There is growing recognition of the effect of peripheral

REVIEW Circ J 2017; 81: 281 – 289 doi: 10.1253/circj.CJ-16-1286 Pathophysiology of Intermittent Claudication in Peripheral Artery Disease Naomi M. H...
Author: Arthur Webster
5 downloads 0 Views 420KB Size
REVIEW

Circ J 2017; 81: 281 – 289 doi: 10.1253/circj.CJ-16-1286

Pathophysiology of Intermittent Claudication in Peripheral Artery Disease Naomi M. Hamburg, MD; Mark A. Creager, MD Peripheral artery disease (PAD) affects more than 200 million adults worldwide. Patients with lower extremity PAD have a heightened risk for cardiovascular events because of the systemic nature of atherosclerosis, and benefit from treatment with risk factor-modifying therapies. Limb symptoms in PAD include intermittent claudication and diminished walking ability. Arterial obstruction from atherosclerotic lesions initiates limb ischemia; however, decreased perfusion incompletely determines the clinical expression of PAD and its response to therapy. Potential mechanistic drivers of claudication in addition to arterial obstruction include inflammation, vascular dysfunction, reduced microvascular flow, impaired angiogenesis, and altered skeletal muscle function. An improved understanding of the pathophysiology of limb symptoms has the potential to accelerate development of novel therapeutic strategies to increase functional capacity in patients with PAD. Key Words: Blood flow; Claudication; Exercise; Ischemia; Peripheral artery disease

T

here is growing recognition of the effect of peripheral artery disease (PAD) on cardiovascular health.1 Recent studies indicate that, globally, over 200 million adults have PAD,2,3 which is an expression of systemic atherosclerosis and well-established as heightening the risk for cardiovascular events.4–6 The limb manifestations of PAD induce considerable suffering. Patients with PAD experience intermittent claudication, characterized as exertional leg pain that limits walking ability, and often times, disability.7–9 Current medical therapies to reduce the burden of lower extremity symptoms in patients with PAD are limited. Revascularization by endovascular intervention or surgical reconstruction is used to treat lifestyle-limiting claudication if patients do not respond adequately to medical therapy including exercise training. Indeed, lower extremity revascularization with endovascular approaches has undergone a dramatic increase in use.10 Though endovascular therapy improves blood flow and function, there are significant associated risks, and durability may be limited, especially in infrainguinal disease. Abundant evidence demonstrates the benefits of exercise therapy in claudication, yet the mechanisms underlying the beneficial effects remain incompletely defined.11 Thus, an improved understanding of the pathophysiology of limb symptoms in PAD is critical to accelerate the development of novel therapies.

Global Epidemiology of PAD A set of recent international studies highlights the worldwide prevalence of PAD. The Global Burden of Disease study

estimates a more than 30% increase in deaths and disability attributable to PAD between 2005 and 2015, which is largely determined by population aging.3,12 PAD prevalence escalates with advancing age, affecting 1 in 10 adults older than 70 years. In addition to age, smoking and diabetes are the strongest risk factors for PAD across the world. A comprehensive global evaluation of PAD found that its prevalence and risk factors are similar in high-income countries to low- and middle-income countries.2,13 Lower socioeconomic status is a newly recognized risk factor for PAD.14 Importantly, there has been a greater increase in PAD prevalence in low- and middle-income countries, rising by 28.7% from 2000 to 2010 as compared with 13.1% in high-income countries.2,15 The changing burden of PAD is also greater in women as compared with men. The health implications of PAD derive from both its limb and cardiovascular manifestations. Consistent with the systemic nature of atherosclerosis, polyvascular disease is common in patients with PAD.16 In the REACH registry, 61% of PAD patients had concomitant coronary artery disease (CAD) and/or cerebrovascular disease.16 Thus, the presence of PAD portends a high risk for subsequent cardiovascular events. A meta-analysis of over 48,000 participants in population-based cohort studies demonstrated that a low ankle-brachial index (ABI) predicted a 2-fold risk of death, cardiovascular death, and major coronary events at all ranges of the Framingham Risk Score.4 Further, the elevated risk of cardiovascular events is present in both symptomatic and asymptomatic patients.17 Patients with atherosclerotic disease in more than 1 vascular territory have particularly poor outcomes.6 In the over 5,000 Japanese

Received December 27, 2016; accepted December 27, 2016; released online January 26, 2017 Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA (N.M.H.); the Section of Vascular Biology, Department of Medicine, Boston Medical Center, Boston, MA (N.M.H.); and Dartmouth-Hitchcock Heart and Vascular Center and the Geisel School of Medicine at Dartmouth, Lebanon, NH (M.A.C.), USA Mailing address:  Mark A. Creager, MD, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA.  E-mail: [email protected] ISSN-1346-9843   All rights are reserved to the Japanese Circulation Society. For permissions, please e-mail: [email protected] Circulation Journal  Vol.81, March 2017

282

HAMBURG NM et al.

Table.  Medical Therapies for Symptomatic Peripheral Artery Disease Cardiovascular risk reduction

Leg symptoms

Antiplatelet therapy

Pharmacologic treatment

    •  Treatment with aspirin or clopidogrel is recommended

    •  Cilostazol is effective to increase walking distance

    •  Benefits of vorapaxar as additional antiplatelet therapy are uncertain Statin agents

Exercise programs

    •  Treatment with a statin medication is indicated

    •  Supervised exercise program is recommended in patients with claudication

Antihypertensive therapy

    •  Structured community or home-based exercise program with behavioral change techniques can be beneficial alternative strategies of exercise therapy, including upper-body ergometry, cycling, and pain-free or low-intensity walking can be beneficial

    •  Antihypertensive therapy should be used to treat hypertension     •  ACE inhibitors or ARBs can be effective in reducing the risk of cardiovascular ischemic events in patients with PAD Smoking cessation     •  Advice to quit and development of a strategy for cessation that includes pharmacotherapy and smoking cessation program

Adapted from the American College of Cardiology/American Heart Association 2016 Guideline on the Management of Patients With Lower Extremity Peripheral Artery Disease. ACE, angiotensin-converting enzyme; ARB, angiotensin-receptor blocker.

patients studied in the REACH registry, the incidence of the composite outcome (cardiovascular death/myocardial infarction/stroke/hospitalization) was highest in patients with PAD, and the presence of PAD in patients with cerebrovascular disease was associated with a doubling of the 1-year rate of stroke and myocardial infarction.18 Thus, PAD is a key atherosclerotic marker, and its diagnosis and treatment may afford an opportunity to reduce the risk of a cardiovascular event. Risk reduction therapy continues to be underutilized in patients with PAD. Successful implementation of medical therapy to target cardiovascular risk is critical to reducing the cardiovascular morbidity and mortality in PAD.19 Medical therapy for cardiovascular risk prevention in PAD has been reviewed elsewhere and updated in the recent AHA/ACC lower extremity guidelines.20,21 As outlined in the Table, several therapies have proven efficacy to reduce cardiovascular risk in PAD and are recommended as guideline-directed therapy.

Functional Effect of PAD Patients with PAD suffer from limb disability. Traditionally, functional limitation has been ascribed to intermittent claudication, the classical PAD symptom characterized by leg pain with activity that resolves with rest.22 Intermittent claudication is present in only 10–20% of PAD patients. However, a large proportion of PAD patients report atypical leg symptoms that restrict walking ability.22–24 Patients with reduced ABI, a ratio of the ankle and brachial systolic pressures, have impaired functional ability regardless of symptom status.25 In prospective studies, both symptomatic and asymptomatic patients with PAD are at risk for progressive functional impairment.7,26,27 The walking limitations in PAD patients are multifactorial in origin and are reflected in the self-restriction of activity to reduce the occurrence of leg discomfort.28 Reduced walking function in patients with PAD is associated with poor clinical outcomes. Quality of life is reduced in patients with PAD, reflecting lower extremity pain and activity restriction.29 Studies that have assessed patients with a walking impairment questionnaire and measurement of walking times demonstrate that reduced

walking ability is associated with higher rates of cardiovascular events and death.30–33 A recent meta-analysis of 10 studies demonstrated that shorter walking distance was associated with higher cardiovascular and all-cause deaths.34 Declines in self-reported exercise performance enhance prediction of mortality risk in PAD.35 Lower physical activity also predicts progression of PAD with greater decline in the ABI over time.36 Thus, therapeutic approaches that restore functional capacity in patients with PAD may have a wide-ranging health-promoting effect.

Pathogenesis of Limb Complications and Walking Impairment in PAD Arterial obstruction by flow-limiting lesions underlies the lower extremity manifestations of PAD. In a demandischemia model, intermittent claudication reflects inadequate augmentation of skeletal muscle perfusion during exercise. Several lines of evidence, however, indicate that the drivers of the limb symptoms in PAD are more complex. Atherosclerotic disease occurs in the context of multiple disease processes that interfere with exercise ability. Potential mechanisms are detailed in the Figure and the current evidence supporting the role of reduced blood flow, vascular dysfunction, altered muscle metabolism, impaired angiogenesis, and inflammatory activation in producing limb discomfort and functional limitation is discussed in this section. Reduced Limb Perfusion Caused by Arterial Obstructive Lesions Reduced limb blood flow because of atherosclerotic disease characterizes PAD. Measurement of the ABI evaluates the severity of atherosclerosis impairing blood flow between the aorta and the ankle. Obstructive lesions create drops in both blood pressure and flow that are additive to reducing the ankle pressure. With exercise, the flow to the lower extremity increases and magnifies the pressure drop across fixed lesions, increasing the sensitivity to detect PAD. Recent studies highlight the clinical import of an abnormal exercise ABI with additive prognostic value for future limb revascularization and death compared with the resting ABI alone.37,38 Inadequate perfusion with exercise, which

Circulation Journal  Vol.81, March 2017

Limb Symptoms in PAD

283

Figure.   Mechanisms of limb symptoms in peripheral artery disease.

is caused by fixed lesions, is clearly a key component underlying limb symptoms in PAD. However, multiple studies indicate that the generation of limb ischemia in PAD has multiple determinants. The anatomic severity of obstruction is an imprecise predictor of clinical status and course. It is well-established that patients with PAD have impaired functional status compared with individuals with a normal ABI.7,39 Yet some studies have shown just modest correlation between ABI and walking ability,40,41 and other studies have found no association between ABI and the magnitude of functional limitation.42 Limb blood flow, assessed by other techniques, has shown inconsistent associations with functional measures. Magnetic resonance imaging-based measurement of calf blood flow was moderately related to walking distance,43 but plethysmography-based calf blood flow showed no association with treadmill walking or change in walking time.42 Prospective studies confirm that individuals with an abnormal ABI have a greater decline in walking ability at 2-year follow-up.44 However, the association between the magnitude of the reduction in ABI and functional outcomes is less clear. Taken together, the available evidence indicates that anatomic disease does not fully govern the clinical status of patients with PAD. Vascular Dysfunction in PAD Arterial insufficiency in PAD reflects both fixed and dynamic reductions in blood flow. A healthy vascular endothelium produces several vasodilator substances, including nitric oxide,45 which has pluripotent vascular benefits such as inhibiting platelets, reducing smooth muscle proliferation, preventing leukocyte adhesion, and promoting angiogenesis.46 Diminished nitric oxide bioactivity in the leg impedes the augmentation of blood flow with exercise.47 Vascular dysfunction may also exacerbate the vasoconstrictive effects of catecholamines and limit flow-mediated dilation.48 Together, the effects of abnormal endothelial function may

worsen clinical symptoms in PAD. Several studies have described the clinical relevance of endothelial dysfunction in PAD. Measures of endotheliumdependent vasodilator function, including brachial artery flow-mediated dilation and acetylcholine-induced vasodilation, are lower in patients with PAD.49–51 Both conduit and microvascular endothelial function were evaluated in 1,320 subjects, including 377 with PAD, and compared with patients with CAD; the patients with PAD had more severe impairment of multiple metrics of vasodilator function.52 The presence of endothelial dysfunction in patients with PAD is consistent with a systemic disruption of vascular function. Impairment of the hyperemic blood flow response is associated with functional impairment in PAD.53 There is evidence connecting greater brachial flowmediated dilation to greater physical activity in daily life in patients with PAD.54 Recent studies related impaired brachial flow-mediated dilation with reduced self-reported walking ability and 6-minute walk test.55,56 Interestingly, patients with PAD have decreased superficial femoral artery flowmediated dilation associated with lesion severity.57 Arterial stiffness measures, including higher pulse pressure and augmentation index, also are associated with reduced walking time in patients with PAD.58,59 Endothelial dysfunction measured by brachial flow-mediated dilation and reactive hyperemia predicts a higher risk of events after vascular surgery.60–62 Whether endothelial dysfunction predicts progressive functional impairment has not been evaluated. Impaired Angiogenesis and Reduced Microcirculatory Flow Chronic limb ischemia initiates several vascular structural adaptations. Insufficient blood supply produced by arterial ischemia induces a complex program of vascular growth.63 Multiple factors have been identified that regulate angiogenesis in animal models, including vascular endothelial growth factor (VEGF), fibroblast growth factor, hepatocyte

Circulation Journal  Vol.81, March 2017

284

HAMBURG NM et al.

