circulation 2012 berger 491 500

Upload: andreia-afloroaei-gherasam

Post on 07-Aug-2018

212 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/20/2019 Circulation 2012 Berger 491 500

    1/11

    Peripheral Arterial Disease

    Medical Therapy in Peripheral Artery Disease

    Jeffrey S. Berger, MD, MS, FAHA; William R. Hiatt, MD, FAHA

    The epidemiology of peripheral artery disease (PAD) iswell described and related to age and, in particular, therisk factors of diabetes mellitus and smoking. Recent data

    from the National Health and Nutrition Examination Survey

    found a 5.9% prevalence in subjects   40 years of age

    resulting in an estimated prevalence of 7.2 million affected

    individuals in the United States.1 These subjects have a

    significant increased risk of all-cause mortality because of the

    underlying atherosclerotic disease process and general under-

    treatment of PAD risk factors.2

    There is a wide spectrum of clinical manifestations for

    PAD: (1) the completely asymptomatic patient found to have

    PAD from a screening ankle-brachial index (ABI), (2) atyp-ical leg symptoms associated with an exercise limitation, (3)

    classic intermittent claudication, and (4) ischemic pain and

    ulceration in the lower extremity from chronic limb ischemia.

    However, despite the level and degree of limb symptoms,

    even asymptomatic persons with PAD have a greatly reduced

    functional capacity. This suggests that occlusive disease in

    the lower extremity is associated with reduced exercise

    capacity and functional status regardless of the symptomatic

    state.

    As noted above, symptomatic and asymptomatic PAD is

    associated with an increased risk for morbidity and mortality,

    and for impairment of quality of life, as well. A prospectivecohort in subjects   65 years of age found a similar high

    ischemic risk in symptomatic and asymptomatic adults with

    PAD.3 Pooled data from 11 studies in 6 countries found that

    PAD, defined by a an ABI of  0.90 was associated with an

    increased risk of subsequent all-cause mortality (relative risk 

    1.60), cardiovascular mortality (relative risk 1.96), coronary

    heart disease (relative risk 1.45), and stroke (relative risk 

    1.35) after adjustment for age, sex, conventional cardiovas-

    cular risk factors, and prevalent cardiovascular disease.4

    The treatment of PAD has evolved over the past decade to

    include a broad approach, focusing on the reduction of 

    adverse cardiovascular events, improving symptoms in clau-dication, and preventing tissue loss in critical limb ischemia.5

    Because quality of life is severely limited in these subjects,

    great emphasis is placed on ameliorating symptoms and

    reducing the risk of PAD progression.

    Established Therapies for Management of theSystemic Atherothrombosis in PAD

    Several sets of guidelines provide physicians taking care of PAD

    subjects with a list of recommendations regarding optimal

    treatment recommendations.6– 8 These include the American

    College of Cardiology Foundation/American Heart Association

    guidelines and the international Inter-Society Consensus

    (TASC) II guidelines. Both are undergoing substantial revision

    at this time. However, only 15% of the American College of 

    Cardiology Foundation/American Heart Association PAD

    guidelines come from evidence that is level A.9 Thus, there is a

    paucity of high-level data to guide decision making.

    Many of the medical therapeutics used in PAD target athero-thrombotic pathways. For example, statins, antiplatelets, and

    angiotensin-converting enzyme inhibitors are used in PAD; yet,

    much of the data supporting their use stem from data derived

    from patients with coronary or cerebrovascular disease. For

    example, the Heart Protection Study (HPS), which evaluated

    simvastatin versus placebo, and the Clopidogrel versus As-

    pirin in Patients at Risk of Ischemic Events (CAPRIE) trial

    enrolled patients with atherosclerosis in several vascular

    territories, including a large subgroup of patients with PAD.

    Both studies found that in general PAD responds similarly to

    non-PAD (except maybe somewhat better to clopidogrel than

    to aspirin).10,11

    Antiplatelet TherapyDespite an abundance of data demonstrating the efficacy of 

    antiplatelet therapy in coronary artery disease and cerebro-

    vascular disease, the data in PAD are less compelling. In the

    Antithrombotic Trialists’ Collaborative meta-analysis, there

    was a significant 23% lowering of cardiovascular events from

    a combined analysis of all antiplatelet agents, but the bene-

    ficial effects were driven by picotamide (an inhibitor of 

    thromboxane A2 synthase and thromboxane A2 receptors).12

    Although antiplatelet therapy is effective at decreasing car-

    diovascular events in patients with PAD, these drugs have no

    effect on symptomatic improvement in subjects with inter-mittent claudication.13

    Aspirin, the most widely used antiplatelet agent in the world,

    does not have compelling evidence supporting a reduction in

    cardiovascular events in patients with PAD (despite a positive

    From the Divisions of Cardiology and Vascular Surgery, New York University School of Medicine, New York, NY (J.S.B.); University of Colorado

    School of Medicine, Division of Cardiology, and CPC Clinical Research, Aurora, CO (W.R.H.).Correspondence to William R. Hiatt, MD, Professor of Medicine/Cardiology, University of Colorado School of Medicine, c/o CPC Clinical Research,

    13199 E Montview Blvd, Suite 200, Aurora, CO 80045. E-mail [email protected]

    (Circulation. 2012;126:491-500.)

    © 2012 American Heart Association, Inc.Circulation  is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.111.033886

     491  by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    2/11

    trend).14 In a recent meta-analysis of 18 prospective randomized

    trials comprising 5269 subjects with PAD, aspirin resulted in a

    12% reduction of the combined end point of nonfatal myocardial

    infarction, nonfatal stroke, and cardiovascular death that failed to

    reach statistical significance.14 Whether a larger sample would

    provide more convincing benefit is unknown, but recent studies

    of aspirin in subjects with asymptomatic PAD or PAD with

    diabetes mellitus were entirely negative.15,16 However, the scar-

    city of randomized data investigating aspirin in symptomatic

    PAD compared with coronary artery disease or cerebrovascular

    disease remains a major limitation in clinical decision making.

    There is some evidence that clopidogrel monotherapy may

    be particularly effective in PAD. In a subgroup analysis of 

    CAPRIE, clopidogrel (versus aspirin) had its greatest effect

    on reducing cardiovascular events in subjects with PAD (P for

    heterogeneity0.045).11 Subsequently, the Clopidogrel for HighAtherothrombotic Risk and Ischemic Stabilization, Management

    and Avoidance (CHARISMA) trial evaluated the effect of 

    aspirin plus clopidogrel versus aspirin alone in patients with

    established and at risk for cardiovascular disease. In a subgroup

    analysis of PAD patients, no benefit was observed for dual-

    antiplatelet therapy with aspirin plus clopidogrel.17

    Current guidelines recommend antiplatelet therapy in patients

    with PAD. Because the majority of patients with PAD have

    concomitant symptomatic coronary artery disease or cerebrovas-

    cular disease, aspirin is an acceptable antiplatelet agent in this

    setting. In addition to aspirin, clopidogrel monotherapy is a

    reasonable alternative strategy. Implicit in these recommenda-

    tions is that a large prospective randomized trial investigating the

    optimal antiplatelet strategy in patients with PAD is long

    overdue.

    Established Therapies forSymptomatic Improvement

    Over the past decade there has been a tremendous growth of 

    PAD limb-specific clinical studies suggesting that treatment

    with metabolic agents, antimicrobials, cell therapy, and gene

    therapy would result in improved lower-extremity function

    and viability in subjects with PAD. Trials in humans have led

    to inconsistent results. This review will provide the current

    state of medical therapy in subjects with PAD (Table 1). Thiswill include the evidence for and against these new modalities

    with a focus on medical treatments including drugs and

    biological agents. Revascularization trials are not within the

    scope of this review.

    Exercise TrainingExercise rehabilitation is a class I, level of evidence A, recom-

    mendation for the treatment of claudication in patients with

    PAD.7,8 A landmark meta-analysis in 1995 demonstrated that

    supervised exercise improves claudication symptoms and in-

    creases pain-free walking distance on a treadmill by 100%.18

    In 2008, a rigorous systematic review by the Cochrane group19

    involving 1200 participants from 22 randomized trials with

    stable claudication demonstrated a significant benefit in improv-

    ing treadmill walking time and walking distance with a super-

    vised exercise program. Although no benefit was seen in

    reducing major adverse cardiovascular events or on improving

    the ABI, subjects randomly assigned to exercise had improve-

    ment in claudication symptoms out to 2 years.19

    Although supervised exercise is very effective in PAD sub-

     jects with claudication, several questions remain: (1) is exerciseeffective in PAD subjects without claudication and (2) what is

    the role of a nonsupervised exercise program? Two recent

    randomized trials address these issues.21,22 The effect of 

    supervised treadmill exercise or lower-extremity resistance

    training on functional performance and quality of life was

    evaluated in a randomized trial of 156 PAD patients, of whom

    20% had symptoms of classic claudication.20 Although any

    exercise (treadmill or resistance training) improved func-

    tional performance, the treadmill exercise group had greater

    increases in the 6-minute walk distance and in the maximum

    treadmill walking time in comparison with the resistance

    group. Of note, the changes in the primary outcomes were

    similar between subjects with and without claudication andbetween asymptomatic and symptomatic participants. Despite

    the efficacy of a supervised exercise program in PAD, lack of 

    reimbursement in the United States remains a major barrier to

    utilization of this treatment.

    Evaluating the role of home-based exercise, Gardner and

    colleagues randomly assigned 119 subjects with claudication

    to either home-based exercise, supervised exercise, or a usual

    care control group.21 Both exercise programs increased clau-

    dication onset time and peak treadmill walking time versus

    the control group; however, the changes in average walking

    cadence time were greater in the home-based exercise group.

    Although this study was small, and nearly 25% of subjects

    did not complete follow-up, it suggests that a quantified

    home-based approach may be useful in this high-risk popu-

    lation with impaired quality of life. However, a meta-analysis

    of home-based exercise training in PAD was negative.22

    Future exercise studies with improved home-training meth-

    ods, larger populations, and clinically meaningful end points

    are certainly warranted. In the meantime, subjects with PAD

    (symptomatic or not) are likely to benefit from an aggressive

    exercise regimen.

    CilostazolCilostazol is a phosphodiesterase type 3 inhibitor approved in

    the United States in 1999 to treat intermittent claudication. Itspossible mechanism of action includes increasing intracellu-

    Table 1. Effect of Medical Therapies in Subjects With

    Peripheral Artery Disease

    Symptomatic

    Improvement

    Reduced

    Cardiovascular Risk 

    Exercise     Not studied

    Cilostazol     Neutral

    Statins    / 

     Antiplatelets  

    L-Carnitine and

    propionyl-L-carnitine

      Not studied

    Pentoxifylline     Not studied

    Naftidrofuryl     Not studied

    Gene therapy    /    Neutral

    Cell therapy    /    Not studied

     492 Circulation   July 24, 2012

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    3/11

    lar concentrations of cAMP, thereby causing vasodilation and

    inhibiting platelet aggregation.23 Although other drugs with

    vasodilating and platelet-inhibiting properties have not been

    demonstrated to improve claudication symptoms, cilostazol is

    effective in ameliorating symptoms. A meta-analysis of 8

    randomized trials including 2702 PAD subjects with claudi-

    cation found that cilostazol improved maximum and pain-free

    treadmill walking distance and quality-of-life measures.24 Of 

    note, cilostazol decreased triglyceride concentration by 16%

    and increased high-density lipoprotein levels by 13%, but

    also increased the incidence of headache, bowel concerns,

    and palpitations.

    Cilostazol may have an additional benefit of reducing reste-

    nosis and repeat revascularization following endovascular ther-

    apy.25 Similar to patients with coronary artery disease,26–28

    patients with PAD have lower rates of target vessel revascular-

    ization following cilostazol therapy.29,30 However, these studies

    have been small and open label in design, thus hypothesis

    generating.

    Milrinone, another phosphodiesterase type 3 inhibitor, wasnoted to increase mortality in subjects with heart failure.31 In

    response, the US Food and Drug Administration mandated a

    long-term safety study of cilostazol. Hiatt and colleagues per-

    formed a phase 4 (postmarketing) randomized, double-blind,

    placebo-controlled trial of cilostazol in 1435 PAD subjects with

    claudication.32 Although underpowered because of a lower

    mortality than projected and poor study drug adherence, this is

    the largest study to date that has evaluated serious adverse events

    and mortality. No difference was observed for overall mortality

    or serious bleeding events between the cilostazol and placebo

    groups; however, the study could not exclude a modest increased

    risk. In comparison with milrinone, cilostazol has fewer cardiac

    inotropic effects but equivalent vasodilating and platelet-inhibiting properties.33 Nonetheless, an advisory from the US

    Food and Drug Administration stated that cilostazol should not

    be used in subjects with congestive heart failure.

    Statin TherapySubgroup analyses of PAD patients from large randomized

    trial data in subjects with PAD found a reduction in cardiovas-

    cular events from statin therapy. Exploratory analyses suggested

    an improvement of claudication symptoms in subjects with

    PAD.34 In a randomized trial of 354 PAD subjects with claudi-

    cation, Mohler and colleagues35 demonstrated that atorvastatin

    (10 or 80 mg daily) improved pain-free walking distance and

    community-based physical activity. However, the functional

    benefits of statins on claudication have not been proven.

    Carnitine and Propionyl-L-Carnitine TherapyL-Carnitine and propionyl-L-carnitine may improve muscle

    metabolism and exercise performance in patients with PAD.

    Initial studies evaluated   L-carnitine given orally as 2 g twice

    daily, and also given as a single 3-g dose intravenously.36

    These early studies in PAD were indicative of clinical benefit

    and therefore led to the development of propionyl-L-carnitine,

    an acyl form of   L-carnitine.

    In 2 published phase 3 trials, propionyl-L-carnitine had a

    significant improvement on claudication-limited treadmillexercise performance. In Europe, a total of 501 subjects were

    randomly assigned to propionyl-L-carnitine 2 g/d for 12 months

    (n248) or placebo (n253).37 The primary analysis was the

    subgroup of 171 patients who had a maximum walking distance

    between 50 and 250 m and baseline variability 25%. After

    12 months of treatment, patients randomly assigned to pla-

    cebo increase maximal walking distance 44% versus 61% on

    drug. This difference had a probability value of 0.055 by the

    primary analysis and 0.048 with a secondary analysis. The

    quality-of-life questionnaire was also positive in favor of 

    drug (P0.002). In the second pivotal trial, a total of 161

    subjects were randomly assigned to propionyl-L-carnitine, 2

    g/d (n85), or placebo (n76).38 After 6 months of therapy,

    treated patients increased peak walking time 78% with a 37%

    increase in controls (P0.002). Patients randomly assigned

    to propionyl-L-carnitine also had significant improvements in

    the physical function scores of the SF-36 (P0.31), but not

    with the Waling Impairment Questionnaire (P0.26). How-

    ever, more recent studies in which propionyl-L-carnitine was

    given on a background of exercise training, although showing

    favorable trends, were not statistically significant on theprimary end point.39 Thus, the overall incremental benefit of 

    propionyl-L-carnitine in PAD remains to be determined.

    Other Medical Therapies

    PentoxifyllinePentoxifylline is a xanthine derivative used to treat patients

    with intermittent claudication. Its mechanism of action is

    thought to be a rheological modifier that includes improving

    the deformability of red blood cells and white blood cells, and

    decreasing fibrinogen concentration, platelet adhesiveness,

    and whole-blood viscosity. A meta-analysis demonstrated a

    modestly improved walking distance, substantially less effec-

    tive than either cilostazol or a supervised exercise program.40

    NaftidrofurylNaftidrofuryl is a serotonin 5-hydroxytryptamine 2 receptor

    antagonist with vasoactive properties in addition to its effect

    on oxidative metabolism and rheological properties on the red

    blood cell and platelet.41 Although not approved in the United

    States, it is currently used in Europe for the treatment of 

    claudication. In a patient-level meta-analysis, naftidrofuryl

    improved symptomatic claudication without any serious ad-

    verse events.42

    Medical Treatment of the PatientUndergoing Revascularization

    Antiplatelet therapy has good evidence for the prevention of 

    graft occlusion after peripheral vascular surgical procedures.

    In the Antithrombotic Trialists’ Collaboration meta-analysis,

    3000 patients with peripheral artery procedures had a 16%

    graft occlusion rate on antiplatelet therapy in comparison

    with 25% in the control group (P0.00001).43 Aspirin with

    or without other antiplatelet therapy was associated with a

    significantly lower risk of graft occlusion.44 Ticlopidine has

    also been shown to promote vein graft patency without any

    difference in cardiovascular events.45 Dual-antiplatelet ther-

    apy with aspirin plus clopidogrel versus aspirin alone was

    tested in the clopidogrel and acetylsalicylic acid in bypasssurgery for peripheral arterial disease (CASPAR) trial.46

    Berger and Hiatt Medical Therapy in Peripheral Artery Disease   493

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    4/11

    There was no significant difference in the primary end point

    (graft occlusion, amputation, or death) between groups. In a

    subgroup analysis, clopidogrel plus aspirin was better than

    aspirin alone among patients who received prosthetic grafts.

    As expected, bleeding was more common in the dual-antiplatelet

    therapy group.47

    Anticoagulation has also been recommended as an adju-

    vant to maintain surgical graft patency.47 The use of heparin

    anticoagulation in the immediate postoperative period, par-

    ticularly low-molecular-weight heparin, may improve the

    patency of infrainguinal bypass grafts (vein and prosthetic),

    but has also been associated with increased bleeding.48,49 The

    role of long-term oral anticoagulation is more controversial.

    In one trial of patients undergoing infrainguinal bypass

    surgery (including the use of vein, prosthetic material, and

    endarterectomy), long-term warfarin was associated with an

    increased bleeding risk but no benefit in patency or survival.50

    In contrast, another study performed in a similar population

    demonstrated that oral anticoagulation improved graft pa-

    tency, limb salvage, and survival.51

    Several trials have compared the benefits of anticoagula-

    tion versus antiplatelet therapy. Among patients treated with

    infrainguinal bypass procedures (predominantly using pros-

    thetic material), randomization to low-molecular-weight hep-

    arin versus aspirin and dipyridamole was associated with

    better graft patency, especially in the patients treated for

    critical limb ischemia.52 The Dutch Bypass Oral Anticoagu-

    lants or Aspirin study evaluated 2690 patients undergoing

    infrainguinal bypass.53 Half the patients were treated for

    claudication and the other half were treated for critical limb

    ischemia with a fairly even distribution of vein versus

    prosthetic material. The aspirin dose was 80 mg/d, and, inpatients randomly assigned to anticoagulation, the interna-

    tional normalized ratio was maintained at 3.0 to 4.5. The

    primary end point of patency was equal between groups after

    21 months follow-up. In subgroup analysis, anticoagulation

    maintained vein graft patency better than aspirin but at a

    higher risk of bleeding complications. In contrast, aspirin

    maintained prosthetic graft patency better than anticoagula-

    tion. Although these results suggest that patients receiving

    vein grafts should be preferentially treated with warfarin and

    those with prosthetic material with aspirin, adequately pow-

    ered studies are needed to determine the optimal antithrom-

    botic and antiplatelet strategy following vascular surgery.

    In the context of promoting patency after a revasculariza-

    tion procedure, antiplatelet and antithrombotic therapy have a

    clear role, but the data are somewhat dated. In terms of 

    selection between type of therapy, aspirin may be favored for

    prosthetic grafts and anticoagulation for vein grafts or for

    higher-risk patients for occlusion.44 Future trials are needed.

    Novel TherapiesTherapeutic angiogenesis is a promising investigational strat-

    egy for the treatment of patients with claudication and

    chronic leg ischemia (CLI). It is an application of biotech-

    nology to stimulate new vessel formation via local adminis-

    tration of proangiogenic growth factors delivered as recom-binant protein or by gene therapy, or by implantation of 

    endothelial or other progenitor cells that will synthesize

    multiple angiogenic cytokines.

    Gene TherapyGene therapy involves transfer of genetic material into cells

    to modify their genetic expression to produce a clinically

    useful effect on angiogenesis, capillary generation, and col-

    lateral formation. Clinical trials have sought to establish the

    role for therapeutic angiogenesis by use of gene transfer with

    proangiogenic factors mediated by plasmids or viral vectors

    in patients with PAD, although results have been inconsistent

    (Table 2). These proangiogenic factors include vascular

    endothelial growth factor (VEGF), fibroblast growth factor

    (FGF), hypoxia-inducible factor (HIF)-1, and hepatocyte

    growth factor (HGF).

    In a meta-analysis evaluating the efficacy and safety of 

    different gene therapies, recombinant protein and cellular-

    based treatment approaches in patients with PAD, therapeutic

    angiogenesis was associated with a modest, albeit significant

    clinical improvement in peak walking time, ulcer healing, restpain relief, and limb salvage versus placebo in subjects with

    PAD (odds ratio 1.44, 95% CI 1.03–2.00).62 The improve-

    ment was most impressive in subjects with CLI. There was

    also a 30% increase in the odds of the adverse events of 

    edema, hypotension, and proteinuria. No significant differ-

    ence was detected for the endpoints of mortality, malignancy,

    or retinopathy.

    Vascular Endothelial Growth FactorVascular endothelial growth factor is expressed under hy-

    poxic conditions and is a potent regulator of endothelial cell

    migration, proliferation, repair, and survival.63 In previous

    studies involving animal studies, VEGF gene transfer inducedrapid production of nitric oxide and prostacyclin from endo-

    thelium causing a vasculoprotective effect.64,65 In a hindlimb

    ischemia animal model, VEGF was demonstrated to induce

    angiogenesis and arteriogenesis.66,67 Initial uncontrolled clin-

    ical trials have suggested promising therapeutic effects with

    naked VEGF plasmid gene transfer.68,69 In a comparison of 

    intra-arterial infusion of adenoviral VEGF165, plasmid lipo-

    some VEGF165, or Ringers lactate placebo, Makinen et al70

    noted that both VEGF165  treatments appeared safe and were

    associated with significant increases in vascularity.

    In a phase I trial, Baumgartner and colleagues70 found that

    intramuscular injection achieves overexpression of VEGF

    sufficient to induce therapeutic angiogenesis in selected

    patients with CLI.

    In the Regional Angiogenesis with VEGF (RAVE) trial, a

    single intramuscular adenoviral delivery of VEGF121   in

    patients with unilateral exercise-limiting claudication failed

    to achieve its primary end point of change in treadmill peak 

    walking time.54 There was no difference in any of the

    secondary end points, and VEGF121   was associated with a

    dose-dependent increase in peripheral edema. Reasons for the

    lack of efficacy could be the population selected (patients

    with bilateral PAD were excluded), the duration of expression

    of the VEGF121  transgene by use of the adenoviral approach

    may be insufficient to induce a phenotypic response, and thesingle injection may be insufficient.71 It is possible that

     494 Circulation   July 24, 2012

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    5/11

  • 8/20/2019 Circulation 2012 Berger 491 500

    6/11

    importance of performing large phase III trials in this new

    area of gene therapeutics.

    In the setting of claudication, Lederman and colleagues57

    performed the Therapeutic Angiogenesis with FGF-2 for

    Intermittent Claudication (TRAFFIC) phase II trial testing the

    efficacy and safety of intra-arterial FGF-2 protein in 190

    patients with moderate-to-severe intermittent claudication.

    Intra-arterial FGF-2 protein resulted in a significant increase

    in peak walking time at 90 days. No follow-up study has been

    performed.

    Hypoxia Inducible Factor-1The transcription factor HIF-1   is important in vascular

    hemostasis; HIF-1 regulates the expression of specific genes

    involved in the response to hypoxia and wound healing.74

    Because HIF-1   involves both physiological and pathologi-

    cal angiogenesis, strategies that target this factor have been

    tested in the setting of PAD. In a phase I dose escalation

    study, HIF-1   delivered intramuscularly with an adenoviral

    vector was safe out to 1-year.75

    There were data to suggestthat certain subjects had amelioration of rest pain symptoms

    and complete ulcer healing. In a larger study (WALK) to

    assess the transcription factor HIF-1  , results demonstrated

    no benefit in the primary end point.71

    Hepatocyte Growth FactorHepatocyte growth factor stimulates the growth of endothe-

    lial cells and promotes angiogenesis.76 HGF is able to induce

    robust collateral formation in preliminary studies.77 Subse-

    quently, a randomized trial was performed to assess the safety

    and potential efficacy of intramuscular injections of HGF

    plasmids in 104 patients with CLI.78 There was no safety

    concern attributed to the HGF therapy. Seventy-three patientswere available for the efficacy component; the highest-dose–

    treated group had a significant increase in transcutaneous

    partial pressure of oxygen at 6 months, which was signifi-

    cantly greater than in the placebo group. There was no

    difference between groups in the ABI, toe-brachial index,

    pain relief, wound healing, or major amputation. In a recent

    phase I/IIa study, Morishita and colleagues79 evaluated the

    safety and potential efficacy of intramuscular injection of 

    plasmid HGF in patients with CLI. No serious adverse events

    were noted. In a nonrandomized comparison, patients had an

    improvement in ABI, toe-brachial index, rest pain, and ulcer

    size at 6 months. There was no difference in transcutaneous

    partial pressure of oxygen. Larger studies are needed to

    determine the efficacy and safety profile of HGF.

    Developmentally Regulated Endothelial LocusDevelopmentally Regulated Endothelial Locus (Del-1) is an

    extracellular matrix protein that accumulates around angio-

    genic blood vessels and promotes angiogenesis even in the

    absence of exogenous growth factors.80 The Del-1 for Ther-

    apeutic Angiogenesis (DELTA) trial randomly assigned 105

    PAD patients with claudication to intramuscular injections of 

    Del-1 plasmid with poloxamer 188 (to enhance transfection)

    or poloxamer 188 alone.56 No significant safety issues were

    associated with Del-1. Peak treadmill walking time improvedsignificantly in both groups without any incremental benefit

    in the Del-1 group. Although ABI, claudication onset time,

    and quality of life improved in both groups, no difference

    emerged between Del-1 and the control group. The improve-

    ment of outcomes in both groups observed in this study

    underscores the substantial placebo effect and the importance

    of having a placebo group.

    Advancement of gene therapy from the safety-and-feasibilitystage to routine clinical use will require carefully designed,

    adequately powered, large-scale, randomized, controlled phase

    III trials that incorporate end points that address methodological

    improvements, long-term safety, and clinically important end

    points.

    Cell TherapyAnother potential novel treatment modality for PAD patients

    with claudication and chronic ischemia is the stimulation of 

    angiogenesis with cell therapy. Endothelial progenitor cells

    are important mediators in postnatal neovascularization after

    mobilization from the bone marrow.81,82 Numerous studies

    have demonstrated that, in the setting of both hindlimb and

    coronary ischemia models, endothelial progenitor cells can be

    harvested, expanded ex vivo, and delivered to augment capillary

    density, perfusion, and organ function.83 Bone marrow–derived

    mononuclear cell implantation stimulates the production of 

    endothelial progenitor cells and increases collateral vessel

    formation in both ischemic limb models and patients with

    limb ischemia.84,85 Implantation of bone marrow mononu-

    clear cells (versus peripheral blood mononuclear cells) im-

    proved the ABI, transcutaneous oxygen pressure, rest pain,

    and pain-free treadmill walking time at 4 and 24 weeks after

    injection.85 In a follow-up study, Higashi and colleagues86

    demonstrated that bone marrow mononuclear cells improvedendothelium-dependent vasodilation in patients with limb

    ischemia in addition to an effect on ischemic symptoms and

    findings of angiography. Several international phase II trials

    are underway to assess the benefit/safety profile of cell therapy

    in patients with PAD.

    In an animal model, intra-arterial infusion of granulocyte-

    macrophage colony-stimulating factor (GM-CSF) stimulates

    the development of arterial collateral blood vessels following

    femoral artery occlusion.87 In a small study of 21 subjects

    with extensive coronary artery disease not eligible for bypass

    surgery, intracoronary injection of GM-CSF improved collat-

    eral flow and led to a reduction in ST-segment changes andepisodes of angina.88 The STimulation of ARTeriogenesis

    (START) study was the first placebo-controlled study in the

    setting of moderate or severe claudication to investigate the

    effect of GM-CSF on walking distance.60 Patients were

    treated with placebo or subcutaneously applied GM-CSF (10

    g/kg) for a period of 14 days. No major side effects were

    observed; however, skin rash, muscle pain, and fever were

    more common in the GM-CSF group. There was no differ-

    ence in the primary end point (increase in maximum walking

    distance) or secondary end point (ABI) either directly after

    treatment or at 90 days of follow-up. Treatment with granu-

    locyte colony-stimulating factor is another promising ap-proach in this setting.61

     496 Circulation   July 24, 2012

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    7/11

    AntimicrobialsObservational data provided the groundwork suggesting that

    an infectious agent may be an important link between chronic

    inflammation and PAD. In a small study, 20 subjects sero-

    positive to   Chlamydia  pneumonia who received roxitrhomy-

    cin (400 mg daily) had better walking distance and fewer

    invasive revascularization procedures than 31 subjects seropos-itive for Chlamydia pneumonia who did not receive antibiotics.89

    Several other small studies failed to demonstrate a benefit of 

    antimicrobial therapy in subjects with PAD.90,91 To better

    understand the role of antimicrobials in PAD, Jaff and col-

    leagues92 randomly assigned 283 PAD subjects with claudica-

    tion to 25 mg rifalazil weekly for 8 weeks or matching placebo.

    All subjects enrolled had  Chlamydia  pneumonia antibody titer

    values 1:128. There was no benefit of rifalazil on the primary

    or any secondary assessment measured.92 No difference in

    cardiovascular events was noted, but the study was underpow-

    ered to detect this difference. Combined with the plethora of 

    negative studies of antimicrobials in subjects with coronary

    disease,93 there is little compelling reason to study this class of compounds in subjects with atherosclerotic disease.

    Future DirectionsMedical management of PAD has gone through important

    innovations over the past several decades with the widespread

    use of exercise, antiplatelet therapy, and statin medications.

    However, many of the recommendations for patients with

    PAD were derived from subgroup analyses of trials per-

    formed in populations with other atherosclerotic disease

    manifestations. Even aspirin, a class I level of recommenda-

    tion A therapy used in the treatment of PAD, has not been

    demonstrated to decrease cardiovascular events in the settingof PAD. There are considerable gaps in knowledge regarding

    the optimal treatment of PAD patients with claudication and

    CLI in comparison with patients with atherosclerotic disease

    in other territories. Current need for improvement in PAD

    patient outcomes include (1) PAD-focused systemic therapies

    designed to reduce the risk of ischemic events in the target

    population, and (2) continued development of limb-specific

    therapies, particularly in CLI.

    ConclusionsDespite the paucity of PAD-specific limb and systemic thera-

    pies, PAD patients are at very high risk for cardiovascularevents and impaired quality of life and should therefore be

    treated aggressively. Until additional prospective, random-

    ized trials are performed in this population, patients should be

    treated with strategies based on current recommendations for

    other manifestations of atherosclerotic disease. Current phar-

    macological options include cilostazol for claudication but

    little else to recommend as other claudication therapies or

    medical approaches to CLI.

    DisclosuresDr Hiatt has received grant support to his research institution (CPC

    Clinical Research, a nonprofit affiliate of the University of Colorado)

    from Sanofi-Aventis, AstraZeneca, Sigma Tau, and Aastrom. DrBerger has received grant support from AstraZeneca.

    References1. Pande RL, Perlstein TS, Beckman JA, Creager MA. Secondary pre-

    vention and mortality in peripheral artery disease: National Health and

    Nutrition Examination Study, 1999 to 2004.   Circulation. 2011;124:

    17–23.

    2. Golomb BA, Dang TT, Criqui MH. Peripheral arterial disease: morbidity

    and mortality implications.  Circulation. 2006;114:688–699.

    3. Diehm C, Allenberg JR, Pittrow D, Mahn M, Tepohl G, Haberl RL,

    Darius H, Burghaus I, Trampisch HJ. Mortality and vascular morbidity inolder adults with asymptomatic versus symptomatic peripheral artery

    disease. Circulation. 2009;120:2053–2061.

    4. Fowkes FG, Murray GD, Butcher I, Heald CL, Lee RJ, Chambless LE,

    Folsom AR, Hirsch AT, Dramaix M, deBacker G, Wautrecht JC, Kor-

    nitzer M, Newman AB, Cushman M, Sutton-Tyrrell K, Lee AJ, Price JF,

    d’Agostino RB, Murabito JM, Norman PE, Jamrozik K, Curb JD, Masaki

    KH, Rodriguez BL, Dekker JM, Bouter LM, Heine RJ, Nijpels G,

    Stehouwer CD, Ferrucci L, McDermott MM, Stoffers HE, Hooi JD,

    Knottnerus JA, Ogren M, Hedblad B, Witteman JC, Breteler MM, Hunink 

    MG, Hofman A, Criqui MH, Langer RD, Fronek A, Hiatt WR, Hamman

    R, Resnick HE, Guralnik J. Ankle brachial index combined with Fra-

    mingham Risk Score to predict cardiovascular events and mortality: a

    meta-analysis.  JAMA. 2008;300:197–208.

    5. Hiatt WR. Medical treatment of peripheral arterial disease and claudi-

    cation.  N Engl J Med . 2001;344:1608–1621.

    6. Norgren L, Hiatt WR, Dormandy JA, Nehler MR, Harris KA, FowkesFG. Inter-Society Consensus for the Management of Peripheral Arterial

    Disease (TASC II).  J Vasc Surg.  2007;45(suppl S):S5–S67.

    7. Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL,

    Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks

    D, Stanley JC, Taylor LM Jr, White CJ, White J, White RA, Antman EM,

    Smith SC Jr, Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ,

    Hunt SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B.

    ACC/AHA 2005 Practice Guidelines for the management of patients with

    peripheral arterial disease (lower extremity, renal, mesenteric, and

    abdominal aortic): a collaborative report from the American Association

    for Vascular Surgery/Society for Vascular Surgery, Society for Cardio-

    vascular Angiography and Interventions, Society for Vascular Medicine

    and Biology, Society of Interventional Radiology, and the ACC/AHA

    Task Force on Practice Guidelines (Writing Committee to Develop

    Guidelines for the Management of Patients With Peripheral Arterial

    Disease): endorsed by the American Association of Cardiovascular and

    Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute;

    Society for Vascular Nursing; TransAtlantic Inter-Society Consensus;

    and Vascular Disease Foundation.  Circulation. 2006;113:e463–e654.

    8. Olin JW, Allie DE, Belkin M, Bonow RO, Casey DE Jr, Creager MA,

    Gerber TC, Hirsch AT, Jaff MR, Kaufman JA, Lewis CA, Martin ET,

    Martin LG, Sheehan P, Stewart KJ, Treat-Jacobson D, White CJ, Zheng

    ZJ, Masoudi FA. ACCF/AHA/ACR/SCAI/SIR/SVM/SVN/SVS 2010

    performance measures for adults with peripheral artery disease: a report

    of the American College of Cardiology Foundation/American Heart

    Association Task Force on performance measures, the American College

    of Radiology, the Society for Cardiac Angiography and Interventions, the

    Society for Interventional Radiology, the Society for Vascular Medicine,

    the Society for Vascular Nursing, and the Society for Vascular Surgery

    (Writing Committee to Develop Clinical Performance Measures for Pe-

    ripheral Artery Disease).  Circulation. 2010;122:2583–2618.

    9. 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.

    10. MRC/BHF Heart Protection Study of cholesterol lowering with simva-

    statin in 20,536 high-risk individuals: a randomised placebo-controlled

    trial.  Lancet.  2002;360:7–22.

    11. A randomised, blinded, trial of clopidogrel versus aspirin in patients at

    risk of ischaemic events (CAPRIE). CAPRIE Steering Committee.

     Lancet.  1996;348:1329–1339.

    12. Collaborative meta-analysis of randomised trials of antiplatelet therapy

    for prevention of death, myocardial infarction, and stroke in high risk 

    patients.   BMJ.   2002;324:71–86.

    13. Collaborative overview of randomised trials of antiplatelet therapy–I:

    Prevention of death, myocardial infarction, and stroke by prolonged

    antiplatelet therapy in various categories of patients. Antiplatelet Tri-

    alists’ Collaboration.  BMJ.  1994;308:81–106.

    14. Berger JS, Krantz MJ, Kittelson JM, Hiatt WR. Aspirin for the prevention

    of cardiovascular events in patients with peripheral artery disease: ameta-analysis of randomized trials.  JAMA. 2009;301:1909–1919.

    Berger and Hiatt Medical Therapy in Peripheral Artery Disease   497 

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    8/11

    15. Fowkes FG, Price JF, Stewart MC, Butcher I, Leng GC, Pell AC,

    Sandercock PA, Fox KA, Lowe GD, Murray GD. Aspirin for prevention

    of cardiovascular events in a general population screened for a low ankle

    brachial index: a randomized controlled trial. JAMA.  303:841–848.

    16. Belch J, MacCuish A, Campbell I, Cobbe S, Taylor R, Prescott R, Lee R,

    Bancroft J, MacEwan S, Shepherd J, Macfarlane P, Morris A, Jung R,

    Kelly C, Connacher A, Peden N, Jamieson A, Matthews D, Leese G,

    McKnight J, O’Brien I, Semple C, Petrie J, Gordon D, Pringle S, Mac-

    Walter R. The prevention of progression of arterial disease and diabetes(POPADAD) trial: factorial randomised placebo controlled trial of aspirin

    and antioxidants in patients with diabetes and asymptomatic peripheral

    arterial disease.  BMJ . 2008;337:a1840.

    17. Cacoub PP, Bhatt DL, Steg PG, Topol EJ, Creager MA. Patients with

    peripheral arterial disease in the CHARISMA trial.   Eur Heart J . 2009;

    30:192–201.

    18. Gardner AW, Poehlman ET. Exercise rehabilitation programs for the

    treatment of claudication pain. A meta-analysis.   JAMA. 1995;274:

    975–980.

    19. Watson L, Ellis B, Leng GC. Exercise for intermittent claudication.

    Cochrane Database Syst Rev.  2008:CD000990.

    20. McDermott MM, Ades P, Guralnik JM, Dyer A, Ferrucci L, Liu K,

    Nelson M, Lloyd-Jones D, Van Horn L, Garside D, Kibbe M, Domanchuk 

    K, Stein JH, Liao Y, Tao H, Green D, Pearce WH, Schneider JR,

    McPherson D, Laing ST, McCarthy WJ, Shroff A, Criqui MH. 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.

    21. Gardner AW, Parker DE, Montgomery PS, Scott KJ, Blevins SM.

    Efficacy of quantified home-based exercise and supervised exercise in

    patients with intermittent claudication: a randomized controlled trial.

    Circulation. 2011;123:491–498.

    22. Bendermacher BL, Willigendael EM, Teijink JA, Prins MH. Supervised

    exercise therapy versus non-supervised exercise therapy for intermittent

    claudication.   Cochrane Database Syst Rev.  2006:CD005263.

    23. Reilly MP, Mohler ER III. Cilostazol. treatment of intermittent claudi-

    cation.  Ann Pharmacother . 2001;35:48–56.

    24. Thompson PD, Zimet R, Forbes WP, Zhang P. Meta-analysis of results

    from eight randomized, placebo-controlled trials on the effect of 

    cilostazol on patients with intermittent claudication.  Am J Cardiol. 2002;

    90:1314–1319.

    25. Soga Y, Iida O, Hirano K, Suzuki K, Yokoi H, Nobuyoshi M. Restenosis

    after stent implantation for superficial femoral artery disease in patients

    treated with cilostazol.   Catheter Cardiovasc Interv.  2012;79:541–548.

    26. Jennings DL, Kalus JS. Addition of cilostazol to aspirin and a thien-

    opyridine for prevention of restenosis after coronary artery stenting: a

    meta-analysis.  J Clin Pharmacol. 2010;50:415–421.

    27. Douglas JS Jr, Holmes DR Jr, Kereiakes DJ, Grines CL, Block E, Ghazzal

    ZM, Morris DC, Liberman H, Parker K, Jurkovitz C, Murrah N, Foster J,

    Hyde P, Mancini GB, Weintraub WS. Coronary stent restenosis in

    patients treated with cilostazol.  Circulation. 2005;112:2826–2832.

    28. Lee SW, Park SW, Kim YH, Yun SC, Park DW, Lee CW, Kang SJ, Park 

    SJ, Lee JH, Choi SW, Seong IW, Lee NH, Cho YH, Shin WY, Lee SJ,

    Hyon MS, Bang DW, Choi YJ, Kim HS, Lee BK, Lee K, Park HK, Park 

    CB, Lee SG, Kim MK, Park KH, Park WJ. A randomized, double-blind,

    multicenter comparison study of triple antiplatelet therapy with dual

    antiplatelet therapy to reduce restenosis after drug-eluting stent implan-

    tation in long coronary lesions: results from the DECLARE-LONG II

    (Drug-Eluting Stenting Followed by Cilostazol Treatment Reduces LateRestenosis in Patients with Long Coronary Lesions) trial.   J Am Coll

    Cardiol. 2011;57:1264–1270.

    29. Iida O, Nanto S, Uematsu M, Morozumi T, Kitakaze M, Nagata S.

    Cilostazol reduces restenosis after endovascular therapy in patients with

    femoropopliteal lesions.  J Vasc Surg. 2008;48:144–149.

    30. Soga Y, Yokoi H, Kawasaki T, Nakashima H, Tsurugida M, Hikichi Y,

    Nobuyoshi M. Efficacy of cilostazol after endovascular therapy for fem-

    oropopliteal artery disease in patients with intermittent claudication. J Am

    Coll Cardiol. 2009;53:48–53.

    31. Packer M, Carver JR, Rodeheffer RJ, Ivanhoe RJ, DiBianco R, Zeldis

    SM, Hendrix GH, Bommer WJ, Elkayam U, Kukin ML, Mallis GI,

    Sollano JA, Shannon J, Tandon PK, DeMets DL. Effect of oral milrinone

    on mortality in severe chronic heart failure. The PROMISE Study

    Research Group.  N Engl J Med . 1991;325:1468 –1475.

    32. Hiatt WR, Money SR, Brass EP. Long-term safety of cilostazol in patients

    with peripheral artery disease: the CASTLE study (Cilostazol: A Study inLong-term Effects).  J Vasc Surg. 2008;47:330–336.

    33. Cone J, Wang S, Tandon N, Fong M, Sun B, Sakurai K, Yoshitake M,

    Kambayashi J, Liu Y. Comparison of the effects of cilostazol and mil-

    rinone on intracellular cAMP levels and cellular function in platelets and

    cardiac cells.   J Cardiovasc Pharmacol. 1999;34:497–504.

    34. Pedersen TR, Kjekshus J, Pyorala K, Olsson AG, Cook TJ, Musliner TA,

    Tobert JA, Haghfelt T. Effect of simvastatin on ischemic signs and

    symptoms in the Scandinavian simvastatin survival study (4S).   Am J 

    Cardiol. 1998;81:333–335.

    35. Mohler ER III, Hiatt WR, Creager MA. Cholesterol reduction withatorvastatin improves walking distance in patients with peripheral arterial

    disease. Circulation. 2003;108:1481–1486.

    36. Brevetti G, Chiariello M, Ferulano G, Policicchio A, Nevola E, Rossini

    A, Attisano T, Ambrosio G, Siliprandi N, Angelini C. Increases in

    walking distance in patients with peripheral vascular disease treated with

    L-carnitine: a double-blind, cross-over study.   Circulation. 1988;77:

    767–773.

    37. Brevetti G, Diehm C, Lambert D. European multicenter study on

    propionyl-L-carnitine in intermittent claudication.   J Am Coll Cardiol.

    1999;34:1618–1624.

    38. Hiatt WR, Regensteiner JG, Creager MA, Hirsch AT, Cooke JP, Olin JW,

    Gorbunov GN, Isner J, Lukjanov YV, Tsitsiashvili MS, Zabelskaya TF,

    Amato A. Propionyl-L-carnitine improves exercise performance and

    functional status in patients with claudication.   Am J Med . 2001;110:

    616–622.

    30. Hiatt WR, Creager MA, Amato A, Brass EP. Effect of propionyl-L-

    carnitine on a background of monitored exercise in patients with claudi-

    cation secondary to peripheral artery disease.  J Cardiopulm Rehabil Prev.

    2011;31:125–132.

    40. Hood SC, Moher D, Barber GG. Management of intermittent claudication

    with pentoxifylline: meta-analysis of randomized controlled trials.  CMAJ .

    1996;155:1053–1059.

    41. Barradell LB, Brogden RN. Oral naftidrofuryl. A review of its pharma-

    cology and therapeutic use in the management of peripheral occlusive

    arterial disease.   Drugs Aging. 1996;8:299–322.

    42. De Backer T, Vander Stichele R, Lehert P, Van Bortel L. Naftidrofuryl

    for intermittent claudication: meta-analysis based on individual patient

    data.  BMJ . 2009;338:b603.

    43. Collaborative overview of randomised trials of antiplatelet therapy–II:

    Maintenance of vascular graft or arterial patency by antiplatelet therapy.

    Antiplatelet Trialists’ Collaboration.   BMJ.   1994;308:159–168.

    44. Tangelder MJ, Lawson JA, Algra A, Eikelboom BC. Systematic review

    of randomized controlled trials of aspirin and oral anticoagulants in the

    prevention of graft occlusion and ischemic events after infrainguinal

    bypass surgery.  J Vasc Surg. 1999;30:701–709.

    45. Becquemin JP. Effect of ticlopidine on the long-term patency of 

    saphenous-vein bypass grafts in the legs. Etude de la Ticlopidine apres

    Pontage Femoro-Poplite and the Association Universitaire de Recherche

    en Chirurgie.  N Engl J Med . 1997;337:1726–1731.

    46. Belch JJ, Dormandy J, Biasi GM, Cairols M, Diehm C, Eikelboom B,

    Golledge J, Jawien A, Lepantalo M, Norgren L, Hiatt WR, Becquemin JP,

    Bergqvist D, Clement D, Baumgartner I, Minar E, Stonebridge P, Ver-

    massen F, Matyas L, Leizorovicz A. Results of the randomized, placebo-

    controlled clopidogrel and acetylsalicylic acid in bypass surgery for

    peripheral arterial disease (CASPAR) trial.   J Vasc Surg.   2010;52:

    825–833, 833.e1–2.

    47. Collins TC, Souchek J, Beyth RJ. Benefits of antithrombotic therapy after

    infrainguinal bypass grafting: a meta-analysis.   Am J Med . 2004;117:

    93–99.48. Samama CM, Gigou F, Ill P. Low-molecular-weight heparin vs. unfrac-

    tionated heparin in femorodistal reconstructive surgery: a multicenter

    open randomized study. Enoxart Study Group.   Ann Vasc Surg.   1995;

    9(suppl):S45–S53.

    49. Sarac TP, Huber TS, Back MR, Ozaki CK, Carlton LM, Flynn TC, Seeger

    JM. Warfarin improves the outcome of infrainguinal vein bypass grafting

    at high risk for failure.  J Vasc Surg. 1998;28:446–457.

    50. Arfvidsson B, Lundgren F, Drott C, Schersten T, Lundholm K. Influence

    of coumarin treatment on patency and limb salvage after peripheral

    arterial reconstructive surgery.  Am J Surg. 1990;159:556–560.

    51. Kretschmer G, Herbst F, Prager M, Sautner T, Wenzl E, Berlakovich GA,

    Zekert F, Marosi L, Schemper M. A decade of oral anticoagulant

    treatment to maintain autologous vein grafts for femoropopliteal athero-

    sclerosis.  Arch Surg. 1992;127:1112–1115.

    52. Edmondson RA, Cohen AT, Das SK, Wagner MB, Kakkar VV. Low-

    molecular weight heparin versus aspirin and dipyridamole after femoro-popliteal bypass grafting.  Lancet . 1994;344:914–918.

     498 Circulation   July 24, 2012

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    9/11

    53. Efficacy of oral anticoagulants compared with aspirin after infrainguinal

    bypass surgery (The Dutch Bypass Oral Anticoagulants or Aspirin

    Study): a randomised trial.  Lancet.   2000;355:346–351.

    54. Rajagopalan S, Mohler ER III, Lederman RJ, Mendelsohn FO, Saucedo

    JF, Goldman CK, Blebea J, Macko J, Kessler PD, Rasmussen HS, Annex

    BH. Regional angiogenesis with vascular endothelial growth factor in

    peripheral arterial disease: a phase II randomized, double-blind, con-

    trolled study of adenoviral delivery of vascular endothelial growth factor

    121 in patients with disabling intermittent claudication.  Circulation

    .2003;108:1933–1938.

    55. Kusumanto YH, van Weel V, Mulder NH, Smit AJ, van den Dungen JJ,

    Hooymans JM, Sluiter WJ, Tio RA, Quax PH, Gans RO, Dullaart RP,

    Hospers GA. Treatment with intramuscular vascular endothelial growth

    factor gene compared with placebo for patients with diabetes mellitus and

    critical limb ischemia: a double-blind randomized trial.  Hum Gene Ther .

    2006;17:683–691.

    56. Grossman PM, Mendelsohn F, Henry TD, Hermiller JB, Litt M, Saucedo

    JF, Weiss RJ, Kandzari DE, Kleiman N, Anderson RD, Gottlieb D,

    Karlsberg R, Snell J, Rocha-Singh K. Results from a phase II multicenter,

    double-blind placebo-controlled study of Del-1 (VLTS-589) for inter-

    mittent claudication in subjects with peripheral arterial disease.   Am

     Heart J . 2007;153:874–880.

    57. Lederman RJ, Mendelsohn FO, Anderson RD, Saucedo JF, Tenaglia AN,

    Hermiller JB, Hillegass WB, Rocha-Singh K, Moon TE, Whitehouse MJ,

    Annex BH. Therapeutic angiogenesis with recombinant fibroblast growth

    factor-2 for intermittent claudication (the TRAFFIC study): a randomised

    trial.  Lancet . 2002;359:2053–2058.

    58. Nikol S, Baumgartner I, Van Belle E, Diehm C, Visona A, Capogrossi

    MC, Ferreira-Maldent N, Gallino A, Wyatt MG, Wijesinghe LD, Fusari

    M, Stephan D, Emmerich J, Pompilio G, Vermassen F, Pham E, Grek V,

    Coleman M, Meyer F. Therapeutic angiogenesis with intramuscular

    NV1FGF improves amputation-free survival in patients with critical limb

    ischemia. Mol Ther . 2008;16:972–978.

    59. Belch J, Hiatt WR, Baumgartner I, Driver IV, Nikol S, Norgren L, Van

    Belle E. Effect of fibroblast growth factor NV1FGF on amputation and

    death: a randomised placebo-controlled trial of gene therapy in critical

    limb ischaemia. Lancet . 2011;377:1929–1937.

    60. van Royen N, Schirmer SH, Atasever B, Behrens CY, Ubbink D,

    Buschmann EE, Voskuil M, Bot P, Hoefer I, Schlingemann RO, Biemond

    BJ, Tijssen JG, Bode C, Schaper W, Oskam J, Legemate DA, Piek JJ,

    Buschmann I. START Trial: a pilot study on STimulation of ARTerio-

    genesis using subcutaneous application of granulocyte-macrophage

    colony-stimulating factor as a new treatment for peripheral vascular

    disease. Circulation. 2005;112:1040–1046.

    61. Arai M, Misao Y, Nagai H, Kawasaki M, Nagashima K, Suzuki K,

    Tsuchiya K, Otsuka S, Uno Y, Takemura G, Nishigaki K, Minatoguchi S,

    Fujiwara H. Granulocyte colony-stimulating factor: a noninvasive regen-

    eration therapy for treating atherosclerotic peripheral artery disease.  Circ

     J . 2006;70:1093–1098.

    62. De Haro J, Acin F, Lopez-Quintana A, Florez A, Martinez-Aguilar E,

    Varela C. Meta-analysis of randomized, controlled clinical trials in angio-

    genesis: gene and cell therapy in peripheral arterial disease.   Heart 

    Vessels. 2009;24:321–328.

    63. Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular permeability

    factor/vascular endothelial growth factor, microvascular hyperperme-

    ability, and angiogenesis.  Am J Pathol. 1995;146:1029–1039.

    64. Yla-Herttuala S, Martin JF. Cardiovascular gene therapy. Lancet . 2000;

    355:213–222.65. Laitinen M, Zachary I, Breier G, Pakkanen T, Hakkinen T, Luoma J,

    Abedi H, Risau W, Soma M, Laakso M, Martin JF, Yla-Herttuala S.

    VEGF gene transfer reduces intimal thickening via increased production

    of nitric oxide in carotid arteries.  Hum Gene Ther . 1997;8:1737–1744.

    66. Gowdak LH, Poliakova L, Wang X, Kovesdi I, Fishbein KW, Zacheo A,

    Palumbo R, Straino S, Emanueli C, Marrocco-Trischitta M, Lakatta EG,

    Anversa P, Spencer RG, Talan M, Capogrossi MC. Adenovirus-mediated

    VEGF(121) gene transfer stimulates angiogenesis in normoperfused

    skeletal muscle and preserves tissue perfusion after induction of ischemia.

    Circulation. 2000;102:565–571.

    67. Mack CA, Magovern CJ, Budenbender KT, Patel SR, Schwarz EA,

    Zanzonico P, Ferris B, Sanborn T, Isom P, Isom OW, Crystal RG,

    Rosengart TK. Salvage angiogenesis induced by adenovirus-mediated

    gene transfer of vascular endothelial growth factor protects against ische-

    mic vascular occlusion.  J Vasc Surg. 1998;27:699–709.

    68. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L, Asahara T,Rosenfield K, Razvi S, Walsh K, Symes JF. Clinical evidence of angio-

    genesis after arterial gene transfer of phVEGF165 in patient with

    ischaemic limb. Lancet . 1996;348:370 –374.

    69. Baumgartner I, Pieczek A, Manor O, Blair R, Kearney M, Walsh K, Isner

    JM. Constitutive expression of phVEGF165 after intramuscular gene

    transfer promotes collateral vessel development in patients with critical

    limb ischemia. Circulation. 1998;97:1114 –1123.

    70. Makinen K, Manninen H, Hedman M, Matsi P, Mussalo H, Alhava E,

    Yla-Herttuala S. Increased vascularity detected by digital subtraction

    angiography after VEGF gene transfer to human lower limb artery: arandomized, placebo-controlled, double-blinded phase II study.  Mol Ther .

    2002;6:127–133.

    71. Creager MA, Olin JW, Belch JJ, Moneta GL, Henry TD, Rajagopalan S,

    Annex BH, Hiatt WR. Effect of hypoxia-inducible factor-1alpha gene

    therapy on walking performance in patients with intermittent claudi-

    cation.  Circulation.  2011;124:1765–1773.

    72. Aviles RJ, Annex BH, Lederman RJ. Testing clinical therapeutic angio-

    genesis using basic fibroblast growth factor (FGF-2).   Br J Pharmacol.

    2003;140:637–646.

    73. Comerota AJ, Throm RC, Miller KA, Henry T, Chronos N, Laird J,

    Sequeira R, Kent CK, Bacchetta M, Goldman C, Salenius JP, Schmieder

    FA, Pilsudski R. Naked plasmid DNA encoding fibroblast growth factor

    type 1 for the treatment of end-stage unreconstructible lower extremity

    ischemia: preliminary results of a phase I trial.   J Vasc Surg. 2002;35:

    930–936.

    74. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of 

    the HIF system.  Nat Med . 2003;9:677–684.

    75. Rajagopalan S, Olin J, Deitcher S, Pieczek A, Laird J, Grossman PM,

    Goldman CK, McEllin K, Kelly R, Chronos N. Use of a constitutively

    active hypoxia-inducible factor-1alpha transgene as a therapeutic strategy

    in no-option critical limb ischemia patients: phase I dose-escalation expe-

    rience.  Circulation. 2007;115:1234–1243.

    76. Bussolino F, Di Renzo MF, Ziche M, Bocchietto E, Olivero M, Naldini

    L, Gaudino G, Tamagnone L, Coffer A, Comoglio PM. Hepatocyte

    growth factor is a potent angiogenic factor which stimulates endothelial

    cell motility and growth.  J Cell Biol. 1992;119:629–641.

    77. Morishita R, Nakamura S, Hayashi S, Taniyama Y, Moriguchi A, Nagano

    T, Taiji M, Noguchi H, Takeshita S, Matsumoto K, Nakamura T, Higaki

    J, Ogihara T. Therapeutic angiogenesis induced by human recombinant

    hepatocyte growth factor in rabbit hind limb ischemia model as cytokine

    supplement therapy.  Hypertension. 1999;33:1379–1384.

    78. Powell RJ, Simons M, Mendelsohn FO, Daniel G, Henry TD, Koga M,

    Morishita R, Annex BH. Results of a double-blind, placebo-controlled

    study to assess the safety of intramuscular injection of hepatocyte growth

    factor plasmid to improve limb perfusion in patients with critical limb

    ischemia. Circulation. 2008;118:58–65.

    79. Morishita R, Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J,

    Taniyama Y, Sawa Y, Kaneda Y, Ogihara T. Phase I/IIa clinical trial of 

    therapeutic angiogenesis using hepatocyte growth factor gene transfer to

    treat critical limb ischemia.   Arterioscler Thromb Vasc Biol. 2011;31:

    713–720.

    80. Zhong J, Eliceiri B, Stupack D, Penta K, Sakamoto G, Quertermous T,

    Coleman M, Boudreau N, Varner JA. Neovascularization of ischemic

    tissues by gene delivery of the extracellular matrix protein Del-1. J Clin

     Invest . 2003;112:30–41.

    81. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T,

    Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor

    endothelial cells for angiogenesis.  Science. 1997;275:964 –967.

    82. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M,Kearne M, Magner M, Isner JM. Bone marrow origin of endothelial

    progenitor cells responsible for postnatal vasculogenesis in physiological

    and pathological neovascularization.  Circ Res. 1999;85:221–228.

    83. Leeper NJ, Hunter AL, Cooke JP. Stem cell therapy for vascular regen-

    eration: adult, embryonic, and induced pluripotent stem cells. Circulation.

    2010;122:517–526.

    84. Shintani S, Murohara T, Ikeda H, Ueno T, Sasaki K, Duan J, Imaizumi T.

    Augmentation of postnatal neovascularization with autologous bone

    marrow transplantation.  Circulation. 2001;103:897–903.

    85. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S,

    Masaki H, Amano K, Kishimoto Y, Yoshimoto K, Akashi H, Shimada K,

    Iwasaka T, Imaizumi T. Therapeutic angiogenesis for patients with limb

    ischaemia by autologous transplantation of bone-marrow cells: a pilot

    study and a randomised controlled trial. Lancet . 2002;360:427–435.

    86. Higashi Y, Kimura M, Hara K, Noma K, Jitsuiki D, Nakagawa K, Oshima

    T, Chayama K, Sueda T, Goto C, Matsubara H, Murohara T, YoshizumiM. Autologous bone-marrow mononuclear cell implantation improves

    Berger and Hiatt Medical Therapy in Peripheral Artery Disease   499

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    10/11

    endothelium-dependent vasodilation in patients with limb ischemia.

    Circulation. 2004;109:1215–1218.

    87. Buschmann IR, Hoefer IE, van Royen N, Katzer E, Braun-Dulleaus R,

    Heil M, Kostin S, Bode C, Schaper W. GM-CSF: a strong arteriogenic

    factor acting by amplification of monocyte function.   Atherosclerosis.

    2001;159:343–356.

    88. Seiler C, Pohl T, Wustmann K, Hutter D, Nicolet PA, Windecker S,

    Eberli FR, Meier B. Promotion of collateral growth by granulocyte-mac-

    rophage colony-stimulating factor in patients with coronary artery dis-

    ease: a randomized, double-blind, placebo-controlled study.  Circulation.

    2001;104:2012–2017.

    89. Krayenbuehl PA, Wiesli P, Maly FE, Vetter W, Schulthess G. Pro-

    gression of peripheral arterial occlusive disease is associated with

    Chlamydia pneumoniae seropositivity and can be inhibited by antibiotic

    treatment.  Atherosclerosis. 2005;179:103–110.

    90. Joensen JB, Juul S, Henneberg E, Thomsen G, Ostergaard L, Lindholt JS.

    Can long-term antibiotic treatment prevent progression of peripheral

    arterial occlusive disease? A large, randomized, double-blinded, placebo-

    controlled trial.  Atherosclerosis. 2008;196:937–942.

    91. Vainas T, Stassen FR, Schurink GW, Tordoir JH, Welten RJ, van den

    Akker LH, Kurvers HA, Bruggeman CA, Kitslaar PJ. Secondary pre-

    vention of atherosclerosis through chlamydia pneumoniae eradication

    (SPACE Trial): a randomised clinical trial in patients with peripheral

    arterial disease.  Eur J Vasc Endovasc Surg. 2005;29:403–411.

    92. Jaff MR, Dale RA, Creager MA, Lipicky RJ, Constant J, Campbell LA,

    Hiatt WR. Anti-chlamydial antibiotic therapy for symptom improvement

    in peripheral artery disease: prospective evaluation of rifalazil effect on

    vascular symptoms of intermittent claudication and other endpoints inChlamydia pneumoniae seropositive patients (PROVIDENCE-1).

    Circulation. 2009;119:452–458.

    93. Andraws R, Berger JS, Brown DL. Effects of antibiotic therapy on

    outcomes of patients with coronary artery disease: a meta-analysis of 

    randomized controlled trials.  JAMA. 2005;293:2641–2647.

    KEY   WORDS: angiogenesis     antiplatelet therapy     arteries     treatment

      vascular disease

     500 Circulation   July 24, 2012

     by guest on July 24, 2015http://circ.ahajournals.org/ Downloaded from 

    http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/

  • 8/20/2019 Circulation 2012 Berger 491 500

    11/11

    Jeffrey S. Berger and William R. HiattMedical Therapy in Peripheral Artery Disease

    Print ISSN: 0009-7322. Online ISSN: 1524-4539Copyright © 2012 American Heart Association, Inc. All rights reserved.

    is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulationdoi: 10.1161/CIRCULATIONAHA.111.033886

    2012;126:491-500Circulation.

    http://circ.ahajournals.org/content/126/4/491

    World Wide Web at:The online version of this article, along with updated information and services, is located on the

     

    http://circ.ahajournals.org//subscriptions/ is online at:CirculationInformation about subscribing toSubscriptions:

     http://www.lww.com/reprints

     Information about reprints can be found online at:Reprints: 

    document.Permissions and Rights Question and Answerthis process is available in theclick Request Permissions in the middle column of the Web page under Services. Further information aboutOffice. Once the online version of the published article for which permission is being requested is located,

    can be obtained via RightsLink, a service of the Copyright Clearance Center, not the EditorialCirculationin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

    by guest on July 24 2015http://circ ahajournals org/Downloaded from

    http://circ.ahajournals.org/content/126/4/491http://circ.ahajournals.org//subscriptions/http://circ.ahajournals.org//subscriptions/http://circ.ahajournals.org//subscriptions/http://www.lww.com/reprintshttp://www.lww.com/reprintshttp://www.lww.com/reprintshttp://www.ahajournals.org/site/rights/http://www.ahajournals.org/site/rights/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org/http://circ.ahajournals.org//subscriptions/http://www.lww.com/reprintshttp://www.ahajournals.org/site/rights/http://circ.ahajournals.org/content/126/4/491