo et al secondary prevention meta-analysis demonstrated a 25% relative reduction in stroke (relative risk 0.75, 95% CI 0.56–0.94, number needed to treat 58, 95% CI 27–177).31

Notably, the stroke outcome in Afilalo included both disabling and nondisabling strokes. For example, in the Heart Protection Study,20 the largest study in the Afilalo et al meta-analysis, approximately 50% of nonfatal, classifiable strokes in the overall study population (ie, both younger and older patients) were not disabling. Including disabling and nondisabling strokes in a composite outcome confounds the clinical meaningfulness of these findings in frailty, as the number needed to treat to prevent one disabling stroke cannot be calculated from the data provided.

 

 

Do statins prevent a second (symptomatic) stroke in people with a previous stroke?

SPARCL27 (Table 3) examined the question of whether statins decrease the risk of recurrent ischemic stroke for patients with a prior history of stroke or transient ischemic attack. There was a statistically significant reduction in the primary composite outcome of fatal and nonfatal stroke, with 11.2% of the treatment group and 13.1% of the placebo group experiencing this outcome, an absolute risk reduction of 1.9% at 5 years (P = .03; number needed to treat 52, 95% CI 26–1,303). However, the difference in nonfatal stroke, which is the outcome of interest for frailty (since mortality has uncertain relevance), was not statistically significant (10.4% with treatment vs 11.8% with placebo, P =.11).

An exploratory subgroup analysis of SPARCL patients based on age30 showed a smaller, nonsignificant reduction in the primary end point of fatal and nonfatal stroke in the group over age 65 (relative risk 0.90, 95% confidence interval 0.73–1.11, P = .33) compared with the younger group (age < 65) (relative risk 0.74, 95% CI 0.57–0.96, P = .02).

The applicability of these results to the frail elderly is uncertain, since the subgroup analysis was not powered to determine outcomes based on age stratification and there were differences between groups in characteristics such as blood pressure and smoking status. In addition, the outcome of interest, nonfatal stroke, is not provided for the elderly subgroup.

In conclusion, in both primary and secondary prevention populations, the evidence that statins reduce nonfatal, symptomatic stroke rates for older adults is uncertain.

Do statins decrease all-cause mortality for primary or secondary prevention?

Due to competing risks for death, the outcome of mortality may not be relevant to those who are frail; however, studies showed the following:

For primary prevention, there was no decrease in mortality in PROSPER13 or in the elderly subgroup of JUPITER.29

For secondary prevention, an analysis of PROSPER trial data by Afilalo et al31 showed a significant 18% decrease in all-cause mortality (relative risk 0.82, 95% CI 0.69–0.98) using pravastatin 40 mg.

A decrease in all-cause mortality with statins was also reported in the pooled result of the Afilalo et al meta-analysis.31

What are the reported composite outcomes for primary and secondary prevention?

While we were most interested in the symptomatic outcomes described above, we recognize that the small numbers of events make it difficult to draw firm conclusions. Therefore, we also considered composite primary outcomes, even though most included multiple measures that have varying associations with disability and relevancy to frail older adults.

For primary prevention, in the PROSPER preplanned subgroup analysis,13 there was no statistical benefit for any outcome, including the primary composite measure. In contrast, the elderly subpopulation in the JUPITER trial28 showed a treatment benefit with rosuvastatin 20 mg compared with placebo for the primary composite outcome of MI, stroke, cardiovascular death, hospitalization for unstable angina, or revascularization. The number needed to treat for 2 years was 62 (95% CI 39–148).

In the CTT meta-analysis,32 patients at all levels of baseline risk showed benefit up to age 70. However, there was no statistically significant benefit in the composite primary outcome of coronary deaths, nonfatal myocardial infarction, ischemic stroke, or revascularization in the population most representative of elderly primary prevention—those who were more than 70 years old with a 5-year baseline risk of less than 20%.

For secondary prevention, in PROSPER,13 the subpopulation of patients treated for secondary prevention experienced benefit in the primary composite outcome of coronary heart disease death, nonfatal MI, or fatal or nonfatal stroke, achieving a 4% absolute risk reduction with a number needed to treat of 23 (95% CI 14–81) over 3 years.

Do statins decrease disability?

PROSPER was the only study that reported on disability. Compared with placebo, pravastatin did not decrease disability in the total population as measured by basic and instrumental activities of daily living scales.

Do statins help patients with heart failure?

Neither GISSI-HF25 nor CORONA26 found significant benefit from rosuvastatin 10 mg, despite LDL-C lowering of 27% in GISSI-HF and 45% in CORONA.

Do ezetimibe or other nonstatin lipid-lowering agents improve outcomes?

There is no definitive evidence that ezetimibe provides clinically meaningful benefit as a single agent.

For combination therapy, the IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial)53 showed that adding ezetimibe 10 mg to simvastatin 40 mg after an acute coronary syndrome reduced the risk of nonfatal myocardial infarction compared with simvastatin monotherapy (event rate 12.8% vs 14.4%; hazard ratio 0.87, 95% CI 0.80–0.95; P = .002) for a population with a mean age of 64. The risk of any stroke was also reduced; strokes occurred in 4.2% of those receiving combination therapy vs 4.8% with monotherapy (hazard ratio 0.86, 95% CI 0.73–1.00, P = .05). After a median of 6 years, 42% of patients in each group had discontinued treatment. Given the very specific clinical scenario of acute coronary syndrome and the young age of the patients in this trial, we do not think that this study justifies the use of ezetimibe for severely frail older adults.

There is no evidence that other combinations (ie, a statin plus another lipid-lowering drug) improve clinical outcomes for either primary or secondary prevention in any population.54

WILL FRAIL PATIENTS LIVE LONG ENOUGH TO BENEFIT?

It is often difficult to determine the number of years that are needed to achieve benefit, as most trials do not provide a statistical analysis of varying time frames.

The PROSPER trial13 lasted 3.2 years. From the Kaplan-Meier curves in PROSPER, we estimate that it took about 1.5 years to achieve a 1% absolute risk reduction and 2.5 years for a 2% absolute risk reduction in coronary heart disease death and nonfatal MI in the combined primary and secondary groups.

JUPITER28 was stopped early at 1.9 years. The Afilalo et al meta-analysis31 was based on follow-up over 4.9 years.

IMPROVE-IT53 reported event rates at 7 years. The authors note that benefit in the primary composite outcome appeared to emerge at 1 year, although no statistical support is given for this statement and divergence in the Kaplan-Meier curves is not visually apparent.

The duration of other studies ranged between 2.7 and 4.9 years (Table 1).26–28

It has been suggested that statins should be considered for elderly patients who have a life expectancy of at least 5 years.3 However, many older adults have already been taking statins for many years, which makes it difficult to interpret the available timeframe evidence.

In a multicenter, unblinded, randomized trial,55 statins were either stopped or continued in older adults who had a short life expectancy and a median survival of approximately 7 months. Causes of death were evenly divided between cancer and noncancer diagnoses, and 22% of the patients were cognitively impaired. Discontinuing statin therapy did not increase mortality or cardiovascular events within 60 days. Nevertheless, stopping statin therapy did not achieve noninferiority for the primary end point, the proportion of participants who died within 60 days. Statin discontinuation was associated with improved quality of life, although the study was not blinded, which could have influenced results.

HAVE THE HARMS BEEN SUFFICIENTLY CONSIDERED?

Frail older adults commonly take multiple medications and are more vulnerable to adverse events.56

Many statins require dose reduction with severe renal impairment (creatinine clearance < 30 mL/min/1.73 m2), which would be a common consideration in severely frail older adults.

Myopathy

Myopathy, which includes myalgias and muscle weakness, is a statin-related adverse event that can impair quality of life. Myopathy typically develops within the first 6 months but can occur at any time during statin treatment.57 When muscle-related adverse effects occur, they may affect the elderly more significantly, particularly their ability to perform activities of daily living, rise from a chair, or mobilize independently. Another concern is that older adults with dementia may not be able to accurately report muscle-related symptoms.

It is difficult to ascertain the true prevalence of myopathy, especially in advanced age and frailty. Randomized controlled trials report incidence rates of 1.5% to 5%, which is comparable to placebo.57,58 However, inconsistent definitions of myopathy and exclusion of subjects with previous statin intolerance or adverse effects during run-in periods limit interpretability.57 Clinical experience suggests that muscle complaints may be relatively common.59–61

Advanced age, female sex, low body mass index, and multisystem disease are all associated with frailty and have also been described as risk factors for statin-associated muscle syndromes.61 Physiologic changes associated with frailty, such as reduced muscle strength, decreased lean body mass, impaired functional mobility, decreased reserve capacity, and altered drug metabolism may increase the risk and severity of myopathy.62

Adverse cognitive events

Meta-analyses of randomized clinical trials and narrative reviews find no definitive relationship between statin therapy and adverse cognitive events.63–67 Nevertheless, there have been case reports of memory loss associated with the use of statins, and the US Food and Drug Administration has issued a warning that statins have been associated with memory loss and confusion.68

It may be difficult to determine whether a statin is causing or aggravating cognitive symptoms among individuals with dementia without a trial withdrawal of the drug.

OUR RECOMMENDATIONS

The recommendations below are intended for adults with severe or very severe frailty (ie, a score of 7 or 8 on the Clinical Frailty Scale11 or FACT5 and therefore apply to most older adults living in long-term care facilities.

Primary prevention

There is no reason to prescribe or continue statins for primary prevention, as it is unlikely that they would provide benefit for outcomes that are relevant in this population.

Secondary prevention

Statin treatment is probably not necessary for secondary prevention in those with severe frailty, although there may be extenuating circumstances that justify statin use.

Heart failure

There is no reason to start or continue statins for heart failure, as there is insufficient evidence that they are effective for this indication in any population.

Ezetimibe

There is no evidence that ezetimibe reduces cardiovascular events in any population when used as monotherapy. For a select population with acute coronary syndromes, ezetimibe has a modest effect. Given the very specific clinical scenario of acute coronary syndrome, we do not think that the available evidence justifies the use of ezetimibe for severely frail older adults.

Agents other than ezetimibe combined with statins

There is no reason to start or continue other lipid-lowering drugs in conjunction with statins.

Statin dosing

As statin adverse effects have the potential to increase with advancing age and frailty, lower doses may be appropriate.68

Adverse events

Consider stopping statins on a trial basis if there is concern regarding myopathy, drug interactions, or other adverse effects.

BOTTOM LINE: DO STATINS IMPROVE QUALITY OF LIFE OR FUNCTION?

In primary prevention for older adults, there is doubt that statins prevent cardiovascular disease and stroke-related events because the main study involving the elderly did not show a benefit in the primary prevention subgroup.13 Additionally, there is no conclusive evidence that statin treatment decreases mortality in primary prevention.13,29

There is insufficient information to determine whether the frail elderly should receive statins for secondary prevention. Although there is evidence that treatment decreases measures of coronary heart disease and stroke, it is unclear whether it improves quality of life or function for those who are frail. To answer this question, we need more information about whether reported outcomes (such as stroke and MI) are associated with disability, which is not provided in many of the studies we reviewed. When disability was specifically considered in the PROSPER trial for the combined population of primary and secondary prevention, treatment with statins had no impact on basic and instrumental activities of daily living.

Some experts may not agree with our interpretation of the complex evidence presented in this article. Others may ask, “What is the harm in using statins, even if there is no definitive benefit?” However, the harms associated with statin therapy for the frail are poorly defined. In the face of these uncertainties and in the absence of definitive improvement in quality of life, we believe that “less is more” in the context of severe frailty.69

The cost of medications should also be considered, especially in long-term care facilities, where there is an added expense of drug administration that diverts human resources away from interactions that are more congruent with respecting the lifestage of frailty.

Careful review of evidence before applying clinical practice guidelines to those who are frail should become the norm. When considering treatment of frail patients, the five questions described in this review shed light on the applicability of clinical trial evidence. Therapies that are highly effective in healthier populations may be less effective when individuals are severely frail. Accordingly, we propose that medications should only be used if they improve quality of life or function.

References
  1. Ontario Pharmacy Research Collaboration. Deprescribing guidelines for the elderly. www.open-pharmacy-research.ca/research-projects/emerging-services/deprescribing-guidelines. Accessed December 28, 2016.
  2. Scott IA, Hilmer SN, Reeve E, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med 2015; 175:827–834.
  3. O’Mahony D, O’Sullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing 2015; 44:213–218.
  4. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc 2012; 60:616–631.
  5. Mallery LH, Allen M, Fleming I, et al. Promoting higher blood pressure targets for frail older adults: a consensus guideline from Canada. Cleve Clin J Med 2014; 81:427–437.
  6. Mallery LH, Ransom T, Steeves B, Cook B, Dunbar P, Moorhouse P. Evidence-informed guidelines for treating frail older adults with type 2 diabetes: from the Diabetes Care Program of Nova Scotia (DCPNS) and the Palliative and Therapeutic Harmonization (PATH) program. J Am Med Dir Assoc 2013; 14:801–808.
  7. Moorhouse P, Mallery L. Palliative and therapeutic harmonization: a model for appropriate decision-making in frail older adults. J Am Geriatr Soc 2012; 60:2326–2332.
  8. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study Collaborative Research Group. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
  9. Morley JE, Malmstrom TK, Miller DK. A simple frailty questionnaire (FRAIL) predicts outcomes in middle aged African Americans. J Nutr Health Aging 2012; 16:601–608.
  10. Rockwood K, Song Z, MacKnight C, et al. A global clinical measure of fitness and frailty in elderly people. CMAJ 2005; 173:489–495.
  11. Morley JE, Vellas B, van Kan GA, et al. Frailty consensus: a call to action. J Am Med Dir Assoc 2013; 14:392–397.
  12. Tricoci P, Allen JM, Kramer JM, Califf RM, Smith SC Jr. Scientific evidence underlying the ACC/AHA clinical practice guidelines. JAMA 2009; 301:831–841.
  13. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  14. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet 1994; 344:1383–1389.
  15. Miettien TA, Pyorala K, Olsson AG, et al. Cholesterol-lowering therapy in women and elderly patients with myocardial infarction or angina pectoris: findings from the Scandinavian Simvastatin Study Group (4S). Circulation 1997; 96:4211–4218.
  16. Lewis SJ, Moye LA, Sacks FM, et al. Effect of pravastatin on cardiovascular events in older patients with myocardial infarction and cholesterol levels in the average range. Results of the Cholesterol and Recurrent Events (CARE) trial. Ann Intern Med 1998; 129:681–689.
  17. Hunt D, Young P, Simes J, et al. Benefits of pravastatin on cardiovascular events and mortality in older patients with coronary heart disease are equal to or exceed those seen in younger patients: results from the LIPID trial. Ann Intern Med 2001; 134:931–940.
  18. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. N Engl J Med 1998; 339:1349–1357.
  19. Heart Protection Study Collaborative Group. The effects of cholesterol lowering with simvastatin on cause-specific mortality and on cancer incidence in 20,536 high-risk people: a randomized placebo-controlled trial. BMC Med 2005; 3:6.
  20. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomized placebo-controlled trial. Lancet 2002; 360:7–22.
  21. Pitt B, Mancini GB, Ellis SG, Rosman HS, Park JS, McGovern ME. Pravastatin limitation of atherosclerosis in the coronary arteries (PLAC 1): reduction in atherosclerosis progression and clinical events. PLAC 1 investigation. J Am Coll Cardiol 1995; 26:1133–1139.
  22. Jukema JW, Bruschke AV, van Boven AJ, et al. Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS). Circulation 1995; 91:2528–2540.
  23. Serruys PW, Foley DP, Jackson G, et al. A randomized placebo-controlled trial of fluvastatin for prevention of restenosis after successful coronary balloon angioplasty; final results of the fluvastatin angiographic restenosis (FLARE) trial. Eur Heart J 1999; 20:58–69.
  24. Serruys PW, de Feyter P, Macaya C, et al; Lescol Intervention Prevention Study (LIPS) Investigators. Fluvastatin for prevention of cardiac events following successful first percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 287:3215–3222.
  25. Tavazzi L, Maggioni AP, Marchioli R, et al; Gissi-HF Investigators. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomized, double-blind, placebo-controlled trial. Lancet 2008; 372:1231–1239.
  26. Kjekshus J, Apatrei E, Barrios V, et al; CORONA Group. Rosuvastatin in older patients with systolic heart failure. N Engl J Med 2007; 357:2248–2261.
  27. Amarenco P, Bogousslavsky J, Callahan A, et al; Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) Investigators. High-dose atorvastatin after stroke or transient ischemic attack. N Engl J Med 2006; 355:549–559.
  28. Ridker PM, Danielson E, Fonseca FA, et al; JUPITER Study Group. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med 2008; 359:2195–2207.
  29. Glynn RJ, Koenig W, Nordestgaard BG, Shepherd J, Ridker PM. Rosuvastatin for primary prevention in older persons with elevated C-reactive protein and low to average low-density lipoprotein cholesterol levels: exploratory analysis of a randomized trial. Ann Intern Med 2010; 152:488–496, W174.
  30. Chaturvedi S, Zivin J, Breazna A, et al; SPARCL Investigators. Effect of atorvastatin in elderly patients with a recent stroke or transient ischemic attack. Neurology 2009; 72:688–694.
  31. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  32. Cholesterol Treatment Trialists’ (CTT) Collaborators; Mihaylova B, Emberson J, Blackwell L, et al. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012; 380:581– 590.
  33. Sacks FM, Pfeffer MA, Moye L, et al. Rationale and design of a secondary prevention trial of lowering normal plasma cholesterol levels after acute myocardial infarction: the Cholesterol and Recurrent Events (CARE). Am J Cardiol 1991; 68:1436–1446.
  34. Armitage J, Collins R. Need for large scale randomised evidence about lowering LDL cholesterol in people with diabetes mellitus: MRC/BHF Heart Protection Study and other major trials. Heart 2000; 84:357–360.
  35. Design features and baseline characteristics of the LIPID (Long-Term Intervention with Pravastatin in Ischemic Disease) study: a randomized trial in patients with previous acute myocardial infarction and/or unstable angina pectoris. Am J Cardiol 1995; 76:474–479.
  36. Shepherd J, Blauw GJ, Murphy MB, et al. The design of a prospective study of Pravastatin in the Elderly at Risk (PROSPER). Am J Cardiol 1999; 84:1192–1197.
  37. Amarenco P, Bogousslavsky J, Callahan AS, et al; SPARCL Investigators. Design and baseline characteristics of the stroke prevention by aggressive reduction in cholesterol levels (SPARCL) study. Cerebrovasc Dis 2003; 16:389–395.
  38. Interpretation of subgroup analyses and meta-regressions. In: Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration, 2011. http://handbook.cochrane.org/chapter_9/9_6_6_interpretation_of_subgroup_analyses_and_meta_regressions.htm. Accessed December 5, 2016.
  39. Borenstein M, Higgins JP. Meta-analysis and subgroups. Prev Sci 2013; 14:134–143.
  40. Savarese G, Gotto AM Jr, Paolillo S, et al. Benefits of statins in elderly subjects without established cardiovascular disease: a meta-analysis. J Am Coll Cardiol 2013; 62:2090–2099.
  41. Sever PS, Dahlof B, Poulter NR, et al. Prevention of coronary and stroke events with atorvastatin in hypertensive patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet 2003; 361:1149–1158.
  42. The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial. Major outcomes in moderately hypercholesterolemic, hypertensive patients randomized to pravastatin vs usual care: The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT-LLT). JAMA 2002; 288:2998–3007.
  43. de Longeril M, Salen P, Abramson J, et al. Cholesterol lowering, cardiovascular diseases, and the rosuvastatin-JUPITER controversy: a critical reappraisal. Arch Intern Med 2010; 170:1032–1036.
  44. Yusuf S, Lonn E, Bosch J. Lipid lowering for primary prevention. Lancet 2009: 373:1152–1155.
  45. Briel M, Bassler D, Wang AT, Guyatt GH, Montori VM. The dangers of stopping a trial too early. J Bone Joint Surg Am 2012; 94(suppl 1):56–60.
  46. Hayward RA, Krumholz HM. Three reasons to abandon low-density lipoprotein targets: an open letter to the Adult Treatment Panel IV of the National Institutes of Health. Circ Cardiovasc Qual Outcomes 2012; 5:2–5.
  47. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical Bayesian meta-analysis. www.ncbi.nlm.nih.gov/pubmedhealth/PMH0026417. Accessed December 5, 2016.
  48. Holmes HM, Hayley DC, Alexander GC, Sachs GA. Reconsidering medication appropriateness for patients late in life. Arch Intern Med 2006; 166:605–609.
  49. Rockwood K, Mitnitski A. Frailty defined by deficit accumulation and geriatric medicine defined by frailty. Clin Geriatr Med 2011; 27:17–26.
  50. Petersen LK, Christensen K, Kragstrup J. Lipid-lowering treatment to the end? A review of observational studies and RCTs on cholesterol and mortality in 80+-year olds. Age Ageing 2010; 39:674–680.
  51. Psaty BM, Anderson M, Kronmal RA, et al. The association between lipid levels and the risks of incident myocardial infarction, stroke, and total mortality: the Cardiovascular Health Study. J Am Geriatr Soc 2004; 52:1639–1647.
  52. de Ruijter W, Westendorp RG, Assendelft WJ, et al. Use of Framingham risk score and new biomarkers to predict cardiovascular mortality in older people: population based observational cohort study. BMJ 2009; 338:a3083.
  53. Canon CP, Blazing MA, Giugliano RP, et al; IMPROVE-IT Investigators. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med 2015; 372:2387–2397.
  54. Anderson TJ, Gregoire J, Hegele RA, et al. 2012 update of the Canadian Cardiovascular Society guidelines for the diagnosis and treatment of dyslipidemia for the prevention of cardiovascular disease in the adult. Can J Cardiol 2013; 29:151–167.
  55. Kutner JS, Blatchford PJ, Taylor DH, et al. Safety and benefit of discontinuing statin therapy in the setting of advanced, life-limiting illness: a randomized clinical trial. JAMA Intern Med 2015; 175:691–700.
  56. Tinetti ME, Bogardus ST Jr, Agostini JV. Potential pitfalls of disease-specific guidelines for patients with multiple conditions. N Engl J Med 2004; 351:2870–2874.
  57. Rosenson RS. Current overview of statin-induced myopathy. Am J Med 2004; 116:408–416.
  58. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: proceedings of a Canadian Working Group Consensus Conference. Can J Cardiol 2011; 27:635–662.
  59. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208–215.
  60. Joy TR, Hegele RA. Narrative review: statin-related myopathy. Ann Intern Med 2009; 150:858–868.
  61. Talbert RL. Safety issues with statin therapy. J Am Pharm Assoc (2003) 2006; 46:479–490.
  62. Sewright KA, Clarkson PM, Thompson PD. Statin myopathy: incidence, risk factors, and pathophysiology. Curr Atheroscler Rep 2007; 9:389–396.
  63. Ott BR, Daiello LA, Dahabreh IJ, et al. Do statins impair cognition? A systematic review and meta-analysis of randomized controlled trials. J Gen Intern Med 2015; 30:348–358.
  64. Mancini GB, Tashakkor AY, Baker S, et al. Diagnosis, prevention and management of statin adverse effects and intolerance: Canadian Working Group Consensus update. Can J Cardiol 2013: 29:1553–1568.
  65. Rojas-Fernandez CH, Cameron JC. Is statin-associated cognitive impairment clinically relevant? A narrative review and clinical recommendations. Ann Pharmacother 2012; 46:549–557.
  66. McGuinness B, O’Hare J, Craig D, Bullock R, Malouf R, Passmore P. Cochrane review on ‘Statins for the treatment of dementia’. Int J Geriatr Psychiatry 2013; 28:119–126.
  67. Pandey RD, Gupta PP, Jha D, Kumar S. Role of statins in Alzheimer’s disease: a retrospective meta-analysis for commonly investigated clinical parameters in RCTs. Int J Neurosci 2013; 123:521–525.
  68. Food and Drug Administration (FDA). FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. www.fda.gov/drugs/ drugsafety/ucm293101.htm. Accessed December 5, 2016.
  69. Garfinkel D, Mangin D. Feasibility study of a systematic approach for discontinuation of multiple medications in older adults: addressing polypharmacy. Arch Intern Med 2010; 170:1648–1654.
References
  1. Ontario Pharmacy Research Collaboration. Deprescribing guidelines for the elderly. www.open-pharmacy-research.ca/research-projects/emerging-services/deprescribing-guidelines. Accessed December 28, 2016.
  2. Scott IA, Hilmer SN, Reeve E, et al. Reducing inappropriate polypharmacy: the process of deprescribing. JAMA Intern Med 2015; 175:827–834.
  3. O’Mahony D, O’Sullivan D, Byrne S, O’Connor MN, Ryan C, Gallagher P. STOPP/START criteria for potentially inappropriate prescribing in older people: version 2. Age Ageing 2015; 44:213–218.
  4. American Geriatrics Society 2012 Beers Criteria Update Expert Panel. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc 2012; 60:616–631.
  5. Mallery LH, Allen M, Fleming I, et al. Promoting higher blood pressure targets for frail older adults: a consensus guideline from Canada. Cleve Clin J Med 2014; 81:427–437.
  6. Mallery LH, Ransom T, Steeves B, Cook B, Dunbar P, Moorhouse P. Evidence-informed guidelines for treating frail older adults with type 2 diabetes: from the Diabetes Care Program of Nova Scotia (DCPNS) and the Palliative and Therapeutic Harmonization (PATH) program. J Am Med Dir Assoc 2013; 14:801–808.
  7. Moorhouse P, Mallery L. Palliative and therapeutic harmonization: a model for appropriate decision-making in frail older adults. J Am Geriatr Soc 2012; 60:2326–2332.
  8. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study Collaborative Research Group. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 2001; 56:M146–M156.
  9. Morley JE, Malmstrom TK, Miller DK. A simple frailty questionnaire (FRAIL) predicts outcomes in middle aged African Americans. J Nutr Health Aging 2012; 16:601–608.
  10. Rockwood K, Song Z, MacKnight C, et al. A global clinical measure of fitness and frailty in elderly people. CMAJ 2005; 173:489–495.
  11. Morley JE, Vellas B, van Kan GA, et al. Frailty consensus: a call to action. J Am Med Dir Assoc 2013; 14:392–397.
  12. Tricoci P, Allen JM, Kramer JM, Califf RM, Smith SC Jr. Scientific evidence underlying the ACC/AHA clinical practice guidelines. JAMA 2009; 301:831–841.
  13. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  14. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet 1994; 344:1383–1389.
  15. Miettien TA, Pyorala K, Olsson AG, et al. Cholesterol-lowering therapy in women and elderly patients with myocardial infarction or angina pectoris: findings from the Scandinavian Simvastatin Study Group (4S). Circulation 1997; 96:4211–4218.
  16. Lewis SJ, Moye LA, Sacks FM, et al. Effect of pravastatin on cardiovascular events in older patients with myocardial infarction and cholesterol levels in the average range. Results of the Cholesterol and Recurrent Events (CARE) trial. Ann Intern Med 1998; 129:681–689.
  17. Hunt D, Young P, Simes J, et al. Benefits of pravastatin on cardiovascular events and mortality in older patients with coronary heart disease are equal to or exceed those seen in younger patients: results from the LIPID trial. Ann Intern Med 2001; 134:931–940.
  18. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. N Engl J Med 1998; 339:1349–1357.
  19. Heart Protection Study Collaborative Group. The effects of cholesterol lowering with simvastatin on cause-specific mortality and on cancer incidence in 20,536 high-risk people: a randomized placebo-controlled trial. BMC Med 2005; 3:6.
  20. Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomized placebo-controlled trial. Lancet 2002; 360:7–22.
  21. Pitt B, Mancini GB, Ellis SG, Rosman HS, Park JS, McGovern ME. Pravastatin limitation of atherosclerosis in the coronary arteries (PLAC 1): reduction in atherosclerosis progression and clinical events. PLAC 1 investigation. J Am Coll Cardiol 1995; 26:1133–1139.
  22. Jukema JW, Bruschke AV, van Boven AJ, et al. Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS). Circulation 1995; 91:2528–2540.
  23. Serruys PW, Foley DP, Jackson G, et al. A randomized placebo-controlled trial of fluvastatin for prevention of restenosis after successful coronary balloon angioplasty; final results of the fluvastatin angiographic restenosis (FLARE) trial. Eur Heart J 1999; 20:58–69.
  24. Serruys PW, de Feyter P, Macaya C, et al; Lescol Intervention Prevention Study (LIPS) Investigators. Fluvastatin for prevention of cardiac events following successful first percutaneous coronary intervention: a randomized controlled trial. JAMA 2002; 287:3215–3222.
  25. Tavazzi L, Maggioni AP, Marchioli R, et al; Gissi-HF Investigators. Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomized, double-blind, placebo-controlled trial. Lancet 2008; 372:1231–1239.
  26. Kjekshus J, Apatrei E, Barrios V, et al; CORONA Group. Rosuvastatin in older patients with systolic heart failure. N Engl J Med 2007; 357:2248–2261.
  27. Amarenco P, Bogousslavsky J, Callahan A, et al; Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) Investigators. High-dose atorvastatin after stroke or transient ischemic attack. N Engl J Med 2006; 355:549–559.
  28. Ridker PM, Danielson E, Fonseca FA, et al; JUPITER Study Group. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med 2008; 359:2195–2207.
  29. Glynn RJ, Koenig W, Nordestgaard BG, Shepherd J, Ridker PM. Rosuvastatin for primary prevention in older persons with elevated C-reactive protein and low to average low-density lipoprotein cholesterol levels: exploratory analysis of a randomized trial. Ann Intern Med 2010; 152:488–496, W174.
  30. Chaturvedi S, Zivin J, Breazna A, et al; SPARCL Investigators. Effect of atorvastatin in elderly patients with a recent stroke or transient ischemic attack. Neurology 2009; 72:688–694.
  31. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  32. Cholesterol Treatment Trialists’ (CTT) Collaborators; Mihaylova B, Emberson J, Blackwell L, et al. The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012; 380:581– 590.
  33. Sacks FM, Pfeffer MA, Moye L, et al. Rationale and design of a secondary prevention trial of lowering normal plasma cholesterol levels after acute myocardial infarction: the Cholesterol and Recurrent Events (CARE). Am J Cardiol 1991; 68:1436–1446.
  34. Armitage J, Collins R. Need for large scale randomised evidence about lowering LDL cholesterol in people with diabetes mellitus: MRC/BHF Heart Protection Study and other major trials. Heart 2000; 84:357–360.
  35. Design features and baseline characteristics of the LIPID (Long-Term Intervention with Pravastatin in Ischemic Disease) study: a randomized trial in patients with previous acute myocardial infarction and/or unstable angina pectoris. Am J Cardiol 1995; 76:474–479.
  36. Shepherd J, Blauw GJ, Murphy MB, et al. The design of a prospective study of Pravastatin in the Elderly at Risk (PROSPER). Am J Cardiol 1999; 84:1192–1197.
  37. Amarenco P, Bogousslavsky J, Callahan AS, et al; SPARCL Investigators. Design and baseline characteristics of the stroke prevention by aggressive reduction in cholesterol levels (SPARCL) study. Cerebrovasc Dis 2003; 16:389–395.
  38. Interpretation of subgroup analyses and meta-regressions. In: Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration, 2011. http://handbook.cochrane.org/chapter_9/9_6_6_interpretation_of_subgroup_analyses_and_meta_regressions.htm. Accessed December 5, 2016.
  39. Borenstein M, Higgins JP. Meta-analysis and subgroups. Prev Sci 2013; 14:134–143.
  40. Savarese G, Gotto AM Jr, Paolillo S, et al. Benefits of statins in elderly subjects without established cardiovascular disease: a meta-analysis. J Am Coll Cardiol 2013; 62:2090–2099.
  41. Sever PS, Dahlof B, Poulter NR, et al. Prevention of coronary and stroke events with atorvastatin in hypertensive patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm (ASCOT-LLA): a multicentre randomised controlled trial. Lancet 2003; 361:1149–1158.
  42. The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial. Major outcomes in moderately hypercholesterolemic, hypertensive patients randomized to pravastatin vs usual care: The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT-LLT). JAMA 2002; 288:2998–3007.
  43. de Longeril M, Salen P, Abramson J, et al. Cholesterol lowering, cardiovascular diseases, and the rosuvastatin-JUPITER controversy: a critical reappraisal. Arch Intern Med 2010; 170:1032–1036.
  44. Yusuf S, Lonn E, Bosch J. Lipid lowering for primary prevention. Lancet 2009: 373:1152–1155.
  45. Briel M, Bassler D, Wang AT, Guyatt GH, Montori VM. The dangers of stopping a trial too early. J Bone Joint Surg Am 2012; 94(suppl 1):56–60.
  46. Hayward RA, Krumholz HM. Three reasons to abandon low-density lipoprotein targets: an open letter to the Adult Treatment Panel IV of the National Institutes of Health. Circ Cardiovasc Qual Outcomes 2012; 5:2–5.
  47. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical Bayesian meta-analysis. www.ncbi.nlm.nih.gov/pubmedhealth/PMH0026417. Accessed December 5, 2016.
  48. Holmes HM, Hayley DC, Alexander GC, Sachs GA. Reconsidering medication appropriateness for patients late in life. Arch Intern Med 2006; 166:605–609.
  49. Rockwood K, Mitnitski A. Frailty defined by deficit accumulation and geriatric medicine defined by frailty. Clin Geriatr Med 2011; 27:17–26.
  50. Petersen LK, Christensen K, Kragstrup J. Lipid-lowering treatment to the end? A review of observational studies and RCTs on cholesterol and mortality in 80+-year olds. Age Ageing 2010; 39:674–680.
  51. Psaty BM, Anderson M, Kronmal RA, et al. The association between lipid levels and the risks of incident myocardial infarction, stroke, and total mortality: the Cardiovascular Health Study. J Am Geriatr Soc 2004; 52:1639–1647.
  52. de Ruijter W, Westendorp RG, Assendelft WJ, et al. Use of Framingham risk score and new biomarkers to predict cardiovascular mortality in older people: population based observational cohort study. BMJ 2009; 338:a3083.
  53. Canon CP, Blazing MA, Giugliano RP, et al; IMPROVE-IT Investigators. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med 2015; 372:2387–2397.
  54. Anderson TJ, Gregoire J, Hegele RA, et al. 2012 update of the Canadian Cardiovascular Society guidelines for the diagnosis and treatment of dyslipidemia for the prevention of cardiovascular disease in the adult. Can J Cardiol 2013; 29:151–167.
  55. Kutner JS, Blatchford PJ, Taylor DH, et al. Safety and benefit of discontinuing statin therapy in the setting of advanced, life-limiting illness: a randomized clinical trial. JAMA Intern Med 2015; 175:691–700.
  56. Tinetti ME, Bogardus ST Jr, Agostini JV. Potential pitfalls of disease-specific guidelines for patients with multiple conditions. N Engl J Med 2004; 351:2870–2874.
  57. Rosenson RS. Current overview of statin-induced myopathy. Am J Med 2004; 116:408–416.
  58. Mancini GB, Baker S, Bergeron J, et al. Diagnosis, prevention, and management of statin adverse effects and intolerance: proceedings of a Canadian Working Group Consensus Conference. Can J Cardiol 2011; 27:635–662.
  59. Cohen JD, Brinton EA, Ito MK, Jacobson TA. Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users. J Clin Lipidol 2012; 6:208–215.
  60. Joy TR, Hegele RA. Narrative review: statin-related myopathy. Ann Intern Med 2009; 150:858–868.
  61. Talbert RL. Safety issues with statin therapy. J Am Pharm Assoc (2003) 2006; 46:479–490.
  62. Sewright KA, Clarkson PM, Thompson PD. Statin myopathy: incidence, risk factors, and pathophysiology. Curr Atheroscler Rep 2007; 9:389–396.
  63. Ott BR, Daiello LA, Dahabreh IJ, et al. Do statins impair cognition? A systematic review and meta-analysis of randomized controlled trials. J Gen Intern Med 2015; 30:348–358.
  64. Mancini GB, Tashakkor AY, Baker S, et al. Diagnosis, prevention and management of statin adverse effects and intolerance: Canadian Working Group Consensus update. Can J Cardiol 2013: 29:1553–1568.
  65. Rojas-Fernandez CH, Cameron JC. Is statin-associated cognitive impairment clinically relevant? A narrative review and clinical recommendations. Ann Pharmacother 2012; 46:549–557.
  66. McGuinness B, O’Hare J, Craig D, Bullock R, Malouf R, Passmore P. Cochrane review on ‘Statins for the treatment of dementia’. Int J Geriatr Psychiatry 2013; 28:119–126.
  67. Pandey RD, Gupta PP, Jha D, Kumar S. Role of statins in Alzheimer’s disease: a retrospective meta-analysis for commonly investigated clinical parameters in RCTs. Int J Neurosci 2013; 123:521–525.
  68. Food and Drug Administration (FDA). FDA drug safety communication: important safety label changes to cholesterol-lowering statin drugs. www.fda.gov/drugs/ drugsafety/ucm293101.htm. Accessed December 5, 2016.
  69. Garfinkel D, Mangin D. Feasibility study of a systematic approach for discontinuation of multiple medications in older adults: addressing polypharmacy. Arch Intern Med 2010; 170:1648–1654.
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Page Number
131-142
Page Number
131-142
Publications
Publications
Topics
Article Type
Display Headline
Severely frail elderly patients do not need lipid-lowering drugs
Display Headline
Severely frail elderly patients do not need lipid-lowering drugs
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, deprescribing, PATH program, Canada, JUPITER trial, PROSPER trial, SPARCL trial, Laurie Mallery, Paige Moorhouse, Pam Veysey, Michael Allen, Isobel Fleming
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, deprescribing, PATH program, Canada, JUPITER trial, PROSPER trial, SPARCL trial, Laurie Mallery, Paige Moorhouse, Pam Veysey, Michael Allen, Isobel Fleming
Sections
Inside the Article

KEY POINTS

  • There is no reason to prescribe or continue statins for primary prevention in severely frail elderly patients, as these drugs are unlikely to provide benefit in terms of outcomes relevant to this population.
  • Statins are probably not necessary for secondary prevention in patients who are severely frail, although there may be extenuating circumstances for their use.
  • There is no reason to start or continue statins for heart failure, as there is insufficient evidence that they are effective for this indication in any population.
  • There is no reason to start or continue other lipid-lowering drugs in conjunction with statins.
  • As the frail elderly may be more vulnerable to the side effects of statins, lower doses may be more appropriate if these drugs are prescribed.
  • If there is concern regarding myopathy, a drug interaction, or other adverse effects, consider a trial of statin discontinuation.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Statin therapy in the frail elderly: A nuanced decision

Article Type
Changed
Mon, 07/31/2017 - 09:23
Display Headline
Statin therapy in the frail elderly: A nuanced decision

The growing elderly population varies widely in functional capacity and mental agility. Age by itself is not a reliable indicator of physiologic performance in patients with cardiovascular disease.1

See related article

The concept of frailty helps to identify elderly patients most susceptible to adverse outcomes. Frailty is a powerful indicator of disability, loss of independence, hospitalization, and death. In a patient whose health is declining, frailty is an appropriate impetus for the clinician and patient to reevaluate the goals of care.

In this issue of the Journal, Mallery et al2 address an important topic: the use of preventive lipid-lowering therapies in frail patients with limited life expectancy. For these patients, they recommend against lipid-lowering therapy for primary prevention, and only in extenuating circumstances for secondary prevention.

No trials have evaluated lipid-lowering therapy specifically in frail older adults, and therefore, these recommendations are based on an evidence-informed appraisal of the literature. Mallery et al2 suggest that in the frail elderly, improvement in function and quality of life are more relevant end points than traditional cardiovascular outcomes. They conclude that available evidence does not support lipid-lowering therapy for most patients with advanced frailty.

POINTS TO CONSIDER

Mallery et al2 effectively articulate the need for frailty-specific care. Multimorbidity, polypharmacy, and increased adverse drug effects require special attention in the frail elderly. The authors make a sound argument against lipid-lowering therapy for primary prevention in the severely frail elderly, in whom the evidence for short-term benefit is not compelling. They also recommend against nonstatin lipid-lowering medications, and against statin therapy for heart failure, which is consistent with major guidelines. In the modern era of reflexive testing and prescribing, the authors’ “less is more” approach provides needed encouragement for thoughtful care in these vulnerable patients.

However, certain points of contention deserve additional consideration, including the imprecise definition of frailty, potential benefits and harms of statin therapy in high-risk patients, and the importance of shared decision-making.

How should frailty be defined?

Frailty biology is a field of ongoing research, and there is a lack of consensus on how best to define the condition.3 Estimates of the prevalence of frailty among older adults with cardiovascular disease range from 10% to 60%, owing to considerable variability in the tools used for frailty assessment.4

Mallery et al2 consider an individual to be severely frail if he or she requires assistance with basic activities of daily living as the result of any physical or cognitive deficit (derived from the Clinical Frailty Scale or Frailty Assessment for Care Planning Tool). While functional dependence may be a consequence of frailty, this generalized definition does not characterize the clinical phenotype, which includes slowness, weakness, low physical activity, exhaustion, and unintentional weight loss.

Furthermore, this definition offers no insight into the unique characteristics, causes, and clinical course related to frailty. Significant heterogeneity among “frail” patients precludes a uniform treatment approach in this population.

 

 

Do statins benefit frail patients at high risk?

Regarding secondary prevention, the authors highlight a meta-analysis by Afilalo et al,5 the most comprehensive assessment to date of statin therapy in elderly patients with documented coronary heart disease. This study included nearly 20,000 elderly patients in nine secondary prevention trials, including the secondary prevention subgroup of the Prospective Study of Pravastatin in the Elderly at Risk (PROSPER) trial.6

Afilalo et al5 calculated that statin therapy reduced the rates of all-cause mortality by 22% and coronary death by 30%, with even greater reductions in the rates of nonfatal myocardial infarction, stroke, and revascularization. Furthermore, the absolute risk reduction was higher and the number needed to treat was lower in those over age 80. Overall, these data convincingly showed that high-risk patients ages 65 to 82 enrolled in clinical trials derive substantial benefit from statin therapy.

Mallery et al2 contend that many of the secondary prevention statin trials evaluated composite outcomes over many years of follow-up and therefore are not generalizable to the frail elderly. However, the Afilalo meta-analysis5 does not provide patient-level data, and therefore the benefit for different clinical and demographic subgroups is unknown. It is only speculative to infer that those with frailty are unlikely to benefit. In fact, the improved outcomes observed with increasing age would argue against this notion.

Given the compelling data supporting statin therapy in the high-risk elderly population, some patients and clinicians may reasonably feel there is value in statin therapy—even in those with advanced frailty.

What about symptoms, disability, quality of life, and short-term benefits? Asymptomatic or “silent” myocardial infarction is associated with angina, congestive heart failure, and subsequent symptomatic myocardial infarction.7,8 Dismissing the importance of these end points in clinical trials fails to recognize potential downstream effects that are directly relevant to a patient’s overall health status.