growth factor, and hypoxia-inducible factor 1-α.64–66 In addition, specific bone marrow-derived cells may target regions of ischemia and promote vessel regeneration.67–69 Genetic regulators, including microRNA, are also important for angiogenesis.70 MicroRNA93 shows lower expression in animals genetically predisposed to severe clinical phenotypes such as hindlimb ischemia.71 In animal models, there is abundant evidence that therapies stimulating angiogenesis increase skeletal muscle perfusion and restore functional status. In patients with PAD, inadequate angiogenesis and collateral formation may potentiate limb ischemia and serve as a mechanism driving functional impairment. One study found that lower capillary density in PAD patients, as assessed by skeletal muscle biopsy, was associated with reduced functional measures including peak walking time.72 Similarly, imaging studies using MRI or contrast-enhanced ultrasound have demonstrated lower microvascular flow in the calf musculature in PAD patients.73,74 Exercise blood flow measured by contrast ultrasound was related to claudication time in a treadmill test.74 Also, reduced skeletal muscle blood flow as measured by MR during exercise was associated with impaired 6-minute walking time.43 Taken together, evidence from clinical investigations supports the concept that microcirculatory dysfunction affects limb function in patients with PAD, and that enhanced calf blood flow may be a therapeutic avenue. Thus, proangiogenic therapy approaches to treating PAD have been used in many studies of growth factor and cell-based therapies. As reviewed recently, these clinical trials have failed to convincingly demonstrate a reduction in limb symptoms, including pain and wound healing.63 There are a number of potential explanations for the disappointment with pro-angiogenic interventions in patients with PAD. Translational studies emphasize the relevance of systemic disease and risk factors to impaired angiogenesis in clinical PAD. Paradoxically, patients with PAD have higher levels of VEGF-A, a key promoter of angiogenesis.75,76 A recent study found evidence that an anti-angiogenic isoform of VEGF, VEGF-165b, is upregulated in both preclinical models and patients with PAD.77 The enhanced expression of anti-angiogenic VEGF is driven by the proinflammatory Wnt5a/JNK pathway, which is activated by obesity. Thus, metabolic dysfunction may mediate inadequate angiogenesis in PAD by generating anti-angiogenic factors and complicate the responses to therapies aimed to stimulate angiogenesis. Resolvin D2, an anti-inflammatory regulator of tissue reparative responses, is important in both hindlimb ischemia models and patients with PAD.78 Resolvin D2 treatment enhances arteriogenesis and reduces inflammation, including in diabetic animals. Thus, the next generation of pro-angiogenic therapies may require evaluation in animal models with metabolic dysfunction and target both inflammation and vascular growth. Skeletal Muscle Alterations and Mitochondrial Dysfunction Repeated episodes of ischemia have deleterious effects on the limb skeletal musculature.79 Altered skeletal muscle structural and metabolic properties magnify ischemiainduced functional impairment.80,81 Imaging studies with CT demonstrate that patients with PAD have a reduced calf muscle area that is not fully explained by inactivity.82 Further, the skeletal muscle displays lower density and higher fat content, which may limit muscle function.82 On muscle biopsy, there is greater muscle cell apoptosis and

reduced type I fiber content, which may interfere with performance.83,84 Ischemia also damages peripheral nerve function, with evidence of poor nerve conduction in patients with severe PAD.85,86 Mitochondrial dysfunction contributes to impaired skeletal muscle metabolism in PAD. Both the circulating and muscle levels of intermediates of oxidative phosphorylation, including acylcarnitines, are higher in PAD, suggesting reduced mitochondrial metabolism.87 In the muscle tissue, mitochondrial mass is higher; however, there is lower activity of several mitochondrial complexes impeding ATP generation and enhancing reactive oxygen species production.88–91 Altered mitochondrial function restricts oxygen utilization and may also promote endothelial dysfunction as mitochondrial-derived oxidants reduce nitric oxide bioactivity.92,93 Muscle fiber degeneration is associated with evidence of oxidative stress, including carbonyl groups and 4-hydroxy-2-nonenal adducts, protein modifications produced by reactive oxygen species.94 Mitochondrial function is also important in angiogenesis, consistent with the notion of coupling of vascular and muscular parameters. In hindlimb ischemia models, peroxisomeproliferator-activated receptor-γ coactivator-1α (PGC-1α), a key regulator of mitochondrial biogenesis, promotes vascular regeneration.95 Altered muscle metabolism also reflects reduced nutrient uptake related to systemic metabolic disturbances in PAD patients. Patients with PAD display insulin resistance and insulin resistance predicts a higher risk of developing clinical PAD.96,97 By evaluating skeletal muscle glucose uptake with PET, it has been shown that PAD patients with intermittent claudication have calf muscle insulin resistance.98 Additional studies are required to link muscle insulin resistance to functional parameters in PAD and to determine whether interventions to promote insulin sensitivity will reduce limb symptoms. Skeletal muscle dysfunction, including mitochondrial abnormalities, affects walking ability in PAD.80 Both decreased calf muscle content and altered fiber type relate to reduced functional parameters.82,84 Importantly, mitochondrial dysfunction assessed by MR spectroscopy to evaluate phosphocreatine recovery is associated with lower treadmill walking time.43 PAD patients with greater amounts of muscle acylcarnitine accumulation have greater degrees of exercise limitation.87 Evidence of myofiber damage is associated with both reduced walking distance and muscle strength in patients with claudication.99 Further, altered regulation of a cytoskeletal protein, desmin, is associated with reduced mitochondrial respiratory function and functional capacity in PAD.100 There is evidence of inadequate mitochondrial clearance through autophagy in the skeletal muscle in PAD that associates with walking parameters, consistent with increased mitochondrial damage.101 Greater levels of daily activity is associated with healthy calf muscle parameters. Several aspects of skeletal muscle phenotype, including increased calf muscle fat and decreased muscle density, predicted 2-year functional decline in a longitudinal study.102 Evidence of reduced mitochondrial biogenesis is associated with higher overall mortality, which is potentially mediated through reduced physical activity.103 Systemic and Local Inflammation Inflammatory activation participates in the development of atherosclerosis and may play a part in the generation of limb symptoms. Circulating biomarkers of systemic

Circulation Journal  Vol.81, March 2017

Limb Symptoms in PAD

285

inflammation, including C-reactive protein (CRP) and soluble intracellular adhesion molecule-1 (sICAM-1), predict a higher risk of developing clinical PAD.104 In patients with established PAD, higher levels of inflammatory biomarker are associated with both progression of lower extremity arterial obstruction and the risk of cardiovascular events.105–107 Skeletal muscle ischemia may drive local inflammation, exacerbating symptoms and altering muscle metabolism. In imaging studies, lower calf muscle area and higher calf muscle fat content were associated with systemic inflammation.108 Vascular inflammation also impairs dynamic responses by reducing nitric oxide bioactivity, which leads to decreased endothelium-mediated vasodilation. Inflammation has been associated with reduced walking capacity in PAD. Markers of vascular inflammation relate to lower functional measures in PAD patients.109,110 Higher peripheral monocyte expression of tumor necrosis factor α is associated with lower treadmill walking time in patients with intermittent claudication.111 In a similar fashion, higher peripheral mononuclear expression of the pro-inflammatory mediator, Wnt5a, is associated with lower ABI.77 In patients with PAD, a greater degree of daily physical activity is associated with lower levels of CRP, IL-6, fibrinogen, sICAM-1, and sVCAM-1.112 In prospective studies, functional decline is less in PAD patients with lower levels of CRP.113

Treatment of Limb Symptoms in PAD Reduction of limb morbidity is a key component of the comprehensive care for PAD patients. Several studies have emphasized the high rate of limb complications in patients with symptomatic PAD. In the REACH registry, 18.2% of patients with symptomatic PAD underwent peripheral revascularization and 3.8% had had an amputation at 4-year follow-up.114 Similarly, recent clinical trials that included patients with symptomatic PAD demonstrated that 1 in 5 patients with PAD undergo revascularization at 3 years.115,116 As discussed before, patients with obstructive PAD, regardless of limb symptom characteristics, have functional impairment. As shown in the Table, available medical treatment modalities to improve limb function and outcome include exercise therapy and antiplatelet therapy.20 Revascularization remains an option for persistent symptoms despite optimal medical therapy.21 Robust evidence supports the utility of exercise training programs to improve walking function in patients with PAD.11 In patients with claudication, exercise is the most effective noninterventional approach to reducing leg symptoms. Based on numerous clinical trials, supervised exercise programs are recommended as initial therapy for claudication.21 Controlled studies and systematic reviews indicate that supervised exercise training improves both walking time and walking distance by clinically relevant amounts.21 Increases in treadmill walking confer improved physical functioning, as exercise rehabilitation augments accelerometer-based daily activity levels and quality of life measures.117,118 Patients with PAD without claudication also derive benefit from supervised exercise. In a trial of 156 PAD patients of whom over 80% had atypical symptoms or were asymptomatic, treadmill exercise intervention increased the 6-minute walk distance.119 Though less beneficial than structured exercise treatment, home-based exercise programs have demonstrated

improvements in functional parameters.120,121 Randomized trials show that home-based walking exercise programs that include measures to enhance adherence increase the 6-minute walk distance as well as quality of life measures.120,122 As shown in the Table, the updated ACCF/AHA practice guidelines support the use of home-based walking and alternate approaches to exercise to enhance walking ability in patients with claudication.21 Ongoing studies are evaluating the use of mobile technology-based activity monitors to deliver exercise training at home and potentially expand access to exercise interventions.123 The benefits of exercise therapy in PAD have not been convincingly linked to gains in peripheral blood flow. In animal models of hindlimb ischemia, restoration of blood flow in the ischemic limb depends on vascular growth and collateral enlargement.124,125 In contrast, studies in patients with PAD do not consistently show changes in blood flow following exercise training. Measurements of hyperemic blood flow, a dynamic response that depends on arterial supply and vasodilation, increased in some but not all studies of exercise training.11,117 Further, the change in calf blood flow is not associated with the improvements in walking ability following exercise, indicating a divergence between arterial flow and functional measures.126 It is possible that more advanced assessment of microvascular flow in the calf will be necessary to evaluate the effects of exercise. However, evaluation of randomized trials of exercise training showed no effect on the resting ABI, suggesting that hemodynamic changes are not the major determinant of the exercise-induced reduction in limb symptoms.127 Exercise induces important vascular and skeletal muscle adaptations that may account for the improvements in functional status. Interventional studies show an improvement in flow-mediated dilation, a measure of conduit artery endothelium-dependent vasodilation, with exercise training in PAD patients.119 Exercise training also has a favorable effect on skeletal muscle, with restoration of carnitine metabolism that is associated with improved treadmill walking time.90,126 Further studies are needed to evaluate whether exercise training improves mitochondrial function or calf muscle phenotype in PAD. Chronic exercise training may also suppress inflammatory activation. Supervised exercise intervention reduced neutrophil activation in a small study of PAD patients with claudication.128 The available trial evidence indicates that exercise training and revascularization are complementary approaches to treating limb symptoms. In patients with claudication, exercise training and lower extremity bypass surgery had similar effects on walking distance. The randomized CLEVER study compared exercise training to endovascular therapy or optimal medical therapy for aorto-iliac disease in 111 patients with claudication.129 Overall, supervised exercise training induced the most robust increase in treadmill walking time at 6 months, without causing a concomitant change in ABI.127 Endovascular therapy also increased treadmill walking time, which was accompanied by a greater improvement in patient-reported quality of life metrics. The combination of supervised exercise training with endovascular revascularization for claudication was evaluated in the ERASE trial.130 Compared with supervised exercise alone, the combined therapy had a greater effect on treadmill walking distance at 1, 6 and 12 months. Thus, the choice of therapeutic approach is optimally applied in a patient-centered fashion with the option of multimodality treatments to maximize functional improvement.

Circulation Journal  Vol.81, March 2017

286

HAMBURG NM et al.

Summary and Conclusions Patients with PAD have both increased cardiovascular risk and decreased functional ability because of their limb symptoms. The recently updated ACC/AHA guidelines emphasize the importance of risk factor modification with antiplatelet therapy, statins, antihypertensive agents, and smoking cessation in patients with PAD. The mechanisms that lead to intermittent claudication in PAD are complex and include reduced limb perfusion, systemic inflammation, vascular dysfunction, impaired angiogenesis, reduced microcirculatory flow, and skeletal muscle dysfunction. An enhanced understanding of the pathophysiology of leg symptoms in PAD is critical for the development of novel therapeutic approaches. Exercise therapy improves walking ability in PAD through multiple beneficial effects on both vascular and skeletal muscle function. A patient-centered approach combining medical treatment, exercise intervention and selective use of revascularization is warranted to reduce limb symptoms and improve the quality of life in PAD. Funding Sources Dr. Hamburg is supported by National Heart, Lung, and Blood Institute grants HL081587 and HL115391.

Conflict of Interest Disclosures Dr. Hamburg and Dr. Creager have no relevant disclosures.

References    1. Creager MA. The crisis of vascular disease and the journey to vascular health: Presidential Address at the American Heart Association 2015 Scientific Sessions. Circulation 2016; 133: 2593 – 2598.   2. Sampson UK, Fowkes FG, McDermott MM, Criqui MH, Aboyans V, Norman PE, et al. Global and regional burden of death and disability from peripheral artery disease: 21 world regions, 1990 to 2010. Glob Heart 2014; 9: 145 – 158.   3. GBD 2015 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016; 388: 1545 – 1602.   4. Fowkes FG, Murray GD, Butcher I, Heald CL, Lee RJ, Chambless LE, et al. Ankle brachial index combined with Framingham Risk Score to predict cardiovascular events and mortality: A meta-analysis. JAMA 2008; 300: 197 – 208.    5. Steg PG, Bhatt DL, Wilson PW, D’Agostino R Sr, Ohman EM, Rother J, et al. One-year cardiovascular event rates in outpatients with atherothrombosis. JAMA 2007; 297: 1197 – 1206.   6. Bhatt DL, Eagle KA, Ohman EM, Hirsch AT, Goto S, Mahoney EM, et al. Comparative determinants of 4-year cardiovascular event rates in stable outpatients at risk of or with atherothrombosis. JAMA 2010; 304: 1350 – 1357.    7. McDermott MM, Greenland P, Liu K, Guralnik JM, Celic L, Criqui MH, et al. The ankle brachial index is associated with leg function and physical activity: The Walking and Leg Circulation Study. Ann Intern Med 2002; 136: 873 – 883.    8. McDermott MM, Liu K, Ferrucci L, Tian L, Guralnik JM, Liao Y, et al. Decline in functional performance predicts later increased mobility loss and mortality in peripheral arterial disease. J Am Coll Cardiol 2011; 57: 962 – 970.    9. McDermott MM. Lower extremity manifestations of peripheral artery disease: The pathophysiologic and functional implications of leg ischemia. Circ Res 2015; 116: 1540 – 1550.  10. Goodney PP, Beck AW, Nagle J, Welch HG, Zwolak RM. National trends in lower extremity bypass surgery, endovascular interventions, and major amputations. J Vasc Surg 2009; 50: 54 – 60.   11. Hamburg NM, Balady GJ. Exercise rehabilitation in peripheral artery disease: Functional impact and mechanisms of benefits. Circulation 2011; 123: 87 – 97.  12. GBD 2015 Mortality and Causes of Death Collaborators.

Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016; 388: 1459 – 1544.   13. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott MM, et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013; 382: 1329 – 1340.   14. Pande RL, Creager MA. Socioeconomic inequality and peripheral artery disease prevalence in US adults. Circ Cardiovasc Qual Outcomes 2014; 7: 532 – 539.   15. Fowkes FG, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott MM, et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013; 382: 1329 – 1340.   16. Bhatt DL, Steg PG, Ohman EM, Hirsch AT, Ikeda Y, Mas JL, et al. International prevalence, recognition, and treatment of cardiovascular risk factors in outpatients with atherothrombosis. JAMA 2006; 295: 180 – 189.   17. Diehm C, Allenberg JR, Pittrow D, Mahn M, Tepohl G, Haberl RL, et al. Mortality and vascular morbidity in older adults with asymptomatic versus symptomatic peripheral artery disease. Circulation 2009; 120: 2053 – 2061.  18. Uchiyama S, Goto S, Matsumoto M, Nagai R, Origasa H, Yamazaki T, et al. Cardiovascular event rates in patients with cerebrovascular disease and atherothrombosis at other vascular locations: Results from 1-year outcomes in the Japanese REACH Registry. J Neurol Sci 2009; 287: 45 – 51.   19. Pande RL, Perlstein TS, Beckman JA, Creager MA. Secondary prevention and mortality in peripheral artery disease: National Health and Nutrition Examination Study, 1999 to 2004. Circulation 2011; 124: 17 – 23.  20. Bonaca MP, Creager MA. Pharmacological treatment and current management of peripheral artery disease. Circ Res 2015; 116: 1579 – 1598.  21. Gerhard-Herman MD, Gornik HL, Barrett C, Barshes NR, Corriere MA, Drachman DE, et al. 2016 AHA/ACC Guideline on the Management of Patients With Lower Extremity Peripheral Artery Disease: A report of the American College of Cardiology/ American Heart Association Task Force on Clinical Practice Guidelines. Circulation, doi:10.1161/CIR.0000000000000471.   22. McDermott MM, Greenland P, Liu K, Guralnik JM, Criqui MH, Dolan NC, et al. Leg symptoms in peripheral arterial disease: Associated clinical characteristics and functional impairment. JAMA 2001; 286: 1599 – 1606.   23. Hirsch AT, Criqui MH, Treat-Jacobson D, Regensteiner JG, Creager MA, Olin JW, et al. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA 2001; 286: 1317 – 1324.  24. McDermott MM, Mehta S, Greenland P. Exertional leg symptoms other than intermittent claudication are common in peripheral arterial disease. Arch Intern Med 1999; 159: 387 – 392.   25. McDermott MM, Guralnik JM, Ferrucci L, Tian L, Liu K, Liao Y, et al. Asymptomatic peripheral arterial disease is associated with more adverse lower extremity characteristics than intermittent claudication. Circulation 2008; 117: 2484 –  2491.   26. McDermott MM, Ferrucci L, Simonsick EM, Balfour J, Fried L, Ling S, et al. The ankle brachial index and change in lower extremity functioning over time: The Women’s Health and Aging Study. J Am Geriatr Soc 2002; 50: 238 – 246.   27. McDermott MM, Guralnik JM, Tian L, Liu K, Ferrucci L, Liao Y, et al. Associations of borderline and low normal anklebrachial index values with functional decline at 5-year followup: The WALCS (Walking and Leg Circulation Study). J Am Coll Cardiol 2009; 53: 1056 – 1062.   28. Gardner AW, Montgomery PS, Scott KJ, Afaq A, Blevins SM. Patterns of ambulatory activity in subjects with and without intermittent claudication. J Vasc Surg 2007; 46: 1208 – 1214.  29. Regensteiner JG, Hiatt WR, Coll JR, Criqui MH, TreatJacobson D, McDermott MM, et al. The impact of peripheral arterial disease on health-related quality of life in the Peripheral Arterial Disease Awareness, Risk, and Treatment: New Resources for Survival (PARTNERS) Program. Vasc Med 2008; 13: 15 –  24.   30. Garg PK, Tian L, Criqui MH, Liu K, Ferrucci L, Guralnik JM, et al. Physical activity during daily life and mortality in patients with peripheral arterial disease. Circulation 2006; 114: 242 – 248.

Circulation Journal  Vol.81, March 2017

Limb Symptoms in PAD

287

  31. McDermott MM, Tian L, Liu K, Guralnik JM, Ferrucci L, Tan J, et al. Prognostic value of functional performance for mortality in patients with peripheral artery disease. J Am Coll Cardiol 2008; 51: 1482 – 1489.   32. Nead KT, Zhou M, Diaz Caceres R, Olin JW, Cooke JP, Leeper NJ. Walking impairment questionnaire improves mortality risk prediction models in a high-risk cohort independent of peripheral arterial disease status. Circ Cardiovasc Qual Outcomes 2013; 6: 255 – 261.  33. McDermott MM, Guralnik JM, Ferrucci L, Tian L, Kibbe MR, Greenland P, et al. Community walking speed, sedentary or lying down time, and mortality in peripheral artery disease. Vasc Med 2016; 21: 120 – 129.  34. Morris DR, Rodriguez AJ, Moxon JV, Cunningham MA, McDermott MM, Myers J, et al. Association of lower extremity performance with cardiovascular and all-cause mortality in patients with peripheral artery disease: A systematic review and meta-analysis. J Am Heart Assoc 2014; 3: e001105.   35. Jain A, Liu K, Ferrucci L, Criqui MH, Tian L, Guralnik JM, et al. Declining walking impairment questionnaire scores are associated with subsequent increased mortality in peripheral artery disease. J Am Coll Cardiol 2013; 61: 1820 – 1829.   36. Delaney JA, Jensky NE, Criqui MH, Whitt-Glover MC, Lima JA, Allison MA. The association between physical activity and both incident coronary artery calcification and ankle brachial index progression: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2013; 230: 278 – 283.   37. Sheikh MA, Bhatt DL, Li J, Lin S, Bartholomew JR. Usefulness of postexercise ankle-brachial index to predict all-cause mortality. Am J Cardiol 2011; 107: 778 – 782.   38. Hammad TA, Strefling JA, Zellers PR, Reed GW, Venkatachalam S, Lowry AM, et al. The effect of post-exercise ankle-brachial index on lower extremity revascularization. JACC Cardiovasc Interv 2015; 8: 1238 – 1244.   39. McDermott MM, Mehta S, Liu K, Guralnik JM, Martin GJ, Criqui MH, et al. Leg symptoms, the ankle-brachial index, and walking ability in patients with peripheral arterial disease. J Gen Intern Med 1999; 14: 173 – 181.  40. Izquierdo-Porrera AM, Gardner AW, Bradham DD, Montgomery PS, Sorkin JD, Powell CC, et al. Relationship between objective measures of peripheral arterial disease severity to self-reported quality of life in older adults with intermittent claudication. J Vasc Surg 2005; 41: 625 – 630.   41. Long J, Modrall JG, Parker BJ, Swann A, Welborn MB III, Anthony T. Correlation between ankle-brachial index, symptoms, and health-related quality of life in patients with peripheral vascular disease. J Vasc Surg 2004; 39: 723 – 727.   42. Szuba A, Oka RK, Harada R, Cooke JP. Limb hemodynamics are not predictive of functional capacity in patients with PAD. Vasc Med 2006; 11: 155 – 163.   43. Anderson JD, Epstein FH, Meyer CH, Hagspiel KD, Wang H, Berr SS, et al. Multifactorial determinants of functional capacity in peripheral arterial disease: Uncoupling of calf muscle perfusion and metabolism. J Am Coll Cardiol 2009; 54: 628 – 635.   44. McDermott MM, Liu K, Greenland P, Guralnik JM, Criqui MH, Chan C, et al. Functional decline in peripheral arterial disease: Associations with the ankle brachial index and leg symptoms. JAMA 2004; 292: 453 – 461.  45. Flammer AJ, Anderson T, Celermajer DS, Creager MA, Deanfield J, Ganz P, et al. The assessment of endothelial function: From research into clinical practice. Circulation 2012; 126: 753 – 767.   46. Vita JA, Hamburg NM. Does endothelial dysfunction contribute to the clinical status of patients with peripheral arterial disease? Can J Cardiol 2010; 26(Suppl A): 45A – 50A.   47. Meredith IT, Currie KE, Anderson TJ, Roddy MA, Ganz P, Creager MA. Postischemic vasodilation in human forearm is dependent on endothelium-derived nitric oxide. Am J Physiol 1996; 270: H1435 – H1440.  48. Gokce N, Vita JA, Bader DS, Sherman DL, Hunter LM, Holbrook M, et al. Effect of exercise on upper and lower extremity endothelial function in patients with coronary artery disease. Am J Cardiol 2002; 90: 124 – 127.   49. Liao JK, Bettmann MA, Sandor T, Tucker JI, Coleman SM, Creager MA. Differential impairment of vasodilator responsiveness of peripheral resistance and conduit vessels in humans with atherosclerosis. Circ Res 1991; 68: 1027 – 1034.   50. Fronek A, DiTomasso DG, Allison M. Noninvasive assessment of endothelial activity in patients with peripheral arterial disease and cardiovascular risk factors. Endothelium 2007; 14: 199 – 205.

  51. Poredos P, Golob M, Jensterle M. Interrelationship between peripheral arterial occlusive disease, carotid atherosclerosis and flow mediated dilation of the brachial artery. Int Angiol 2003; 22: 83 – 87.  52. Kiani S, Aasen JG, Holbrook M, Khemka A, Sharmeen F, LeLeiko RM, et al. Peripheral artery disease is associated with severe impairment of vascular function. Vasc Med 2013; 18: 72 – 78.  53. Robbins JL, Jones WS, Duscha BD, Allen JD, Kraus WE, Regensteiner JG, et al. Relationship between leg muscle capillary density and peak hyperemic blood flow with endurance capacity in peripheral artery disease. J Appl Physiol 2011; 111: 81 – 86.   54. Payvandi L, Dyer A, McPherson D, Ades P, Stein J, Liu K, et al. Physical activity during daily life and brachial artery flowmediated dilation in peripheral arterial disease. Vasc Med 2009; 14: 193 – 201.  55. Grenon SM, Chong K, Alley H, Nosova E, Gasper W, Hiramoto J, et al. Walking disability in patients with peripheral artery disease is associated with arterial endothelial function. J Vasc Surg 2014; 59: 1025 – 1034.  56. Silva R de C, Wolosker N, Yugar-Toledo JC, ConsolimColombo FM. Vascular reactivity is impaired and associated with walking ability in patients with intermittent claudication. Angiology 2015; 66: 680 – 686.   57. Heinen Y, Stegemann E, Sansone R, Benedens K, Wagstaff R, Balzer J, et al. Local association between endothelial dysfunction and intimal hyperplasia: Relevance in peripheral artery disease. J Am Heart Assoc 2015; 4: e001472.   58. Brewer LC, Chai HS, Bailey KR, Kullo IJ. Measures of arterial stiffness and wave reflection are associated with walking distance in patients with peripheral arterial disease. Atherosclerosis 2007; 191: 384 – 390.   59. Amoh-Tonto CA, Malik AR, Kondragunta V, Ali Z, Kullo IJ. Brachial-ankle pulse wave velocity is associated with walking distance in patients referred for peripheral arterial disease evaluation. Atherosclerosis 2009; 206: 173 – 178.   60. Gokce N, Keaney JF Jr, Menzoian JO, Watkins M, Hunter L, Duffy SJ, et al. Risk stratification for postoperative cardiovascular events via noninvasive assessment of endothelial function. Circulation 2002; 105: 1567 – 1572.   61. Gokce N, Keaney JF Jr, Hunter LM, Watkins MT, Nedeljkovic ZS, Menzoian JO, et al. Predictive value of non-invasivelydetermined endothelial dysfunction for long-term cardiovascular events in patients with peripheral vascular disease. J Am Coll Cardiol 2003; 41: 1769 – 1775.   62. Huang AL, Silver AE, Shvenke E, Schopfer DW, Jahangir E, Titas MA, et al. Predictive value of reactive hyperemia for cardiovascular events in patients with peripheral arterial disease undergoing vascular surgery. Arterioscler Thromb Vasc Biol 2007; 27: 2113 – 2119.   63. Cooke JP, Losordo DW. Modulating the vascular response to limb ischemia: Angiogenic and cell therapies. Circ Res 2015; 116: 1561 – 1578.   64. Semenza GL. Vasculogenesis, angiogenesis, and arteriogenesis: Mechanisms of blood vessel formation and remodeling. J Cell Biochem 2007; 102: 840 – 847.   65. Ito WD, Arras M, Scholz D, Winkler B, Htun P, Schaper W. Angiogenesis but not collateral growth is associated with ischemia after femoral artery occlusion. Am J Physiol 1997; 273: H1255 – H1265.   66. Patel TH, Kimura H, Weiss CR, Semenza GL, Hofmann LV. Constitutively active HIF-1alpha improves perfusion and arterial remodeling in an endovascular model of limb ischemia. Cardiovasc Res 2005; 68: 144 – 154.  67. Aicher A, Rentsch M, Sasaki K, Ellwart JW, Fandrich F, Siebert R, et al. Nonbone marrow-derived circulating progenitor cells contribute to postnatal neovascularization following tissue ischemia. Circ Res 2007; 100: 581 – 589.   68. Asahara T, Murohara T, Sullivan A, Silver M, van der ZR, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997; 275: 964 – 967.  69. Murasawa S, Asahara T. Endothelial progenitor cells for vasculogenesis. Physiology (Bethesda) 2005; 20: 36 – 42.   70. Hamburg NM, Leeper NJ. Therapeutic potential of modulating microRNA in peripheral artery disease. Curr Vasc Pharmacol 2015; 13: 316 – 323.   71. Hazarika S, Farber CR, Dokun AO, Pitsillides AN, Wang T, Lye RJ, et al. MicroRNA-93 controls perfusion recovery after hindlimb ischemia by modulating expression of multiple genes in the cell cycle pathway. Circulation 2013; 127: 1818 – 1828.

Circulation Journal  Vol.81, March 2017

288

HAMBURG NM et al.

 72. Robbins JL, Jones WS, Duscha BD, Allen JD, Kraus WE, Regensteiner JG, et al. Relationship between leg muscle capillary density and peak hyperemic blood flow with endurance capacity in peripheral artery disease. J Appl Physiol 2011; 111: 81 – 86.   73. Wu WC, Mohler E 3rd, Ratcliffe SJ, Wehrli FW, Detre JA, Floyd TF. Skeletal muscle microvascular flow in progressive peripheral artery disease: Assessment with continuous arterial spin-labeling perfusion magnetic resonance imaging. J Am Coll Cardiol 2009; 53: 2372 – 2377.  74. Lindner JR, Womack L, Barrett EJ, Weltman J, Price W, Harthun NL, et al. Limb stress-rest perfusion imaging with contrast ultrasound for the assessment of peripheral arterial disease severity. JACC Cardiovasc Imaging 2008; 1: 343 – 350.   75. Findley CM, Mitchell RG, Duscha BD, Annex BH, Kontos CD. Plasma levels of soluble Tie2 and vascular endothelial growth factor distinguish critical limb ischemia from intermittent claudication in patients with peripheral arterial disease. J Am Coll Cardiol 2008; 52: 387 – 393.   76. Jones WS, Duscha BD, Robbins JL, Duggan NN, Regensteiner JG, Kraus WE, et al. Alteration in angiogenic and anti-angiogenic forms of vascular endothelial growth factor-A in skeletal muscle of patients with intermittent claudication following exercise training. Vasc Med 2012; 17: 94 – 100.   77. Kikuchi R, Nakamura K, MacLauchlan S, Ngo DT, Shimizu I, Fuster JJ, et al. An antiangiogenic isoform of VEGF-A contributes to impaired vascularization in peripheral artery disease. Nat Med 2014; 20: 1464 – 1471.  78. Zhang MJ, Sansbury BE, Hellmann J, Baker JF, Guo L, Parmer CM, et al. Resolvin D2 enhances postischemic revascularization while resolving inflammation. Circulation 2016; 134: 666 – 680.   79. Paradis S, Charles AL, Meyer A, Lejay A, Scholey JW, Chakfe N, et al. Chronology of mitochondrial and cellular events during skeletal muscle ischemia-reperfusion. Am J Physiol Cell Physiol 2016; 310: C968 – C982.  80. Brass EP, Hiatt WR, Green S. Skeletal muscle metabolic changes in peripheral arterial disease contribute to exercise intolerance: A point-counterpoint discussion. Vasc Med 2004; 9: 293 – 301.   81. Regensteiner JG, Wolfel EE, Brass EP, Carry MR, Ringel SP, Hargarten ME, et al. Chronic changes in skeletal muscle histology and function in peripheral arterial disease. Circulation 1993; 87: 413 – 421.   82. McDermott MM, Hoff F, Ferrucci L, Pearce WH, Guralnik JM, Tian L, et al. Lower extremity ischemia, calf skeletal muscle characteristics, and functional impairment in peripheral arterial disease. J Am Geriatr Soc 2007; 55: 400 – 406.   83. Mitchell RG, Duscha BD, Robbins JL, Redfern SI, Chung J, Bensimhon DR, et al. Increased levels of apoptosis in gastrocnemius skeletal muscle in patients with peripheral arterial disease. Vasc Med 2007; 12: 285 – 290.   84. Askew CD, Green S, Walker PJ, Kerr GK, Green AA, Williams AD, et al. Skeletal muscle phenotype is associated with exercise tolerance in patients with peripheral arterial disease. J Vasc Surg 2005; 41: 802 – 807.   85. Garg PK, Liu K, Ferrucci L, Guralnik JM, Criqui MH, Tian L, et al. Lower extremity nerve function, calf skeletal muscle characteristics, and functional performance in peripheral arterial disease. J Am Geriatr Soc 2011; 59: 1855 – 1863.   86. McDermott MM, Sufit R, Nishida T, Guralnik JM, Ferrucci L, Tian L, et al. Lower extremity nerve function in patients with lower extremity ischemia. Arch Intern Med 2006; 166: 1986 –  1992.  87. Hiatt WR, Wolfel EE, Regensteiner JG, Brass EP. Skeletal muscle carnitine metabolism in patients with unilateral peripheral arterial disease. J Appl Physiol 1992; 73: 346 – 353.  88. Pipinos II, Sharov VG, Shepard AD, Anagnostopoulos PV, Katsamouris A, Todor A, et al. Abnormal mitochondrial respiration in skeletal muscle in patients with peripheral arterial disease. J Vasc Surg 2003; 38: 827 – 832.   89. Pipinos II, Judge AR, Zhu Z, Selsby JT, Swanson SA, Johanning JM, et al. Mitochondrial defects and oxidative damage in patients with peripheral arterial disease. Free Radic Biol Med 2006; 41: 262 – 269.   90. Hiatt WR, Regensteiner JG, Wolfel EE, Carry MR, Brass EP. Effect of exercise training on skeletal muscle histology and metabolism in peripheral arterial disease. J Appl Physiol 1996; 81: 780 – 788.   91. Greiner A, Esterhammer R, Messner H, Biebl M, Muhlthaler H, Fraedrich G, et al. High-energy phosphate metabolism during

incremental calf exercise in patients with unilaterally symptomatic peripheral arterial disease measured by phosphor 31 magnetic resonance spectroscopy. J Vasc Surg 2006; 43: 978 – 986.   92. Bauer TA, Brass EP, Hiatt WR. Impaired muscle oxygen use at onset of exercise in peripheral arterial disease. J Vasc Surg 2004; 40: 488 – 493.   93. Bauer TA, Brass EP, Barstow TJ, Hiatt WR. Skeletal muscle StO2 kinetics are slowed during low work rate calf exercise in peripheral arterial disease. Eur J Appl Physiol 2007; 100: 143 –  151.   94. Weiss DJ, Casale GP, Koutakis P, Nella AA, Swanson SA, Zhu Z, et al. Oxidative damage and myofiber degeneration in the gastrocnemius of patients with peripheral arterial disease. J Transl Med 2013; 11: 230.   95. Arany Z, Foo SY, Ma Y, Ruas JL, Bommi-Reddy A, Girnun G, et al. HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha. Nature 2008; 451: 1008 – 1012.   96. Pande RL, Perlstein TS, Beckman JA, Creager MA. Association of insulin resistance and inflammation with peripheral arterial disease: The National Health and Nutrition Examination Survey, 1999 to 2004. Circulation 2008; 118: 33 – 41.   97. Britton KA, Mukamal KJ, Ix JH, Siscovick DS, Newman AB, de Boer IH, et al. Insulin resistance and incident peripheral artery disease in the Cardiovascular Health Study. Vasc Med 2012; 17: 85 – 93.  98. Pande RL, Park MA, Perlstein TS, Desai AS, Doyle J, Navarrete N, et al. Impaired skeletal muscle glucose uptake by [18F]fluorodeoxyglucose-positron emission tomography in patients with peripheral artery disease and intermittent claudication. Arterioscler Thromb Vasc Biol 2011; 31: 190 – 196.  99. Koutakis P, Myers SA, Cluff K, Ha DM, Haynatzki G, McComb RD, et al. Abnormal myofiber morphology and limb dysfunction in claudication. J Surg Res 2015; 196: 172 – 179. 100. Koutakis P, Miserlis D, Myers SA, Kim JK, Zhu Z, Papoutsi E, et al. Abnormal accumulation of desmin in gastrocnemius myofibers of patients with peripheral artery disease: Associations with altered myofiber morphology and density, mitochondrial dysfunction and impaired limb function. J Histochem Cytochem 2015; 63: 256 – 269. 101. White SH, McDermott MM, Sufit RL, Kosmac K, Bugg AW, Gonzalez-Freire M, et al. Walking performance is positively correlated to calf muscle fiber size in peripheral artery disease subjects, but fibers show aberrant mitophagy: An observational study. J Transl Med 2016; 14: 284. 102. McDermott MM, Ferrucci L, Guralnik J, Tian L, Liu K, Hoff F, et al. Pathophysiological changes in calf muscle predict mobility loss at 2-year follow-up in men and women with peripheral arterial disease. Circulation 2009; 120: 1048 – 1055. 103. Thompson JR, Swanson SA, Haynatzki G, Koutakis P, Johanning JM, Reppert PR, et al. Protein concentration and mitochondrial content in the gastrocnemius predicts mortality rates in patients with peripheral arterial disease. Ann Surg 2015; 261: 605 – 610. 104. Pradhan AD, Shrivastava S, Cook NR, Rifai N, Creager MA, Ridker PM. Symptomatic peripheral arterial disease in women: Nontraditional biomarkers of elevated risk. Circulation 2008; 117: 823 – 831. 105. Tzoulaki I, Murray GD, Lee AJ, Rumley A, Lowe GD, Fowkes FG. C-reactive protein, interleukin-6, and soluble adhesion molecules as predictors of progressive peripheral atherosclerosis in the general population: Edinburgh Artery Study. Circulation 2005; 112: 976 – 983. 106. Vidula H, Tian L, Liu K, Criqui MH, Ferrucci L, Pearce WH, et al. Biomarkers of inflammation and thrombosis as predictors of near-term mortality in patients with peripheral arterial disease: A cohort study. Ann Intern Med 2008; 148: 85 – 93. 107. Beckman JA, Preis O, Ridker PM, Gerhard-Herman M. Comparison of usefulness of inflammatory markers in patients with versus without peripheral arterial disease in predicting adverse cardiovascular outcomes (myocardial infarction, stroke, and death). Am J Cardiol 2005; 96: 1374 – 1378. 108. McDermott MM, Ferrucci L, Guralnik JM, Tian L, Green D, Liu K, et al. Elevated levels of inflammation, d-dimer, and homocysteine are associated with adverse calf muscle characteristics and reduced calf strength in peripheral arterial disease. J Am Coll Cardiol 2007; 50: 897 – 905. 109. McDermott MM, Liu K, Ferrucci L, Tian L, Guralnik JM, Green D, et al. Circulating blood markers and functional impairment in peripheral arterial disease. J Am Geriatr Soc

Circulation Journal  Vol.81, March 2017

Limb Symptoms in PAD

289

2008; 56: 1504 – 1510. 110. Nylaende M, Kroese A, Stranden E, Morken B, Sandbaek G, Lindahl AK, et al. Markers of vascular inflammation are associated with the extent of atherosclerosis assessed as angiographic score and treadmill walking distances in patients with peripheral arterial occlusive disease. Vasc Med 2006; 11: 21 – 28. 111. Pande RL, Brown J, Buck S, Redline W, Doyle J, Plutzky J, et al. Association of monocyte tumor necrosis factor alpha expression and serum inflammatory biomarkers with walking impairment in peripheral artery disease. J Vasc Surg 2015; 61: 155 –  161. 112. Craft LL, Guralnik JM, Ferrucci L, Liu K, Tian L, Criqui MH, et al. Physical activity during daily life and circulating biomarker levels in patients with peripheral arterial disease. Am J Cardiol 2008; 102: 1263 – 1268. 113. McDermott MM, Ferrucci L, Liu K, Criqui MH, Greenland P, Green D, et al. D-dimer and inflammatory markers as predictors of functional decline in men and women with and without peripheral arterial disease. J Am Geriatr Soc 2005; 53: 1688 –  1696. 114. Kumbhani DJ, Steg PG, Cannon CP, Eagle KA, Smith SC Jr, Goto S, et al. Statin therapy and long-term adverse limb outcomes in patients with peripheral artery disease: Insights from the REACH registry. Eur Heart J 2014; 35: 2864 – 2872. 115. Bonaca MP, Scirica BM, Creager MA, Olin J, Bounameaux H, Dellborg M, et al. Vorapaxar in patients with peripheral artery disease: Results from TRA2ºP-TIMI 50. Circulation 2013; 127: 1522 – 1529. 116. Bonaca MP, Bhatt DL, Storey RF, Steg PG, Cohen M, Kuder J, et al. Ticagrelor for prevention of ischemic events after myocardial infarction in patients with peripheral artery disease. J Am Coll Cardiol 2016; 67: 2719 – 2728. 117. Gardner AW, Katzel LI, Sorkin JD, Bradham DD, Hochberg MC, Flinn WR, et al. Exercise rehabilitation improves functional outcomes and peripheral circulation in patients with intermittent claudication: A randomized controlled trial. J Am Geriatr Soc 2001; 49: 755 – 762. 118. Tsai JC, Chan P, Wang CH, Jeng C, Hsieh MH, Kao PF, et al. The effects of exercise training on walking function and perception of health status in elderly patients with peripheral arterial occlusive disease. J Intern Med 2002; 252: 448 – 455. 119. McDermott MM, Ades P, Guralnik JM, Dyer A, Ferrucci L, Liu K, et al. Treadmill exercise and resistance training in patients with peripheral arterial disease with and without intermittent claudication: A randomized controlled trial. JAMA

2009; 301: 165 – 174. 120. McDermott MM, Liu K, Guralnik JM, Criqui MH, Spring B, Tian L, et al. Home-based walking exercise intervention in peripheral artery disease: A randomized clinical trial. JAMA 2013; 310: 57 – 65. 121. Fokkenrood HJ, Bendermacher BL, Lauret GJ, Willigendael EM, Prins MH, Teijink JA. Supervised exercise therapy versus non-supervised exercise therapy for intermittent claudication. Cochrane Database Syst Rev 2013; (8): CD005263. 122. McDermott MM, Guralnik JM, Criqui MH, Ferrucci L, Zhao L, Liu K, et al. Home-based walking exercise in peripheral artery disease: 12-month follow-up of the GOALS randomized trial. J Am Heart Assoc 2014; 3: e000711. 123. McDermott MM, Polonsky TS. Home-based exercise: A therapeutic option for peripheral artery disease. Circulation 2016; 134: 1127 – 1129. 124. Prior BM, Lloyd PG, Ren J, Li H, Yang HT, Laughlin MH, et al. Time course of changes in collateral blood flow and isolated vessel size and gene expression after femoral artery occlusion in rats. Am J Physiol Heart Circ Physiol 2004; 287: H2434 – H2447. 125. Yang HT, Ren J, Laughlin MH, Terjung RL. Prior exercise training produces NO-dependent increases in collateral blood flow after acute arterial occlusion. Am J Physiol Heart Circ Physiol 2002; 282: H301 – H310. 126. Hiatt WR, Regensteiner JG, Hargarten ME, Wolfel EE, Brass EP. Benefit of exercise conditioning for patients with peripheral arterial disease. Circulation 1990; 81: 602 – 609. 127. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER, Cohen DJ, Reynolds MR, et al. Supervised exercise versus primary stenting for claudication resulting from aortoiliac peripheral artery disease: Six-month outcomes from the claudication: Exercise versus endoluminal revascularization (CLEVER) study. Circulation 2012; 125: 130 – 139. 128. Turton EP, Coughlin PA, Kester RC, Scott DJ. Exercise training reduces the acute inflammatory response associated with claudication. Eur J Vasc Endovasc Surg 2002; 23: 309 – 316. 129. Murphy TP, Hirsch AT, Ricotta JJ, Cutlip DE, Mohler E, Regensteiner JG, et al. The Claudication: Exercise Vs. Endoluminal Revascularization (CLEVER) study: Rationale and methods. J Vasc Surg 2008; 47: 1356 – 1363. 130. Fakhry F, Spronk S, van der Laan L, Wever JJ, Teijink JA, Hoffmann WH, et al. Endovascular revascularization and supervised exercise for peripheral artery disease and intermittent claudication: A randomized clinical trial. JAMA 2015; 314: 1936 – 1944.

Circulation Journal  Vol.81, March 2017

Suggest Documents