1,047
Views
30
CrossRef citations to date
0
Altmetric
Perspectives

Paclitaxel: new uses for an old drug

, , &
Pages 279-284 | Published online: 20 Feb 2014

Abstract

Paclitaxel (Taxol), one of the most important anticancer drugs, has been used for therapy of different types of cancers. Mechanistically, paclitaxel arrests cell cycle and induces cell death by stabilizing microtubules and interfering with microtubule disassembly in cell division. Recently, it has been found that low-dose paclitaxel seems promising in treating non-cancer diseases, such as skin disorders, renal and hepatic fibrosis, inflammation, axon regeneration, limb salvage, and coronary artery restenosis. Future studies need to understand the mechanisms underlying these effects in order to design therapies with specificity.

Introduction

Taxol, a natural diterpene alkaloid (), was originally isolated from the bark of Taxus brevifolia tree in the western region of the United States. When it was commercially developed by the Bristol-Myers Squibb (BMS; New York, NY, USA) Taxol was renamed to paclitaxel. Melting point of paclitaxel is around 216°C–217°C, and it has highly lipophilic, low water solubility; higher protein binding rate; and mainly disturbs the structure of the inner part of the cell membrane.Citation1,Citation2 The role of paclitaxel has been the subject of study on anticancer agents for almost half a century. It is one of the most widely used anticancer drugs, and has been used for the treatment of various cancers from metastatic breast cancer, advanced ovarian cancer, non-small-cell lung cancer, to Kaposi’s sarcoma.Citation3,Citation4 Recent studies, however, have demonstrated using low-dose paclitaxel to treat non-cancer human diseases, such as skin disorders, renal and hepatic fibrosis, inflammation, axon regeneration, limb salvage, and coronary artery restenosisCitation5Citation12 ().

Figure 1 Chemical structure of paclitaxel.

Figure 1 Chemical structure of paclitaxel.

Figure 2 Reported effects of paclitaxel in non-cancer diseases.

Abbreviations: TLR-4, toll-like receptor 4; TGF-β, transforming growth factor-beta.
Figure 2 Reported effects of paclitaxel in non-cancer diseases.

Basic mechanism of the anti-cancer effect of paclitaxel

Paclitaxel belongs to the family of cytoskeletal drugs that target tubulin. As a result, paclitaxel treatment leads to abnormality of the mitotic spindle assembly, chromosome segregation, and consequently defects of cell division. By stabilizing the microtubule polymer and preventing microtubules from disassembly, paclitaxel arrests cell cycle in the G0/G1 and G2/M phases and induces cell death in cancerCitation13Citation14 (). It has been known that inhibition of mitotic spindle using paclitaxel usually depends on its suppression of microtubule dynamics.Citation15 However, recent studies demonstrated that only low-dose paclitaxel can do so, in contrast, high-dose paclitaxel might suppress microtubule detachment from the centrosomes.Citation16 The binding site for paclitaxel has been identified to be the subunit of beta-tubulin.Citation17 Paclitaxel has other mechanisms of action than for microtubule targeting. Panis et alCitation18 found that the breast cancer patients after acute paclitaxel treatment exhibited immunosuppressive status by a strong type 2 helper T-cell (Th2) profile demonstrated by high levels of interleukin (IL)-10. Alexandre et alCitation19 and Hadzic et alCitation20 reported paclitaxel induced reactive oxygen species generation by enhancing the activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which contributed to the potent anticancer activity of paclitaxel. The antineoplastic mechanisms to the non-chemotherapeutic use of paclitaxel were found. For example, Sevko et alCitation21,Citation22 reported that paclitaxel enhanced the efficacy of chemotherapy by blocking the immunosuppressive potential of myeloid-derived suppressor cells. Gan et alCitation23 discovered that paclitaxel inhibited the androgen receptor by inducing nuclear accumulation of FOXO1 (forkhead box protein O1) as one of the anticancer mechanisms ().

Figure 3 Anti-cancer actions of paclitaxel.

Note: By stabilizing microtubules, paclitaxel arrests cell cycle in the G0/G1 and G2/M phases and induces cell death.
Abbreviations: ROS, reactive oxygen species; MDSCs, myeloid-derived suppressor cells; AR, androgen receptor.
Figure 3 Anti-cancer actions of paclitaxel.

Treatment of fibrotic diseases

Transforming growth factor-beta (TGF-β) is one of the most important profibrotic growth factors, which can bind to and activate cell surface-specific receptors and in turn promotes diverse cellular responses. The activated TGF-β receptors activate mothers against decapentaplegic homolog (Smad)2 and Smad3 proteins, which further form a protein complex with Smad4. This protein complex then translocates from the cytoplasm into the nucleus to regulate the transcription of target genes.Citation24 It has been reported that in some cell lines, binding of endogenous Smad2, Smad3 and Smad4 to microtubules negatively modulates TGF-β activity.Citation25 Paclitaxel, by stabilizing microtubules, may therefore be inhibitory to TGF-β signaling in fibrosis (). Interestingly, low-dose paclitaxel has been shown to inhibit collagen-induced arthritis, hepatic fibrosis, and fibrosis associated with systemic sclerosis in severe combined immunodeficiency (SCID) mice.Citation5Citation6,Citation26 In kidneys, Zhang et alCitation7 reported that low-dose paclitaxel (0.3 mg/kg, twice a week) significantly reduced tubulointerstitial fibrosis in a rat model of unilateral ureteral obstruction. Karbalay-Doust et alCitation27 found that both taurine and paclitaxel (0.3 mg/kg/d) had a renoprotective role in the unilateral ureteral obstruction model and the latter was more effective. Following subtotal renal ablation in rats, low-dose paclitaxel (0.3 mg/kg, twice a week) showed a significant renoprotective role by modulating TGF-β/Smad/miR-192 signaling.Citation8 In respect of lungs, Wang et alCitation28 recently reported that low-dose paclitaxel (0.6 mg/kg/d) reduced pulmonary fibrosis in rat (bleomycin) BLM-instilled pulmonary fibrosis model. In this model, paclitaxel blocked the TGF-β1/Smad3 pathway via up regulation of miR-140. Interestingly, low-dose paclitaxel (5–10 nM) also reduced stromal fibrosis in gastric cancer.Citation29 High-dose paclitaxel (175 mg/mm2) may inhibit tumor cell proliferation, however, some cancer patients with prolonged paclitaxel (175 mg/mm2) treatment suffered from scleroderma-like changes or pulmonary fibrosis.Citation30Citation33 These results suggest that low-dose paclitaxel has the potential to treat or prevent tissue fibrosis in experimental animal models, but more work is needed to carefully assess the effect in human patients.

Figure 4 Paclitaxel inhibits TGF-β/Smad signaling via enhancing endogenous Smad2, Smad3 and Smad4 binding to microtubules, and thereby ameliorates fibrosis.

Abbreviations: TGF-β, transforming growth factor-beta; Smad, mothers against decapentaplegic homolog; p, phosphorylated; R-I, TGFβ receptor I; R-II, TGFβ receptor II.
Figure 4 Paclitaxel inhibits TGF-β/Smad signaling via enhancing endogenous Smad2, Smad3 and Smad4 binding to microtubules, and thereby ameliorates fibrosis.

Regulation of inflammation

It was reported that paclitaxel attenuated tumor necrosis factor (TNF)-α and thrombin-induced solute permeability, by enhancing the endothelial monolayer and paracellular gap junction formation.Citation34,Citation35 Paclitaxel (10, 25, and 50 μg/mL) also decreased leukocyte transmigration through endothelial monolayers by stabilization of endothelial microtubules.Citation36 Pretreatment with paclitaxel (5–10 M) markedly inhibited chemotaxis induced by endotoxin-activated serum.Citation37 Moreover, paclitaxel (plasma concentration of 10 μM) was shown to attenuate the vascular leak and inflammation in lipopolysaccharide (LPS)-induced acute lung injury in a mouse model.Citation9 However, the molecular mechanism underlying the inhibitory effect of paclitaxel on inflammation is not fully understood. One study indicated that, like LPS, paclitaxel (≥5 µM) was able to activate nuclear factor-kappaB (NF-κB) signaling in 70Z/3 pre-B cells;Citation38 however, pretreatment with paclitaxel might completely inhibit LPS-induced NF-κB activation,Citation38 suggesting that LPS and paclitaxel may share and compete for a common receptor/signaling pathway.Citation39,Citation40 LPS can directly interact with toll-like receptor (TLR)-4-associated MD-2 which has a critical role in LPS recognition.Citation41 Of note, in murine cells and tissues the effect of paclitaxel is highly dose-dependent. At 3 μM or lower concentrations, paclitaxel binds to MD-2 to block TLR-4 signaling and inflammation in murine cells as human cells; however at 3.25 μM or higher concentrations, paclitaxel binding to murine MD-2 promoted inflammation by activation of MD-2/TLR-4Citation42,Citation43 (). Our recent research demonstrated that paclitaxel at 2 μM had a significant protective effect in kidneys by competitive binding to MD-2 to block MD-2/TLR-4 signaling during LPS treatment, resulting in the suppression of NF-κB activation and pro-inflammatory cytokine production.Citation44 Use of low-dose paclitaxel may therefore offer a treatment for inflammatory diseases including sepsis and related complications.

Figure 5 Paclitaxel inhibits inflammation by blocking TLR-4 signaling via binding to MD-2.

Notes: Binding of paclitaxel (≥3.125 μM) to murine MD-2 results in the activation of MD-2/TLR-4 and promotes inflammation, whereas binding of paclitaxel to human MD-2, does not.
Abbreviations: TLR-4, toll-like receptor 4; LPS, lipopolysaccharide; hMD-2, human MD-2; mMD-2, mouse MD-2.
Figure 5 Paclitaxel inhibits inflammation by blocking TLR-4 signaling via binding to MD-2.

Facilitation of axon regeneration in injured central nervous system

Poor regeneration of damaged axons in traumatic injuries of the central nervous system (CNS) usually causes permanent, devastating disabilities. Sengottuvel et alCitation45 demonstrated that low-dose paclitaxel (1, 10, 100, and 1,000 μM) treatment enabled axons to regenerate without affecting the intrinsic regenerative state of mature retinal ganglion cells in rats. Furthermore, paclitaxel treatment promoted lens injury-mediated axon regeneration by microtubule stabilization in the growth cone. The beneficial effects of paclitaxel were accompanied by a reduction of the infiltration of macrophages and a delay in glial scar formation at the injury site. These findings are supported by another study, which demonstrated that paclitaxel (256 ng/day) could reduce fibrotic scarring and enhance the capacity of axons to grow after spinal cord injury in rodents.Citation10 Mechanistically, it was suggested that paclitaxel may reduce accumulation of some inhibitory substances from scar tissue by dampening TGF-β signaling.Citation10 These data suggest that paclitaxel may be beneficial in facilitating axon regeneration in different areas of the CNS.

Paclitaxel use in critical limb ischemia

Critical limb ischemia (CLI) typically presents as rest pain, tissue ulceration, or frank tissue loss with gangrene, often accompanied by infection. A clinical trial tested the effect of local paclitaxel application on restenosis, a disease condition of narrowing of blood vessels leading to restricted blood flow. In this study, 154 patients with stenosis were randomly divided into three groups for treatment with balloon; bare balloon with paclitaxel dissolved in contrast media; or a paclitaxel-coated balloon (3 μg/mm2). The late lumen loss was significantly lower in the group treated with paclitaxel-coated angioplasty balloons.Citation11 Similarly, in another study, 87 patients were randomized to bare balloon and paclitaxel (3 μg/mm2)-coated balloon groups. Both late lumen loss and target lesion revascularization were significantly lower in the paclitaxel-coated balloon-treated patients.Citation46 In a third study, 29 patients with 32 limbs with CLI were treated by infrapopliteal application of paclitaxel-eluting stents (PES) procedures and acceptable clinical results were achieved in CLI, although they failed to prevent vascular restenosis and reduce repeat interventions.Citation47 In a fourth study, 104 patients were treated with paclitaxel-eluting balloons (PEBs), compared with historical data using uncoated balloons, the early restenosis rate of long-segment infrapopliteal disease was significantly lower.Citation48 Furthermore, paclitaxel delivered by drug-coated balloons or drug-eluting stents enhanced durability of lower extremity endovascular procedures, and had a particular benefit for diabetic limb salvage.Citation49 Mechanically, paclitaxel has a role in anti-proliferation and migration for muscle smooth cells.Citation49 Together, these clinical trials have demonstrated the beneficial effect of paclitaxel in treating stenosis and related limb ischemia conditions.

Treatment for patients with coronary artery restenosis

Percutaneous coronary intervention was a major and minimally invasive way to treat coronary artery disease. However, cell hyperproliferation of local artery after percutaneous coronary intervention may result in lumen narrowing,Citation50 which limited the use of this technology. The recent development of paclitaxel was considered as one of the most promising ways for reducing restenosis. Paclitaxel at a dose of 175 mg/mm2 is recommended for tumor therapy, a broad range of doses (1.3–10 μg/mm2) is found to be safe and efficacious for reducing restenosis.Citation51 In a randomized study it was demonstrated that restenosis occurred at a lower rate in patients with high-dose paclitaxel stents than those receiving low-dose paclitaxel stents at 6 months.Citation52 After 9 months follow-up, the rate of angiographic restenosis was significantly reduced by paclitaxel-eluting stent.Citation12 Gershlick et alCitation53 reported that the angiographic indicators of in-stent restenosis were reduced without short-term or medium-term side effects by paclitaxel-coated stents at a dose density of 2.7 μg/mm2. Furthermore, Milewski et alCitation54 demonstrated that paclitaxel-coated balloons had a dose (1–3 μg/mm2)-dependent effect on the inhibition of neointimal proliferation. Byrne et alCitation55 investigated the efficacy among PEBs, PESs, and balloon angioplasty in restenosis patients; the results indicated that PEB could be a useful treatment. Mechanically, the smooth muscle cell cycle was interrupted by low dose paclitaxel via stabilizing microtubules, thereby arresting mitosis.Citation53,Citation56 However, Pires et alCitation57 demonstrated high dose (53.5 μg) paclitaxel-eluting cuffs had adverse vascular pathology and transcriptional responses. Hence, low dose paclitaxel is better than high dose paclitaxel for reducing restenosis.

Conclusion

Recent research from both clinical trials and preclinical work in animal models has demonstrated the therapeutic effects of paclitaxel in several non-cancer diseases. While some of the effects depend on the tubulin-stabilizing action of paclitaxel, others apparently may not. Further investigation is needed to gain insights into the cellular and molecular mechanisms of the effects of paclitaxel in various disease conditions. A thorough understanding of the actions and targets of paclitaxel within a cell would guide the design of therapies with efficacy and specificity. High dose paclitaxel induces inflammation and partly organ fibrosis, hence, before clinical application of paclitaxel, we need to carefully assess what dose is safe.

Acknowledgments

The study was supported in part by grants from National Natural Science Foundation of China [81370791; 81100507] and the National Institutes of Health and Department of Veterans Administration of USA.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Goldspiel BR Clinical overview of the taxanes Pharmacotherapy 1997 17 110S 125S 9322878
  • Ledwitch K Ogburn R Cox J Taxol: efficacy against oral squamous cell carcinoma Mini Rev Med Chem 2013 13 509 521 23373651
  • Rowinsky EK The development and clinical utility of the taxane class of antimicrotubule chemotherapy agents Annu Rev Med 1997 48 353 374 9046968
  • Mekhail TM Markman M Paclitaxel in cancer therapy Expert Opin Pharmacother 2002 3 6 755 766 12036415
  • Zhou J Zhong DW Wang QW Paclitaxel ameliorates fibrosis in hepatic stellate cells via inhibition of TGF-β/Smad activity World J Gasteroenterol 2010 16 26 3330 3334
  • Liu X Zhu S Wang T Paclitaxel modulates TGF-β signaling in scleroderma skin grafts in immunodeficient mice PLoS Med 2005 2 12 e354 16250671
  • Zhang D Sun L Xian W Low-dose paclitaxel ameliorates renal fibrosis in rat UUO model by inhibition of TGF-β/Smad activity Lab Invest 2010 90 3 436 447 20142807
  • Sun L Zhang D Fuyou Liu Low-dose paclitaxel ameliorates fibrosis in the remnant kidney model by down-regulating miR-192 J Pathol 2011 225 3 364 377 21984124
  • Mirzapoiazova T Kolosova IA Moreno L Sammani S Garcia JG Verin AD Suppression of endotoxin-induced inflammation by paclitaxel Eur Respir J 2007 30 3 429 435 17537765
  • Hellal F Hurtado A Ruschel J Microtubule stabilization reduces scarring and causes axon regeneration after spinal cord injury Science 2011 331 6019 928 931 21273450
  • Tepe G Zeller T Albrecht T Local delivery of paclitaxel to inhibit restenosis during angioplasty of the leg N Engl J Med 2008 358 7 689 699 18272892
  • Stone GW Ellis SG Cox DA A polymer-based, paclitaxel-eluting stent in patients with coronary artery disease N Engl J Med 2004 350 3 221 231 14724301
  • Bharadwaj R Yu H The spindle checkpoint, aneuploidy, and cancer Oncogene 2004 23 11 2016 2027 15021889
  • Brito DA Yang Z Rieder CL Microtubules do not promote mitotic slippage when the spindle assembly checkpoint cannot be satisfied The Journal of Cell Biology 2008 182 4 623 629 18710927
  • Jordan MA Wilson L Microtubules as a target for anticancer drugs Nat Rev Cancer 2004 4 4 253 265 15057285
  • Ganguly A Yang H Cabral F Paclitaxel-dependent cell lines reveal a novel drug activity Mol Cancer Ther 2010 9 11 2914 2923 20978163
  • Löwe J Li H Downing KH Refined structure of alpha beta-tubulin at 3.5 A resolution J Mol Biol 2001 313 5 1045 1057 11700061
  • Panis C Lemos LG Victorino VJ Immunological effects of taxol and adryamicin in breast cancer patients Cancer Immunol Immunother 2012 61 4 481 488 21959683
  • Alexandre J Hu Y Lu W Pelicano H Huang P Novel action of paclitaxel against cancer cells: bystander effect mediated by reactive oxygen species Cancer Res 2007 67 8 3512 3517 17440056
  • Hadzic T Aykin-Burns N Zhu Y Paclitaxel combined with inhibitors of glucose and hydroperoxide metabolism enhances breast cancer cell killing via H2O2-mediated oxidative stress Free Radic Biol Med 2010 48 8 1024 1033 20083194
  • Sevko A Michels T Vrohlings M Antitumor effect of paclitaxel is mediated by inhibition of myeloid-derived suppressor cells and chronic inflammation in the spontaneous melanoma model J Immunol 2013 190 5 2464 2471 23359505
  • Sevko A Kremer V Falk C Application of paclitaxel in low non-cytotoxic doses supports vaccination with melanoma antigens in normal mice J Immunotoxicol 2012 9 3 275 281 22449053
  • Gan L Chen S Wang Y Inhibition of the androgen receptor as a novel mechanism of taxol chemotherapy in prostate cancer Cancer Res 2009 69 21 8386 8394 19826044
  • Derynck R Zhang YE Smad-dependent and Smad-independent pathways in TGF-beta family signalling Nature 2003 425 6958 577 584 14534577
  • Dong C Li Z Alvarez R Microtubule binding to Smads may regulate TGF beta activity Mol Cell 2000 5 1 27 34 10678166
  • Brahn E Tang C Banquerigo ML Regression of collagen-induced arthritis with Taxol, a microtubule stabilizer Arthritis Rheum 1994 37 6 839 845 7911665
  • Karbalay-Doust S Noorafshan A Pourshahid SM Taxol and taurine protect the renal tissue of rats after unilateral ureteral obstruction: a stereological survey Korean J Urol 2012 53 5 360 367 22670197
  • Wang C Song X Li Y Low-dose paclitaxel ameliorates pulmonary fibrosis by suppressing TGF-β1/Smad3 pathway via miR-140 upregulation PLoS One 2013 8 8 e70725 23967091
  • Tsukada T Fushida S Harada S Low-dose paclitaxel modulates tumour fibrosis in gastric cancer Int J Oncol 2013 42 4 1167 1174 23443842
  • Kupfer I Balguerie X Courville P Scleroderma-like cutaneous lesions induced by paclitaxel: a case study J Am Acad Dermatol 2003 48 2 279 281 12582404
  • Eisenbeilss C Weizel J Wolff H Scleroderma-like skin changes following administration of paclitaxel for the treatment of ovarian carcinoma J Dtsch Dermatol Ges 2003 1 6 468 470 16295141
  • Ostoros G Pretz A Fillinger J Fatal pulmonary fibrosis induced by paclitaxel: a case report and review of the literature Int J Gynecol Cancer 2006 16 1 391 393 16515630
  • Sako T Burioka N Watanabe M A case of stage IV advanced lung adenocarcinoma treated effectively using paclitaxel with the fibrosing radiographically on the chest CT Gan To Kagaku Ryoho 2001 28 7 989 992 Japanese 11478149
  • Petrache I Birukova A Ramirez SI Garcia JG Verin AD The role of the microtubules in tumor necrosis factor-ainduced endothelial cell permeability Am J Respir Cell Mol Biol 2003 28 5 574 581 12707013
  • Birukova AA Smurova K Birukov KG Microtubule disassembly induces cytoskeletal remodeling and lung vascular barrier dysfunction: role of Rho-dependent mechanisms J Cell Physiol 2004 201 1 55 70 15281089
  • Kielbassa K Schmitz C Gerke V Disruption of endothelial microfilaments selectively reduces the transendothelial migration of monocytes Exp Cell Res 1998 243 1 129 141 9716457
  • Roberts RL Nath J Friedman MM Gallin JI Effects of taxol on human neutrophils J Immunol 1982 129 5 2134 2141 6126502
  • Lee M Jeon YJ Paclitaxel-Induced Immune Suppression is sociated with NF-κB Activation Via Conventional PKC Isotypes in Lipopolysaccharide-Stimulated 70Z/3 Pre-B Lymphocyte Tumor Cells Mol Pharmacol 2001 59 2 248 253 11160860
  • Byrd CA Bornmann W Erdjument-Bromage H Heat shock protein 90 mediates macrophage activation by Paclitaxel and bacterial lipopolysaccharide Proc Natl Acad Sci U S A 1999 96 10 5645 5650 10318938
  • Lee M Yea SS Jeon YJ Paclitaxel causes mouse splenic lymphocytes to a state hyporesponsive to lipopolysaccharide stimulation Int J Immunopharmacol 2000 22 8 615 621 10988356
  • Shimazu R Akashi S Ogata H MD-2, a molecule that confers lipopolysaccharide responsiveness on toll-like receptor 4 J Exp Med 1999 189 11 1777 1782 10359581
  • Zimmer SM Liu J Clayton JL Paclitaxel binding to human and murine MD-2 J Biol Chem 2008 283 41 27916 27926 18650420
  • Resman N Gradisar H Vasl J Taxanes inhibit human TLR4 signaling by binding to MD-2 FEBS Lett 2008 582 28 3929 3934 18977229
  • Zhang D Li Y Liu Y Paclitaxel ameliorates lipopolysaccharide-induced kidney injury by binding myeloid differentiation protein-2 to block Toll-like receptor 4-mediated nuclear factor-κB activation and cytokine production J Pharmacol Exp Ther 2013 345 1 69 75 23318472
  • Sengottuvel V Leibinger M Pfreimer M Andreadaki A Fischer D Taxol Facilitates Axon Regeneration in the Mature CNS J Neurosci 2011 31 7 2688 2699 21325537
  • Werk M Langner S Reinkensmeier B Inhibition of restenosis in femoropopliteal arteries: paclitaxel-coated versus uncoated balloon: femoral paclitaxel randomized pilot trial Circulation 2008 118 13 1358 1365 18779447
  • Siablis D Karnabatidis D Katsanos K Diamantopoulos A Christeas N Kaqadis GC Infrapopliteal application of paclitaxel-eluting stents for critical limb ischemia: midterm angiographic and clinical results J Vasc Interv Radiol 2007 18 11 1351 1361 18003984
  • Schmidt A Piorkowski M Werner M First experience with drug-eluting balloons in infrapopliteal arteries: restenosis rate and clinical outcome J Am Coll Cardiol 2011 58 11 1105 1109 21884945
  • Cafasso D Schneider P How paclitaxel can improve results in diabetics J Cardiovasc Surg (Torino) 2012 53 1 13 21
  • Indolfi C Coppola C Torella D Arcucci O Chiariello M Gene therapy for restenosis after balloon angioplasty and stenting Cardiol Rev 1999 7 6 324 331 11208244
  • Tepe Gunnar Scheller Bruno Speck Ulrich Paclitaxel-coated angioplasty catheters for local drug delivery Diagn Interv Radiol 2007 13 2 61 63 17562508
  • Park SJ Shim WH Ho DS A paclitaxel-eluting stent for the prevention of coronary restenosis N Engl J Med 2003 348 16 1537 1545 12700373
  • Gershlick A De Scheerder I Chevalier B Inhibition of restenosis with a paclitaxel-eluting, polymer-free coronary stent: the European evaLUation of pacliTaxel Eluting Stent (ELUTES) trial Circulation 2004 109 4 487 493 14744971
  • Milewski K Afari ME Tellez A Evaluation of efficacy and dose response of different paclitaxel-coated balloon formulations in a novel swine model of iliofemoral in-stent restenosis JACC Cardiovasc Interv 2012 5 10 1081 1088 23078739
  • Byrne RA Neumann FJ Mehilli J Paclitaxel-eluting balloons, paclitaxel-eluting stents, and balloon angioplasty in patients with restenosis after implantation of a drug-eluting stent (ISAR-DESIRE 3): a randomised, open-label trial Lancet 2013 381 9865 461 467 23206837
  • Sonoda S Honda Y Kataoka T Bonneau HN Taxol-based eluting stents from theory to human validation: clinical and intravascular ultrasound observations J Invasive Cardiol 2003 15 3 109 114 12612382
  • Pires NMM Eefting D de Vries MR Quax PHA Jukema JW Sirolimus and paclitaxel provoke different vascular pathological responses after local delivery in a murine model for restenosis on underlying atherosclerotic arteries Heart 2007 93 8 922 927 17449502