The Study Assessing Goals in the Elderly (SAGE) trial9 assessed the effect of statin therapy on ischemia burden in patients ages 65 to 85 with stable coronary disease. The results showed that both moderate and intensive statin dosing significantly reduced myocardial ischemia at 3 and 12 months, as detected by continuous electrocardiographic monitoring.

More research is needed to determine the effect of statin therapy on functional capacity and quality of life. Currently, it is premature to conclude that statins have no relevance to these important patient-centered outcomes.

What are the potential harms?

Mallery et al2 cite numerous articles that emphasize the potential adverse effects of statin therapy in the elderly. Unfortunately, data supporting the safety of statin therapy in the elderly were not included. This should be stressed, given that older statin-eligible patients are often undertreated in contemporary practice.10

A 2015 systematic review and meta-analysis indicated that statin-related events are relatively rare in the elderly.11 Another study showed elderly patients who started statin therapy after a myocardial infarction had no change in short-term cognitive or physical function.12

Older age and low body mass index are risk factors for statin myopathy, underscoring the need for close monitoring in frail patients. However, it is important to maintain an objective and balanced approach when considering potential harms.

Need for shared decision-making

Mallery et al2 make no mention of shared decision-making. Best practice guidelines for the management of frailty support a holistic medical review to establish an individualized care plan for each patient.13 Firm recommendations based on indeterminate evidence undermine the patient-physician relationship and do not allow for personal preferences of care. In an environment of uncertain benefit and harm, the patient’s priorities and values should serve as the cornerstone for clinical decisions.

References
  1. Barakat K, Wilkinson P, Deaner A, Fluck D, Ranjadayalan K, Timmis A. How should age affect management of acute myocardial infarction? A prospective cohort study. Lancet 1999; 353:955–959.
  2. Mallery L, Moorhouse P, McLea Veysey P, Allen M, Fleming I. Frail elderly patients do not need lipid-lowering drugs. Cleve Clin J Med 2016; 83:131–142.
  3. Bergman H, Ferrucci L, Guralnik J, et al. Frailty: an emerging research and clinical paradigm—issues and controversies. J Gerontol A Biol Sci Med Sci 2007; 62:731–737.
  4. Afilalo J, Alexander KP, Mack MJ, et al. Frailty assessment in the cardiovascular care of older adults. J Am Coll Cardiol 2014; 63:747–762.
  5. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  6. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  7. Nadelmann J, Frishman WH, Ooi WL, et al. Prevalence, incidence and prognosis of recognized and unrecognized myocardial infarction in persons aged 75 years or older: The Bronx Aging Study. Am J Cardiol 1990; 66:533–537.
  8. Sheifer SE, Gersh BJ, Yanez ND 3rd, Ades PA, Burke GL, Manolio TA. Prevalence, predisposing factors, and prognosis of clinically unrecognized myocardial infarction in the elderly. J Am Coll Cardiol 2000; 35:119–126.
  9. Deedwania P, Stone PH, Bairey Merz CN, et al. Effects of intensive versus moderate lipid-lowering therapy on myocardial ischemia in older patients with coronary heart disease: results of the Study Assessing Goals in the Elderly (SAGE). Circulation 2007; 115:700–707.
  10. Maddox TM, Borden WB, Tang F, et al. Implications of the 2013 ACC/AHA cholesterol guidelines for adults in contemporary cardiovascular practice: insights from the NCDR PINNACLE registry. J Am Coll Cardiol 2014; 64:2183–2192.
  11. Iwere RB, Hewitt J. Myopathy in older people receiving statin therapy: a systematic review and meta-analysis. Br J Clin Pharmacol 2015; 80:363–371.
  12. Swiger KJ, Martin SS, Tang F, et al. Cognitive and physical function by statin exposure in elderly individuals following acute myocardial infarction. Clin Cardiol 2015; 38:455–461.
  13. Turner G, Clegg A; British Geriatrics Society; Age UK; Royal College of General Practioners. Best practice guidelines for the management of frailty: a British Geriatrics Society, Age UK and Royal College of General Practitioners report. Age Ageing 2014; 43:744–747.
Article PDF
Author and Disclosure Information

Donald Clark III, MD
Department of Cardiovascular Medicine, Cleveland Clinic

Leslie S. Cho, MD
Section Head, Preventive Cardiology & Rehabilitation, and Director, Women’s Cardiovascular Center, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Leslie Cho, MD, Preventive Cardiovascular Medicine, JB1, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; chol@ccf.org

Issue
Cleveland Clinic Journal of Medicine - 84(2)
Publications
Topics
Page Number
143-145
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, Donald Clark, Leslie Cho
Sections
Author and Disclosure Information

Donald Clark III, MD
Department of Cardiovascular Medicine, Cleveland Clinic

Leslie S. Cho, MD
Section Head, Preventive Cardiology & Rehabilitation, and Director, Women’s Cardiovascular Center, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Leslie Cho, MD, Preventive Cardiovascular Medicine, JB1, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; chol@ccf.org

Author and Disclosure Information

Donald Clark III, MD
Department of Cardiovascular Medicine, Cleveland Clinic

Leslie S. Cho, MD
Section Head, Preventive Cardiology & Rehabilitation, and Director, Women’s Cardiovascular Center, Department of Cardiovascular Medicine, Cleveland Clinic

Address: Leslie Cho, MD, Preventive Cardiovascular Medicine, JB1, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; chol@ccf.org

Article PDF
Article PDF
Related Articles

The growing elderly population varies widely in functional capacity and mental agility. Age by itself is not a reliable indicator of physiologic performance in patients with cardiovascular disease.1

See related article

The concept of frailty helps to identify elderly patients most susceptible to adverse outcomes. Frailty is a powerful indicator of disability, loss of independence, hospitalization, and death. In a patient whose health is declining, frailty is an appropriate impetus for the clinician and patient to reevaluate the goals of care.

In this issue of the Journal, Mallery et al2 address an important topic: the use of preventive lipid-lowering therapies in frail patients with limited life expectancy. For these patients, they recommend against lipid-lowering therapy for primary prevention, and only in extenuating circumstances for secondary prevention.

No trials have evaluated lipid-lowering therapy specifically in frail older adults, and therefore, these recommendations are based on an evidence-informed appraisal of the literature. Mallery et al2 suggest that in the frail elderly, improvement in function and quality of life are more relevant end points than traditional cardiovascular outcomes. They conclude that available evidence does not support lipid-lowering therapy for most patients with advanced frailty.

POINTS TO CONSIDER

Mallery et al2 effectively articulate the need for frailty-specific care. Multimorbidity, polypharmacy, and increased adverse drug effects require special attention in the frail elderly. The authors make a sound argument against lipid-lowering therapy for primary prevention in the severely frail elderly, in whom the evidence for short-term benefit is not compelling. They also recommend against nonstatin lipid-lowering medications, and against statin therapy for heart failure, which is consistent with major guidelines. In the modern era of reflexive testing and prescribing, the authors’ “less is more” approach provides needed encouragement for thoughtful care in these vulnerable patients.

However, certain points of contention deserve additional consideration, including the imprecise definition of frailty, potential benefits and harms of statin therapy in high-risk patients, and the importance of shared decision-making.

How should frailty be defined?

Frailty biology is a field of ongoing research, and there is a lack of consensus on how best to define the condition.3 Estimates of the prevalence of frailty among older adults with cardiovascular disease range from 10% to 60%, owing to considerable variability in the tools used for frailty assessment.4

Mallery et al2 consider an individual to be severely frail if he or she requires assistance with basic activities of daily living as the result of any physical or cognitive deficit (derived from the Clinical Frailty Scale or Frailty Assessment for Care Planning Tool). While functional dependence may be a consequence of frailty, this generalized definition does not characterize the clinical phenotype, which includes slowness, weakness, low physical activity, exhaustion, and unintentional weight loss.

Furthermore, this definition offers no insight into the unique characteristics, causes, and clinical course related to frailty. Significant heterogeneity among “frail” patients precludes a uniform treatment approach in this population.

 

 

Do statins benefit frail patients at high risk?

Regarding secondary prevention, the authors highlight a meta-analysis by Afilalo et al,5 the most comprehensive assessment to date of statin therapy in elderly patients with documented coronary heart disease. This study included nearly 20,000 elderly patients in nine secondary prevention trials, including the secondary prevention subgroup of the Prospective Study of Pravastatin in the Elderly at Risk (PROSPER) trial.6

Afilalo et al5 calculated that statin therapy reduced the rates of all-cause mortality by 22% and coronary death by 30%, with even greater reductions in the rates of nonfatal myocardial infarction, stroke, and revascularization. Furthermore, the absolute risk reduction was higher and the number needed to treat was lower in those over age 80. Overall, these data convincingly showed that high-risk patients ages 65 to 82 enrolled in clinical trials derive substantial benefit from statin therapy.

Mallery et al2 contend that many of the secondary prevention statin trials evaluated composite outcomes over many years of follow-up and therefore are not generalizable to the frail elderly. However, the Afilalo meta-analysis5 does not provide patient-level data, and therefore the benefit for different clinical and demographic subgroups is unknown. It is only speculative to infer that those with frailty are unlikely to benefit. In fact, the improved outcomes observed with increasing age would argue against this notion.

Given the compelling data supporting statin therapy in the high-risk elderly population, some patients and clinicians may reasonably feel there is value in statin therapy—even in those with advanced frailty.

What about symptoms, disability, quality of life, and short-term benefits? Asymptomatic or “silent” myocardial infarction is associated with angina, congestive heart failure, and subsequent symptomatic myocardial infarction.7,8 Dismissing the importance of these end points in clinical trials fails to recognize potential downstream effects that are directly relevant to a patient’s overall health status.

The Study Assessing Goals in the Elderly (SAGE) trial9 assessed the effect of statin therapy on ischemia burden in patients ages 65 to 85 with stable coronary disease. The results showed that both moderate and intensive statin dosing significantly reduced myocardial ischemia at 3 and 12 months, as detected by continuous electrocardiographic monitoring.

More research is needed to determine the effect of statin therapy on functional capacity and quality of life. Currently, it is premature to conclude that statins have no relevance to these important patient-centered outcomes.

What are the potential harms?

Mallery et al2 cite numerous articles that emphasize the potential adverse effects of statin therapy in the elderly. Unfortunately, data supporting the safety of statin therapy in the elderly were not included. This should be stressed, given that older statin-eligible patients are often undertreated in contemporary practice.10

A 2015 systematic review and meta-analysis indicated that statin-related events are relatively rare in the elderly.11 Another study showed elderly patients who started statin therapy after a myocardial infarction had no change in short-term cognitive or physical function.12

Older age and low body mass index are risk factors for statin myopathy, underscoring the need for close monitoring in frail patients. However, it is important to maintain an objective and balanced approach when considering potential harms.

Need for shared decision-making

Mallery et al2 make no mention of shared decision-making. Best practice guidelines for the management of frailty support a holistic medical review to establish an individualized care plan for each patient.13 Firm recommendations based on indeterminate evidence undermine the patient-physician relationship and do not allow for personal preferences of care. In an environment of uncertain benefit and harm, the patient’s priorities and values should serve as the cornerstone for clinical decisions.

The growing elderly population varies widely in functional capacity and mental agility. Age by itself is not a reliable indicator of physiologic performance in patients with cardiovascular disease.1

See related article

The concept of frailty helps to identify elderly patients most susceptible to adverse outcomes. Frailty is a powerful indicator of disability, loss of independence, hospitalization, and death. In a patient whose health is declining, frailty is an appropriate impetus for the clinician and patient to reevaluate the goals of care.

In this issue of the Journal, Mallery et al2 address an important topic: the use of preventive lipid-lowering therapies in frail patients with limited life expectancy. For these patients, they recommend against lipid-lowering therapy for primary prevention, and only in extenuating circumstances for secondary prevention.

No trials have evaluated lipid-lowering therapy specifically in frail older adults, and therefore, these recommendations are based on an evidence-informed appraisal of the literature. Mallery et al2 suggest that in the frail elderly, improvement in function and quality of life are more relevant end points than traditional cardiovascular outcomes. They conclude that available evidence does not support lipid-lowering therapy for most patients with advanced frailty.

POINTS TO CONSIDER

Mallery et al2 effectively articulate the need for frailty-specific care. Multimorbidity, polypharmacy, and increased adverse drug effects require special attention in the frail elderly. The authors make a sound argument against lipid-lowering therapy for primary prevention in the severely frail elderly, in whom the evidence for short-term benefit is not compelling. They also recommend against nonstatin lipid-lowering medications, and against statin therapy for heart failure, which is consistent with major guidelines. In the modern era of reflexive testing and prescribing, the authors’ “less is more” approach provides needed encouragement for thoughtful care in these vulnerable patients.

However, certain points of contention deserve additional consideration, including the imprecise definition of frailty, potential benefits and harms of statin therapy in high-risk patients, and the importance of shared decision-making.

How should frailty be defined?

Frailty biology is a field of ongoing research, and there is a lack of consensus on how best to define the condition.3 Estimates of the prevalence of frailty among older adults with cardiovascular disease range from 10% to 60%, owing to considerable variability in the tools used for frailty assessment.4

Mallery et al2 consider an individual to be severely frail if he or she requires assistance with basic activities of daily living as the result of any physical or cognitive deficit (derived from the Clinical Frailty Scale or Frailty Assessment for Care Planning Tool). While functional dependence may be a consequence of frailty, this generalized definition does not characterize the clinical phenotype, which includes slowness, weakness, low physical activity, exhaustion, and unintentional weight loss.

Furthermore, this definition offers no insight into the unique characteristics, causes, and clinical course related to frailty. Significant heterogeneity among “frail” patients precludes a uniform treatment approach in this population.

 

 

Do statins benefit frail patients at high risk?

Regarding secondary prevention, the authors highlight a meta-analysis by Afilalo et al,5 the most comprehensive assessment to date of statin therapy in elderly patients with documented coronary heart disease. This study included nearly 20,000 elderly patients in nine secondary prevention trials, including the secondary prevention subgroup of the Prospective Study of Pravastatin in the Elderly at Risk (PROSPER) trial.6

Afilalo et al5 calculated that statin therapy reduced the rates of all-cause mortality by 22% and coronary death by 30%, with even greater reductions in the rates of nonfatal myocardial infarction, stroke, and revascularization. Furthermore, the absolute risk reduction was higher and the number needed to treat was lower in those over age 80. Overall, these data convincingly showed that high-risk patients ages 65 to 82 enrolled in clinical trials derive substantial benefit from statin therapy.

Mallery et al2 contend that many of the secondary prevention statin trials evaluated composite outcomes over many years of follow-up and therefore are not generalizable to the frail elderly. However, the Afilalo meta-analysis5 does not provide patient-level data, and therefore the benefit for different clinical and demographic subgroups is unknown. It is only speculative to infer that those with frailty are unlikely to benefit. In fact, the improved outcomes observed with increasing age would argue against this notion.

Given the compelling data supporting statin therapy in the high-risk elderly population, some patients and clinicians may reasonably feel there is value in statin therapy—even in those with advanced frailty.

What about symptoms, disability, quality of life, and short-term benefits? Asymptomatic or “silent” myocardial infarction is associated with angina, congestive heart failure, and subsequent symptomatic myocardial infarction.7,8 Dismissing the importance of these end points in clinical trials fails to recognize potential downstream effects that are directly relevant to a patient’s overall health status.

The Study Assessing Goals in the Elderly (SAGE) trial9 assessed the effect of statin therapy on ischemia burden in patients ages 65 to 85 with stable coronary disease. The results showed that both moderate and intensive statin dosing significantly reduced myocardial ischemia at 3 and 12 months, as detected by continuous electrocardiographic monitoring.

More research is needed to determine the effect of statin therapy on functional capacity and quality of life. Currently, it is premature to conclude that statins have no relevance to these important patient-centered outcomes.

What are the potential harms?

Mallery et al2 cite numerous articles that emphasize the potential adverse effects of statin therapy in the elderly. Unfortunately, data supporting the safety of statin therapy in the elderly were not included. This should be stressed, given that older statin-eligible patients are often undertreated in contemporary practice.10

A 2015 systematic review and meta-analysis indicated that statin-related events are relatively rare in the elderly.11 Another study showed elderly patients who started statin therapy after a myocardial infarction had no change in short-term cognitive or physical function.12

Older age and low body mass index are risk factors for statin myopathy, underscoring the need for close monitoring in frail patients. However, it is important to maintain an objective and balanced approach when considering potential harms.

Need for shared decision-making

Mallery et al2 make no mention of shared decision-making. Best practice guidelines for the management of frailty support a holistic medical review to establish an individualized care plan for each patient.13 Firm recommendations based on indeterminate evidence undermine the patient-physician relationship and do not allow for personal preferences of care. In an environment of uncertain benefit and harm, the patient’s priorities and values should serve as the cornerstone for clinical decisions.

References
  1. Barakat K, Wilkinson P, Deaner A, Fluck D, Ranjadayalan K, Timmis A. How should age affect management of acute myocardial infarction? A prospective cohort study. Lancet 1999; 353:955–959.
  2. Mallery L, Moorhouse P, McLea Veysey P, Allen M, Fleming I. Frail elderly patients do not need lipid-lowering drugs. Cleve Clin J Med 2016; 83:131–142.
  3. Bergman H, Ferrucci L, Guralnik J, et al. Frailty: an emerging research and clinical paradigm—issues and controversies. J Gerontol A Biol Sci Med Sci 2007; 62:731–737.
  4. Afilalo J, Alexander KP, Mack MJ, et al. Frailty assessment in the cardiovascular care of older adults. J Am Coll Cardiol 2014; 63:747–762.
  5. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  6. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  7. Nadelmann J, Frishman WH, Ooi WL, et al. Prevalence, incidence and prognosis of recognized and unrecognized myocardial infarction in persons aged 75 years or older: The Bronx Aging Study. Am J Cardiol 1990; 66:533–537.
  8. Sheifer SE, Gersh BJ, Yanez ND 3rd, Ades PA, Burke GL, Manolio TA. Prevalence, predisposing factors, and prognosis of clinically unrecognized myocardial infarction in the elderly. J Am Coll Cardiol 2000; 35:119–126.
  9. Deedwania P, Stone PH, Bairey Merz CN, et al. Effects of intensive versus moderate lipid-lowering therapy on myocardial ischemia in older patients with coronary heart disease: results of the Study Assessing Goals in the Elderly (SAGE). Circulation 2007; 115:700–707.
  10. Maddox TM, Borden WB, Tang F, et al. Implications of the 2013 ACC/AHA cholesterol guidelines for adults in contemporary cardiovascular practice: insights from the NCDR PINNACLE registry. J Am Coll Cardiol 2014; 64:2183–2192.
  11. Iwere RB, Hewitt J. Myopathy in older people receiving statin therapy: a systematic review and meta-analysis. Br J Clin Pharmacol 2015; 80:363–371.
  12. Swiger KJ, Martin SS, Tang F, et al. Cognitive and physical function by statin exposure in elderly individuals following acute myocardial infarction. Clin Cardiol 2015; 38:455–461.
  13. Turner G, Clegg A; British Geriatrics Society; Age UK; Royal College of General Practioners. Best practice guidelines for the management of frailty: a British Geriatrics Society, Age UK and Royal College of General Practitioners report. Age Ageing 2014; 43:744–747.
References
  1. Barakat K, Wilkinson P, Deaner A, Fluck D, Ranjadayalan K, Timmis A. How should age affect management of acute myocardial infarction? A prospective cohort study. Lancet 1999; 353:955–959.
  2. Mallery L, Moorhouse P, McLea Veysey P, Allen M, Fleming I. Frail elderly patients do not need lipid-lowering drugs. Cleve Clin J Med 2016; 83:131–142.
  3. Bergman H, Ferrucci L, Guralnik J, et al. Frailty: an emerging research and clinical paradigm—issues and controversies. J Gerontol A Biol Sci Med Sci 2007; 62:731–737.
  4. Afilalo J, Alexander KP, Mack MJ, et al. Frailty assessment in the cardiovascular care of older adults. J Am Coll Cardiol 2014; 63:747–762.
  5. Afilalo J, Duque G, Steele R, Jukema JW, de Craen AJ, Eisenberg MJ. Statins for secondary prevention in elderly patients: a hierarchical bayesian meta-analysis. J Am Coll Cardiol 2008; 51:37–45.
  6. Shepherd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002; 360:1623–1630.
  7. Nadelmann J, Frishman WH, Ooi WL, et al. Prevalence, incidence and prognosis of recognized and unrecognized myocardial infarction in persons aged 75 years or older: The Bronx Aging Study. Am J Cardiol 1990; 66:533–537.
  8. Sheifer SE, Gersh BJ, Yanez ND 3rd, Ades PA, Burke GL, Manolio TA. Prevalence, predisposing factors, and prognosis of clinically unrecognized myocardial infarction in the elderly. J Am Coll Cardiol 2000; 35:119–126.
  9. Deedwania P, Stone PH, Bairey Merz CN, et al. Effects of intensive versus moderate lipid-lowering therapy on myocardial ischemia in older patients with coronary heart disease: results of the Study Assessing Goals in the Elderly (SAGE). Circulation 2007; 115:700–707.
  10. Maddox TM, Borden WB, Tang F, et al. Implications of the 2013 ACC/AHA cholesterol guidelines for adults in contemporary cardiovascular practice: insights from the NCDR PINNACLE registry. J Am Coll Cardiol 2014; 64:2183–2192.
  11. Iwere RB, Hewitt J. Myopathy in older people receiving statin therapy: a systematic review and meta-analysis. Br J Clin Pharmacol 2015; 80:363–371.
  12. Swiger KJ, Martin SS, Tang F, et al. Cognitive and physical function by statin exposure in elderly individuals following acute myocardial infarction. Clin Cardiol 2015; 38:455–461.
  13. Turner G, Clegg A; British Geriatrics Society; Age UK; Royal College of General Practioners. Best practice guidelines for the management of frailty: a British Geriatrics Society, Age UK and Royal College of General Practitioners report. Age Ageing 2014; 43:744–747.
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Page Number
143-145
Page Number
143-145
Publications
Publications
Topics
Article Type
Display Headline
Statin therapy in the frail elderly: A nuanced decision
Display Headline
Statin therapy in the frail elderly: A nuanced decision
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, Donald Clark, Leslie Cho
Legacy Keywords
frailty, statins, lipids, elderly, frail elderly, Donald Clark, Leslie Cho
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Submassive pulmonary embolism

Article Type
Changed
Mon, 07/31/2017 - 09:38
Display Headline
Submassive pulmonary embolism

To the Editor: I read with interest the review on submassive pulmonary embolism by Ataya et al1 in the December 2016 issue. I had 3 questions or observations for the authors

First, systemic thrombolytic therapy for massive or hemodynamically unstable pulmonary embolism is given a grade 2C recommendation, similar to the level for select patients with submassive pulmonary embolism with low bleeding risk but at high risk of developing hypotension. The reference for this is the 2012 American College of Chest Physicians guidelines.2 I would like to point out that these guidelines were updated and published in February 2016,3 and systemic thrombolytic therapy for massive pulmonary embolism now carries a grade 2B recommendation. Thrombolytic therapy still has a grade 2C recommendation for select patients with submassive pulmonary embolism.

Second, the Moderate Pulmonary Embolism Treated With Thrombolysis (MOPETT) trial is described as a randomized trial in patients with moderate pulmonary hypertension and right ventricular dysfunction. I would like to point out that right ventricular dysfunction was not a criterion for enrollment in the trial.4

Finally, catheter-directed thrombolytic therapy is mentioned as an option for select patients with submassive and massive pulmonary embolism. The advantage is believed to be due to local action of the drug with fewer systemic effects. Since the protocol involves alteplase for 12 or 24 hours with a maximum dose of 24 mg, and since in most cases pulmonary embolism originates in the lower extremity, are we not exposing these patients to further clot propagation for 12 or 24 hours without the benefit of concomitant systemic anticoagulation or an inferior vena cava filter?

References
  1. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  4. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
Article PDF
Author and Disclosure Information

Anup Katyal, MD
Mercy Hospital, St. Louis, MO

Issue
Cleveland Clinic Journal of Medicine - 84(2)
Publications
Topics
Page Number
94-95
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Anup Katyal
Sections
Author and Disclosure Information

Anup Katyal, MD
Mercy Hospital, St. Louis, MO

Author and Disclosure Information

Anup Katyal, MD
Mercy Hospital, St. Louis, MO

Article PDF
Article PDF
Related Articles

To the Editor: I read with interest the review on submassive pulmonary embolism by Ataya et al1 in the December 2016 issue. I had 3 questions or observations for the authors

First, systemic thrombolytic therapy for massive or hemodynamically unstable pulmonary embolism is given a grade 2C recommendation, similar to the level for select patients with submassive pulmonary embolism with low bleeding risk but at high risk of developing hypotension. The reference for this is the 2012 American College of Chest Physicians guidelines.2 I would like to point out that these guidelines were updated and published in February 2016,3 and systemic thrombolytic therapy for massive pulmonary embolism now carries a grade 2B recommendation. Thrombolytic therapy still has a grade 2C recommendation for select patients with submassive pulmonary embolism.

Second, the Moderate Pulmonary Embolism Treated With Thrombolysis (MOPETT) trial is described as a randomized trial in patients with moderate pulmonary hypertension and right ventricular dysfunction. I would like to point out that right ventricular dysfunction was not a criterion for enrollment in the trial.4

Finally, catheter-directed thrombolytic therapy is mentioned as an option for select patients with submassive and massive pulmonary embolism. The advantage is believed to be due to local action of the drug with fewer systemic effects. Since the protocol involves alteplase for 12 or 24 hours with a maximum dose of 24 mg, and since in most cases pulmonary embolism originates in the lower extremity, are we not exposing these patients to further clot propagation for 12 or 24 hours without the benefit of concomitant systemic anticoagulation or an inferior vena cava filter?

To the Editor: I read with interest the review on submassive pulmonary embolism by Ataya et al1 in the December 2016 issue. I had 3 questions or observations for the authors

First, systemic thrombolytic therapy for massive or hemodynamically unstable pulmonary embolism is given a grade 2C recommendation, similar to the level for select patients with submassive pulmonary embolism with low bleeding risk but at high risk of developing hypotension. The reference for this is the 2012 American College of Chest Physicians guidelines.2 I would like to point out that these guidelines were updated and published in February 2016,3 and systemic thrombolytic therapy for massive pulmonary embolism now carries a grade 2B recommendation. Thrombolytic therapy still has a grade 2C recommendation for select patients with submassive pulmonary embolism.

Second, the Moderate Pulmonary Embolism Treated With Thrombolysis (MOPETT) trial is described as a randomized trial in patients with moderate pulmonary hypertension and right ventricular dysfunction. I would like to point out that right ventricular dysfunction was not a criterion for enrollment in the trial.4

Finally, catheter-directed thrombolytic therapy is mentioned as an option for select patients with submassive and massive pulmonary embolism. The advantage is believed to be due to local action of the drug with fewer systemic effects. Since the protocol involves alteplase for 12 or 24 hours with a maximum dose of 24 mg, and since in most cases pulmonary embolism originates in the lower extremity, are we not exposing these patients to further clot propagation for 12 or 24 hours without the benefit of concomitant systemic anticoagulation or an inferior vena cava filter?

References
  1. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  4. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
References
  1. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  4. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Page Number
94-95
Page Number
94-95
Publications
Publications
Topics
Article Type
Display Headline
Submassive pulmonary embolism
Display Headline
Submassive pulmonary embolism
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Anup Katyal
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Anup Katyal
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

In reply: Submassive pulmonary embolism

Article Type
Changed
Mon, 07/31/2017 - 09:40
Display Headline
In reply: Submassive pulmonary embolism

In Reply: We thank Dr. Katyal for his thoughtful comments.

Dr. Katyal points out that the grade of recommendation for thrombolysis in patients with massive pulmonary embolism was upgraded from 2C to 2B in the 2016 American College of Chest Physicians (ACCP) guidelines1 compared with the 2012 guidelines2 that we cited. The upgrade in this recommendation was owing to 2 small trials and 1 large randomized controlled trial that included patients with submassive pulmonary embolism.3–5 Interestingly, these 3 studies led to an upgrade in the level of recommendation for thrombolysis in the treatment of massive pulmonary embolism, perhaps more from a safety aspect (in view of the incidence of major bleeding vs mortality). Regardless, Dr. Katyal is correct in highlighting that the new 2016 ACCP guidelines now give a grade of 2B for thrombolytic therapy in the treatment of massive pulmonary embolism. These guidelines had not been published at the time of submission of our manuscript.

Dr. Katyal is also correct that patients were not required to have right ventricular dysfunction to be enrolled in the MOPETT trial.3 As we pointed out, “Only 20% of the participants were enrolled on the basis of right ventricular dysfunction on echocardiography, whereas almost 60% had elevated cardiac biomarkers.”6

Regarding catheter-directed therapy, patients who received low-dose catheter-directed alteplase were also concurrently anticoagulated with systemic unfractionated heparin in the Ultrasound-Assisted, Catheter-Directed Thrombolysis for Acute Intermediate-Risk Pulmonary Embolism (ULTIMA) trial.7 The ULTIMA trial authors commented that unfractionated heparin was started with an 80-U/kg bolus followed by an 18-U/kg/hour infusion to target an anti-factor Xa level of 0.3 to 0.7 μg/mL, which is considered therapeutic anticoagulation. The investigators in the SEATTLE II trial8 continued systemic unfractionated heparin but targeted a lower “intermediate” anticoagulation target (an augmented partial thromboplastin time of 40–60 seconds), so these patients weren’t completely without systemic anticoagulation either. At our institution, the current practice is to target an anti-Xa level of 0.3 to 0.7 μg/mL in patients receiving catheter-directed therapy for large-volume pulmonary embolism.

References
  1. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
  4. Meyer G, Vicaut E, Danays T, et al; PEITHO Investigators. Fibrinolysis for patients with intermediate-risk pulmonary embolism. N Engl J Med 2014; 370:1402–1411.
  5. Kline JA, Nordenholz KE, Courtney DM, et al. Treatment of submassive pulmonary embolism with tenecteplase or placebo: cardiopulmonary outcomes at 3 months: multicenter double-blind, placebo-controlled randomized trial. J Thromb Haemost 2014; 12:459–468.
  6. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  7. Kucher N, Boekstegers P, Muller OJ, et al. Randomized, controlled trial of ultrasound-assisted catheter-directed thrombolysis for acute intermediate-risk pulmonary embolism. Circulation 2014; 129:479–486.
  8. Piazza G, Hohlfelder B, Jaff MR, et al; SEATTLE II Investigators. A prospective, single-arm, multicenter trial of ultrasound-facilitated, catheter-directed, low-dose fibrinolysis for acute massive and submassive pulmonary embolism (The SEATTLE II Study). JACC Cardiovasc Interv 2015; 8:1382–1392.
Article PDF
Author and Disclosure Information

Ali Ataya, MD
University of Florida, Gainesville

Jessica Cope, PharmD
University of Florida, Gainesville

Abbas Shahmohammadi, MD
University of Florida, Gainesville

Hassan Alnuaimat, MD
University of Florida, Gainesville

Issue
Cleveland Clinic Journal of Medicine - 84(2)
Publications
Topics
Page Number
94, 95
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Ali Ataya, Jessica Cope, Abbas Shahmohammadi, Hassan Alnuaimat
Sections
Author and Disclosure Information

Ali Ataya, MD
University of Florida, Gainesville

Jessica Cope, PharmD
University of Florida, Gainesville

Abbas Shahmohammadi, MD
University of Florida, Gainesville

Hassan Alnuaimat, MD
University of Florida, Gainesville

Author and Disclosure Information

Ali Ataya, MD
University of Florida, Gainesville

Jessica Cope, PharmD
University of Florida, Gainesville

Abbas Shahmohammadi, MD
University of Florida, Gainesville

Hassan Alnuaimat, MD
University of Florida, Gainesville

Article PDF
Article PDF
Related Articles

In Reply: We thank Dr. Katyal for his thoughtful comments.

Dr. Katyal points out that the grade of recommendation for thrombolysis in patients with massive pulmonary embolism was upgraded from 2C to 2B in the 2016 American College of Chest Physicians (ACCP) guidelines1 compared with the 2012 guidelines2 that we cited. The upgrade in this recommendation was owing to 2 small trials and 1 large randomized controlled trial that included patients with submassive pulmonary embolism.3–5 Interestingly, these 3 studies led to an upgrade in the level of recommendation for thrombolysis in the treatment of massive pulmonary embolism, perhaps more from a safety aspect (in view of the incidence of major bleeding vs mortality). Regardless, Dr. Katyal is correct in highlighting that the new 2016 ACCP guidelines now give a grade of 2B for thrombolytic therapy in the treatment of massive pulmonary embolism. These guidelines had not been published at the time of submission of our manuscript.

Dr. Katyal is also correct that patients were not required to have right ventricular dysfunction to be enrolled in the MOPETT trial.3 As we pointed out, “Only 20% of the participants were enrolled on the basis of right ventricular dysfunction on echocardiography, whereas almost 60% had elevated cardiac biomarkers.”6

Regarding catheter-directed therapy, patients who received low-dose catheter-directed alteplase were also concurrently anticoagulated with systemic unfractionated heparin in the Ultrasound-Assisted, Catheter-Directed Thrombolysis for Acute Intermediate-Risk Pulmonary Embolism (ULTIMA) trial.7 The ULTIMA trial authors commented that unfractionated heparin was started with an 80-U/kg bolus followed by an 18-U/kg/hour infusion to target an anti-factor Xa level of 0.3 to 0.7 μg/mL, which is considered therapeutic anticoagulation. The investigators in the SEATTLE II trial8 continued systemic unfractionated heparin but targeted a lower “intermediate” anticoagulation target (an augmented partial thromboplastin time of 40–60 seconds), so these patients weren’t completely without systemic anticoagulation either. At our institution, the current practice is to target an anti-Xa level of 0.3 to 0.7 μg/mL in patients receiving catheter-directed therapy for large-volume pulmonary embolism.

In Reply: We thank Dr. Katyal for his thoughtful comments.

Dr. Katyal points out that the grade of recommendation for thrombolysis in patients with massive pulmonary embolism was upgraded from 2C to 2B in the 2016 American College of Chest Physicians (ACCP) guidelines1 compared with the 2012 guidelines2 that we cited. The upgrade in this recommendation was owing to 2 small trials and 1 large randomized controlled trial that included patients with submassive pulmonary embolism.3–5 Interestingly, these 3 studies led to an upgrade in the level of recommendation for thrombolysis in the treatment of massive pulmonary embolism, perhaps more from a safety aspect (in view of the incidence of major bleeding vs mortality). Regardless, Dr. Katyal is correct in highlighting that the new 2016 ACCP guidelines now give a grade of 2B for thrombolytic therapy in the treatment of massive pulmonary embolism. These guidelines had not been published at the time of submission of our manuscript.

Dr. Katyal is also correct that patients were not required to have right ventricular dysfunction to be enrolled in the MOPETT trial.3 As we pointed out, “Only 20% of the participants were enrolled on the basis of right ventricular dysfunction on echocardiography, whereas almost 60% had elevated cardiac biomarkers.”6

Regarding catheter-directed therapy, patients who received low-dose catheter-directed alteplase were also concurrently anticoagulated with systemic unfractionated heparin in the Ultrasound-Assisted, Catheter-Directed Thrombolysis for Acute Intermediate-Risk Pulmonary Embolism (ULTIMA) trial.7 The ULTIMA trial authors commented that unfractionated heparin was started with an 80-U/kg bolus followed by an 18-U/kg/hour infusion to target an anti-factor Xa level of 0.3 to 0.7 μg/mL, which is considered therapeutic anticoagulation. The investigators in the SEATTLE II trial8 continued systemic unfractionated heparin but targeted a lower “intermediate” anticoagulation target (an augmented partial thromboplastin time of 40–60 seconds), so these patients weren’t completely without systemic anticoagulation either. At our institution, the current practice is to target an anti-Xa level of 0.3 to 0.7 μg/mL in patients receiving catheter-directed therapy for large-volume pulmonary embolism.

References
  1. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
  4. Meyer G, Vicaut E, Danays T, et al; PEITHO Investigators. Fibrinolysis for patients with intermediate-risk pulmonary embolism. N Engl J Med 2014; 370:1402–1411.
  5. Kline JA, Nordenholz KE, Courtney DM, et al. Treatment of submassive pulmonary embolism with tenecteplase or placebo: cardiopulmonary outcomes at 3 months: multicenter double-blind, placebo-controlled randomized trial. J Thromb Haemost 2014; 12:459–468.
  6. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  7. Kucher N, Boekstegers P, Muller OJ, et al. Randomized, controlled trial of ultrasound-assisted catheter-directed thrombolysis for acute intermediate-risk pulmonary embolism. Circulation 2014; 129:479–486.
  8. Piazza G, Hohlfelder B, Jaff MR, et al; SEATTLE II Investigators. A prospective, single-arm, multicenter trial of ultrasound-facilitated, catheter-directed, low-dose fibrinolysis for acute massive and submassive pulmonary embolism (The SEATTLE II Study). JACC Cardiovasc Interv 2015; 8:1382–1392.
References
  1. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016; 149:315–352.
  2. Kearon C, Akl EA, Comerota AJ, et al; American College of Chest Physicians. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141(suppl 2):e419S–e494S.
  3. Sharifi M, Bay C, Skrocki L, Rahimi F, Mehdipour M; “MOPETT” Investigators. Moderate pulmonary embolism treated with thrombolysis (from the “MOPETT” Trial). Am J Cardiol 2013; 111:273–277.
  4. Meyer G, Vicaut E, Danays T, et al; PEITHO Investigators. Fibrinolysis for patients with intermediate-risk pulmonary embolism. N Engl J Med 2014; 370:1402–1411.
  5. Kline JA, Nordenholz KE, Courtney DM, et al. Treatment of submassive pulmonary embolism with tenecteplase or placebo: cardiopulmonary outcomes at 3 months: multicenter double-blind, placebo-controlled randomized trial. J Thromb Haemost 2014; 12:459–468.
  6. Ataya A, Cope J, Shahmohammadi A, Alnuaimat H. Do patients with submassive pulmonary embolism benefit from thrombolytic therapy? Cleve Clin J Med 2016; 83:923–932.
  7. Kucher N, Boekstegers P, Muller OJ, et al. Randomized, controlled trial of ultrasound-assisted catheter-directed thrombolysis for acute intermediate-risk pulmonary embolism. Circulation 2014; 129:479–486.
  8. Piazza G, Hohlfelder B, Jaff MR, et al; SEATTLE II Investigators. A prospective, single-arm, multicenter trial of ultrasound-facilitated, catheter-directed, low-dose fibrinolysis for acute massive and submassive pulmonary embolism (The SEATTLE II Study). JACC Cardiovasc Interv 2015; 8:1382–1392.
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Issue
Cleveland Clinic Journal of Medicine - 84(2)
Page Number
94, 95
Page Number
94, 95
Publications
Publications
Topics
Article Type
Display Headline
In reply: Submassive pulmonary embolism
Display Headline
In reply: Submassive pulmonary embolism
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Ali Ataya, Jessica Cope, Abbas Shahmohammadi, Hassan Alnuaimat
Legacy Keywords
pulmonary embolism, thrombolytic therapy, anticoagulation, Ali Ataya, Jessica Cope, Abbas Shahmohammadi, Hassan Alnuaimat
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

An alternative device for ESRD patients with central venous obstruction

Article Type
Changed
Wed, 01/02/2019 - 09:46

 

– Catheter dependence is often the final option available for hemodialysis patients who have exhausted upper extremity access because of central venous obstruction. But an alternative device that combines a standard expanded polytetrafluoroethylene (ePTFE) arterial graft component with an entirely internalized central venous catheter component may provide an additional option that can help avoid catheters in selected patients, according to pooled results reported at a symposium on vascular surgery sponsored by Northwestern University.

Virginia L. Wong, MD, of University Hospitals Cleveland Medical Center, reported on her group’s and others’ experience using the Hemodialysis Reliable Outflow (HeRO) graft (Merit Medical) to gain access to the superior vena cava (SVC), thus allowing for further upper extremity access options. The device has its limitations in patients with CVO, Dr. Wong noted, “but it can be an important tool for the dedicated access surgeon who is likely to be referred the most complicated patients who have run out of just about every other option.”

Dr. Virginia Wong
Dr. Virginia Wong

The Food and Drug Administration approved the HeRO graft for CVO in 2008, but a recent pooled analysis (Eur J Vasc Endovasc Surg. 2015;50[1]:108-13), which showed a 1-year primary patency rate of 22% and a secondary patency rate of 60%, may provide clarity on how the device can be used to treat CVO in end-stage renal disease (ESRD) patients when the care team desires an alternative to femoral arteriovenous graft, Dr. Wong said. “The 1-year primary patency rate overall was not very good, but with aggressive thrombectomy programs the 1-year patency rate was decent,” she said.

The pooled analysis involved eight series from 2009 to 2015, but the largest series, which involved 164 patients, reported primary and secondary patency rates of 48.8% and 90.8%, respectively (Eur J Vasc Endovasc Surg. 2012;44[1]:93-9). “Patency for these alternative accesses may not be quite what we can achieve with standard upper-extremity access,” Dr. Wong said, “but these patients do not have the standard access as an option.”

Dr. Wong explained where the HeRO fits into the existing vascular practice. “The current data suggest that we should try to exhaust all traditional upper extremity access options before considering anything else, but the HeRO could be considered as an acceptable option for suitable patients,” she said. However, to achieve those outcomes, “you need to have an aggressive thrombectomy program.”

HeRO may be an option for salvage of an existing arm access, plagued by recalcitrant CVO, while still preserving the femoral sites and for future hemodialysis access and/or renal transplantation, Dr. Wong said.

The HeRO also has been used in alternative configurations, taking advantage of axillary or subclavian routes to the SVC when both internal jugular veins are occluded. Dr. Wong has used the femoral route to the inferior vena cava (IVC) for salvaging the femoral AV graft in which iliofemoral venous outflow has been compromised.

Anatomically, the patient must be able to accept a large-bore (19-Fr) access catheter into the central vein. Physiologically, the patient must be able to maintain patency of the long, low-resistance HeRO circuit, which can be up to 50 cm in length, she said. The protocol at Dr. Wong’s institution recommends an inflow arterial diameter of at least 3 mm, along with a left ventricular ejection fraction of 20% or greater and a minimum systolic blood pressure of 100 mm Hg for HeRO on the right side, and possibly higher when coming from the left.

Chronic hypotension is a frequent disqualifier, although some of these patients may benefit from midodrine hydrochloride, she said. In any event, a review of medications and consultation with nephrology and the dialysis unit are mandatory elements of patient screening. “I usually request hemodialysis run sheets from the last three sessions to see what systolic blood pressure excursion is like over the course of treatment,” she said.

The basic principles of hemo-access care are important when considering the HeRO for CVO patients, Dr. Wong said. These include site/side preservation, catheter avoidance and “not to burn any bridges” for future access. “Individualization of care and careful patient selection are probably the best bets if you’re just starting out,” she said. “Choose good patients before resorting to HeRO as the last option for a fairly marginal candidate.”

Dr. Wong had no relevant financial relationships to disclose.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Catheter dependence is often the final option available for hemodialysis patients who have exhausted upper extremity access because of central venous obstruction. But an alternative device that combines a standard expanded polytetrafluoroethylene (ePTFE) arterial graft component with an entirely internalized central venous catheter component may provide an additional option that can help avoid catheters in selected patients, according to pooled results reported at a symposium on vascular surgery sponsored by Northwestern University.

Virginia L. Wong, MD, of University Hospitals Cleveland Medical Center, reported on her group’s and others’ experience using the Hemodialysis Reliable Outflow (HeRO) graft (Merit Medical) to gain access to the superior vena cava (SVC), thus allowing for further upper extremity access options. The device has its limitations in patients with CVO, Dr. Wong noted, “but it can be an important tool for the dedicated access surgeon who is likely to be referred the most complicated patients who have run out of just about every other option.”

Dr. Virginia Wong
Dr. Virginia Wong

The Food and Drug Administration approved the HeRO graft for CVO in 2008, but a recent pooled analysis (Eur J Vasc Endovasc Surg. 2015;50[1]:108-13), which showed a 1-year primary patency rate of 22% and a secondary patency rate of 60%, may provide clarity on how the device can be used to treat CVO in end-stage renal disease (ESRD) patients when the care team desires an alternative to femoral arteriovenous graft, Dr. Wong said. “The 1-year primary patency rate overall was not very good, but with aggressive thrombectomy programs the 1-year patency rate was decent,” she said.

The pooled analysis involved eight series from 2009 to 2015, but the largest series, which involved 164 patients, reported primary and secondary patency rates of 48.8% and 90.8%, respectively (Eur J Vasc Endovasc Surg. 2012;44[1]:93-9). “Patency for these alternative accesses may not be quite what we can achieve with standard upper-extremity access,” Dr. Wong said, “but these patients do not have the standard access as an option.”

Dr. Wong explained where the HeRO fits into the existing vascular practice. “The current data suggest that we should try to exhaust all traditional upper extremity access options before considering anything else, but the HeRO could be considered as an acceptable option for suitable patients,” she said. However, to achieve those outcomes, “you need to have an aggressive thrombectomy program.”

HeRO may be an option for salvage of an existing arm access, plagued by recalcitrant CVO, while still preserving the femoral sites and for future hemodialysis access and/or renal transplantation, Dr. Wong said.

The HeRO also has been used in alternative configurations, taking advantage of axillary or subclavian routes to the SVC when both internal jugular veins are occluded. Dr. Wong has used the femoral route to the inferior vena cava (IVC) for salvaging the femoral AV graft in which iliofemoral venous outflow has been compromised.

Anatomically, the patient must be able to accept a large-bore (19-Fr) access catheter into the central vein. Physiologically, the patient must be able to maintain patency of the long, low-resistance HeRO circuit, which can be up to 50 cm in length, she said. The protocol at Dr. Wong’s institution recommends an inflow arterial diameter of at least 3 mm, along with a left ventricular ejection fraction of 20% or greater and a minimum systolic blood pressure of 100 mm Hg for HeRO on the right side, and possibly higher when coming from the left.

Chronic hypotension is a frequent disqualifier, although some of these patients may benefit from midodrine hydrochloride, she said. In any event, a review of medications and consultation with nephrology and the dialysis unit are mandatory elements of patient screening. “I usually request hemodialysis run sheets from the last three sessions to see what systolic blood pressure excursion is like over the course of treatment,” she said.

The basic principles of hemo-access care are important when considering the HeRO for CVO patients, Dr. Wong said. These include site/side preservation, catheter avoidance and “not to burn any bridges” for future access. “Individualization of care and careful patient selection are probably the best bets if you’re just starting out,” she said. “Choose good patients before resorting to HeRO as the last option for a fairly marginal candidate.”

Dr. Wong had no relevant financial relationships to disclose.

 

– Catheter dependence is often the final option available for hemodialysis patients who have exhausted upper extremity access because of central venous obstruction. But an alternative device that combines a standard expanded polytetrafluoroethylene (ePTFE) arterial graft component with an entirely internalized central venous catheter component may provide an additional option that can help avoid catheters in selected patients, according to pooled results reported at a symposium on vascular surgery sponsored by Northwestern University.

Virginia L. Wong, MD, of University Hospitals Cleveland Medical Center, reported on her group’s and others’ experience using the Hemodialysis Reliable Outflow (HeRO) graft (Merit Medical) to gain access to the superior vena cava (SVC), thus allowing for further upper extremity access options. The device has its limitations in patients with CVO, Dr. Wong noted, “but it can be an important tool for the dedicated access surgeon who is likely to be referred the most complicated patients who have run out of just about every other option.”

Dr. Virginia Wong
Dr. Virginia Wong

The Food and Drug Administration approved the HeRO graft for CVO in 2008, but a recent pooled analysis (Eur J Vasc Endovasc Surg. 2015;50[1]:108-13), which showed a 1-year primary patency rate of 22% and a secondary patency rate of 60%, may provide clarity on how the device can be used to treat CVO in end-stage renal disease (ESRD) patients when the care team desires an alternative to femoral arteriovenous graft, Dr. Wong said. “The 1-year primary patency rate overall was not very good, but with aggressive thrombectomy programs the 1-year patency rate was decent,” she said.

The pooled analysis involved eight series from 2009 to 2015, but the largest series, which involved 164 patients, reported primary and secondary patency rates of 48.8% and 90.8%, respectively (Eur J Vasc Endovasc Surg. 2012;44[1]:93-9). “Patency for these alternative accesses may not be quite what we can achieve with standard upper-extremity access,” Dr. Wong said, “but these patients do not have the standard access as an option.”

Dr. Wong explained where the HeRO fits into the existing vascular practice. “The current data suggest that we should try to exhaust all traditional upper extremity access options before considering anything else, but the HeRO could be considered as an acceptable option for suitable patients,” she said. However, to achieve those outcomes, “you need to have an aggressive thrombectomy program.”

HeRO may be an option for salvage of an existing arm access, plagued by recalcitrant CVO, while still preserving the femoral sites and for future hemodialysis access and/or renal transplantation, Dr. Wong said.

The HeRO also has been used in alternative configurations, taking advantage of axillary or subclavian routes to the SVC when both internal jugular veins are occluded. Dr. Wong has used the femoral route to the inferior vena cava (IVC) for salvaging the femoral AV graft in which iliofemoral venous outflow has been compromised.

Anatomically, the patient must be able to accept a large-bore (19-Fr) access catheter into the central vein. Physiologically, the patient must be able to maintain patency of the long, low-resistance HeRO circuit, which can be up to 50 cm in length, she said. The protocol at Dr. Wong’s institution recommends an inflow arterial diameter of at least 3 mm, along with a left ventricular ejection fraction of 20% or greater and a minimum systolic blood pressure of 100 mm Hg for HeRO on the right side, and possibly higher when coming from the left.

Chronic hypotension is a frequent disqualifier, although some of these patients may benefit from midodrine hydrochloride, she said. In any event, a review of medications and consultation with nephrology and the dialysis unit are mandatory elements of patient screening. “I usually request hemodialysis run sheets from the last three sessions to see what systolic blood pressure excursion is like over the course of treatment,” she said.

The basic principles of hemo-access care are important when considering the HeRO for CVO patients, Dr. Wong said. These include site/side preservation, catheter avoidance and “not to burn any bridges” for future access. “Individualization of care and careful patient selection are probably the best bets if you’re just starting out,” she said. “Choose good patients before resorting to HeRO as the last option for a fairly marginal candidate.”

Dr. Wong had no relevant financial relationships to disclose.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Combined graft-catheter device may preserve femoral access for hemodialysis for patients with central venous obstruction.

Major finding: One-year primary potency rate was 22% and secondary patency rate 60% for device recipients.

Data source: Literature review, including pooled results from eight studies involving 408 subjects.

Disclosures: Dr. Wong reported having no financial disclosures.

Now is time to embrace emerging PAD interventions

Article Type
Changed
Wed, 01/02/2019 - 09:46

 

– Bioresorbable scaffolds, new drugs, adjuvant interventions, and stem and progenitor cell therapy will change how vascular surgeons treat peripheral artery disease in the next 5 years, so they must embrace these emerging treatments or run the risk of being displaced by other specialists, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

“Vascular surgeons must position their practices to be the nexus for the evaluation and treatment of the patient and proactively engage in the critical trials of these new technologies,” said Patrick J. Geraghty, MD, of Washington University, St. Louis. “If our specialty fails to adapt to new treatment options, we risk getting sidelined as critical limb ischemia (CLI) treatment moves into a multimodality model.”

Dr. Patrick Geraghty
Dr. Patrick Geraghty


Dr. Geraghty focused on several future directions for PAD treatment: improved drug-eluting stents (DES) for superficial femoral artery disease; drug-coated balloons and modified DES for infrapopliteal disease; biologic modifiers for claudication and CLI; and bioresorbable, drug-eluting scaffolds for infrainguinal interventions.

“You’re not simply a plumber anymore; you’re a biological response modifier,” Dr. Geraghty said, explaining that biologic response modification technologies are the logical successor where standard surgical and endovascular techniques have either fallen short (as in early patency loss due to restenosis) or failed to offer effective alternatives (as in no-option advanced CLI patients). “And that takes many of us out of our comfort zone,” he said.

Dr. Geraghty noted the VIBRANT trial (J Vasc Surg. 2013;58[2]:386-95) and similar studies of non–drug eluting constructs identified early restenosis as the primary culprit in endovascular patency loss. “If you could reduce those early patency losses, you’d have an admirable primary patency rate for these complex lesions,” he said. “We’re able to reconstruct a vessel lumen. The question is, how to best maintain it?”

To answer that, Dr. Geraghty noted that the SIROCCO II trial (J Vasc Interv Radiol. 2005;16[3]:331-8) failed to show an advantage for a sirolimus-eluting stent over bare nitinol stent for superficial femoral artery (SFA) disease, but the subsequent Zilver PTX trial showed the benefits of paclitaxel-eluting stents over 5 years (Circulation. 2016;133[15]:1472-83).

He noted that drug-coated balloons (DCBs) trials have yielded mixed results in infrapopliteal intervention. Most notably, the multicenter In.Pact DEEP trial (Circulation. 2015;131[5]:495-502) failed to show treatment efficacy, Dr. Geraghty said. “The In.Pact DEEP results sharply contrasted with the positive data from trials of similar DCBs in the SFA” (N Engl J Med. 2015;373[2]:145-53).

With regard to DES for infrapopliteal disease, Dr. Geraghty noted the promise of positive results of the ACHILLES (J Am Coll Cardiol. 2012;60[22]:2290-5) and DESTINY (J Vasc Surg. 2012;55[2]:390-9) trials, along with the modest structural changes needed to convert from coronary to proximal tibial applications.

Bioresorbable vascular scaffolds (BVS) for CLI have also made recent advances. “It has been a slow road, but I’m happy that industry has pursued this aggressively,” Dr. Geraghty said. He pointed out that the ESPRIT I trial of bioresorbable everolimus-eluting vascular scaffolds in PAD involving the external iliac artery and SFA reported restenosis rates of 12.1% and 16.1% at 1 and 2 years, respectively (JACC Cardiovasc Interv. 2016;9[11]:1178-87). A trial of the Absorb BVS (Abbott) for short infrapopliteal lesions showed primary patency rates of 96% and 85% at 1 and 2 years, he said (JACC Cardiovasc Interv. 2016;9[7]:715-24).

“Vascular surgeons should be tracking BVS technology closely,” Dr. Geraghty said. “It achieves multiple desirable goals: immediate scaffolding for luminal restoration; mitigation of the restenotic stimulus via stent resorption; drug delivery for inhibition of restenosis; and the prospect of simpler re-interventions.”

Stem/progenitor cell therapies may also provide new solutions for no-option vasculature. One trial that showed “promising trends,” Dr. Geraghty said, is the RESTORE-CLI study of bone marrow aspiration (Mol Ther. 2012;20[6]:1280-6). “This trial reported a trend toward improved time to failure and reduced amputation-free survival, but did not meet its primary endpoint,” he said. “Likewise, the recently presented Biomet MOBILE data failed to meet its primary endpoint, but showed favorable trends in some treatment subgroups” (J Vasc Surg. 2011;54[6]:1650-8).

Dr. Geraghty noted that trial design in this field may need to change directions. “Look at the Delphi consensus matrices for the WIfI (Wound, Ischemia, foot Infection) Threatened limb Classification System (J Vasc Surg. 2014;59[1]:220-34). These show that complex wounds bear a significant risk of amputation, perhaps unmitigated by successful revascularization.” In addition, he called amputation-free survival “a rather blunt instrument” for evaluating how therapies impact limb outcomes and said it can confound the analysis of their effectiveness.

“Instead of confining the progenitor-cell therapies to no-option CLI trials, I’m eager to also see them investigated for treatment of claudication,” Dr. Geraghty said. “Can cell-based therapies possibly displace endovascular interventions as the first-line, least-harmful option for claudication?”

Dr. Geraghty also touched on intra/extravascular adjuvant therapies: antithrombin nanoparticles; inhibitory nanoparticles and polymeric wraps; and adventitial drug delivery techniques, among others.

“It’s critically important for vascular surgeons to position themselves for continued success in CLI treatment,” he said. “That involves aggressive practice branding, active trial participation, critical analysis of new technologies, and adoption of new, even disruptive, treatment modalities that show patient benefit.”

Dr. Geraghty disclosed stock ownership in Pulse Therapeutics; consultant fees from Bard Peripheral Vascular, Boston Scientific, Intact Vascular, Bard/Lutonix and Spectranetics; and serving as principal investigator for trials by Cook Medical, Bard/Lutonix, and Intact Vascular, with fees going to Washington University Medical School.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Bioresorbable scaffolds, new drugs, adjuvant interventions, and stem and progenitor cell therapy will change how vascular surgeons treat peripheral artery disease in the next 5 years, so they must embrace these emerging treatments or run the risk of being displaced by other specialists, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

“Vascular surgeons must position their practices to be the nexus for the evaluation and treatment of the patient and proactively engage in the critical trials of these new technologies,” said Patrick J. Geraghty, MD, of Washington University, St. Louis. “If our specialty fails to adapt to new treatment options, we risk getting sidelined as critical limb ischemia (CLI) treatment moves into a multimodality model.”

Dr. Patrick Geraghty
Dr. Patrick Geraghty


Dr. Geraghty focused on several future directions for PAD treatment: improved drug-eluting stents (DES) for superficial femoral artery disease; drug-coated balloons and modified DES for infrapopliteal disease; biologic modifiers for claudication and CLI; and bioresorbable, drug-eluting scaffolds for infrainguinal interventions.

“You’re not simply a plumber anymore; you’re a biological response modifier,” Dr. Geraghty said, explaining that biologic response modification technologies are the logical successor where standard surgical and endovascular techniques have either fallen short (as in early patency loss due to restenosis) or failed to offer effective alternatives (as in no-option advanced CLI patients). “And that takes many of us out of our comfort zone,” he said.

Dr. Geraghty noted the VIBRANT trial (J Vasc Surg. 2013;58[2]:386-95) and similar studies of non–drug eluting constructs identified early restenosis as the primary culprit in endovascular patency loss. “If you could reduce those early patency losses, you’d have an admirable primary patency rate for these complex lesions,” he said. “We’re able to reconstruct a vessel lumen. The question is, how to best maintain it?”

To answer that, Dr. Geraghty noted that the SIROCCO II trial (J Vasc Interv Radiol. 2005;16[3]:331-8) failed to show an advantage for a sirolimus-eluting stent over bare nitinol stent for superficial femoral artery (SFA) disease, but the subsequent Zilver PTX trial showed the benefits of paclitaxel-eluting stents over 5 years (Circulation. 2016;133[15]:1472-83).

He noted that drug-coated balloons (DCBs) trials have yielded mixed results in infrapopliteal intervention. Most notably, the multicenter In.Pact DEEP trial (Circulation. 2015;131[5]:495-502) failed to show treatment efficacy, Dr. Geraghty said. “The In.Pact DEEP results sharply contrasted with the positive data from trials of similar DCBs in the SFA” (N Engl J Med. 2015;373[2]:145-53).

With regard to DES for infrapopliteal disease, Dr. Geraghty noted the promise of positive results of the ACHILLES (J Am Coll Cardiol. 2012;60[22]:2290-5) and DESTINY (J Vasc Surg. 2012;55[2]:390-9) trials, along with the modest structural changes needed to convert from coronary to proximal tibial applications.

Bioresorbable vascular scaffolds (BVS) for CLI have also made recent advances. “It has been a slow road, but I’m happy that industry has pursued this aggressively,” Dr. Geraghty said. He pointed out that the ESPRIT I trial of bioresorbable everolimus-eluting vascular scaffolds in PAD involving the external iliac artery and SFA reported restenosis rates of 12.1% and 16.1% at 1 and 2 years, respectively (JACC Cardiovasc Interv. 2016;9[11]:1178-87). A trial of the Absorb BVS (Abbott) for short infrapopliteal lesions showed primary patency rates of 96% and 85% at 1 and 2 years, he said (JACC Cardiovasc Interv. 2016;9[7]:715-24).

“Vascular surgeons should be tracking BVS technology closely,” Dr. Geraghty said. “It achieves multiple desirable goals: immediate scaffolding for luminal restoration; mitigation of the restenotic stimulus via stent resorption; drug delivery for inhibition of restenosis; and the prospect of simpler re-interventions.”

Stem/progenitor cell therapies may also provide new solutions for no-option vasculature. One trial that showed “promising trends,” Dr. Geraghty said, is the RESTORE-CLI study of bone marrow aspiration (Mol Ther. 2012;20[6]:1280-6). “This trial reported a trend toward improved time to failure and reduced amputation-free survival, but did not meet its primary endpoint,” he said. “Likewise, the recently presented Biomet MOBILE data failed to meet its primary endpoint, but showed favorable trends in some treatment subgroups” (J Vasc Surg. 2011;54[6]:1650-8).

Dr. Geraghty noted that trial design in this field may need to change directions. “Look at the Delphi consensus matrices for the WIfI (Wound, Ischemia, foot Infection) Threatened limb Classification System (J Vasc Surg. 2014;59[1]:220-34). These show that complex wounds bear a significant risk of amputation, perhaps unmitigated by successful revascularization.” In addition, he called amputation-free survival “a rather blunt instrument” for evaluating how therapies impact limb outcomes and said it can confound the analysis of their effectiveness.

“Instead of confining the progenitor-cell therapies to no-option CLI trials, I’m eager to also see them investigated for treatment of claudication,” Dr. Geraghty said. “Can cell-based therapies possibly displace endovascular interventions as the first-line, least-harmful option for claudication?”

Dr. Geraghty also touched on intra/extravascular adjuvant therapies: antithrombin nanoparticles; inhibitory nanoparticles and polymeric wraps; and adventitial drug delivery techniques, among others.

“It’s critically important for vascular surgeons to position themselves for continued success in CLI treatment,” he said. “That involves aggressive practice branding, active trial participation, critical analysis of new technologies, and adoption of new, even disruptive, treatment modalities that show patient benefit.”

Dr. Geraghty disclosed stock ownership in Pulse Therapeutics; consultant fees from Bard Peripheral Vascular, Boston Scientific, Intact Vascular, Bard/Lutonix and Spectranetics; and serving as principal investigator for trials by Cook Medical, Bard/Lutonix, and Intact Vascular, with fees going to Washington University Medical School.

 

– Bioresorbable scaffolds, new drugs, adjuvant interventions, and stem and progenitor cell therapy will change how vascular surgeons treat peripheral artery disease in the next 5 years, so they must embrace these emerging treatments or run the risk of being displaced by other specialists, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

“Vascular surgeons must position their practices to be the nexus for the evaluation and treatment of the patient and proactively engage in the critical trials of these new technologies,” said Patrick J. Geraghty, MD, of Washington University, St. Louis. “If our specialty fails to adapt to new treatment options, we risk getting sidelined as critical limb ischemia (CLI) treatment moves into a multimodality model.”

Dr. Patrick Geraghty
Dr. Patrick Geraghty


Dr. Geraghty focused on several future directions for PAD treatment: improved drug-eluting stents (DES) for superficial femoral artery disease; drug-coated balloons and modified DES for infrapopliteal disease; biologic modifiers for claudication and CLI; and bioresorbable, drug-eluting scaffolds for infrainguinal interventions.

“You’re not simply a plumber anymore; you’re a biological response modifier,” Dr. Geraghty said, explaining that biologic response modification technologies are the logical successor where standard surgical and endovascular techniques have either fallen short (as in early patency loss due to restenosis) or failed to offer effective alternatives (as in no-option advanced CLI patients). “And that takes many of us out of our comfort zone,” he said.

Dr. Geraghty noted the VIBRANT trial (J Vasc Surg. 2013;58[2]:386-95) and similar studies of non–drug eluting constructs identified early restenosis as the primary culprit in endovascular patency loss. “If you could reduce those early patency losses, you’d have an admirable primary patency rate for these complex lesions,” he said. “We’re able to reconstruct a vessel lumen. The question is, how to best maintain it?”

To answer that, Dr. Geraghty noted that the SIROCCO II trial (J Vasc Interv Radiol. 2005;16[3]:331-8) failed to show an advantage for a sirolimus-eluting stent over bare nitinol stent for superficial femoral artery (SFA) disease, but the subsequent Zilver PTX trial showed the benefits of paclitaxel-eluting stents over 5 years (Circulation. 2016;133[15]:1472-83).

He noted that drug-coated balloons (DCBs) trials have yielded mixed results in infrapopliteal intervention. Most notably, the multicenter In.Pact DEEP trial (Circulation. 2015;131[5]:495-502) failed to show treatment efficacy, Dr. Geraghty said. “The In.Pact DEEP results sharply contrasted with the positive data from trials of similar DCBs in the SFA” (N Engl J Med. 2015;373[2]:145-53).

With regard to DES for infrapopliteal disease, Dr. Geraghty noted the promise of positive results of the ACHILLES (J Am Coll Cardiol. 2012;60[22]:2290-5) and DESTINY (J Vasc Surg. 2012;55[2]:390-9) trials, along with the modest structural changes needed to convert from coronary to proximal tibial applications.

Bioresorbable vascular scaffolds (BVS) for CLI have also made recent advances. “It has been a slow road, but I’m happy that industry has pursued this aggressively,” Dr. Geraghty said. He pointed out that the ESPRIT I trial of bioresorbable everolimus-eluting vascular scaffolds in PAD involving the external iliac artery and SFA reported restenosis rates of 12.1% and 16.1% at 1 and 2 years, respectively (JACC Cardiovasc Interv. 2016;9[11]:1178-87). A trial of the Absorb BVS (Abbott) for short infrapopliteal lesions showed primary patency rates of 96% and 85% at 1 and 2 years, he said (JACC Cardiovasc Interv. 2016;9[7]:715-24).

“Vascular surgeons should be tracking BVS technology closely,” Dr. Geraghty said. “It achieves multiple desirable goals: immediate scaffolding for luminal restoration; mitigation of the restenotic stimulus via stent resorption; drug delivery for inhibition of restenosis; and the prospect of simpler re-interventions.”

Stem/progenitor cell therapies may also provide new solutions for no-option vasculature. One trial that showed “promising trends,” Dr. Geraghty said, is the RESTORE-CLI study of bone marrow aspiration (Mol Ther. 2012;20[6]:1280-6). “This trial reported a trend toward improved time to failure and reduced amputation-free survival, but did not meet its primary endpoint,” he said. “Likewise, the recently presented Biomet MOBILE data failed to meet its primary endpoint, but showed favorable trends in some treatment subgroups” (J Vasc Surg. 2011;54[6]:1650-8).

Dr. Geraghty noted that trial design in this field may need to change directions. “Look at the Delphi consensus matrices for the WIfI (Wound, Ischemia, foot Infection) Threatened limb Classification System (J Vasc Surg. 2014;59[1]:220-34). These show that complex wounds bear a significant risk of amputation, perhaps unmitigated by successful revascularization.” In addition, he called amputation-free survival “a rather blunt instrument” for evaluating how therapies impact limb outcomes and said it can confound the analysis of their effectiveness.

“Instead of confining the progenitor-cell therapies to no-option CLI trials, I’m eager to also see them investigated for treatment of claudication,” Dr. Geraghty said. “Can cell-based therapies possibly displace endovascular interventions as the first-line, least-harmful option for claudication?”

Dr. Geraghty also touched on intra/extravascular adjuvant therapies: antithrombin nanoparticles; inhibitory nanoparticles and polymeric wraps; and adventitial drug delivery techniques, among others.

“It’s critically important for vascular surgeons to position themselves for continued success in CLI treatment,” he said. “That involves aggressive practice branding, active trial participation, critical analysis of new technologies, and adoption of new, even disruptive, treatment modalities that show patient benefit.”

Dr. Geraghty disclosed stock ownership in Pulse Therapeutics; consultant fees from Bard Peripheral Vascular, Boston Scientific, Intact Vascular, Bard/Lutonix and Spectranetics; and serving as principal investigator for trials by Cook Medical, Bard/Lutonix, and Intact Vascular, with fees going to Washington University Medical School.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

AT THE NORTHWESTERN VASCULAR SYMPOSIUM

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Emerging treatments for lower-extremity interventions range from improved drug-eluting stents for the superficial femoral artery and infrapopliteal disease to bioresorbable, drug-eluting scaffolds for infrainguinal interventions.

Major finding: The future of minimally invasive revascularization hinges on reliably reopening stenosed or occluded arteries, maintaining vessel patency and using therapies to stimulate arteriogenesis or angiogenesis without reintervention.

Data source: Review of literature.

Disclosures: Dr. Geraghty disclosed stock ownership in Pulse Therapeutics; consultant fees from Bard Peripheral Vascular, Boston Scientific, Intact Vascular, Bard/Lutonix and Spectranetics; and serving as principal investigator for trials by Cook Medical, Bard/Lutonix, and Intact Vascular, with fees going to Washington University Medical School.

New data signal paradigm shift in FMD and arterial disease

Article Type
Changed
Wed, 01/02/2019 - 09:45

– New data have shown that fibromuscular dysplasia is associated with high rates of dissection and/or aneurysm, and emerging recommendations call for routine imaging early on in the diagnosis of FMD to monitor for these vascular events, a researcher who developed those recommendations reported at a symposium on vascular surgery sponsored by Northwestern University.

“Given the very high rate of aneurysms in this population, it is now recommended that all patients with FMD should undergo at least one-time head-to pelvis imaging with CT angiography or MR angiography to screen for the presence of an aneurysm or to identify other areas of FMD involvement,” said Daniella Kadian-Dodov, MD, of Icahn School of Medicine at Mount Sinai in New York (J Am Coll Cardiol. 2016;68:176-85).

Dr. Daniella Kadian-Dodov
Dr. Daniella Kadian-Dodov
Dr. Kadian-Dodov and her colleagues further refined their recommendations for imaging in FMD in a JACC Cardiovascular Imaging article in press. “Our group tends to prefer CTA for the chest-abdomen-pelvis over MRA because MRA may miss FMD due to its lower resolution or the chance of a false-positive reading because of a motion artifact mimicking multifocal FMD,” she said. Dr. Kadian-Dodov cited a Cleveland Clinic study showing that chest-abdomen-pelvis CTA had a reproducibility rate of around 90% for diagnosis of FMD-related pathology.

First described in 1938, FMD is a non-atherosclerotic, noninflammatory disease that had been thought to be a rare cause of renovascular hypertension with a classic “string-of-beads” appearance upon imaging, Dr. Kadian-Dodov noted. However, recent data from the Fibromuscular Dysplasia Society of America–sponsored U.S. registry has changed that thinking. “We now know it occurs more frequently in the carotid and renal arteries, although it has been observed in almost every artery,” she said. “The pathogenesis is still unknown but up to 10% of cases are familial.”

And manifestations of disease now extend beyond the “string-of-beads” appearance to include aneurysm, dissection, and arterial tortuosity, she said (Circulation. 2012;125:3182-90; Circulation. 2014;129:1048-78; J Am Coll Cardiol. 2016;68:176-85). The classification system for FMD has also undergone a recent change, according to Dr. Kadian-Dodov. “Traditionally, a histopathologic scheme was used to classify FMD,” she said. “Nowadays, fewer and fewer patients are undergoing surgical procedures, so the classification has changed to an angiographic system,” the most common of which is the American Heart Association system adopted in 2014 that distinguishes between multifocal, characterized by the classic “string-of-beads” appearance, and focal FMD with a single area of stenosis.

But the diagnosis of either variant of FMD is not exclusive. “Patients may have multiple areas of disease involvement and the same patient may have both focal and multifocal FMD findings,” Dr. Kadian-Dodov said.

The U.S. registry has helped clarify the thinking on FMD, Dr. Kadian-Dodov said. More than 1,400 patients are in the registry, 90% of whom are women with multifocal disease. The average age of onset of symptoms is 47 years, but 52 is the average age for diagnosis. “So these patients are experiencing several years delay to FMD diagnosis,” she said.

Manifestations depend on the vascular bed involved. “In the case of cervical artery FMD, headaches and pulsatile tinnitus are commonly reported, whereas with renal artery involvement hypertension is the most common symptom,” she said. A recent analysis showed 41.7% of the FMD population have either aneurysm and dissection or both (J Am Coll Cardiol. 2016;68:176-185).

But no specific guidelines for treatment of FMD yet exist, Dr. Kadian-Dodov said. “General guidelines should be applied for the management of dissection and aneurysm in patients with FMD,” she said. For patients with arterial dissection, that means conservative therapy comprising either anticoagulation or antiplatelet agents for 3-6 months followed by daily low-dose aspirin therapy. “Revascularization is rarely required for these patients,” she said. “Endovascular or surgical modalities should be reserved for those with continued ischemia despite conservative management or more complicated pseudoaneurysm formations.”

Daily aspirin therapy is likewise the recommendation for patients with cervical artery multifocal or focal FMD involvement without dissection or aneurysms. “We follow up with imaging every 6 months for 2 years,” Dr. Kadian-Dodov said. “If they’re stable, we switch over to annual surveillance; and if the patient has an aneurysm or dissection, that might alter the imaging and surveillance program.”

During angioplasty, determining the severity of stenosis upon visual inspection is difficult, especially in multifocal FMD. She advised measuring the gradient across the area of FMD involvement with a pressure wire to determine if angioplasty has adequately treated the lesion. “You should see obliteration of the gradient with successful treatment; you don’t have to target your therapy to a perfect angiographic result,” she said.

In patients with FMD and hypertension, she recommended renal artery angioplasty for hypertension of less than 5 years duration or in resistant or labile hypertension. “In this setting, stents are only reserved for complicated or refractory cases; angioplasty alone is sufficient,” Dr. Kadian-Dodov said. Cure rates decline with age, and hypertension in focal disease has a higher cure rate than does multifocal disease, she said (Hypertension. 2010;56:525-32).

Dr. Kadian-Dodov had no relevant financial relationships to disclose.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

– New data have shown that fibromuscular dysplasia is associated with high rates of dissection and/or aneurysm, and emerging recommendations call for routine imaging early on in the diagnosis of FMD to monitor for these vascular events, a researcher who developed those recommendations reported at a symposium on vascular surgery sponsored by Northwestern University.

“Given the very high rate of aneurysms in this population, it is now recommended that all patients with FMD should undergo at least one-time head-to pelvis imaging with CT angiography or MR angiography to screen for the presence of an aneurysm or to identify other areas of FMD involvement,” said Daniella Kadian-Dodov, MD, of Icahn School of Medicine at Mount Sinai in New York (J Am Coll Cardiol. 2016;68:176-85).

Dr. Daniella Kadian-Dodov
Dr. Daniella Kadian-Dodov
Dr. Kadian-Dodov and her colleagues further refined their recommendations for imaging in FMD in a JACC Cardiovascular Imaging article in press. “Our group tends to prefer CTA for the chest-abdomen-pelvis over MRA because MRA may miss FMD due to its lower resolution or the chance of a false-positive reading because of a motion artifact mimicking multifocal FMD,” she said. Dr. Kadian-Dodov cited a Cleveland Clinic study showing that chest-abdomen-pelvis CTA had a reproducibility rate of around 90% for diagnosis of FMD-related pathology.

First described in 1938, FMD is a non-atherosclerotic, noninflammatory disease that had been thought to be a rare cause of renovascular hypertension with a classic “string-of-beads” appearance upon imaging, Dr. Kadian-Dodov noted. However, recent data from the Fibromuscular Dysplasia Society of America–sponsored U.S. registry has changed that thinking. “We now know it occurs more frequently in the carotid and renal arteries, although it has been observed in almost every artery,” she said. “The pathogenesis is still unknown but up to 10% of cases are familial.”

And manifestations of disease now extend beyond the “string-of-beads” appearance to include aneurysm, dissection, and arterial tortuosity, she said (Circulation. 2012;125:3182-90; Circulation. 2014;129:1048-78; J Am Coll Cardiol. 2016;68:176-85). The classification system for FMD has also undergone a recent change, according to Dr. Kadian-Dodov. “Traditionally, a histopathologic scheme was used to classify FMD,” she said. “Nowadays, fewer and fewer patients are undergoing surgical procedures, so the classification has changed to an angiographic system,” the most common of which is the American Heart Association system adopted in 2014 that distinguishes between multifocal, characterized by the classic “string-of-beads” appearance, and focal FMD with a single area of stenosis.

But the diagnosis of either variant of FMD is not exclusive. “Patients may have multiple areas of disease involvement and the same patient may have both focal and multifocal FMD findings,” Dr. Kadian-Dodov said.

The U.S. registry has helped clarify the thinking on FMD, Dr. Kadian-Dodov said. More than 1,400 patients are in the registry, 90% of whom are women with multifocal disease. The average age of onset of symptoms is 47 years, but 52 is the average age for diagnosis. “So these patients are experiencing several years delay to FMD diagnosis,” she said.

Manifestations depend on the vascular bed involved. “In the case of cervical artery FMD, headaches and pulsatile tinnitus are commonly reported, whereas with renal artery involvement hypertension is the most common symptom,” she said. A recent analysis showed 41.7% of the FMD population have either aneurysm and dissection or both (J Am Coll Cardiol. 2016;68:176-185).

But no specific guidelines for treatment of FMD yet exist, Dr. Kadian-Dodov said. “General guidelines should be applied for the management of dissection and aneurysm in patients with FMD,” she said. For patients with arterial dissection, that means conservative therapy comprising either anticoagulation or antiplatelet agents for 3-6 months followed by daily low-dose aspirin therapy. “Revascularization is rarely required for these patients,” she said. “Endovascular or surgical modalities should be reserved for those with continued ischemia despite conservative management or more complicated pseudoaneurysm formations.”

Daily aspirin therapy is likewise the recommendation for patients with cervical artery multifocal or focal FMD involvement without dissection or aneurysms. “We follow up with imaging every 6 months for 2 years,” Dr. Kadian-Dodov said. “If they’re stable, we switch over to annual surveillance; and if the patient has an aneurysm or dissection, that might alter the imaging and surveillance program.”

During angioplasty, determining the severity of stenosis upon visual inspection is difficult, especially in multifocal FMD. She advised measuring the gradient across the area of FMD involvement with a pressure wire to determine if angioplasty has adequately treated the lesion. “You should see obliteration of the gradient with successful treatment; you don’t have to target your therapy to a perfect angiographic result,” she said.

In patients with FMD and hypertension, she recommended renal artery angioplasty for hypertension of less than 5 years duration or in resistant or labile hypertension. “In this setting, stents are only reserved for complicated or refractory cases; angioplasty alone is sufficient,” Dr. Kadian-Dodov said. Cure rates decline with age, and hypertension in focal disease has a higher cure rate than does multifocal disease, she said (Hypertension. 2010;56:525-32).

Dr. Kadian-Dodov had no relevant financial relationships to disclose.

– New data have shown that fibromuscular dysplasia is associated with high rates of dissection and/or aneurysm, and emerging recommendations call for routine imaging early on in the diagnosis of FMD to monitor for these vascular events, a researcher who developed those recommendations reported at a symposium on vascular surgery sponsored by Northwestern University.

“Given the very high rate of aneurysms in this population, it is now recommended that all patients with FMD should undergo at least one-time head-to pelvis imaging with CT angiography or MR angiography to screen for the presence of an aneurysm or to identify other areas of FMD involvement,” said Daniella Kadian-Dodov, MD, of Icahn School of Medicine at Mount Sinai in New York (J Am Coll Cardiol. 2016;68:176-85).

Dr. Daniella Kadian-Dodov
Dr. Daniella Kadian-Dodov
Dr. Kadian-Dodov and her colleagues further refined their recommendations for imaging in FMD in a JACC Cardiovascular Imaging article in press. “Our group tends to prefer CTA for the chest-abdomen-pelvis over MRA because MRA may miss FMD due to its lower resolution or the chance of a false-positive reading because of a motion artifact mimicking multifocal FMD,” she said. Dr. Kadian-Dodov cited a Cleveland Clinic study showing that chest-abdomen-pelvis CTA had a reproducibility rate of around 90% for diagnosis of FMD-related pathology.

First described in 1938, FMD is a non-atherosclerotic, noninflammatory disease that had been thought to be a rare cause of renovascular hypertension with a classic “string-of-beads” appearance upon imaging, Dr. Kadian-Dodov noted. However, recent data from the Fibromuscular Dysplasia Society of America–sponsored U.S. registry has changed that thinking. “We now know it occurs more frequently in the carotid and renal arteries, although it has been observed in almost every artery,” she said. “The pathogenesis is still unknown but up to 10% of cases are familial.”

And manifestations of disease now extend beyond the “string-of-beads” appearance to include aneurysm, dissection, and arterial tortuosity, she said (Circulation. 2012;125:3182-90; Circulation. 2014;129:1048-78; J Am Coll Cardiol. 2016;68:176-85). The classification system for FMD has also undergone a recent change, according to Dr. Kadian-Dodov. “Traditionally, a histopathologic scheme was used to classify FMD,” she said. “Nowadays, fewer and fewer patients are undergoing surgical procedures, so the classification has changed to an angiographic system,” the most common of which is the American Heart Association system adopted in 2014 that distinguishes between multifocal, characterized by the classic “string-of-beads” appearance, and focal FMD with a single area of stenosis.

But the diagnosis of either variant of FMD is not exclusive. “Patients may have multiple areas of disease involvement and the same patient may have both focal and multifocal FMD findings,” Dr. Kadian-Dodov said.

The U.S. registry has helped clarify the thinking on FMD, Dr. Kadian-Dodov said. More than 1,400 patients are in the registry, 90% of whom are women with multifocal disease. The average age of onset of symptoms is 47 years, but 52 is the average age for diagnosis. “So these patients are experiencing several years delay to FMD diagnosis,” she said.

Manifestations depend on the vascular bed involved. “In the case of cervical artery FMD, headaches and pulsatile tinnitus are commonly reported, whereas with renal artery involvement hypertension is the most common symptom,” she said. A recent analysis showed 41.7% of the FMD population have either aneurysm and dissection or both (J Am Coll Cardiol. 2016;68:176-185).

But no specific guidelines for treatment of FMD yet exist, Dr. Kadian-Dodov said. “General guidelines should be applied for the management of dissection and aneurysm in patients with FMD,” she said. For patients with arterial dissection, that means conservative therapy comprising either anticoagulation or antiplatelet agents for 3-6 months followed by daily low-dose aspirin therapy. “Revascularization is rarely required for these patients,” she said. “Endovascular or surgical modalities should be reserved for those with continued ischemia despite conservative management or more complicated pseudoaneurysm formations.”

Daily aspirin therapy is likewise the recommendation for patients with cervical artery multifocal or focal FMD involvement without dissection or aneurysms. “We follow up with imaging every 6 months for 2 years,” Dr. Kadian-Dodov said. “If they’re stable, we switch over to annual surveillance; and if the patient has an aneurysm or dissection, that might alter the imaging and surveillance program.”

During angioplasty, determining the severity of stenosis upon visual inspection is difficult, especially in multifocal FMD. She advised measuring the gradient across the area of FMD involvement with a pressure wire to determine if angioplasty has adequately treated the lesion. “You should see obliteration of the gradient with successful treatment; you don’t have to target your therapy to a perfect angiographic result,” she said.

In patients with FMD and hypertension, she recommended renal artery angioplasty for hypertension of less than 5 years duration or in resistant or labile hypertension. “In this setting, stents are only reserved for complicated or refractory cases; angioplasty alone is sufficient,” Dr. Kadian-Dodov said. Cure rates decline with age, and hypertension in focal disease has a higher cure rate than does multifocal disease, she said (Hypertension. 2010;56:525-32).

Dr. Kadian-Dodov had no relevant financial relationships to disclose.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Fibromuscular dysplasia was thought to be a rare cause of renovascular hypertension, but new data has challenged this thinking.

Major finding: FMD accounts for 15%-20% of patients with spontaneous carotid or vertebral artery dissection and 45%-86% of patients with spontaneous coronary artery dissection.

Data source: U.S. Registry for FMD maintained by the Fibromuscular Dystrophy Society of America.

Disclosures: Dr. Kadian-Dodov reported having no financial disclosures.

High-intensity statins cut amputations and mortality in PAD

Article Type
Changed
Fri, 01/18/2019 - 16:27

 

– High-intensity statin therapy in patients with peripheral artery disease was associated with significant reductions in amputations as well as mortality during up to 5 years of follow-up in the first large study to examine the relationship, Shipra Arya, MD, reported at the American Heart Association scientific sessions.

Low- or moderate-intensity statin therapy also improved survival compared to no statin, albeit to a significantly lesser magnitude than high-intensity therapy. But high- and low/intermediate-intensity statins were similarly effective in reducing amputation risk, according to Dr. Arya, a vascular surgeon at Emory University in Atlanta.

Incidence of key outcomes by statin use in patients with PAD
She presented a retrospective observational study of 208,194 patients with clinical peripheral artery disease (PAD) in the national Veterans Affairs database for 2003-2014, of whom 98% were men. In her study, Dr. Arya examined the association between the highest statin dose prescribed within 1 year of PAD diagnosis and rates of amputation and all-cause mortality at 1, 3, and 5 years of follow-up. During this interval there were 17,643 amputations, of which 14,981 were considered major amputations, as well as 99,870 deaths.

The 2013 AHA/American College of Cardiology treatment guidelines recommend high-intensity statins for all patients with clinical atherosclerotic disease, including those with PAD (Circulation. 2014 Jun 24;129[25 Suppl 2]:S1-45). (Updated PAD guidelines unveiled at the AHA meeting strongly recommend statin medication for all patients with PAD [Circulation. 2016 Nov 13. doi: 10.1161/CIR.0000000000000470]).However, the bulk of patients in Dr. Arya’s study were captured in the database prior to release of the 2013 guidelines. That may account for the sparse use of high-intensity statin therapy in the study cohort. Indeed, only 11.3% of the PAD patients were on a high-intensity statin. Another 36.2% were on moderate-intensity statin therapy, 3.5% were on low-intensity therapy, and 27.6% weren’t on a statin at all.

The relationship between statin therapy and mortality was strongly dose-dependent.

This study was funded by the AHA and the Atlanta Veterans Affairs Medical Center. Dr. Arya reported having no financial conflicts of interest.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– High-intensity statin therapy in patients with peripheral artery disease was associated with significant reductions in amputations as well as mortality during up to 5 years of follow-up in the first large study to examine the relationship, Shipra Arya, MD, reported at the American Heart Association scientific sessions.

Low- or moderate-intensity statin therapy also improved survival compared to no statin, albeit to a significantly lesser magnitude than high-intensity therapy. But high- and low/intermediate-intensity statins were similarly effective in reducing amputation risk, according to Dr. Arya, a vascular surgeon at Emory University in Atlanta.

Incidence of key outcomes by statin use in patients with PAD
She presented a retrospective observational study of 208,194 patients with clinical peripheral artery disease (PAD) in the national Veterans Affairs database for 2003-2014, of whom 98% were men. In her study, Dr. Arya examined the association between the highest statin dose prescribed within 1 year of PAD diagnosis and rates of amputation and all-cause mortality at 1, 3, and 5 years of follow-up. During this interval there were 17,643 amputations, of which 14,981 were considered major amputations, as well as 99,870 deaths.

The 2013 AHA/American College of Cardiology treatment guidelines recommend high-intensity statins for all patients with clinical atherosclerotic disease, including those with PAD (Circulation. 2014 Jun 24;129[25 Suppl 2]:S1-45). (Updated PAD guidelines unveiled at the AHA meeting strongly recommend statin medication for all patients with PAD [Circulation. 2016 Nov 13. doi: 10.1161/CIR.0000000000000470]).However, the bulk of patients in Dr. Arya’s study were captured in the database prior to release of the 2013 guidelines. That may account for the sparse use of high-intensity statin therapy in the study cohort. Indeed, only 11.3% of the PAD patients were on a high-intensity statin. Another 36.2% were on moderate-intensity statin therapy, 3.5% were on low-intensity therapy, and 27.6% weren’t on a statin at all.

The relationship between statin therapy and mortality was strongly dose-dependent.

This study was funded by the AHA and the Atlanta Veterans Affairs Medical Center. Dr. Arya reported having no financial conflicts of interest.
 

 

– High-intensity statin therapy in patients with peripheral artery disease was associated with significant reductions in amputations as well as mortality during up to 5 years of follow-up in the first large study to examine the relationship, Shipra Arya, MD, reported at the American Heart Association scientific sessions.

Low- or moderate-intensity statin therapy also improved survival compared to no statin, albeit to a significantly lesser magnitude than high-intensity therapy. But high- and low/intermediate-intensity statins were similarly effective in reducing amputation risk, according to Dr. Arya, a vascular surgeon at Emory University in Atlanta.

Incidence of key outcomes by statin use in patients with PAD
She presented a retrospective observational study of 208,194 patients with clinical peripheral artery disease (PAD) in the national Veterans Affairs database for 2003-2014, of whom 98% were men. In her study, Dr. Arya examined the association between the highest statin dose prescribed within 1 year of PAD diagnosis and rates of amputation and all-cause mortality at 1, 3, and 5 years of follow-up. During this interval there were 17,643 amputations, of which 14,981 were considered major amputations, as well as 99,870 deaths.

The 2013 AHA/American College of Cardiology treatment guidelines recommend high-intensity statins for all patients with clinical atherosclerotic disease, including those with PAD (Circulation. 2014 Jun 24;129[25 Suppl 2]:S1-45). (Updated PAD guidelines unveiled at the AHA meeting strongly recommend statin medication for all patients with PAD [Circulation. 2016 Nov 13. doi: 10.1161/CIR.0000000000000470]).However, the bulk of patients in Dr. Arya’s study were captured in the database prior to release of the 2013 guidelines. That may account for the sparse use of high-intensity statin therapy in the study cohort. Indeed, only 11.3% of the PAD patients were on a high-intensity statin. Another 36.2% were on moderate-intensity statin therapy, 3.5% were on low-intensity therapy, and 27.6% weren’t on a statin at all.

The relationship between statin therapy and mortality was strongly dose-dependent.

This study was funded by the AHA and the Atlanta Veterans Affairs Medical Center. Dr. Arya reported having no financial conflicts of interest.
 

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

AT THE AHA SCIENTIFIC SESSIONS

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: High-intensity statin therapy in patients with peripheral artery disease saves lives and limbs.

Major finding: The 5-year all-cause mortality rate after diagnosis of peripheral artery disease was 20.4% in patients on high-intensity statin therapy, 28.6% in those on a low- or moderate-intensity statin, and 33.9% in patients not on a statin.

Data source: A retrospective observational study of 208,194 patients with clinical peripheral artery disease in the national Veterans Affairs database for 2003-2014.

Disclosures: The AHA and the Atlanta Veterans Affairs Medical Center funded the study. The presenter reported having no financial conflicts of interest.

How to reduce readmissions after vascular procedures

Article Type
Changed
Wed, 04/03/2019 - 10:29

 

– As Medicare ratchets up penalties for readmissions and hospitals scrutinize procedures such as carotid interventions and lower extremity bypass that have traditionally high readmission rates, a four-phase model that assesses readmission risks could help vascular surgeons and their institutions keep patients from returning after vascular procedures, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

Dr. Karen Ho
Dr. Karen Ho
“Total Medicare penalties assessed on hospitals for readmissions will increase to $528 million in fiscal year 2017,” Dr. Ho said, quoting data that Kaiser Health News has reported. “Postoperative complications, including wound complications, are among the most common reasons for readmissions and thus are appropriate targets to focus on to decrease readmissions.”

Dr. Ho said one model vascular specialists and hospitals could employ to curtail readmissions was first reported in 2012 by Benjamin S. Brooke, MD, of Dartmouth-Hitchcock Medical Center, Lebanon, N.H., and his colleagues (J Vasc Surg. 2012;56:556-62). This model focuses on the following four phases: 1) develop a deeper understanding of the patient’s preexisting health conditions before surgery; 2) target the in-hospital postoperative period for possible intervention; 3) focus on discharge planning; and 4) determine at the actual readmission event itself if the patient should go to an alternative setting.

“I think as surgeons we often focus on the patients, the procedure, and what goes on in the hospital, but discharge planning and execution is potentially just as important in preventing readmissions,” Dr. Ho said. “This includes things like medication reconciliation, involvement of family, the primary care doctor, the nursing home or rehab facility, and the timing and scheduling of follow-up visits.”

However, she noted that unaccounted factors can also contribute to readmission risk. These can include availability of family to provide support; history of substance abuse; functional status; socioeconomic status; and medical history.

Dr. Ho noted that understanding the reasons for readmission can help vascular specialists gain a deeper understanding of their underlying causes. For example, wound complications top the list in readmissions of numerous vascular procedures, including AAA repair and lower extremity revascularization, but other causes are linked to specific procedures. “If you look at the endovascular repair group in AAA repairs, aneurysm and graft complications were the third most common reason for unplanned 30-day readmission,” she said. A multivariate analysis showed that while preoperative comorbidities had a modest effect on readmission rates after AAA repair, postoperative factors such as complications extending patients’ length of stay and discharge to a setting other than home had a profound effect (Ann Surg. 2012;256:595-605).

In carotid procedures, Dr. Ho noted that carotid artery stenting and CEA had 30-day readmission rates of around 10% (Stroke. 2012;43:2408-16), although CEA seemed to have a slight advantage. Cardiac complications, headache, and bleeding were the top reasons for readmissions for carotid procedures, Dr. Ho said. “In a multivariate analysis, a history of coronary artery bypass and any postoperative complication were associated with readmission,” she said (Vasc Endovascular Surg. 2014;48:217-23).

However, many risk factors for readmission are nonmodifiable, such as patient age 80 and up, or a history of renal failure, heart failure, or diabetes – all characteristics that made patients more prone to readmission after carotid procedures.

Likewise in lower extremity revascularization, nonmodifiable risk factors – end-stage renal disease, heart failure, or tissue loss indication – were prime culprits for readmissions, Dr. Ho noted (J Vasc Surg. 2013;57:955-62). “But also the strongest predictors for readmission included surgical site infections postoperatively as well as graft complications,” she said.

“Risk prediction models for readmissions perform poorly, which makes it difficult to identify high risk and to implement clinically actionable plans to reduce readmissions,” Dr. Ho said. “It also raises the question of whether other important variables, such as social determinants, which may disproportionately affect disadvantaged patients, maybe should be included in these risk prevention models to increase their predicative value.”

Until or if Medicare adjusts its risk evaluation measures accordingly to more accurately reflect the influence of underlying variables such as socioeconomic status and medical history, vascular specialists and their institutions will be pressed to develop programs to reduce readmissions.

Dr. Ho had no relevant financial relationships to disclose.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– As Medicare ratchets up penalties for readmissions and hospitals scrutinize procedures such as carotid interventions and lower extremity bypass that have traditionally high readmission rates, a four-phase model that assesses readmission risks could help vascular surgeons and their institutions keep patients from returning after vascular procedures, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

Dr. Karen Ho
Dr. Karen Ho
“Total Medicare penalties assessed on hospitals for readmissions will increase to $528 million in fiscal year 2017,” Dr. Ho said, quoting data that Kaiser Health News has reported. “Postoperative complications, including wound complications, are among the most common reasons for readmissions and thus are appropriate targets to focus on to decrease readmissions.”

Dr. Ho said one model vascular specialists and hospitals could employ to curtail readmissions was first reported in 2012 by Benjamin S. Brooke, MD, of Dartmouth-Hitchcock Medical Center, Lebanon, N.H., and his colleagues (J Vasc Surg. 2012;56:556-62). This model focuses on the following four phases: 1) develop a deeper understanding of the patient’s preexisting health conditions before surgery; 2) target the in-hospital postoperative period for possible intervention; 3) focus on discharge planning; and 4) determine at the actual readmission event itself if the patient should go to an alternative setting.

“I think as surgeons we often focus on the patients, the procedure, and what goes on in the hospital, but discharge planning and execution is potentially just as important in preventing readmissions,” Dr. Ho said. “This includes things like medication reconciliation, involvement of family, the primary care doctor, the nursing home or rehab facility, and the timing and scheduling of follow-up visits.”

However, she noted that unaccounted factors can also contribute to readmission risk. These can include availability of family to provide support; history of substance abuse; functional status; socioeconomic status; and medical history.

Dr. Ho noted that understanding the reasons for readmission can help vascular specialists gain a deeper understanding of their underlying causes. For example, wound complications top the list in readmissions of numerous vascular procedures, including AAA repair and lower extremity revascularization, but other causes are linked to specific procedures. “If you look at the endovascular repair group in AAA repairs, aneurysm and graft complications were the third most common reason for unplanned 30-day readmission,” she said. A multivariate analysis showed that while preoperative comorbidities had a modest effect on readmission rates after AAA repair, postoperative factors such as complications extending patients’ length of stay and discharge to a setting other than home had a profound effect (Ann Surg. 2012;256:595-605).

In carotid procedures, Dr. Ho noted that carotid artery stenting and CEA had 30-day readmission rates of around 10% (Stroke. 2012;43:2408-16), although CEA seemed to have a slight advantage. Cardiac complications, headache, and bleeding were the top reasons for readmissions for carotid procedures, Dr. Ho said. “In a multivariate analysis, a history of coronary artery bypass and any postoperative complication were associated with readmission,” she said (Vasc Endovascular Surg. 2014;48:217-23).

However, many risk factors for readmission are nonmodifiable, such as patient age 80 and up, or a history of renal failure, heart failure, or diabetes – all characteristics that made patients more prone to readmission after carotid procedures.

Likewise in lower extremity revascularization, nonmodifiable risk factors – end-stage renal disease, heart failure, or tissue loss indication – were prime culprits for readmissions, Dr. Ho noted (J Vasc Surg. 2013;57:955-62). “But also the strongest predictors for readmission included surgical site infections postoperatively as well as graft complications,” she said.

“Risk prediction models for readmissions perform poorly, which makes it difficult to identify high risk and to implement clinically actionable plans to reduce readmissions,” Dr. Ho said. “It also raises the question of whether other important variables, such as social determinants, which may disproportionately affect disadvantaged patients, maybe should be included in these risk prevention models to increase their predicative value.”

Until or if Medicare adjusts its risk evaluation measures accordingly to more accurately reflect the influence of underlying variables such as socioeconomic status and medical history, vascular specialists and their institutions will be pressed to develop programs to reduce readmissions.

Dr. Ho had no relevant financial relationships to disclose.

 

– As Medicare ratchets up penalties for readmissions and hospitals scrutinize procedures such as carotid interventions and lower extremity bypass that have traditionally high readmission rates, a four-phase model that assesses readmission risks could help vascular surgeons and their institutions keep patients from returning after vascular procedures, according to a presentation at a symposium on vascular surgery sponsored by Northwestern University.

Dr. Karen Ho
Dr. Karen Ho
“Total Medicare penalties assessed on hospitals for readmissions will increase to $528 million in fiscal year 2017,” Dr. Ho said, quoting data that Kaiser Health News has reported. “Postoperative complications, including wound complications, are among the most common reasons for readmissions and thus are appropriate targets to focus on to decrease readmissions.”

Dr. Ho said one model vascular specialists and hospitals could employ to curtail readmissions was first reported in 2012 by Benjamin S. Brooke, MD, of Dartmouth-Hitchcock Medical Center, Lebanon, N.H., and his colleagues (J Vasc Surg. 2012;56:556-62). This model focuses on the following four phases: 1) develop a deeper understanding of the patient’s preexisting health conditions before surgery; 2) target the in-hospital postoperative period for possible intervention; 3) focus on discharge planning; and 4) determine at the actual readmission event itself if the patient should go to an alternative setting.

“I think as surgeons we often focus on the patients, the procedure, and what goes on in the hospital, but discharge planning and execution is potentially just as important in preventing readmissions,” Dr. Ho said. “This includes things like medication reconciliation, involvement of family, the primary care doctor, the nursing home or rehab facility, and the timing and scheduling of follow-up visits.”

However, she noted that unaccounted factors can also contribute to readmission risk. These can include availability of family to provide support; history of substance abuse; functional status; socioeconomic status; and medical history.

Dr. Ho noted that understanding the reasons for readmission can help vascular specialists gain a deeper understanding of their underlying causes. For example, wound complications top the list in readmissions of numerous vascular procedures, including AAA repair and lower extremity revascularization, but other causes are linked to specific procedures. “If you look at the endovascular repair group in AAA repairs, aneurysm and graft complications were the third most common reason for unplanned 30-day readmission,” she said. A multivariate analysis showed that while preoperative comorbidities had a modest effect on readmission rates after AAA repair, postoperative factors such as complications extending patients’ length of stay and discharge to a setting other than home had a profound effect (Ann Surg. 2012;256:595-605).

In carotid procedures, Dr. Ho noted that carotid artery stenting and CEA had 30-day readmission rates of around 10% (Stroke. 2012;43:2408-16), although CEA seemed to have a slight advantage. Cardiac complications, headache, and bleeding were the top reasons for readmissions for carotid procedures, Dr. Ho said. “In a multivariate analysis, a history of coronary artery bypass and any postoperative complication were associated with readmission,” she said (Vasc Endovascular Surg. 2014;48:217-23).

However, many risk factors for readmission are nonmodifiable, such as patient age 80 and up, or a history of renal failure, heart failure, or diabetes – all characteristics that made patients more prone to readmission after carotid procedures.

Likewise in lower extremity revascularization, nonmodifiable risk factors – end-stage renal disease, heart failure, or tissue loss indication – were prime culprits for readmissions, Dr. Ho noted (J Vasc Surg. 2013;57:955-62). “But also the strongest predictors for readmission included surgical site infections postoperatively as well as graft complications,” she said.

“Risk prediction models for readmissions perform poorly, which makes it difficult to identify high risk and to implement clinically actionable plans to reduce readmissions,” Dr. Ho said. “It also raises the question of whether other important variables, such as social determinants, which may disproportionately affect disadvantaged patients, maybe should be included in these risk prevention models to increase their predicative value.”

Until or if Medicare adjusts its risk evaluation measures accordingly to more accurately reflect the influence of underlying variables such as socioeconomic status and medical history, vascular specialists and their institutions will be pressed to develop programs to reduce readmissions.

Dr. Ho had no relevant financial relationships to disclose.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

AT THE NORTHWESTERN VASCULAR SYMPOSIUM

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: A four-phase model that assesses readmission risks could help vascular surgeons and their institutions keep patients from returning after procedures.

Major finding: Readmission rates for cardiac and vascular interventions range from around 13% for aortic aneurysm repair to 20% after lower extremity revascularization.

Data source: Literature review.

Disclosures: Dr. Ho reported having no financial disclosures.

Point/Counterpoint: Is limb salvage always best in diabetes?

Article Type
Changed
Tue, 05/03/2022 - 15:31

 

Salvage limbs at all costs

Aggressive limb salvage in people with diabetes leads to an overall reduction in cost not only economically, but also from the patient’s perspective. The vast majority of diabetic patients with critical ischemia are actually good candidates for limb salvage. Tragically, many of these patients are never referred for evaluation for limb salvage because of misconceptions about the pathophysiology of the disease.

An argument against limb salvage is that primary amputation prevents or shortens the course of wound care and enables patients to become ambulatory, albeit with a prosthesis, faster. However, in the modern era of vascular surgery, revascularization can be performed successfully with minimal mortality and excellent rates of limb salvage, especially when it’s done within a team-based approach.

Dr. Trissa A. Babrowski
Dr. Trissa A. Babrowski
Limb salvage in people with diabetes is a urgent public health issue. Today 29 million U.S. adults have diabetes, nearly a quarter of whom are undiagnosed. Ninety million U.S. adults are glucose intolerant. Diabetes is by far the most common cause of nontraumatic amputation in the United States; the Centers for Disease Control and Prevention estimated 73,000 nontraumatic adult amputations in 2010 – about 44,000 in diabetics. That means nearly 2 million Americans are living with an amputation today. That will rise to 3.6 million by 2050.

The mortality in primary amputation is shockingly high, anywhere from 5% to 23% higher than revascularization alone, and the major complication rate of amputation associated with diabetes is also unacceptably high – up to 37%. This is in contrast to a 17% rate in major nonamputation vascular surgery and 1%-5% in endovascular procedures (BMC Nephrol. 2005;6:3).

We can’t ignore the economic burden this places on the country. In 2014, primary amputations cost the health care system $11 billion annually, and that is expected to grow to more than $25 billion in the next several years, according to the SAGE Group. It’s important to keep in mind that Medicare covers over 80% of this cost.

A number of studies have shown that conservative management with wound care and amputation is more cost effective than primary amputation in ambulatory, independent adults. Data can be difficult to interpret because of different recording strategies for all the costs associated with amputation, but a single-institution study concluded that revascularization costs almost $5,280 more than expectant management, but $33,900 less than primary amputation alone (Cardiovasc Surg. 1999;7;62-9).

We must also consider the costs of revision after primary amputation; above-the-knee amputation has a 12% in-hospital revision rate, and below-the-knee amputation about 20%. Endovascular interventions, on the other hand, have a 1%-9% in-hospital revision rate, and only 2%-4% of these patients will go on to require an amputation during the same admission (Eur J Vasc Endovasc Surg. 2006;32:484-90; Arch Phys Med Rehabil. 2005;86:480-6).This does not include the costs of those complications as well as other indirect costs of amputation, such as nursing home care and living situation modification (Int J Behav Med. 2016;23:714-21; Pak J Med Sci. 2014; 30:1044-9). They quickly add up to that $25 billion.

The proponents of primary amputation tell us that it leads to quicker recovery time and an earlier time to ambulation. However, only 47% of patients will actually ambulate after amputation, in contrast to 97% who will ambulate after limb salvage as a primary procedure. In a nonambulatory cohort, 21% of those patients go on to regain functional status that was lost prior to surgery (J Vasc Surg. 1997;25;287-95).

Many question if our success with vascular surgery over the past few decades can translate to helping the most difficult subset of patients. An Italian study reported on a cohort of diabetic vs. nondiabetic patients and determined both groups have similar amputation-free rates after infrainguinal arterial reconstruction for critical limb ischemia, with excellent primary and secondary patency rates and a limb salvage rate of 88% at 5 years (J Vasc Surg. 2014;59:708-19). This tells us that we do have the skill set necessary to save these limbs.

A multidisciplinary limb preservation team is paramount to the success of any limb salvage program. A revascularization team should be in place which uses early intervention to achieve the highest limb salvage rates possible. Wound care needs to be an integrated part of it. Advanced podiatric reconstructive surgery also is key because this can provide complex foot reconstructions and help ambulatory patients return home.

Dr. Trissa A. Babrowski is an assistant professor of surgery, specializing in vascular surgery and endovascular therapy, at the University of Chicago Heart and Vascular Center. She had no financial relationships to disclose.

 

 

Primary amputation can be OK

I am not an amputationalist. I do practice limb salvage. In fact I’m probably the most aggressive limb salvage surgeon in my hospital. But primary amputation is a completely acceptable option for a selected group of patients with diabetes. We should not try to do limb salvage “at all costs.”

I do not find this to be a contradictory position. In fact, I think it adds credence to my support of limb salvage that I think primary amputation can be OK. In all honesty, there are very few things in life that should be done at all costs.

Dr. Timothy J. Nypaver
Dr. Timothy J. Nypaver
Bypass revascularization efforts to prevent amputation can fail, and many of them do so frequently in the first year. These failures can often be predicted preoperatively. Appropriate application of primary amputation based on data not only improves cost-effectiveness and patients’ quality of life, but improves our outcomes overall for critical limb ischemia (CLI).

A study out of Loma Linda University involving patients with CLI compared primary amputation vs. revascularization; 43% of patients had a primary amputation (Ann Vasc Surg. 2007;21:458-63). A multivariate analysis showed that patients with major tissue loss, end-stage renal disease (ESRD), diabetes and nonambulatory status were more likely to undergo primary amputation rather than revascularization.

While major tissue loss (Rutherford category 6) is certainly an indication for primary amputation, ambulatory status can represent a gray area in determining the best course. ESRD and diabetes are much more nonspecific factors; probably more than 10% of the patients that we see with CLI have ESRD. Also, 50%-70% of these patients with CLI, and in some series even higher percentages, have diabetes. Thus, these factors by themselves do not assist us in determining which patients potentially should be offered primary amputation vs. revascularization.

In general, we know that we can get good results in limb bypass or revascularization in patients with CLI: The PREVENT III multicenter trial, with the use of the vein as the conduit, showed 1-year limb salvage rates of 88% in these high-risk patients (J Vasc Surg. 2006;43:742-51). However, one of the major risk factors that adversely affected outcome was ESRD.

We know that ESRD is a significant predictor of lowering our chances of saving a limb successfully. Knowing the cost of multiple continued episodes of revascularization in these patients prior to proceeding with an amputation, it’s intuitive that these patients would benefit from a more precise process in their treatment from the beginning. A number of papers have concluded that a primary amputation may be the preferred approach in patients with ESRD.

Can we preoperatively predict which patients with CLI will fail operative revascularization? Data from the New England Vascular Quality Initiative identified eight variables associated with failure of revascularization, among them age younger than 59, ESRD, diabetes, CLI, conduit requiring venovenostomy, tarsal target, and nursing home residence (Ann Vasc Surg. 2010;24:57-68). The presence of three or more risk factors has a 27.7% risk of limb loss and/or graft thrombosis within 1 year.

Postponing amputation is a major cost issue. Direct costs of bypass for critical limb ischemia were $3.6 billion in 2004 (J Vasc Surg. 2011;54:1021-31), and we know that a functional outcome can be problematic in this patient group. Factors associated with a poor functional outcome include dementia, dependent-living situation preoperatively and nonambulatory status.

Unfortunately, there are not a lot of data that deal with quality of life outcomes for patients with CLI who have undergone bypass. Using a point system comprised of dialysis (4 points), tissue loss (3 points), age above 75 (2 points), hematocrit less than or equal to 30 (2 points), and coronary artery disease (1 point), a follow-up study of patients in the PREVENT III trial found that a high-risk group (greater than or equal to 8 points) had an amputation-free survival of only 45% (J Vasc Surg. 2009;50:769-75). Again, these results do not justify the effort and costs of limb salvage in this high-risk patient group.

We should consider the following options carefully in selecting a cost-effective patient-focused approach in patients with CLI: wound care, primary amputation, bypass revascularization, or endovascular revascularization. I would argue that the vascular surgeon who is qualified as an expert in all of the above is best positioned to select an appropriate plan of treatment based upon the patient’s risk factors, wound factors, ambulatory ability, pattern of disease, severity of ischemia, and living status.

Thus, upon presentation, a patient with CLI should undergo confirmatory tests and optimize his or her risk factors. The vascular surgeon then has the option, in discussion with the patient and family, to pursue an appropriate treatment plan inclusive of primary amputation – not one of limb salvage “at all costs.”

Primary amputation should be used in situations where there is dementia and nonambulatory status, and in patients who are poor candidates for revascularization because of high risk of failure and limited life expectancy. The recently developed WIfI (wound, ischemia, and foot infection) classification can also be utilized, as stage 4 WIfI classification is associated with high risk of limb loss – 38%-40% at 1 year.

Primary amputation is an option that can result in better care overall, and it is a cost-effective approach for a selected group of patients. We should not try to do limb salvage at all cost. Primary amputation, in selected patients, is OK.

 

 

 

Dr. Timothy J. Nypaver is head of vascular surgery at Henry Ford Hospital, Detroit. He had no financial relationships to disclose.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

Salvage limbs at all costs

Aggressive limb salvage in people with diabetes leads to an overall reduction in cost not only economically, but also from the patient’s perspective. The vast majority of diabetic patients with critical ischemia are actually good candidates for limb salvage. Tragically, many of these patients are never referred for evaluation for limb salvage because of misconceptions about the pathophysiology of the disease.

An argument against limb salvage is that primary amputation prevents or shortens the course of wound care and enables patients to become ambulatory, albeit with a prosthesis, faster. However, in the modern era of vascular surgery, revascularization can be performed successfully with minimal mortality and excellent rates of limb salvage, especially when it’s done within a team-based approach.

Dr. Trissa A. Babrowski
Dr. Trissa A. Babrowski
Limb salvage in people with diabetes is a urgent public health issue. Today 29 million U.S. adults have diabetes, nearly a quarter of whom are undiagnosed. Ninety million U.S. adults are glucose intolerant. Diabetes is by far the most common cause of nontraumatic amputation in the United States; the Centers for Disease Control and Prevention estimated 73,000 nontraumatic adult amputations in 2010 – about 44,000 in diabetics. That means nearly 2 million Americans are living with an amputation today. That will rise to 3.6 million by 2050.

The mortality in primary amputation is shockingly high, anywhere from 5% to 23% higher than revascularization alone, and the major complication rate of amputation associated with diabetes is also unacceptably high – up to 37%. This is in contrast to a 17% rate in major nonamputation vascular surgery and 1%-5% in endovascular procedures (BMC Nephrol. 2005;6:3).

We can’t ignore the economic burden this places on the country. In 2014, primary amputations cost the health care system $11 billion annually, and that is expected to grow to more than $25 billion in the next several years, according to the SAGE Group. It’s important to keep in mind that Medicare covers over 80% of this cost.

A number of studies have shown that conservative management with wound care and amputation is more cost effective than primary amputation in ambulatory, independent adults. Data can be difficult to interpret because of different recording strategies for all the costs associated with amputation, but a single-institution study concluded that revascularization costs almost $5,280 more than expectant management, but $33,900 less than primary amputation alone (Cardiovasc Surg. 1999;7;62-9).

We must also consider the costs of revision after primary amputation; above-the-knee amputation has a 12% in-hospital revision rate, and below-the-knee amputation about 20%. Endovascular interventions, on the other hand, have a 1%-9% in-hospital revision rate, and only 2%-4% of these patients will go on to require an amputation during the same admission (Eur J Vasc Endovasc Surg. 2006;32:484-90; Arch Phys Med Rehabil. 2005;86:480-6).This does not include the costs of those complications as well as other indirect costs of amputation, such as nursing home care and living situation modification (Int J Behav Med. 2016;23:714-21; Pak J Med Sci. 2014; 30:1044-9). They quickly add up to that $25 billion.

The proponents of primary amputation tell us that it leads to quicker recovery time and an earlier time to ambulation. However, only 47% of patients will actually ambulate after amputation, in contrast to 97% who will ambulate after limb salvage as a primary procedure. In a nonambulatory cohort, 21% of those patients go on to regain functional status that was lost prior to surgery (J Vasc Surg. 1997;25;287-95).

Many question if our success with vascular surgery over the past few decades can translate to helping the most difficult subset of patients. An Italian study reported on a cohort of diabetic vs. nondiabetic patients and determined both groups have similar amputation-free rates after infrainguinal arterial reconstruction for critical limb ischemia, with excellent primary and secondary patency rates and a limb salvage rate of 88% at 5 years (J Vasc Surg. 2014;59:708-19). This tells us that we do have the skill set necessary to save these limbs.

A multidisciplinary limb preservation team is paramount to the success of any limb salvage program. A revascularization team should be in place which uses early intervention to achieve the highest limb salvage rates possible. Wound care needs to be an integrated part of it. Advanced podiatric reconstructive surgery also is key because this can provide complex foot reconstructions and help ambulatory patients return home.

Dr. Trissa A. Babrowski is an assistant professor of surgery, specializing in vascular surgery and endovascular therapy, at the University of Chicago Heart and Vascular Center. She had no financial relationships to disclose.

 

 

Primary amputation can be OK

I am not an amputationalist. I do practice limb salvage. In fact I’m probably the most aggressive limb salvage surgeon in my hospital. But primary amputation is a completely acceptable option for a selected group of patients with diabetes. We should not try to do limb salvage “at all costs.”

I do not find this to be a contradictory position. In fact, I think it adds credence to my support of limb salvage that I think primary amputation can be OK. In all honesty, there are very few things in life that should be done at all costs.

Dr. Timothy J. Nypaver
Dr. Timothy J. Nypaver
Bypass revascularization efforts to prevent amputation can fail, and many of them do so frequently in the first year. These failures can often be predicted preoperatively. Appropriate application of primary amputation based on data not only improves cost-effectiveness and patients’ quality of life, but improves our outcomes overall for critical limb ischemia (CLI).

A study out of Loma Linda University involving patients with CLI compared primary amputation vs. revascularization; 43% of patients had a primary amputation (Ann Vasc Surg. 2007;21:458-63). A multivariate analysis showed that patients with major tissue loss, end-stage renal disease (ESRD), diabetes and nonambulatory status were more likely to undergo primary amputation rather than revascularization.

While major tissue loss (Rutherford category 6) is certainly an indication for primary amputation, ambulatory status can represent a gray area in determining the best course. ESRD and diabetes are much more nonspecific factors; probably more than 10% of the patients that we see with CLI have ESRD. Also, 50%-70% of these patients with CLI, and in some series even higher percentages, have diabetes. Thus, these factors by themselves do not assist us in determining which patients potentially should be offered primary amputation vs. revascularization.

In general, we know that we can get good results in limb bypass or revascularization in patients with CLI: The PREVENT III multicenter trial, with the use of the vein as the conduit, showed 1-year limb salvage rates of 88% in these high-risk patients (J Vasc Surg. 2006;43:742-51). However, one of the major risk factors that adversely affected outcome was ESRD.

We know that ESRD is a significant predictor of lowering our chances of saving a limb successfully. Knowing the cost of multiple continued episodes of revascularization in these patients prior to proceeding with an amputation, it’s intuitive that these patients would benefit from a more precise process in their treatment from the beginning. A number of papers have concluded that a primary amputation may be the preferred approach in patients with ESRD.

Can we preoperatively predict which patients with CLI will fail operative revascularization? Data from the New England Vascular Quality Initiative identified eight variables associated with failure of revascularization, among them age younger than 59, ESRD, diabetes, CLI, conduit requiring venovenostomy, tarsal target, and nursing home residence (Ann Vasc Surg. 2010;24:57-68). The presence of three or more risk factors has a 27.7% risk of limb loss and/or graft thrombosis within 1 year.

Postponing amputation is a major cost issue. Direct costs of bypass for critical limb ischemia were $3.6 billion in 2004 (J Vasc Surg. 2011;54:1021-31), and we know that a functional outcome can be problematic in this patient group. Factors associated with a poor functional outcome include dementia, dependent-living situation preoperatively and nonambulatory status.

Unfortunately, there are not a lot of data that deal with quality of life outcomes for patients with CLI who have undergone bypass. Using a point system comprised of dialysis (4 points), tissue loss (3 points), age above 75 (2 points), hematocrit less than or equal to 30 (2 points), and coronary artery disease (1 point), a follow-up study of patients in the PREVENT III trial found that a high-risk group (greater than or equal to 8 points) had an amputation-free survival of only 45% (J Vasc Surg. 2009;50:769-75). Again, these results do not justify the effort and costs of limb salvage in this high-risk patient group.

We should consider the following options carefully in selecting a cost-effective patient-focused approach in patients with CLI: wound care, primary amputation, bypass revascularization, or endovascular revascularization. I would argue that the vascular surgeon who is qualified as an expert in all of the above is best positioned to select an appropriate plan of treatment based upon the patient’s risk factors, wound factors, ambulatory ability, pattern of disease, severity of ischemia, and living status.

Thus, upon presentation, a patient with CLI should undergo confirmatory tests and optimize his or her risk factors. The vascular surgeon then has the option, in discussion with the patient and family, to pursue an appropriate treatment plan inclusive of primary amputation – not one of limb salvage “at all costs.”

Primary amputation should be used in situations where there is dementia and nonambulatory status, and in patients who are poor candidates for revascularization because of high risk of failure and limited life expectancy. The recently developed WIfI (wound, ischemia, and foot infection) classification can also be utilized, as stage 4 WIfI classification is associated with high risk of limb loss – 38%-40% at 1 year.

Primary amputation is an option that can result in better care overall, and it is a cost-effective approach for a selected group of patients. We should not try to do limb salvage at all cost. Primary amputation, in selected patients, is OK.

 

 

 

Dr. Timothy J. Nypaver is head of vascular surgery at Henry Ford Hospital, Detroit. He had no financial relationships to disclose.

 

Salvage limbs at all costs

Aggressive limb salvage in people with diabetes leads to an overall reduction in cost not only economically, but also from the patient’s perspective. The vast majority of diabetic patients with critical ischemia are actually good candidates for limb salvage. Tragically, many of these patients are never referred for evaluation for limb salvage because of misconceptions about the pathophysiology of the disease.

An argument against limb salvage is that primary amputation prevents or shortens the course of wound care and enables patients to become ambulatory, albeit with a prosthesis, faster. However, in the modern era of vascular surgery, revascularization can be performed successfully with minimal mortality and excellent rates of limb salvage, especially when it’s done within a team-based approach.

Dr. Trissa A. Babrowski
Dr. Trissa A. Babrowski
Limb salvage in people with diabetes is a urgent public health issue. Today 29 million U.S. adults have diabetes, nearly a quarter of whom are undiagnosed. Ninety million U.S. adults are glucose intolerant. Diabetes is by far the most common cause of nontraumatic amputation in the United States; the Centers for Disease Control and Prevention estimated 73,000 nontraumatic adult amputations in 2010 – about 44,000 in diabetics. That means nearly 2 million Americans are living with an amputation today. That will rise to 3.6 million by 2050.

The mortality in primary amputation is shockingly high, anywhere from 5% to 23% higher than revascularization alone, and the major complication rate of amputation associated with diabetes is also unacceptably high – up to 37%. This is in contrast to a 17% rate in major nonamputation vascular surgery and 1%-5% in endovascular procedures (BMC Nephrol. 2005;6:3).

We can’t ignore the economic burden this places on the country. In 2014, primary amputations cost the health care system $11 billion annually, and that is expected to grow to more than $25 billion in the next several years, according to the SAGE Group. It’s important to keep in mind that Medicare covers over 80% of this cost.

A number of studies have shown that conservative management with wound care and amputation is more cost effective than primary amputation in ambulatory, independent adults. Data can be difficult to interpret because of different recording strategies for all the costs associated with amputation, but a single-institution study concluded that revascularization costs almost $5,280 more than expectant management, but $33,900 less than primary amputation alone (Cardiovasc Surg. 1999;7;62-9).

We must also consider the costs of revision after primary amputation; above-the-knee amputation has a 12% in-hospital revision rate, and below-the-knee amputation about 20%. Endovascular interventions, on the other hand, have a 1%-9% in-hospital revision rate, and only 2%-4% of these patients will go on to require an amputation during the same admission (Eur J Vasc Endovasc Surg. 2006;32:484-90; Arch Phys Med Rehabil. 2005;86:480-6).This does not include the costs of those complications as well as other indirect costs of amputation, such as nursing home care and living situation modification (Int J Behav Med. 2016;23:714-21; Pak J Med Sci. 2014; 30:1044-9). They quickly add up to that $25 billion.

The proponents of primary amputation tell us that it leads to quicker recovery time and an earlier time to ambulation. However, only 47% of patients will actually ambulate after amputation, in contrast to 97% who will ambulate after limb salvage as a primary procedure. In a nonambulatory cohort, 21% of those patients go on to regain functional status that was lost prior to surgery (J Vasc Surg. 1997;25;287-95).

Many question if our success with vascular surgery over the past few decades can translate to helping the most difficult subset of patients. An Italian study reported on a cohort of diabetic vs. nondiabetic patients and determined both groups have similar amputation-free rates after infrainguinal arterial reconstruction for critical limb ischemia, with excellent primary and secondary patency rates and a limb salvage rate of 88% at 5 years (J Vasc Surg. 2014;59:708-19). This tells us that we do have the skill set necessary to save these limbs.

A multidisciplinary limb preservation team is paramount to the success of any limb salvage program. A revascularization team should be in place which uses early intervention to achieve the highest limb salvage rates possible. Wound care needs to be an integrated part of it. Advanced podiatric reconstructive surgery also is key because this can provide complex foot reconstructions and help ambulatory patients return home.

Dr. Trissa A. Babrowski is an assistant professor of surgery, specializing in vascular surgery and endovascular therapy, at the University of Chicago Heart and Vascular Center. She had no financial relationships to disclose.

 

 

Primary amputation can be OK

I am not an amputationalist. I do practice limb salvage. In fact I’m probably the most aggressive limb salvage surgeon in my hospital. But primary amputation is a completely acceptable option for a selected group of patients with diabetes. We should not try to do limb salvage “at all costs.”

I do not find this to be a contradictory position. In fact, I think it adds credence to my support of limb salvage that I think primary amputation can be OK. In all honesty, there are very few things in life that should be done at all costs.

Dr. Timothy J. Nypaver
Dr. Timothy J. Nypaver
Bypass revascularization efforts to prevent amputation can fail, and many of them do so frequently in the first year. These failures can often be predicted preoperatively. Appropriate application of primary amputation based on data not only improves cost-effectiveness and patients’ quality of life, but improves our outcomes overall for critical limb ischemia (CLI).

A study out of Loma Linda University involving patients with CLI compared primary amputation vs. revascularization; 43% of patients had a primary amputation (Ann Vasc Surg. 2007;21:458-63). A multivariate analysis showed that patients with major tissue loss, end-stage renal disease (ESRD), diabetes and nonambulatory status were more likely to undergo primary amputation rather than revascularization.

While major tissue loss (Rutherford category 6) is certainly an indication for primary amputation, ambulatory status can represent a gray area in determining the best course. ESRD and diabetes are much more nonspecific factors; probably more than 10% of the patients that we see with CLI have ESRD. Also, 50%-70% of these patients with CLI, and in some series even higher percentages, have diabetes. Thus, these factors by themselves do not assist us in determining which patients potentially should be offered primary amputation vs. revascularization.

In general, we know that we can get good results in limb bypass or revascularization in patients with CLI: The PREVENT III multicenter trial, with the use of the vein as the conduit, showed 1-year limb salvage rates of 88% in these high-risk patients (J Vasc Surg. 2006;43:742-51). However, one of the major risk factors that adversely affected outcome was ESRD.

We know that ESRD is a significant predictor of lowering our chances of saving a limb successfully. Knowing the cost of multiple continued episodes of revascularization in these patients prior to proceeding with an amputation, it’s intuitive that these patients would benefit from a more precise process in their treatment from the beginning. A number of papers have concluded that a primary amputation may be the preferred approach in patients with ESRD.

Can we preoperatively predict which patients with CLI will fail operative revascularization? Data from the New England Vascular Quality Initiative identified eight variables associated with failure of revascularization, among them age younger than 59, ESRD, diabetes, CLI, conduit requiring venovenostomy, tarsal target, and nursing home residence (Ann Vasc Surg. 2010;24:57-68). The presence of three or more risk factors has a 27.7% risk of limb loss and/or graft thrombosis within 1 year.

Postponing amputation is a major cost issue. Direct costs of bypass for critical limb ischemia were $3.6 billion in 2004 (J Vasc Surg. 2011;54:1021-31), and we know that a functional outcome can be problematic in this patient group. Factors associated with a poor functional outcome include dementia, dependent-living situation preoperatively and nonambulatory status.

Unfortunately, there are not a lot of data that deal with quality of life outcomes for patients with CLI who have undergone bypass. Using a point system comprised of dialysis (4 points), tissue loss (3 points), age above 75 (2 points), hematocrit less than or equal to 30 (2 points), and coronary artery disease (1 point), a follow-up study of patients in the PREVENT III trial found that a high-risk group (greater than or equal to 8 points) had an amputation-free survival of only 45% (J Vasc Surg. 2009;50:769-75). Again, these results do not justify the effort and costs of limb salvage in this high-risk patient group.

We should consider the following options carefully in selecting a cost-effective patient-focused approach in patients with CLI: wound care, primary amputation, bypass revascularization, or endovascular revascularization. I would argue that the vascular surgeon who is qualified as an expert in all of the above is best positioned to select an appropriate plan of treatment based upon the patient’s risk factors, wound factors, ambulatory ability, pattern of disease, severity of ischemia, and living status.

Thus, upon presentation, a patient with CLI should undergo confirmatory tests and optimize his or her risk factors. The vascular surgeon then has the option, in discussion with the patient and family, to pursue an appropriate treatment plan inclusive of primary amputation – not one of limb salvage “at all costs.”

Primary amputation should be used in situations where there is dementia and nonambulatory status, and in patients who are poor candidates for revascularization because of high risk of failure and limited life expectancy. The recently developed WIfI (wound, ischemia, and foot infection) classification can also be utilized, as stage 4 WIfI classification is associated with high risk of limb loss – 38%-40% at 1 year.

Primary amputation is an option that can result in better care overall, and it is a cost-effective approach for a selected group of patients. We should not try to do limb salvage at all cost. Primary amputation, in selected patients, is OK.

 

 

 

Dr. Timothy J. Nypaver is head of vascular surgery at Henry Ford Hospital, Detroit. He had no financial relationships to disclose.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME