Next Article in Journal
Machine Learning Analysis of the Anatomical Parameters of the Upper Airway Morphology: A Retrospective Study from Cone-Beam CT Examinations in a French Population
Previous Article in Journal
Impact of Sodium Zirconium Cyclosilicate Therapy on Nutrition Status in Patients with Hyperkalemia
Previous Article in Special Issue
Long-Term Outcomes following Common Femoral Endarterectomy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Drug-Coated Balloon versus Plain Balloon Angioplasty in the Treatment of Infrainguinal Vein Bypass Stenosis: A Systematic Review and Meta-Analysis

Department of Cardiocascular surgery, Kyorin University, 6-20-2, Shinkawa, Mitaka 181-8611, Tokyo, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(1), 87; https://doi.org/10.3390/jcm12010087
Submission received: 17 November 2022 / Revised: 10 December 2022 / Accepted: 19 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue Clinical Frontiers in Peripheral Artery Disease)

Abstract

:
The optimal endovascular therapy for vein graft stenosis (VGS) following infrainguinal arterial bypass is yet to be established. Drug-coated balloons (DCB) have rapidly improved the inferior patency outcomes of angioplasty using a conventional plain balloon (PB). This study compares the efficacy of DCBs and PBs for the treatment of infrainguinal VGS. This systematic review and meta-analysis was performed according to the PRISMA statement. Multiple electronic searches were conducted in consultation with a health science librarian in September 2022. Studies describing the comparative outcomes of angioplasty using DCBs and PBs in the treatment of infrainguinal VGS were eligible. Datasets from one randomized controlled trial and two cohort studies with a total of 179 patients were identified. The results indicated no significant difference in target lesion revascularization between DCBs and PBs (OR, 0.64; 95% CI, 0.32–1.28; p = 0.21), with no significant heterogeneity between studies. Additionally, differences in primary patency, assisted primary patency, secondary patency, and graft occlusion were not significant. Subgroup analysis showed similar effects for different DCB devices. In conclusion, DCBs showed no significant benefit in the treatment of VGS compared to PBs. Given the small population size of this meta-analysis, future trials with a larger population are desired.

1. Introduction

Infrainguinal arterial bypass (IAB) using an autologous vein graft is a well-established surgical technique that remains the gold standard for femoropopliteal arterial occlusive disease, particularly for patients with chronic limb-threatening ischemia [1]. In an era of endovascular therapy (EVT), IAB for long arterial occlusion extending to the crural arteries [2], advanced limb threat, or highly complex anatomy resistant to EVT [3] are the best revascularization methods.
Autologous IAB, typically using the great saphenous vein, showed higher longevity (60–80% of five-year patency rates [2,4,5,6]) than IAB with prosthetic grafts. Graft surveillance after autologous IAB occasionally reveals vein graft stenosis (VGS) mostly within two years after the bypass, potentially causing graft occlusion. Regular surveillance and intervention for VGS are essential to further improve long-term graft patency [7,8]. Therapeutic interventions for VGS are classified as surgical and endovascular treatments. Although the efficacy of the former is well established [9,10], the latter is increasingly reported and can be performed less invasively and repeatedly [11]. Angioplasty using balloons is more beneficial than stenting in the absence of a permanent metallic scaffold [12].
Drug-coated balloons (DCB), mostly paclitaxel-coated balloons, are increasingly used in endovascular therapies, improving the inferior patency outcomes of angioplasty with conventional plain balloons (PB). Neointimal hyperplasia (NIH), caused by vascular smooth muscle cell (VSMC) proliferation, is the main etiology of vein graft failure in the intermediate period [13]. Several studies indicate that paclitaxel, a highly lipophilic antineoplastic drug, inhibits the proliferation of VSMC after administration to cell culture and swine tissues for a few minutes [14]. DCBs are valuable in many vascular diseases, including coronary artery disease [15], arteriovenous fistula (AVF) as blood access [16,17], and superficial femoral artery disease [18]. However, the superiority of DCB in the treatment of VGS after IAB remains unclear. The objective of this systematic review and meta-analysis is to assess the efficacy of DCBs compared with PBs in the treatment of infrainguinal vein bypass stenosis.

2. Materials and Methods

This systematic review and meta-analysis was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The registration number in PROSPERO is CRD42022352017.

2.1. Search Strategy

A comprehensive search of MEDLINE and the Cochrane Library in consultation with a health science librarian (Y.S.) was conducted electronically from 16–26 September 2022. The complete details of the search strategy are provided in the Supplementary Materials. A combination of search words, including bypass graft, drug-coated balloon, drug-eluting balloon, paclitaxel-coated balloon, peripheral arterial disease, femoropopliteal artery disease, and infrainguinal artery disease, were used to probe for eligible studies.

2.2. Study Eligibility and Risk of Bias Assessment

Two independent authors (T.I. (Toshihiko Isaji) and Y.H.) reviewed the search results and selected studies. No publication dates or language restrictions were included. Randomized controlled trials (RCTs) and observational studies describing the comparative outcomes of percutaneous transluminal angioplasty (PTA) using drug-coated balloons and plain balloons in the treatment of infrainguinal VGS were considered for inclusion. Studies that focused on balloon angioplasty for the treatment of native arteries were excluded. Case reports were excluded from the study. Outcomes of target lesion revascularization (TLR) and graft patency during routine follow-up after PTA for vein graft stenosis had to be reported. Full-text articles were reviewed for eligibility (T.I. (Toshihiko Isaji) and Y.H.). The Risk of Bias Assessment Tool for Nonrandomized Studies (RoBANS) was used to assess the risk of bias [19]. Two authors (T.I. (Toshihiko Isaji) and Y.H.) independently assessed the RoB.

2.3. Data Extraction and Synthesis

Original data were extracted according to the PRISMA checklist. Two independent authors (T.I. (Toshihiko Isaji) and Y.H.) reviewed the eligible data for analysis. Patient demographics, operative characteristics, TLR, primary patency (PP), assisted primary patency (APP), secondary patency (SP), and graft occlusion were obtained from each study. For the systematic review, outcomes were extracted to estimate the odds ratios (OR). If the study reported outcomes using only the Kaplan–Meier curve, the numerical results at certain time points were considered as previously reported [20].

2.4. Study Outcomes

The outcomes were assessed in accordance with the reporting standards of the Society for Vascular Surgery [21]. To assess the validity of drug-coated balloons, TLR and graft occlusion in each study were incorporated. Additionally, PP, APP, and SP at one year were assessed. TLR was defined as any revascularization of the same stenotic lesion. PP was expressed as uninterrupted patency after revascularization without additional procedures. APP was defined as the patency achieved with any additional intervention to prevent bypass occlusion. SP was described as the patency achieved with any additional intervention to restore patency after occlusion. A subgroup analysis was performed to assess the outcome differences between the DCB devices.

2.5. Statistical Analysis

RevMan version 5.4.1 (Nordic Cochrane Centre, the Cochrane Collaboration, 2020, Copenhagen, Denmark) software was used to incorporate the pooled rates and ORs from each study with 95% CI using the random effects model. Any results with an I2 > 50% or the p value for heterogeneity < 0.05 were considered to represent significant heterogeneity between the studies. A random-effects model, which accounts for the variance between studies, was used to analyze the pooled data. Publication bias was assessed using funnel plot asymmetry. A leave-one-out sensitivity analysis was conducted with the exclusion of one study to confirm that our findings were not driven by any single study.

3. Results

3.1. Search Results

We initially screened 149 articles (131 from MEDLINE (Washington, DC, USA.) and 18 from the Cochrane Library (London, United Kingdom)). After excluding five duplicate records and 134 records for other reasons, 107 records of coronary artery disease, 10 records of other endovascular therapies, 2 records comparing bypass surgery and angioplasty for VGS, 11 literature reviews, and 4 case reports, 10 articles were screened for their abstracts. Three irrelevant articles and one article that assessed identical data as other papers were excluded. Six articles were screened for full text. We identified three titles, including one RCT and two cohort studies, with a total of 179 patients meeting the inclusion criteria [22,23,24], after excluding the following three papers. The first study compared the outcomes of angioplasty for both failing peripheral arterial vein grafts and synthetic bypass grafts; the data of the vein graft group could not be extracted [25]. The second study was a single-arm study of drug-coated balloon treatment for infrainguinal bypass graft stenosis, including both vein grafts and prosthetic grafts [26]. The third study showed severe bias in the selection of participants. Each patient belonged to both the drug-coated balloon group and the plain balloon group simultaneously, as only restenosis or occlusion in vein grafts that had been previously treated with plain balloons was indicated for drug-coated balloon treatment [27]. The PRISMA flow diagram for systematic reviews is depicted in Figure 1.

3.2. Study Characteristics

Of the 179 patients, 92 (51%) were treated with DCBs, and 87 (49%) were treated with PBs. The baseline characteristics of the three studies are shown in Table 1. The treatment characteristics of angioplasty for vein graft stenosis are presented in Table 2. The follow-up methods and indications for the intervention are depicted in Table 3. In all studies, duplex ultrasound was performed when symptomatic or regularly after arterial bypass, and angioplasty was considered for graft stenosis that was defined by a peak systolic velocity or peak systolic velocity ratio on duplex ultrasound. All DCBs used in the studies were coated with paclitaxel at a dose of 3.0 µg/mm2 [21] or 3.5 µg/mm2 [22,24] on the balloon surface. The balloon diameters of the DCBs and PBs were similar. The treatment location was described in two studies [22,23]. As shown in Figure 2, the RoBANS assessment demonstrated a high risk of bias regarding the participant selection (33%) and confounding variables (33%).

3.3. Outcomes

Two studies reported TLR at one year. TLR during the whole study period was reported in one study, of which the mean follow-up was 2.5 years [22]. The difference in TLR between DCBs and PBs was not significant (OR, 0.64; 95% CI, 0.32–1.28; p = 0.21) (Figure 3A), with no significant heterogeneity between studies (I2 = 0%; p = 0.94). Two studies reported the outcomes of PP at one year. The difference in PP between DCBs and PBs was not significant (OR, 1.26; 95% CI, 0.54–2.94; p = 0.59; I2 = 0%) (Figure 3B), and there was no significant heterogeneity between studies (I2 = 0%; p = 0.42). Three studies reported the outcomes of APP at one year. The difference in APP between DCBs and PBs was not significant (OR, 1.55; 95% CI, 0.55–4.34; p = 0.40) (Figure 3C), without significant heterogeneity between studies (I2 = 0%; p = 0.74). Three studies reported the outcomes of SP at one year. The difference in SP between DCBs and PBs was not significant (OR, 1.45; 95% CI, 0.44–4.79; p = 0.54) (Figure 3D), with no significant heterogeneity between studies (I2 = 0%; p = 0.46). Three studies reported the outcomes of graft occlusion. The difference in graft occlusion between DCBs and PBs was not significant (OR, 0.65; 95% CI, 0.25–1.68; p = 0.38) (Figure 3E), and there was no significant heterogeneity between studies (I2 = 0%; p = 0.56). Subgroup analysis classifying the studies according to the DCB devices showed similar effects of DCB in studies using InPact and Passeo-19 Lux at one-year assisted primary patency. Funnel plots revealed no evidence of publication bias (Figure 4).

4. Discussion

The results of the systematic review indicate that DCBs are not significantly superior to PBs in reducing the occurrence of TLR in the treatment of infrainguinal VGS, and no significant differences were observed in the graft patency of PP, APP, and SP at one year and graft occlusion. Additionally, the one-year APP was not significantly different between the two DCB devices, and no significant heterogeneity was observed in any of these outcomes.
No meta-analysis or multicenter trial comparing the outcomes of DCB and PB in the endovascular treatment of infrainguinal vein bypass stenosis has been reported previously as far as we know. Two cohort studies and one randomized controlled study were included in this review. In the RoBANS assessment, one study was considered to have a high risk of participant selection bias. In this study, consecutive patients with VGS and treated with DCBs were prospectively enrolled after the treatment regimen of VGS had changed from PBs. Data from patients treated with PBs before the treatment regimen change were retrospectively collected [23]. The other study presented a high risk of bias in confounding variables because significantly more patients presented Rutherford category 2 ischemia in the PB group than in the DCB group [22].
In contrast to this meta-analysis, previously reported studies concerning the EVT of other vascular diseases showed the benefit of DCBs in preventing the restenosis of target lesions compared to PBs [15,16,17,18]. Among them, AVF has similar aspects to vein grafts because both require venous adaptation to the arterial environment to achieve a long-term duration. Venous adaptation integrates outward remodeling and wall thickening. Vein grafts adapt mainly via increased wall thickening with less outward dilation, whereas AVF adapts mainly through outward dilation and less intimal thickening [28]. Venous wall thickening leading to stenosis in both AVF and vein grafts is mainly caused by NIH in the venous wall, which typically occurs within the first year, which was consistent with the observational periods of this meta-analysis. DCB inhibits the proliferation of VSMC after intimal injury by delivering an antiproliferative drug with an excipient substance that promotes drug transfer to the vessel wall [29]. However, progressive NIH in vein graft stenosis may reduce the effect of paclitaxel on DCB, which inhibits VSMC proliferation.
In addition, the mechanisms of vein graft failure appear more complex than those of VSMC proliferation [30,31]. PREVENT trials have been performed to overcome vein graft failure by regulating VSMC proliferation. E2F decoy, which blocks the activation of genes mediating cell cycle progression and intimal hyperplasia, successfully inhibited VSMC proliferation in a rat carotid injury model [32]. However, the PREVENT-III study showed no predominance of the patient group treated with the E2F decoy in time to graft reintervention or major amputation, suggesting that strategies inhibiting VSMC growth do not prevent vein graft stenosis. Furthermore, technical errors in anastomoses, graft routes, or intimal injuries during graft harvesting can cause VGS [33].
Drug-eluting stents (DES), including paclitaxel-eluting stents, are reportedly associated with a small increase in stent thrombosis after percutaneous coronary interventions compared with bare-metal stents [34]. Double antiplatelet therapy (DAPT) after DES placement or DCB angioplasty has been widely accepted. The studies included in this meta-analysis did not strictly follow the DAPT regimen; instead, they adopted different regimens of postprocedural antithrombotic medication. This may have prevented DCB angioplasty from achieving superior results in terms of inhibiting restenosis and repeat angioplasty.
Hence, we need to further explore the treatment options to improve the outcomes of VGS. DES therapy for the treatment of infrainguinal VGS has better primary patency than cutting balloon and plain balloon angioplasty [35]. However, external forces on stents due to the anatomic condition of vein grafts, such as subcutaneous tunneled routes and crossing of joint space, may cause stent fracture. In contrast, the advantage of DCB therapy is that it does not require a permanent metallic implant, thus preserving the subsequent treatment options. As such, VGS lesions suitable for DES therapy may be limited. Further studies comparing DESs and DCBs in the treatment of VGS are required to assess the benefit of non-metallic implants. In addition, circulating biomarkers as predictors of postinterventional restenosis for VGS could help optimize the use of DCBs. For instance, proinflammatory markers and high plasma levels of micro-RNA have been reported to potentially influence the outcomes after peripheral stenting [36].
This meta-analysis had several limitations. First, except for one RCT, two of the three included studies were retrospective cohort studies, leading to a low level of evidence. Second, each study was underpowered by sample size. Therefore, a lack of statistically significant difference does not mean no difference between the two groups. As EVT therapy is first recommended for most lesions of infrainguinal artery diseases, it is difficult to increase the entry of VGS cases after IAB in additional studies. Third, the treatment methods of VGS after IAB intrinsically include complexity and heterogeneity between studies; especially with regard to IAB procedures such as anastomosis fashion and grafting route as well as EVT procedures such as the predilatation method. As mentioned above, the included studies adopted different regimens of post-procedural antithrombotic medication.

5. Conclusions

Comparing DCB with PB angioplasty in the treatment of VGS, a meta-analysis showed no significant difference in outcomes with regard to TLR and graft patency at one year. No significant heterogeneity between the studies was observed. Given the small population of this meta-analysis and the ever-evolving nature of endovascular techniques and devices, future trials with a large population from multiple institutions are required to change or reinforce the results.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12010087/s1.

Author Contributions

Conceptualization, T.I. (Toshihiko Isaji) and Y.H.; methodology, T.I. (Toshihiko Isaji) and Y.H.; software, T.I. (Toshihiko Isaji); validation, T.I. (Toshihiko Isaji), Y.H., K.K., Y.I., K.F., T.I. (Toru Ikezoe), M.N. and H.K.; formal analysis, T.I. (Toshihiko Isaji) and Y.H.; investigation, T.I. (Toshihiko Isaji); resources, T.I. (Toshihiko Isaji); data curation, T.I. (Toshihiko Isaji); writing—original draft preparation, T.I. (Toshihiko Isaji) and Y.H.; writing—review and editing, K.K., Y.I., K.F. and T.I. (Toru Ikezoe); visualization, T.I. (Toshihiko Isaji); supervision, M.N. and H.K.; project administration, T.I. (Toshihiko Isaji). All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Informed Consent Statement

Patient consent was waived because this study was a systematic review and meta-analysis of published data.

Acknowledgments

We thank Yukari Shimizu (Kyorin University Library) for her cooperation with this systematic review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Farber, A.; Menard, M.T.; Conte, M.S.; Kaufman, J.A.; Powell, R.J.; Choudhry, N.K.; Hamza, T.H.; Assmann, S.F.; Creager, M.A.; Cziraky, M.J.; et al. Surgery or Endovascular Therapy for Chronic Limb-Threatening Ischemia. N. Engl. J. Med. 2022. [Google Scholar] [CrossRef] [PubMed]
  2. Norgren, L.; Hiatt, W.R.; Dormandy, J.A.; Nehler, M.R.; Harris, K.A.; Fowkes, F.G.; Rutherford, R.B.; TASC II Working Group. Inter-Society Consensus for the management of peripheral arterial disease. Int. Angiol. 2007, 26, 82–157. [Google Scholar]
  3. Conte, M.S.; Bradbury, A.W.; Kolh, P.; White, J.V.; Dick, F.; Fitridge, R.; Mills, J.L.; Suresh, K.R.; Murad, M.H.; GVG Writing Group. Global vascular guidelines on the management of chronic limb-threatening ischemia. J. Vasc. Surg. 2019, 69, 3S–125S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Klinkert, P.; Post, P.N.; Breslau, P.J.; van Bockel, J.H. Saphenous Vein Versus PTFE for Above-Knee Femoropopliteal Bypass. A Review of the Literature. Eur. J. Vasc. Endovasc. Surg. 2004, 27, 4357–4362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mii, S.; Tanaka, K.; Kyuragi, R.; Kyuragi, R.; Kuma, S.; Kodama, A.; Fukunaga, R.; Masaki, I.; Okazaki, J.; Eguchi, D.; et al. Raison d’etre of Tibial Artery Bypass for Intermittent Claudication in the Era of Endovascular Therapy. Circ. J. 2016, 80, 1460–1469. [Google Scholar] [CrossRef] [Green Version]
  6. Londrey, G.L.; Ramsey, D.E.; Hodgson, K.J.; Barkmeier, L.D.; Sumner, D.S. Infrapopliteal bypass for severe ischemia: Comparison of autogenous vein, composite, and prosthetic grafts. J. Vasc. Surg. 1991, 13, 631–636. [Google Scholar] [CrossRef] [Green Version]
  7. Mills Sr, J.L.; Wixon, C.L.; James, D.C.; Devine, J.; Westerband, A.; Hughes, J.D. The natural history of intermediate and critical vein graft stenosis: Recommendations for continued surveillance or repair. J. Vasc. Surg. 2001, 33, 273–278, discussion 278–280. [Google Scholar] [CrossRef]
  8. Humphries, M.D.; Pevec, W.C.; Laird, J.R.; Yeo, K.K.; Hedayati, N.; Dawson, D.L. Early duplex scanning after infrainguinal endovascular therapy. J. Vasc. Surg. 2011, 53, 353–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Nguyen, L.L.; Conte, M.S.; Menard, M.T.; Gravereaux, E.C.; Chew, D.K.; Donaldson, M.C.; Whittemore, A.D.; Belkin, M. Infrainguinal vein bypass graft revision: Factors affecting long-term outcome. J. Vasc. Surg. 2004, 40, 916–923. [Google Scholar] [CrossRef] [Green Version]
  10. Berceli, S.A.; Hevelone, N.D.; Lipsitz, S.R.; Bandyk, D.F.; Clowes, A.W.; Moneta, G.L.; Conte, M.S. Surgical and endovascular revision of infrainguinal vein bypass grafts: Analysis of midterm outcomes from the PREVENT III trial. J. Vasc. Surg. 2007, 46, 1173–1179. [Google Scholar] [CrossRef] [Green Version]
  11. Alexander, J.Q.; Katz, S.G. The Efficacy of Percutaneous Transluminal Angioplasty in the Treatment of Infrainguinal Vein Bypass Graft Stenosis. Arch. Surg. 2003, 138, 510–513. [Google Scholar] [CrossRef] [PubMed]
  12. Soga, Y.; Iida, O.; Fujihara, M.; Kawasaki, D.; Saito, S.; Urasawa, K.; Yokoi, H.; Fernandez, E.J.; Guo, J.; Nakamura, M.; et al. Real-World Clinical Outcomes of IN.PACT Admiral Drug-Coated Balloon for Femoropopliteal Artery Disease—12-Month Results From Japan Post-Market Surveillance Study. Circ. J. 2021, 85, 2149–2156. [Google Scholar] [CrossRef] [PubMed]
  13. Isaji, T.; Hashimoto, T.; Yamamoto, K.; Santana, J.M.; Yatsula, B.; Hu, H.; Bai, H.; Jianming, G.; Kudze, T.; Nishibe, T.; et al. Improving the Outcome of Vein Grafts: Should Vascular Surgeons Turn Veins into Arteries? Ann. Vasc. Dis. 2017, 10, 8–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tepe, G.; Zeller, T.; Albrecht, T.; Heller, S.; Schwarzwälder, U.; Beregi, J.P.; Claussen, C.D.; Oldenburg, A.; Scheller, B.; Speck, U. Local Delivery of Paclitaxel to Inhibit Restenosis during Angioplasty of the Leg. N. Engl. J. Med. 2008, 358, 689–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Scheller, B.; Hehrlein, C.; Bocksch, W.; Rutsch, W.; Haghi, D.; Dietz, U.; Böhm, M.; Speck, U. Treatment of Coronary In-Stent Restenosis with a Paclitaxel-Coated Balloon Catheter. N. Engl. J. Med. 2006, 355, 2113–2124. [Google Scholar] [CrossRef]
  16. Trerotola, S.O.; Lawson, J.; Roy-Chaudhury, P.; Saad, T.F.; Lutonix, A.V. Clinical Trial Investigators. Drug Coated Balloon Angioplasty in Failing AV Fistulas: A Randomized Controlled Trial. Clin. J. Am. Soc. Nephrol. 2018, 13, 1215–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Han, A.; Park, T.; Kim, H.J.; Min, S.; Ha, J.; Min, S.K. Editor’s Choice-Paclitaxel Coated Balloon Angioplasty vs. Plain Balloon Angioplasty for Haemodialysis Arteriovenous Access Stenosis: A Systematic Review and a Time to Event Meta-Analysis of Randomised Controlled Trials. Eur. J. Vasc. Endovasc. Surg. 2021, 62, 597–609. [Google Scholar] [CrossRef]
  18. Giacoppo, D.; Cassese, S.; Harada, Y.; Colleran, R.; Michel, J.; Fusaro, M.; Kastrati, A.; Byrne, R.A. Drug-Coated Balloon Versus Plain Balloon Angioplasty for the Treatment of Femoropopliteal Artery Disease. JACC Cardiovasc. Interv. 2016, 9, 1731–1742. [Google Scholar] [CrossRef]
  19. Kim, S.Y.; Park, J.E.; Lee, Y.J.; Seo, H.J.; Sheen, S.S.; Hahn, S.; Jang, B.H.; Son, H.J. Testing a tool for assessing the risk of bias for nonrandomized studies showed moderate reliability and promising validity. J. Clin. Epidemiol. 2013, 66, 408–414. [Google Scholar] [CrossRef]
  20. Kim, S.Y.; Park, J.E.; Lee, Y.J.; Seo, H.J.; Sheen, S.S.; Hahn, S.; Jang, B.H.; Son, H.J. Patency Rates of the Arteriovenous Fistula for Hemodialysis: A Systematic Review and Meta-analysis. Am. J. Kidney Dis. 2014, 63, 464–478. [Google Scholar] [CrossRef]
  21. Stoner, M.C.; Calligaro, K.D.; Chaer, R.A.; Dietzek, A.M.; Farber, A.; Guzman, R.J.; Hamdan, A.D.; Landry, G.J.; Yamaguchi, D.J.; Society for Vascular Surgery. Reporting standards of the Society for Vascular Surgery for endovascular treatment of chronic lower extremity peripheral artery disease. J. Vasc. Surg. 2016, 64, e1–e21. [Google Scholar] [CrossRef] [PubMed]
  22. Linni, K.; Ugurluoglu, A.; Aspalter, M.; Hitzl, W.; Hölzenbein, T. Paclitaxel-coated versus plain balloon angioplasty in the treatment of infrainguinal vein bypass stenosis. J. Vasc. Surg. 2016, 63, 391–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jongsma, H.; Akkersdijk, G.P.; de Smet, A.A.E.A.; Vroegindeweij, D.; de Vries, J.P.M.; Fioole, B. Drug-eluting balloons and uncoated balloons perform equally to rescue infrainguinal autologous bypasses at risk. J. Vasc. Surg. 2017, 66, 454–460. [Google Scholar] [CrossRef]
  24. Björkman, P.; Kokkonen, T.; Albäck, A.; Venermo, M. Drug-Coated versus Plain Balloon Angioplasty in Bypass Vein Grafts (the DRECOREST I-Study). Ann. Vasc. Surg. 2019, 55, 36–44. [Google Scholar] [CrossRef] [PubMed]
  25. Kitrou, P.; Parthipun, A.; Diamantopoulos, A.; Padayachee, S.; Karunanithy, N.; Ahmed, I.; Zayed, H.; Katsanos, K. Paclitaxel-coated balloons for failing peripheral bypass grafts: The BYPACS study. J. Cardiovasc. Surg. 2014, 55, 217–224. [Google Scholar]
  26. San Norberto, E.M.; Taylor, J.H.; Carrera, S.; Vaquero, C. Percutaneous Transluminal Angioplasty With Drug-Eluting Balloons for Salvage of Infrainguinal Bypass Grafts. J. Endovasc. Ther. 2014, 21, 12–21. [Google Scholar] [CrossRef] [PubMed]
  27. Björkman, P.; Peltola, E.; Albäck, A.; Venermo, M. Peripheral Vascular Restenosis: A Retrospective Study on the Use of Drug-Eluting Balloons in Native Arteries, Vein Grafts and Dialysis Accesses. Scand. J. Surg. 2017, 106, 158–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lu, D.Y.; Chen, E.Y.; Wong, D.J.; Yamamoto, K.; Protack, C.D.; Williams, W.T.; Assi, R.; Hall, M.R.; Sadaghianloo, N.; Dardik, A. Vein graft adaptation and fistula maturation in the arterial environment. J. Surg. Res. 2014, 188, 162–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. A Byrne, R.A.; Joner, M.; Alfonso, F.; Kastrati, A. Drug-coated balloon therapy in coronary and peripheral artery disease. Nat. Rev. Cardiol. 2014, 11, 1. [Google Scholar] [CrossRef]
  30. Conte, M.S.; Bandyk, D.F.; Clowes, A.W.; Moneta, G.L.; Seely, L.; Lorenz, T.J.; Namini, H.; Hamdan, A.D.; Roddy, S.P.; Belkin, M. Results of PREVENT III: A multicenter, randomized trial of edifoligide for the prevention of vein graft failure in lower extremity bypass surgery. J. Vasc. Surg. 2006, 43, 742–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Alexander, J.H.; Hafley, G.; Harrington, R.A.; Peterson, E.D.; Ferguson, T.B., Jr.; Lorenz, T.J.; Goyal, A.; Gibson, M.; Mack, M.J.; Gennevois, D.; et al. Efficacy and Safety of Edifoligide, an E2F Transcription Factor Decoy, for Prevention of Vein Graft Failure Following Coronary Artery Bypass Graft Surgery: PREVENT IV: A Randomized Controlled Trial. JAMA 2005, 294, 2446–2454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Morishita, R.; Gibbons, G.H.; Horiuchi, M.; Ellison, K.E.; Nakama, M.; Zhang, L.; Kaneda, Y.; Ogihara, T.; Dzau, V.J. A gene therapy strategy using a transcription factor decoy of the E2F binding site inhibits smooth muscle proliferation in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 5855–5859. [Google Scholar] [CrossRef] [PubMed]
  33. Szilagyi, D.E.; Elliott, J.P.; Hageman, J.H.; Smith, R.F.; Dall’olmo, C.A. Biologic Fate of Autogenous Vein lmplants as Arterial Substitutes: Clinical, Angiographic and Histopathologic Observations in Femoro-Popliteal Operations for Atherosc erosis. Ann. Surg. 1973, 178, 232–246. [Google Scholar] [CrossRef]
  34. Mohammad, R.A.; Goldberg, T.; Dorsch, M.P.; Cheng, J.W. Antiplatelet Therapy after Placement of a Drug-Eluting Stent: A Review of Efficacy and Safety Studies. Clin. Ther. 2010, 32, 2265–2281. [Google Scholar] [CrossRef] [PubMed]
  35. Hicks, C.W.; Canner, J.K.; Lum, Y.W.; Perler, B.A.; Black, J.H., 3rd; Abularrage, C.J. Drug-eluting stents are associated with improved outcomes for the treatment of infrainguinal bypass graft stenosis. J. Vasc. Surg. 2019, 69, 875–882. [Google Scholar] [CrossRef]
  36. Montelione, N.; Catanese, V.; Nenna, A.; Jawabra, M.; Verghi, E.; Loreni, F.; Nappi, F.; Lusini, M.; Mastroianni, C.; Jiritano, F.; et al. The Diagnostic Value of Circulating Biomarkers and Role of Drug-Coated Balloons for In-Stent Restenosis in Patients with Peripheral Arterial Disease. Diagnostics 2022, 12, 2207. [Google Scholar] [CrossRef]
Figure 1. Preferred reporting items for systematic reviews and meta-analyses diagram for article selection.
Figure 1. Preferred reporting items for systematic reviews and meta-analyses diagram for article selection.
Jcm 12 00087 g001
Figure 2. Risk of Bias Assessment Tool for Nonrandomized Studies (RoBANS) [22,23,24]. + in green circle meants “High”. − in red circle means “Low”.
Figure 2. Risk of Bias Assessment Tool for Nonrandomized Studies (RoBANS) [22,23,24]. + in green circle meants “High”. − in red circle means “Low”.
Jcm 12 00087 g002
Figure 3. Forest plots of the meta-analysis results of drug-coated balloons (DCB) vs. plain balloons (PB). Angioplasty in studies. (A). Target lesion vascularization. (B). One-year primary patency. (C). One-year assisted primary patency. (D). One-year secondary patency. (E). Graft occlusion. (F,G). Subgroup analysis of one-year assisted primary patency according to DCB device. (F). In.Pact. (G). Passeo-18 Lux. M-H, Mantel-Haenszel; CI, confidence interval; I2, study heterogeneity; Z, Z-score test statistic [22,23,24].
Figure 3. Forest plots of the meta-analysis results of drug-coated balloons (DCB) vs. plain balloons (PB). Angioplasty in studies. (A). Target lesion vascularization. (B). One-year primary patency. (C). One-year assisted primary patency. (D). One-year secondary patency. (E). Graft occlusion. (F,G). Subgroup analysis of one-year assisted primary patency according to DCB device. (F). In.Pact. (G). Passeo-18 Lux. M-H, Mantel-Haenszel; CI, confidence interval; I2, study heterogeneity; Z, Z-score test statistic [22,23,24].
Jcm 12 00087 g003aJcm 12 00087 g003b
Figure 4. Funnel plots of the meta-analysis results of drug-coated balloons (DCB) vs. plain balloons (PB) angioplasty in studies. (A). Target lesion vascularization. (B). One-year primary patency. (C). One-year assisted primary patency. (D). One-year secondary patency. (E). Graft occlusion.
Figure 4. Funnel plots of the meta-analysis results of drug-coated balloons (DCB) vs. plain balloons (PB) angioplasty in studies. (A). Target lesion vascularization. (B). One-year primary patency. (C). One-year assisted primary patency. (D). One-year secondary patency. (E). Graft occlusion.
Jcm 12 00087 g004
Table 1. Baseline characteristics.
Table 1. Baseline characteristics.
First Author, YearLinni,
2016 [22]
Jongsma,
2017 [23]
Björkman,
2019 [24]
CountryAustriaNetherlandsFinland
Study designCTCTRCT
Patients (No.) (total)42 vs. 41 (83)21 vs. 18 (39)29 vs. 28 (57)
Mean age (years)70 vs. 7170 vs. 7170 vs. 72
Male (%)21 vs. 3962 vs. 3961 vs. 52
DM (%)40 vs. 4629 vs. 2838 vs 61 *
Smokers (%)36 vs. 3481 vs. 7859 vs. 50
HTN (%)95 vs. 9557 vs. 8379 vs. 71
DL (%)55 vs. 6352 vs. 7890 vs. 96
CKD (%) NA10 vs. 021 vs. 14
CAD (%)55 vs. 5648 vs. 3948 vs. 39
CLTI before PTA (%)26 vs. 4210 vs. 1755 vs. 36
pre ABI0.57 vs. 0.51NA0.6 vs. 0.74
Bypass distal anastomosis
(%)
Above knee popliteal 33 vs. 3776 vs. 8914 vs. 14
Below knee popliteal67 vs.6338 vs. 46
crural24 vs. 1148 vs. 39
Bypass with spliced vein graft (%)NA14 vs. 11 31 vs. 32
CT, cohort study; RCT, randomized control study; DCB, drug-coated balloon; PB, plain balloon; DM, diabetes mellitus; HTN, hypertension; DL, dyslipidemia; CKD, chronic kidney disease; CAD, coronary artery disease; CLTI, chronic limb-threatening ischemia; PTA, percutaneous transluminal angioplasty; ABI, ankle-brachial pressure index; NA, not available. Each column shows the comparison data of the DCB and PB. * Significant difference (p = 0.012).
Table 2. Treatment characteristics.
Table 2. Treatment characteristics.
First Author, YearLinni,
2016 [22]
Jongsma,
2017 [23]
Björkman,
2019 [24]
DCB balloonIn.PactPasseo-18 Lux In.Pact
Balloon diameter (DCB vs. PB; mm)NA3.8 vs. 3.74.9 vs. 5.3
DCB, drug-coated balloon; PB, plain balloon; NA, not available.
Table 3. Follow-up methods and indications for intervention.
Table 3. Follow-up methods and indications for intervention.
First Author, YearLinni, 2016 [22]Jongsma, 2017 [23]Björkman, 2019 [24]
Indication for PTA>70% bypass stenosis verified as a PSV < 45 cm/s or 300 cm/s or PSVR > 4 in DUS imaging.>70% bypass stenosis verified as a PSV > 300 cm/s or PSVR > 3.0 in DUS imaging.PSVR > 2.5.
Follow-up protocolDUS was performed in case of recurrent symptoms or decrease of ABI > 0.15 at 1, 3, 6, and 12 months after PTA and at 12-month intervals thereafter. Reintervention was performed for
restenosis of >70%.
DUS was performed at 6 and 12 months after PTA.
Reintervention was performed for
restenosis of >70%.
DUS and clinical evaluation (symptoms, ABI) were performed at 1, 6, and 12 months after PTA. Reintervention was performed for
PSVR >2.5
Antithrombotic medication (AM)No specific regimen. Aspirin 100 mg for patients taking no AM at admission.Anticoagulants or aspirin for at least 2 years after bypass surgery.aspirin 100 mg + clopidogrel 75 mg or warfarin + aspirin 50 mg for three months for all patients
PTA, percutaneous transluminal angioplasty; DUS, Doppler ultrasonography; PSV, peak systolic velocity; PSVR, peak systolic velocity ratio.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Isaji, T.; Hosoi, Y.; Kogure, K.; Ichikawa, Y.; Fujimaki, K.; Ikezoe, T.; Nunokawa, M.; Kubota, H. Drug-Coated Balloon versus Plain Balloon Angioplasty in the Treatment of Infrainguinal Vein Bypass Stenosis: A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 87. https://doi.org/10.3390/jcm12010087

AMA Style

Isaji T, Hosoi Y, Kogure K, Ichikawa Y, Fujimaki K, Ikezoe T, Nunokawa M, Kubota H. Drug-Coated Balloon versus Plain Balloon Angioplasty in the Treatment of Infrainguinal Vein Bypass Stenosis: A Systematic Review and Meta-Analysis. Journal of Clinical Medicine. 2023; 12(1):87. https://doi.org/10.3390/jcm12010087

Chicago/Turabian Style

Isaji, Toshihiko, Yutaka Hosoi, Kota Kogure, Yohei Ichikawa, Keisuke Fujimaki, Toru Ikezoe, Masao Nunokawa, and Hiroshi Kubota. 2023. "Drug-Coated Balloon versus Plain Balloon Angioplasty in the Treatment of Infrainguinal Vein Bypass Stenosis: A Systematic Review and Meta-Analysis" Journal of Clinical Medicine 12, no. 1: 87. https://doi.org/10.3390/jcm12010087

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop