6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Is Paclitaxel Causing Mortality During Lower‐Extremity Revascularization?

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Drug‐coated devices for treatment of lower‐extremity peripheral artery disease have been used worldwide for nearly a decade. Before their debut, endovascular treatment options for peripheral artery disease revascularization were limited to conventional balloon angioplasty and bare‐metal stenting, both associated with high rates of restenosis, recoil, and vessel closure, yielding primary patency rates of only 30% to 70% by 1 year.1, 2 Vascular specialists quickly embraced drug‐coated devices when early trial data revealed 1‐year primary patency rates in the range of 70% to 90%.3, 4, 5, 6 In addition, the use of drug‐coated balloons (DCBs) avoided the need for a permanent metal scaffold in lower‐extremity vessels, where high degrees of shear stress, torsion, and flexion exist. Experts considered the technology to be a significant breakthrough in the field, not only for potentially improving patient outcomes, but also for being cost‐effective.7 Hence, the surprising results presented in the recent systematic review and meta‐analysis by Katsanos et al, published in the Journal of the American Heart Association (JAHA), 8 have led some to have a clinical pause. The analysis was performed with a primary safety measure of all‐cause mortality across 28 randomized controlled trials (RCTs), including 4432 cases of drug‐coated device use in the femoropopliteal artery of the lower limbs. The increased risk of late death after application of paclitaxel in these vessels was found to be alarmingly high, and the number needed to harm at 5 years was only 14 patients. In the current Viewpoint, we explore the scientific data behind paclitaxel‐coated devices in the context of recent mortality concerns. We also discuss newly generated patient‐level analyses and preclinical animal data on potential mechanisms of paclitaxel toxicity to address whether there is true causation or simply a correlation. Finally, we provide a perspective on use of drug‐coated devices and how to apply the results to clinical practice and patient‐centered decision making. Efficacy and Safety in Early Drug‐Coated Device Trials The Zilver PTX drug‐eluting stent (DES) (Cook Medical, Bloomington, IN) was the first paclitaxel‐coated device to gain US Food and Drug Administration (FDA) approval in 2012. The stent is directly coated with crystalline paclitaxel at a concentration of 3 μg/mm2 without any polymer, binder, or excipient. The 12‐month Zilver PTX study3 randomized 474 patients with predominantly claudication and femoropopliteal disease to the DES (n=236) or standard percutaneous transluminal angioplasty (PTA) (n=238). Nearly half of the patients in the PTA group underwent a second randomization because of PTA failure to provisional DES (n=61) or bare‐metal stenting (n=59). The primary end points included a safety and efficacy measure, defined as the rate of event‐free survival and patency. Twelve‐month event‐free survival and primary patency were significantly higher in the primary DES group compared with the PTA group (90.4% versus 82.6% [P<0.001] and 83.1% versus 32.8% [P<0.001], respectively).3 Five‐year follow‐up of the Zilver PTX randomized trial was published in 2016 and concluded that the initial results were sustained through this extended follow‐up period.9 The authors maintain this position despite recently correcting an article error that inadvertently reversed the all‐cause mortality rates between the 2 groups. The accurate all‐cause mortality rates at 5 years were significantly higher at 16.9% in the primary DES group compared with 10.2% in the PTA group (P=0.03).9 Exact causes of death were not discussed at that time, although none was adjudicated to be device or procedure related. Initial DCBs gained FDA approval in 2014 (Lutonix; C.R. Bard, Tempe, AZ) and 2015 (IN.PACT Admiral; Medtronic, Santa Rosa, CA). The Lutonix DCB is semicompliant, is coated with paclitaxel at 2 μg/mm2, and contains excipients (polysorbate and sorbitol) to control drug release and tissue deposition. The IN.PACT DCB is coated with paclitaxel at 3.5 μg/mm2 and has an excipient that allows for the antiproliferative drug to remain in the vessel at the treatment site for up to 180 days.10 LEVANT (Lutonix Paclitaxel‐Coated Balloon for the Prevention of Femoropopliteal Restenosis) 2 randomized 476 patients with claudication or rest leg pain with femoropopliteal disease in a 2:1 manner to the Lutonix DCB versus plain balloon angioplasty.4 The primary efficacy end point was primary patency of the target lesion at 12 months (defined as freedom from binary restenosis or from the need for target‐lesion revascularization). Safety end points were secondary and included freedom from perioperative all‐cause death and 12‐month freedom from limb‐related death, amputation, and reintervention. Primary patency was superior in the Lutonix DCB group compared with the conventional angioplasty group (65.2% versus 52.6%; P=0.02), and for the safety end points measured, there were no significant differences between devices. Continued benefits of patency and similar safety were reported with longer‐term follow‐up within the LEVANT 2 randomized trial and the Lutonix Global SFA (Superficial Femoral Artery) Registry.11 The initial IN.PACT SFA trial5 randomized 331 patients with similar clinical presentations as in LEVANT 2. The randomization was also 2:1, yielding a final sample size analyzed of 207 patients in the DCB group and 107 patients in the PTA group. The primary end point was primary patency, defined as freedom from restenosis or clinically driven target lesion revascularization at 12 months. The IN.PACT DCB had significantly higher 12‐month primary patency at 82.2% versus 52.4% for the standard PTA (P<0.001). Safety events were reported and numerically low at 12 months. There were 4 deaths in the DCB group attributed to cerebral infarction, biliary sepsis, sudden death, and a perforated colon. The Clinical Events Committee did not determine that any safety issues were device or procedure related. Longer‐term outcomes of the IN.PACT SFA trial to 2 years showed durable patency results of the DCB compared with the PTA.12 All‐cause mortality increased to 16 deaths in the DCB group and 1 death in the PTA group. The DCB deaths were again broad, affecting various organ systems, and were delayed in onset (median time, 565 days) relative to PTA (397 days). When followed out to 3 years, relative efficacy of the DCB was maintained; however, the mortality signal increased further, with 21 deaths in the DCB group and only 2 deaths in the PTA group.13 A more recent low‐dose paclitaxel DCB (Stellarex DCB; Spectranetics Corp, Colorado Springs, CO) gained FDA approval in 2017. The Stellarex DCB is coated with paclitaxel at 2 μg/mm2 and has a novel excipient, polyethylene glycol. The ILLUMENATE (Prospective, Randomized, Single‐Blind, U.S. Multi‐Center Study to Evaluate Treatment of Obstructive Superficial Femoral Artery or Popliteal Lesions With A Novel Paclitaxel‐Coated Percutaneous Angioplasty Balloon) pivotal trial6 randomized 300 symptomatic patients with femoropopliteal disease to DCB (n=200) or conventional angioplasty (n=100). Approximately half the patients enrolled were diabetics, were women, and had calcified disease. At 1 year, the primary patency by Kaplan‐Meier estimates was 82.3% (DCB) versus 70.9% (PTA) (P=0.002). The primary safety end point included freedom from device‐ or procedure‐related death through 30 days, target limb major amputation, and clinically driven target lesion revascularization through 12 months and was superior for DCB versus PTA (92.1% versus 83.2%; P=0.025). A pharmacokinetics evaluation within this study showed that detectable levels of circulating paclitaxel declined to low levels within the first hour of DCB deployment, from 54.4 to 1.4 ng/mL.6 All‐cause deaths were no different between groups: 5 of 192 patients (2.6%) in the DCB group and 2 of 96 patients (2.1%) in the PTA group. The 2‐year data from the ILLUMENATE European RCT14 likewise showed a sustained treatment effect and no statistical difference between all‐cause mortality: 13 of 199 patients (6.5%) in the DCB group and 3 of 59 patients (5.1%) in the PTA group. No deaths were adjudicated as being device or procedure related. The Katsanos Meta‐Analysis Given clustering of late mortality in several of the RCTs with long‐term follow‐up, Katsanos et al performed an important systematic review and meta‐analysis of the major paclitaxel‐coated trials with a focus on all‐cause mortality.8 The number of RCTs evaluating paclitaxel‐coated devices since the early FDA approval studies has grown steadily and involves use of the devices in various locations (above and below the knee) and in a range of clinical presentations, from intermittent claudication to critical limb ischemia. Several studies included in the meta‐analysis by Katsanos et al8 are multicenter, are global, and use PTA as the control arm. In total, 28 RCTs with 4663 patients were analyzed. A careful examination of the demographics of the total patient population included in the meta‐analysis is pertinent to review in the context of all‐cause mortality. The average age ranged from 67 to 76 years, two thirds of patients were men, and the prevalence of diabetes mellitus ranged from 21% to 77%. In addition, there was a high incidence of smoking, hypertension, and hyperlipidemia across all studies. All‐cause death was no different between paclitaxel‐coated devices and control at 1 year (2.3% versus 2.3% crude risk of death; risk ratio, 1.08; 95% CI, 0.72–1.61). At 2 years, the all‐cause risk of death became significant, with higher risk in the paclitaxel group (7.2% versus 3.8% crude risk of death; risk ratio, 1.68; 95% CI, 1.15–2.47). Finally, at 5 years, the all‐cause risk of death in paclitaxel‐treated patients was nearly doubled (14.7% versus 8.1% crude risk of death; risk ratio, 1.93; 95% CI, 1.27–2.93). This resulted in a number needed to harm of only 14 patients.8 Paclitaxel Biological Effects There are no obvious biological mechanisms that explain a direct link between paclitaxel and mortality. Paclitaxel is a known cytotoxic agent that inhibits smooth muscle cell proliferation and neointimal hyperplasia through binding of microtubules and prevention of tubulin disassembly. The arrest of mitosis reduces vascular restenosis when applied locally, an effect well demonstrated historically in the coronary vasculature. Paclitaxel doses on peripheral devices are an order of magnitude higher than what was used in relatively smaller coronary stents. Furthermore, when long lesions are involved in the femoropopliteal region, it is common for multiple paclitaxel‐coated devices to be used, increasing patient exposure and dose. Interestingly, Katsanos et al performed a meta‐regression of all‐cause death against paclitaxel exposure and found a 0.4±0.1% excess risk of death for every paclitaxel milligram‐year (95% CI, 0.1%–0.6%; P<0.001).8 Peripheral paclitaxel‐coated devices vary in terms of paclitaxel dose per surface area, and some use proprietary excipients to allow for effective drug transfer. The excipients themselves can be involved in hypersensitivity reactions, although the mortality signal was also seen in studies of devices without excipients. Peripheral paclitaxel devices use the crystalline form of the cytotoxic drug, which aids in tissue uptake and retention. Despite these features, drug transfer remains inefficient and ≈80% to 90% of paclitaxel is lost in the systemic circulation. Animal studies have shown evidence of distal embolization to downstream vessels. Histopathological analysis has confirmed crystalline paclitaxel remnants and fibrinoid necrosis, particularly with DCBs over DES.15, 16 Paclitaxel transferred to the vessel wall may cause positive remodeling, arterial wall dilation, and medial wall necrosis. The half‐life of paclitaxel is generally in the range of months. The peak plasma concentration occurs soon after the procedure and is thought to be below the level known to cause systemic adverse effects.17 For these reasons, one would hypothesize that any mortality linked directly to paclitaxel would occur sooner than the divergence in event rates seen at 2 years and beyond. The causes of deaths in the RCTs were also broad and included cardiovascular causes, infectious causes, pulmonary causes, and malignancy, among others. Establishing biological plausibility is difficult without a clear signal of one type of adverse event. Further mechanisms and markers of systemic inflammation should be considered. Considerations and Opportunities Although the pooled mortality signal is concerning given the statistical power afforded by a meta‐analysis, the overall results are subject to limitations of the imputed data from the individual trials. The individual trials were not primarily designed to look at safety, but rather efficacy. As a result, analyses were performed on the basis of the intention‐to‐treat principle. Factoring in crossovers and now focusing on safety (all‐cause mortality), it would be prudent to review and analyze the data when reclassified according to as‐treated groups. Another consideration is the number of patients who were lost to follow‐up. Even in the setting of RCTs, several studies had a noteworthy number of patients who were lost, impacting the numerator and denominator when recording safety event rates and calculating frequencies. In the 5‐year Zilver PTX data,9 for example, ≈20% and 17% of the as‐treated patients for DES and non‐DES therapies, respectively, were lost during the study period. In the 5‐year follow‐up of THUNDER (Local Taxan With Short Time Contact for Reduction of Restenosis in Distal Arteries),18 15% and 24% of patients in the paclitaxel‐coated balloon and control groups, respectively, were lost to follow‐up, yielding a small sample size for final analysis. These 2 studies were the only 5‐year follow‐up RCTs available for inclusion in the meta‐analysis. Although Katsanos and colleagues8 conducted meticulous and complex statistical models, there can be skewed and biased results within a meta‐analysis when the included studies have unbalanced groups, small numbers in the control arms, and low event rates, particularly with zero events or a single event in some groups. For hard end points, such as all‐cause mortality, efforts to complete follow‐up or use the social security death index database could assist in tracking accurate mortality rates. Clinical event committees, when established, did not deem any deaths as device or procedure related. Adjudication of death is, of course, limited by not always knowing potential underlying mechanisms of systemic paclitaxel toxicities. Analyzing these data at the patient level would offer additional insights, particularly when trying to draw conclusions about drug dose and mortality. Emerging Data New patient‐level data are emerging rapidly and will take time to completely synthesize and review in an unbiased manner. Secemsky et al evaluated 16 560 Centers for Medicare and Medicaid Services beneficiaries admitted for femoropopliteal interventions and reported no differences in all‐cause mortality between patients treated with drug‐coated versus non–drug‐coated devices.19 However, there were important differences between this data set and the meta‐analysis of Katsanos et al.8 The former were inpatients, with most having critical limb ischemia, and the median follow‐up time period was just over a year at 389 days (a time point at which even the meta‐analysis of Katsanos et al8 did not find a signal for mortality difference). Another Medicare analysis, by Long et al, with >83 000 all‐treated patients (inpatients, outpatients, patients with critical limb ischemia, and patients with claudication), showed that patients treated with drug‐coated devices had lower mortality and amputation risk compared with those treated with noncoated devices.20 Schneider et al performed an independent patient‐level meta‐analysis of 1980 patients treated with DCB (n=1837) and standard PTA (n=143) from 4 independently adjudicated prospective studies, with most patients coming from IN.PACT Global (n=1230), a real‐world, prospective, multicenter, single‐arm study in which patients were more likely to have long lesions, chronic total occlusions, and in‐stent restenosis.21 There was no difference in all‐cause mortality through 5 years (9.3% versus 11.2%; P=0.40). In addition, time to survival by paclitaxel dose tercile was performed and showed no statistically significant difference in all‐cause mortality between the 3 groups. This analysis is limited by the small number of control patients and variations in the setting and baseline characteristics of included patients. Summary and Future Investigations The FDA has released a “Letter to Health Care Providers”22 that recommends that providers use these devices only in the highest‐risk patients and ensure close postprocedure follow‐up. The recommendations also emphasize discussing risks and benefits with patients during the consent process. The concerns have already impacted clinical care and ongoing trials; the BASIL‐3 (Balloon Versus Stenting in Severe Ischemia of the Leg‐3) and SWEDEPAD (Swedish Drug‐elution Trial in Peripheral Arterial Disease) studies were immediately paused after the meta‐analysis publication of Katsanos et al.8 As confusion remains about the current safety of paclitaxel devices during lower‐extremity revascularization procedures, it is important to convene stakeholders for open discussions as new data emerge. The Vascular Leaders Forum and the DCB Safety Town Hall meetings are important steps. In the meantime, a reasonable approach toward patient care is to use caution and have straightforward conversations with patients. Perhaps the same as low as reasonably achievable concept used for radiation protection should be applied herein (essentially, to use the lowest dose of paclitaxel necessary to get an effective result). Simultaneously, research and development of nonpaclitaxel treatment strategies should continue. Sirolimus‐eluting balloons, for example, are under development. When currently faced with patients who we think will benefit from the technology (long lesions, chronic total occlusions, or in‐stent restenosis), we should use our best clinical judgement and be as transparent as possible with our patients, assessing each individual case's risk and benefit. Finally, the current example of drug‐coated devices highlights the shortcoming of the vascular medicine field, specifically on the lack of consistent longitudinal clinical outcome data (Figure). Many patients with vascular disease get numerous touchpoints by multiple providers without ongoing clinical data capture. We must learn how to study devices in peripheral arterial disease effectively, pragmatically, and with adequate follow‐up. This is the only path to ultimately making a safe decision for our patients. Figure 1 Paclitaxel‐coated devices as the focus of various types of clinical outcome data. Initial randomized controlled trials of these devices were focused on efficacy end points and not powered for safety events. A safety alert signal from a recent systematic review and meta‐analysis has led to analyses of larger registries and Centers for Medicare and Medicaid Services (CMS) data. Further studies at the patient level and with longer‐term follow‐up are pending. This highlights the future importance of capturing longitudinal clinical outcome data via a pragmatic approach. DCB indicates drug‐coated balloon; DES, drug‐eluting stent. Disclosures Swaminathan reports consulting for Medtronic, Inc; and receiving a research grant from ACIST Medical. Jones reports receiving a research grant from Medtronic, Inc. Patel reports receiving research grants from Medtronic, Inc, Bayer, Janssen, AstraZeneca, the National Heart, Lung, and Blood Institute, and Heartflow; and is on the advisory board for Bayer, Janssen, and Amgen.

          Related collections

          Most cited references20

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Risk of Death Following Application of Paclitaxel‐Coated Balloons and Stents in the Femoropopliteal Artery of the Leg: A Systematic Review and Meta‐Analysis of Randomized Controlled Trials

          Background Several randomized controlled trials (RCTs) have already shown that paclitaxel‐coated balloons and stents significantly reduce the rates of vessel restenosis and target lesion revascularization after lower extremity interventions. Methods and Results A systematic review and meta‐analysis of RCTs investigating paclitaxel‐coated devices in the femoral and/or popliteal arteries was performed. The primary safety measure was all‐cause patient death. Risk ratios and risk differences were pooled with a random effects model. In all, 28 RCTs with 4663 patients (89% intermittent claudication) were analyzed. All‐cause patient death at 1 year (28 RCTs with 4432 cases) was similar between paclitaxel‐coated devices and control arms (2.3% versus 2.3% crude risk of death; risk ratio, 1.08; 95% CI, 0.72–1.61). All‐cause death at 2 years (12 RCTs with 2316 cases) was significantly increased in the case of paclitaxel versus control (7.2% versus 3.8% crude risk of death; risk ratio, 1.68; 95% CI, 1.15–2.47; —number‐needed‐to‐harm, 29 patients [95% CI, 19–59]). All‐cause death up to 5 years (3 RCTs with 863 cases) increased further in the case of paclitaxel (14.7% versus 8.1% crude risk of death; risk ratio, 1.93; 95% CI, 1.27–2.93; —number‐needed‐to‐harm, 14 patients [95% CI, 9–32]). Meta‐regression showed a significant relationship between exposure to paclitaxel (dose‐time product) and absolute risk of death (0.4±0.1% excess risk of death per paclitaxel mg‐year; P<0.001). Trial sequential analysis excluded false‐positive findings with 99% certainty (2‐sided α, 1.0%). Conclusions There is increased risk of death following application of paclitaxel‐coated balloons and stents in the femoropopliteal artery of the lower limbs. Further investigations are urgently warranted. Clinical Trial Registration URL: www.crd.york.ac.uk/PROSPERO. Unique identifier: CRD42018099447.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Trial of a Paclitaxel-Coated Balloon for Femoropopliteal Artery Disease.

            The treatment of peripheral artery disease with percutaneous transluminal angioplasty is limited by the occurrence of vessel recoil and restenosis. Drug-coated angioplasty balloons deliver antiproliferative agents directly to the artery, potentially improving vessel patency by reducing restenosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Drug-Coated Balloon Versus Standard Percutaneous Transluminal Angioplasty for the Treatment of Superficial Femoral and Popliteal Peripheral Artery Disease

              Endovascular treatment of symptomatic atherosclerotic peripheral artery disease (PAD) has gained widespread acceptance and is now recommended as the primary revascularization strategy in many clinical and anatomic scenarios. 1–3 Percutaneous transluminal angioplasty (PTA) of the superficial femoral and popliteal artery has a high initial success rate, but restenosis occurs in up to 60% of cases. 4 Although randomized trials have demonstrated patency rates with bare metal stents and drug-eluting stents superior to those observed with PTA, 5–8 the optimal treatment for superficial femoral and popliteal artery disease remains controversial. Some practice guidelines advise against primary stenting in patients with intermittent claudication, 9 whereas others recommend primary stenting in short- or intermediate-length lesions 3 or in the event of acute PTA failure. 1–2 Despite the improved outcomes reported in some trials with stenting, the dynamic stresses applied by the superficial femoral and popliteal artery may result in stent fracture 10–11 or in-stent restenosis. 12 Given the limitations of stenting, there has been considerable interest in identifying the approaches that could improve patency without the need for a permanent metallic implant. Clinical Perspective on p 502 One approach to this challenge has been the development of the drug-coated balloon (DCB), which combines balloon dilatation with local delivery of an antiproliferative drug. Proof-of-concept evidence has demonstrated the utility of different DCB technologies in reducing both restenosis and the need for reintervention in comparison with PTA. 13–17 Promising primary patency and target lesion revascularization rates up to 2 years postimplantation have been reported. 18 However, robust evidence from large randomized, controlled trials is lacking. The IN.PACT SFA Trial was designed to test the safety and efficacy of the IN.PACT Admiral DCB for the treatment of patients with symptomatic PAD in the superficial femoral and proximal popliteal artery. Methods Study Design The IN.PACT SFA Trial is a multicenter, international, single-blinded, randomized, controlled trial to assess the safety and efficacy of the IN.PACT Admiral DCB (Medtronic Inc, Santa Rosa, CA) versus standard PTA balloons in patients with symptomatic superficial femoral and proximal popliteal artery disease. The trial was prospectively designed to be conducted in 2 phases: IN.PACT SFA I (conducted in Europe) and IN.PACT SFA II (conducted in the United States), which are jointly referred to as IN.PACT SFA. The IN.PACT SFA Trial was prospectively analyzed according to a single statistical analysis plan. The 2 phases occurred sequentially in time with enrollment completed in the IN.PACT SFA I phase before the initiation of the IN.PACT SFA II phase. Minor differences between the IN.PACT SFA I phase and the IN.PACT SFA II phase eligibility criteria exist and include subtle variations in concomitant inflow and contralateral limb treatment, along with differences in predilatation requirements. A prespecified poolability test for treatment-by-trial phase interaction was established, with planned data pooling across the 2 phases in the event that there was no significant treatment-by-trial interaction. Both protocols were approved by the institutional review boards or ethics committees at each trial site. All patients provided written informed consent before enrollment. Both trial phases were conducted in accordance with the Declaration of Helsinki, good clinical practice guidelines, and applicable laws as specified by all relevant governmental bodies. An independent clinical events committee adjudicated all major adverse events. Independent core laboratories analyzed all images, including duplex ultrasonography (VasCore, Massachusetts General Hospital, Boston, MA) and angiography (SynvaCor, Springfield, IL). Patient Population Patients were eligible for enrollment if they had moderate to severe intermittent claudication or ischemic rest pain (Rutherford 2–4) and stenosis of 70% to 99% with lesion lengths between 4 and 18 cm or occlusion with lengths of ≤10 cm involving the superficial femoral and proximal popliteal arteries, and met all other eligibility criteria. Randomization and Blinding Randomization occurred after successful crossing of the lesion in the IN.PACT SFA I phase and after successful crossing and predilatation with a standard PTA balloon 1 mm smaller than the reference vessel diameter in the IN.PACT SFA II phase. A patient was considered enrolled at the time of randomization. Subjects were randomly assigned by an Interactive Voice Response System with the use of a method of permuted blocks to ensure that a 2:1 ratio was maintained across sites (Figure 1). Figure 1. Trial flow diagram. The IN.PACT SFA Trial used a 2:1 randomized, control design, and intent-to-treat (ITT) analysis was conducted at 12 months. Three hundred thirty-one (331) patients with de novo or nonstented restenotic lesions in the superficial femoral and proximal popliteal artery were randomly assigned either to the IN.PACT Admiral drug-coated balloon or standard PTA treatment group. All subjects enrolled in the IN.PACT SFA Trial (n=331) will be followed for up to 5 years. Analysis at 1 year included subjects that provided end point data at the time of data snapshot. A subject was excluded under the following circumstances: (1) consent was withdrawn before the 1-year visit and no event had occurred before withdrawal or (2) there was no contact with the subject permitting a 1-year evaluation and no events had occurred before the 1-year evaluation. DCB indicates drug-coated balloon; PTA, percutaneous transluminal angioplasty; and SFA, superficial femoral artery. The patients and the trial sponsor were blinded to the treatment assignments through the completion of all 12-month follow-up evaluations. The independent core laboratories and clinical events committee will remain blinded to the treatment assignments throughout the 60-month follow-up duration. Because of the visual difference between the IN.PACT DCB and standard PTA balloon, treating physicians, research coordinators, and catheterization laboratory staff were not blinded to the treatment assignment. Treating physicians, research coordinators, and catheterization laboratory staff received detailed and specific instructions and training on how to preserve the patients’ blinded status. Treatment and Medical Therapy Patients randomly assigned to the experimental arm were treated with the IN.PACT Admiral DCB. The IN.PACT DCB has a dual mode of action, comprising mechanical dilatation by the angioplasty balloon plus local drug delivery to the arterial wall intended to inhibit restenosis. The IN.PACT DCB coating includes paclitaxel as the antiproliferative agent at a dose of 3.5 μg/mm2, with urea as the excipient. Available IN.PACT Admiral DCB sizes included 4-, 5-, 6-, and 7-mm diameters and 20-, 40-, 60-, 80 and 120-mm lengths (the 7-mm diameter device was not available in the 120-mm length). A minimum balloon inflation time of 180 seconds was required for both treatment groups. To avoid geographic miss, DCB length was chosen to exceed the target lesion length by 10 mm at the proximal and distal edges. The IN.PACT DCB is a single-inflation device, and, when treatment required multiple balloons, an overlap of 10 mm was applied for contiguous balloon inflations. Premedication included a loading dose of aspirin 300 to 325 mg and clopidogrel 300 mg within 24 hours of the index procedure or 2 hours postprocedure. Heparin was administered at the time of the procedure to maintain an activated clotting time ≥250 seconds. Postdilatation with a standard PTA balloon was allowed at the discretion of the operator. In both treatment groups, provisional stenting was allowed only in the case of PTA failure after repeated and prolonged PTA inflations. PTA failure was defined as a residual stenosis ≥50% or major (≥grade D) flow-limiting dissection confirmed by a peak translesional systolic pressure gradient of >10 mm Hg. In both arms, postprocedure medical therapy included aspirin 81 to 325 mg daily (for a minimum of 6 months) and clopidogrel 75 mg daily for a minimum duration of 1 month for nonstented patients and 3 months for patients who received stents. Usage of aspirin and antiplatelet drugs did not differ between treatment arms at discharge (97.6%), 30 days (87.6%), or 12 months (51.5%). Follow-Up For the primary end point analysis, patients were followed by the treating physician at 30 days, 6 months, and 12 months, including office visits with duplex ultrasonography functional testing and adverse event assessment. Reinterventions, if required within 12 months of the procedure, were performed according to standard practice by using PTA balloons and provisional stenting. Study End Points The primary efficacy end point was primary patency at 12 months following the index procedure, defined as freedom from clinically driven target lesion revascularization and restenosis as determined by a duplex ultrasonography–derived peak systolic velocity ratio of ≤2.4. 19 Each component of the primary efficacy end point was independently adjudicated by the blinded Clinical Events Committee (for clinically driven target lesion revascularization) or by the core laboratories (for restenosis). Specifically, the independent Clinical Events Committee determined whether reinterventions at the target lesion were clinically driven on the basis of objective testing (ankle-brachial index decrease ≥20% or >0.15 in comparison with postprocedure ankle-brachial index) or symptoms of exertional limb discomfort. Safety end points included 30-day device- and procedure-related death, all-cause death, major target limb amputation, and target vessel thrombosis. These events were site reported and Clinical Events Committee adjudicated. Additional efficacy end points included acute procedural success, target vessel revascularization at 12 months, and primary sustained clinical improvement (defined as freedom from target limb amputation, target vessel revascularization, and increase in Rutherford class at 12 months). Functional assessments included general appraisal through administration of a 5-dimension (EQ-5D) health-related quality-of-life questionnaire and specific evaluation of walking capacity by using a Walking Impairment Questionnaire. A Six-Minute Walk Test was additionally conducted in the IN.PACT SFA II phase only. Statistical Analysis The planned enrollment of 330 subjects provided a power of 80% to detect a 50% improvement in the primary end point at 12 months (from 40% 4 in the PTA group to 60% in the DCB group) with a 1-sided type I error of 2.5%. From its inception, the trial was intended to have 2 phases under a single statistical analysis plan. Poolability of subjects across trial phases for the primary end point analysis was tested by using Cox proportional hazards regression. For this poolability analysis, model covariates included treatment group, phase, and a treatment-by-phase interaction effect. Because the treatment-by-trial phase interaction value for the primary end point was nonsignificant (P=0.428), the 2 trial phases were pooled for all analyses. All analyses were based on the intention-to-treat principle. Continuous variables are described as mean±standard deviation and were compared by t tests. Categorical variables are described as proportions and were compared by the Z test owing to the 1-sided testing. The Z test was used to test the hypothesis of equality of proportions in achieving the primary end point. Multiple imputation was performed by using the logistic regression approach for patients with missing primary end point data (29 DCB, 7 PTA). The following variables were included in the imputation model as covariates: age, sex, diabetes mellitus, lesion length, total occlusion, and Rutherford class at baseline. Five data sets were imputed from these covariates that mimic different realizations of the missing data. Within each imputed data set for the end point, the proportion experiencing the end point was statistically compared between treatment groups by using the 2-sample Z test. From these, an overall test statistic for the end point and its associated P value were calculated for the imputed data. The imputed difference (95% confidence interval) and P value are reported along with the as-observed numerator and denominator. A sensitivity analysis of the as-observed rates revealed a similar highly significant P value (P 70% at 1 to 2 years, although recent results have suggested improved outcomes with peripheral drug-eluting stents 28 and DCB. 29,30 Use of a DCB (and avoidance of stent implantation) does not limit future treatment options, an important consideration given the chronic and progressive nature of PAD. These findings compare favorably with other randomized clinical trials in this patient population. Despite the inclusion of longer lesion lengths that are at a higher risk of treatment failure, the 2.4% target lesion revascularization rate experienced in this trial is the lowest reported for an SFA device trial at 12 months. Clinically driven target lesion revascularization rates of 12.7% and 9.5% were reported in 2 recent randomized trials of bare metal and drug-eluting stents, despite their inclusion of shorter lesions (average lesion lengths of 7.0 and 5.4 cm, respectively). 7,8 IN.PACT DCB was associated with a low complication rate, including the absence of major amputations and a low rate of vessel thrombosis. Study Limitations The trial was deliberately and prospectively conducted in 2 sequential phases. The blinding of phase I was rigorously maintained until the completion of phase II. When the data were analyzed, there were no statistical differences between the 2 phases. Although improvements in the functional assessments of quality of life, walking impairment, and walking distance were observed in both treatment groups, the interpretation of these measures is complicated by the subjective nature of patient questionnaires and the influence of comorbidities, including progressive disease in nontreated vessels. The results of this trial cannot be generalized to patients not included in this trial. Future studies should encompass longer lesions and consider comparison with bare metal stents, drug-eluting stents, and bypass, and optimal medical therapy and exercise, as well. Longer-term follow-up is needed to confirm the durability of the benefit. Conclusions In conclusion, in this large, prospective, multicenter, international, randomized trial, DCB was superior to PTA and had a favorable safety profile for the treatment of patients with symptomatic superficial femoral and proximal popliteal artery PAD. The IN.PACT DCB demonstrated impressive patency rates with low repeat revascularization rates in comparison with other modern endovascular therapies. DCB stands to become an important treatment option for patients with superficial femoral and popliteal artery disease. Acknowledgments We thank Judith Greengard, Victoria Rendon, and Melissa Hasenbank for editing assistance. Contributions: Drs Tepe, Jaff, and Laird prepared the first draft of this manuscript, which was then reviewed and edited by the other coauthors. Drs Snead and Cohen undertook the statistical analysis. All the authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; and also contributed to drafting the article or revising it critically for important intellectual content. Source of Funding The study was funded by Medtronic, Inc. Disclosures Dr Tepe holds research grants from Bard Peripheral Vascular, B. Braun, Biotronic, Covidien, Medrad, and Medtronic. He is a compensated advisory board member for Medtronic, and receives speaking honoraria from Bard Peripheral Vascular, Biotronic, Covidien, Medrad, and Medtronic. Dr Laird holds research grants from W.L. Gore and Medtronic. He is a compensated advisory board member and consultant for Abbott Vascular, Bard Peripheral Vascular, Boston Scientific, Covidien, and Medtronic. Dr Micari holds research grants from Medtronic, and is a compensated consultant for Medtronic. Dr Metzger receives speaking honoraria from Bard Peripheral Vascular and Medtronic and is compensated for participation in training courses sponsored by Abbott Vascular and Medtronic. He is a compensated consultant for Abbott Vascular. Dr Scheinert holds research grants from Abbott Vascular, Angioslide, Atheromed, Biotronik, Boston Scientific, Cook Medical, Cordis, Covidien, CR Bard, Gardia Medical, Heoteq, Intact Vascular, Medtronic, TriReme Medical, and Upstream Peripheral Technologies. He is a compensated consultant/advisory board member for Abbott Vascular, Angioslide, Atheromed, Biotronik, Boston Scientific, Cook Medical, Cordis, Covidien, CR Bard, Gardia Medical, Heoteq, Intact Vascular, Medtronic, TriReme Medical, and Upstream Peripheral Technologies. Dr Zeller holds research grants from Bard-Lutonix, Biotronik, Cook Medical, and Medtronic. He receives speaking honoraria from Bard-Lutonix, Biotronik, Cook Medical, and Medtronic. Dr Cohen holds research grants from Abbott Vascular, Boston Scientific, Covidien, and Medtronic and is a compensated consultant for Abbott Vascular and Medtronic. Dr Snead, B. Alexander, and M. Landini are full-time employees of Medtronic. Dr Jaff is a noncompensated Advisor for Medtronic and is the Medical Director of VasCore, the Vascular Ultrasound Core Laboratory. He is a compensated member of VIVA Physicians, a not-for-profit 501c3 education/research organization. The other authors report no conflicts.
                Bookmark

                Author and article information

                Contributors
                manesh.patel@duke.edu
                Journal
                J Am Heart Assoc
                J Am Heart Assoc
                10.1002/(ISSN)2047-9980
                JAH3
                ahaoa
                Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
                John Wiley and Sons Inc. (Hoboken )
                2047-9980
                16 May 2019
                21 May 2019
                : 8
                : 10 ( doiID: 10.1002/jah3.2019.8.issue-10 )
                : e012523
                Affiliations
                [ 1 ] Division of Cardiology Department of Medicine Duke University School of Medicine and the Duke Clinical Research Institute Durham NC
                Author notes
                [*] [* ] Correspondence to: Manesh R. Patel, MD, Duke University Medical, 2301 Erwin Rd, Hafs Bldg, Room 8695, Durham, NC 27710. E‐mail: manesh.patel@ 123456duke.edu
                Article
                JAH34095
                10.1161/JAHA.119.012523
                6585334
                31094266
                4cd8777d-26f4-448a-aaa2-c3953b8ee6b2
                © 2019 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 04 April 2019
                : 10 April 2019
                Page count
                Figures: 1, Tables: 0, Pages: 5, Words: 7990
                Categories
                Viewpoints
                Viewpoints
                Custom metadata
                2.0
                jah34095
                21 May 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.6.3 mode:remove_FC converted:23.05.2019

                Cardiovascular Medicine
                drug‐coated balloon,drug‐eluting stent,paclitaxel,mortality,peripheral vascular disease,restenosis

                Comments

                Comment on this article