• Keine Ergebnisse gefunden

6.1 Conclusion

The increasing risk of coronary heart disease throughout the global population has been of critical concern. The increasing population with a requirement for stent-based interventions will ultimately lead to a large population experiencing stent-related pathologies such as restenosis and thrombosis.

The main aim of stent manufacturers is to design a stent that eradicates these complications. This can only be achieved through a therapeutic option that promotes rapid endothelialisation whilst simultaneously supressing smooth muscle cell proliferation. This study developed and used systematic assessment techniques to identify potential therapies and coating platforms for use in drug eluting stents. A static and dynamic platform was designed and developed in order to facilitate these coatings and therapies to assess ex vivo performance.

Although initially hypothesised, not all drugs performed as expected for the purpose of dual-functional therapies for combating restenosis and stent thrombosis through in vitro assessments. Within the concentration ranges tested in vitro, magnolol and curcumin demonstrated no optimal effect on the endothelial and smooth muscle cells. Ferulic acid and exendin-4 did provide a level of suitability for this application due to an increase in endothelial cell activity relative to the maintenance of smooth muscle cells. The end goal was to have a drug that promotes endothelialisation faster than the rate of smooth muscle cell proliferation and thus inhibiting the possibility of stent thrombosis associated with current drug eluting stents. It was also observed that at the same concentration, the current commercially available drugs used in drug eluting stents demonstrate that endothelial cells show a higher sensitivity to the anti-proliferatives than the smooth muscle cells, which may contribute to the poor endothelialisation observed in a physiological setting upon insertion of a drug eluting stent. The retention of endothelial functionality and phenotype was observed upon dosage with exendin-4 and ferulic acid.

The assessment of the polymer coatings yielded several insights. It was observed that the rat tail collagen blended with PLGA provided an appropriate surface for cell seeding. The fast adhesion time of 30 minutes upon seeding shows that the hybrid blends of polymer are capable of rapid endothelial cell adhesion and can be incorporated into an aspect of in situ endothelialisation. The tobacco derived recombinant human type I collagen demonstrated a cytotoxic behaviour upon increasing concentrations of the collagen with PLGA. Upon neutralisation of the collagen within the blend, the coatings exhibited improved endothelial cell proliferation. The recombinant human collagen based blends exhibited a higher increase in cell activity as compared to the rat tail collagen and thus indicating

information of the drug which can be attributed to the low concentrations used. The improved outcome of using recombinant human type I collagen over animal-sourced type I collagen attests that the former type of collagen should be implemented into more biomaterials and coatings.

The commissioning and optimisation of the custom multi-station pulsatile flow bioreactor was achieved during this study. The bioreactor was set up in conjunction with the chemostat for optimising culture media properties whilst maintaining temperature within the tissue chamber. The user handling experience from tissue source to bioreactor was also improved by reducing the handling time of affixing the tissue to the tissue chamber. A thermal control system was employed within the device to compensate for temperature loss throughout the series of tubing from the chemostat to the bioreactor. Most importantly, the pressure waveform generated by the linear actuator based on a series of calculations did achieve near-physiological behaviour.

The newly developed custom static tissue holder and specialised electropolished stainless steel meshes allowed for a simplified planar stented artery in order to improve tissue and cell visualisation upon static culture. The observation of cell migration and proliferation from the tissue and endothelialised sheets onto the struts demonstrated proof of concept of the static system in order to identify endothelialisation rates of different coated mesh samples. The flow system demonstrated cell viability; although poor when using the endothelialised sheet and shows that optimisation of the system needs to be carried out. The preliminary tissue study phase in static indicated possibility of cold storage in tiprotec with the antibiotic solution as cell viability was observed of up to 1 week of culture.

The exemplary static assessment of the coated mesh structure affixed atop the endothelialised sheet established cell viability and the early process of cell migration/proliferation to the struts. Once the flow system is optimised, it is hypothesised that the endothelialisation procedure will occur at an improved rate.

This thesis covers the assessment of potential therapeutic drugs dosed on endothelial and smooth muscle cells in order to identify the optimal safe concentration that provides dual functionality on the respective cell types for the purpose of drug eluting stents. Different coating options were optimised and evaluated for endothelial cell interactions and the feasibility of in situ endothelialisation. The optimisation and setup of the multi-station pulsatile flow bioreactor as well as the design and setup of the static and dynamic stent testing platform was achieved. Final porcine tissue studies indicated proof of principle of these systems indicative of feasibility. This project encompasses an assortment of various critical stent-based parameters on an individual basis and developed a system to evaluate these aspects once they are to be assimilated as a final coated mesh structure.

6.2 Future Recommendations

The work reported in this thesis has paved the way as a development tool for a multitude of future research. Apart from continuing the stent focused research, the bioreactor aspect can dwell into further tissue engineering principles with regards to vascular tissue regeneration and graft synthesis.

Using the systematic assessment of drugs and polymers provides an initial indication towards its potential for use in drug eluting stents. Encompassing these two parameters requires a high degree of optimisation in order to match the drug release/polymer degradation with that of the effective drug concentration. The heterogeneity of the collagen dispersion within the PLGA might be an issue and would require verification. The use of an atomic force microscopy (AFM)-Raman system could provide a visualisation of the dispersion of collagen throughout the coating. The optimisation of the coating procedure is also a required point of study as homogenous thickness of the polymer coating would govern well dispersed drug release.

The static and flow system demonstrated a solid proof of concept and needs validation to ensure that the observable cells on the struts are in fact endothelial cells. The flow system requires further optimisation and would benefit from computation fluid dynamic assessments to identify the optimal flow required to generate physiological shear stresses. One important aspect with regards to stent development is the inhibition of smooth muscle cells and this study focused predominantly on the endothelial aspect. The next stage in the project would be to demonstrate the ability to induce observable smooth muscle cell proliferation over the struts in order to assess the efficacy of SMC inhibition of the coated meshes. It would also be of interest to utilise cobalt chromium mesh samples with thinner struts as this would increase endothelialisation time and provide a more accurate physiological representation. The observable differences of endothelialisation of different coated struts are the primary target for the future of this study.

The bioreactor, with its capability to produce near-physiological pulsatile flow conditions, can be used to house a stented artery in order to observe stent endothelialisation in an improved ex vivo format.

The bespoke multistation pulsatile flow bioreactor has the potential to assess the effects of atypical conditions on ex vivo arteries as a biophysical drug model. Conditions like arrhythmia and abnormal blood pressures can be simulated by the highly programmable system. The work presented in this thesis, indicating the possibility of rapid cell adhesion on the polymeric surface, showed that in situ endothelialisation might be a viable option.

References

[1] Van Buuren F, Dahm JB, Horskotte D. Stent restenosis and thrombosis: etiology, treatment, and outcomes. Minerva Med 2012;103:503–11.

[2] Lee JH, Kim E Do, Jun EJ, Yoo HS, Lee JW. Analysis of trends and prospects regarding stents for human blood vessels. Biomater Res 2018;22:8. doi:10.1186/s40824-018-0114-1.

[3] Van Beusekom HMM, Serruys PW. Drug-Eluting Stent Endothelium: Presence or Dysfunction.

JACC Cardiovasc Interv 2010;3:76–7. doi:10.1016/J.JCIN.2009.10.016.

[4] Reejhsinghani R, Lotfi AS. Prevention of stent thrombosis: challenges and solutions. Vasc Health Risk Manag 2015;11:93–106. doi:10.2147/VHRM.S43357.

[5] Saladin K. Human Anatomy. Hum. Anat. 3rd ed., New York: McGraw-Hill Higher Education;

2011, p. 564–71.

[6] OpenStax. Structure and Function of Blood Vessels 2013:1–21.

http://cnx.org/contents/58db2cce-b3d9-4904-9049-80a6cd89264b@4/Structure_and_Function_of_Bloo#tbl-ch21_02 (accessed June 1, 2015).

[7] Secomb TW. Hemodynamics. Compr Physiol 2016;6:975–1003. doi:10.1002/cphy.c150038.

[8] Elad D, Einav S. PHYSICAL AND FLOW PROPERTIES OF BLOOD David Elad and Shmuel Einav.

Stand Handb Biomed Eng Des 2004:1–25.

[9] Crystal GJ, Pagel PS. Right Ventricular Perfusion. Anesthesiology 2018;128:202–18.

doi:10.1097/ALN.0000000000001891.

[10] Sumpio BE, Riley JT, Dardik A. Cells in focus: endothelial cell. Int J Biochem Cell Biol 2002;34:1508–12.

[11] Liu H-B, Gong Y-F, Yu C-J, Sun Y-Y, Li X-Y, Zhao D, et al. Endothelial progenitor cells in cardiovascular diseases: from biomarker to therapeutic agent. Regen Med Res 2013;1:9.

doi:10.1186/2050-490X-1-9.

[12] Walenta K, Bohm M, Schiegl T, Link a., Werner N, Ahlers P, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med 2005;353:999–1007.

doi:353/10/999 [pii] 10.1056/NEJMoa043814.

[13] Ghani U, Shuaib A, Salam A, Nasir A, Shuaib U, Jeerakathil T, et al. Endothelial progenitor cells during cerebrovascular disease. Stroke 2005;36:151–3.

doi:10.1161/01.STR.0000149944.15406.16.

[14] Lacolley P, Regnault V, Nicoletti A, Li Z, Michel JB. The vascular smooth muscle cell in arterial pathology: A cell that can take on multiple roles. Cardiovasc Res 2012;95:194–204.

doi:10.1093/cvr/cvs135.

[15] Saleh Al-Shehabi T, Iratni R, Eid AH. Anti-atherosclerotic plants which modulate the

[16] Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, et al. Heart Disease and Stroke Statistics-2015 Update: A Report From the American Heart Association. Circulation 2014;131:e29–322. doi:10.1161/CIR.0000000000000152.

[17] Frostegård J. Immunity, atherosclerosis and cardiovascular disease. BMC Med 2013;11:117.

doi:10.1186/1741-7015-11-117.

[18] Libby P. Current Concepts of the Pathogenesis of the Acute Coronary Syndromes. Circulation 2001;104:365–72. doi:10.1161/01.CIR.104.3.365.

[19] Tesfamariam B. Drug release kinetics from stent device-based delivery systems. J Cardiovasc Pharmacol 2008;51:118–25. doi:10.1097/FJC.0b013e318158540f.

[20] Iakovou I. Thrombosis after stent implantation: how much of a problem is there? Future Cardiol 2008;4:261–7. doi:10.2217/14796678.4.3.261.

[21] Lemesle G, Delhaye C, Bonello L, de Labriolle A, Waksman R, Pichard A. Stent thrombosis in 2008: definition, predictors, prognosis and treatment. Arch Cardiovasc Dis 2008;101:769–77.

doi:10.1016/j.acvd.2008.10.006.

[22] Serruys PW, de Jaegere P, Kiemeneij F, Macaya C, Rutsch W, Heyndrickx G, et al. A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. Benestent Study Group. N Engl J Med 1994;331:489–95.

doi:10.1056/NEJM199408253310801.

[23] Fischman DL, Leon MB, Baim DS, Schatz R a, Savage MP, Penn I, et al. A randomized comparison of coronary-stent placement and balloon angioplasty in the treatment of coronary artery disease. Stent Restenosis Study Investigators. N Engl J Med 1994;331:496–

501. doi:10.1056/NEJM199408253310802.

[24] Dotter CT, Buschmann RW, McKinney MK, Rösch J. Transluminal expandable nitinol coil stent grafting: preliminary report. Radiology 1983;147:259–60.

[25] Roguin A. Stent: The man and word behind the coronary metal prosthesis. Circ Cardiovasc Interv 2011;4:206–9. doi:10.1161/CIRCINTERVENTIONS.110.960872.

[26] Ako J, Bonneau HN, Honda Y, Fitzgerald PJ. Design Criteria for the Ideal Drug-Eluting Stent. Am J Cardiol 2007;100:3M – 9M. doi:10.1016/j.amjcard.2007.08.016.

[27] Regar E, Sianos G, Serruys PW. Stent development and local drug delivery. [Review] [78 refs].

Br Med Bull 2001;59:227–48.

[28] Wilson Nicholas LM Cruden WM. Advances in coronary stent technology: current expectations and new developments. Res Reports Clin Cardiol 2013:4–85. doi:10.2147/RRCC.S34408.

[29] Whittaker DR, Fillinger MF. The engineering of endovascular stent technology: a review. Vasc Endovascular Surg 2006;40:85–94. doi:10.1177/153857440604000201.

[30] Taylor A, Sigwart U. Endoluminal stenting. London Co Ltd 1996.

[31] Yang K, Ren Y, Wan P. High nitrogen nickel-free austenitic stainless steel: A promising coronary stent material. Sci China Technol Sci 2012;55:329–40. doi:10.1007/s11431-011-4679-3.

[32] Stoeckel D, Bonsignore C, Duda S, Dunitz M. A survey of stent designs. Minim Invasive Ther Allied Technol 2002;11:137–47. doi:10.1080/136457002760273340.

[33] Williams DF. Tissue-biomaterial interactions. J Mater Sci 1987;22:3421–45.

[34] Halwani DO, Anderson PG, Lemons JE, Jordan WD, Anayiotos AS, Brott BC. In-vivo corrosion and local release of metallic ions from vascular stents into surrounding tissue. J Invasive Cardiol 2010;22:528–35.

[35] Köster R, Vieluf D, Kiehn M, Sommerauer M, Kähler J, Baldus S, et al. Nickel and molybdenum contact allergies in patients with coronary in-stent restenosis. Lancet 2000;356:1895–7.

doi:10.1016/S1062-1458(01)00267-7.

[36] Saito T, Hokimoto S, Oshima S, Noda K, Kojyo Y, Matsunaga K. Metal allergic reaction in chronic refractory in-stent restenosis. Cardiovasc Revascularization Med 2009;10:17–22.

doi:10.1016/j.carrev.2008.01.004.

[37] Poncin P, Proft J. Stent tubing: understanding the desired attributes. In: Poncin P, Proft J, editors., ASM Internationa; 2003, p. 253–9.

[38] Fujiu K, Manabe I, Sasaki M, Inoue M, Iwata H, Hasumi E, et al. Nickel-free stainless steel avoids neointima formation following coronary stent implantation. Sci Technol Adv Mater 2012;13:064218. doi:10.1088/1468-6996/13/6/064218.

[39] Inoue M, Sasaki M, Katada Y, Taguchi T. Quantitative biocompatibility evaluation of nickel-free high-nitrogen stainless steel in vitro/in vivo. J Biomed Mater Res - Part B Appl Biomater 2014;102:68–72. doi:10.1002/jbm.b.32982.

[40] Medtronic. Advantages of Cobalt Alloy for Coronary Stents. Medtronic Vascular; 2003.

[41] Tahir H, Hoekstra a. G, Lorenz E, Lawford P V., Hose DR, Gunn J, et al. Multi-scale simulations of the dynamics of in-stent restenosis: impact of stent deployment and design. Interface Focus 2011;1:365–73. doi:10.1098/rsfs.2010.0024.

[42] Kauffman GB, Mayo I. The Story of Nitinol: The Serendipitous Discovery of the Memory Metal and Its Applications. Chem Educ 1997;2:1–21. doi:10.1007/s00897970111a.

[43] Duerig T, Pelton a, Stöckel D. An overview of nitinol medical applications. Mater Sci Eng A 1999;273-275:149–60. doi:10.1016/S0921-5093(99)00294-4.

[44] Cohen-Inbar O. Nitinol Stenting in Post-Traumatic Pseudo-Aneurysm of Internal Carotid Artery. Open J Mod Neurosurg 2012;02:45–9. doi:10.4236/ojmn.2012.23009.

[45] Stöckel D. The Shape Memory Effect - Phenomenon, Alloys, Applications. EUROflex; 2000.

[46] Barrett RD, Bishara SE, Quinn JK. Biodegradation of orthodontic appliances. Part I.

[47] Mani G, Feldman MD, Patel D, Agrawal CM. Coronary stents: a materials perspective.

Biomaterials 2007;28:1689–710.

[48] Virmani R, Farb A. Pathology of in-stent restenosis. Curr Opin Lipidol 1999;10:499–506.

[49] Martin DM, Boyle FJ. Drug-eluting stents for coronary artery disease: a review. Med Eng Phys 2011;33:148–63. doi:10.1016/j.medengphy.2010.10.009.

[50] Stone GW, Ellis SG, Colombo A, Grube E, Popma JJ, Uchida T, et al. Long-term safety and efficacy of paclitaxel-eluting stents: Final 5-year analysis from the TAXUS clinical trial program.

JACC Cardiovasc Interv 2011;4:530–42. doi:10.1016/j.jcin.2011.03.005.

[51] Iqbal J, Gunn J, Serruys PW. Coronary stents: historical development, current status and future directions. Br Med Bull 2013;106:193–211.

[52] Silber S. Cypher versus taxus: Are there differences? J Interv Cardiol 2005;18:441–6.

doi:10.1111/j.1540-8183.2005.00084.x.

[53] Shafiq N, Malhotra S, Pandhi P, Behl A, Sharma YP. Cypher versus Taxus: the stent war. Expert Opin Investig Drugs 2006;15:1537–44. doi:10.1517/13543784.15.12.1537.

[54] Chen JL, Gao RL, Yang YJ, Qiao SB, Qin XW, Yao M, et al. Comparison of short- and mid-term outcomes between CYPHER and TAXUS stents in patients with complex lesions of the coronary arteries. Chin Med J (Engl) 2006;119:21–5.

[55] Mehilli J, Dibra A, Kastrati A, Pache J, Dirschinger J, Schömig A. Randomized trial of paclitaxel- and sirolimus-eluting stents in small coronary vessels. Eur Heart J 2006;27:260–6.

doi:10.1093/eurheartj/ehi721.

[56] Kastrati A, Mehilli J, von Beckerath N, Dibra A, Hausleiter J, Pache J, et al. Sirolimus-eluting stent or paclitaxel-eluting stent vs balloon angioplasty for prevention of recurrences in patients with coronary in-stent restenosis: a randomized controlled trial. JAMA 2005;293:165–71. doi:10.1016/j.accreview.2005.02.086.

[57] Dibra A, Kastrati A, Mehilli J, Pache J, Schühlen H, von Beckerath N, et al. Paclitaxel-eluting or sirolimus-eluting stents to prevent restenosis in diabetic patients. N Engl J Med

2005;353:663–70. doi:10.1056/NEJMoa044372.

[58] Hossainy S, Prabhu S. A mathematical model for predicting drug release from a biodurable drug-eluting stent coating. J Biomed Mater Res Part A 2008;87A:487–93.

doi:10.1002/jbm.a.31787.

[59] Simon C, Palmaz JC, Sprague EA. Influence of topography on endothelialization of stents: clues for new designs. J Long Term Eff Med Implants 2000;10:143–51.

[60] Tsyganov I, Maitz MF, Wieser E. Blood compatibility of titanium-based coatings prepared by metal plasma immersion ion implantation and deposition. Appl Surf Sci 2004;235:156–63.

doi:10.1016/j.apsusc.2004.05.134.

[61] Meredith IT, Ormiston J, Whitbourn R, Kay IP, Muller D, Bonan R, et al. First-in-human study of the Endeavor ABT-578-eluting phosphorylcholine-encapsulated stent system in de novo

native coronary artery lesions: Endeavor I Trial. EuroIntervention 2005;1:157–64.

doi:EIJV1I2A26 [pii].

[62] Singh K, Sun S, Vézina C. Rapamycin (AY-22,989), a new antifungal antibiotic. IV. Mechanism of action. J Antibiot (Tokyo) 1979;32:630–45.

[63] Chang JY, Sehgal SN, Bansbach CC. FK506 and rapamycin: Novel pharmacological probes of the immune response. Trends Pharmacol Sci 1991;12:218–23.

doi:10.1016/0165-6147(91)90555-7.

[64] Gallo R, Padurean a, Jayaraman T, Marx S, Roque M, Adelman S, et al. Inhibition of intimal thickening after balloon angioplasty in porcine coronary arteries by targeting regulators of the cell cycle. Circulation 1999;99:2164–70. doi:10.1161/01.CIR.99.16.2164.

[65] Duda SH, Pusich B, Richter G, Landwehr P, Oliva VL, Tielbeek A, et al. Sirolimus-Eluting Stents for the Treatment of Obstructive Superficial Femoral Artery Disease: Six-Month Results.

Circulation 2002;106:1505–9. doi:10.1161/01.CIR.0000029746.10018.36.

[66] Zohlnhöfer D, Klein C a, Richter T, Brandl R, Murr a, Nührenberg T, et al. Gene expression profiling of human stent-induced neointima by cDNA array analysis of microscopic specimens retrieved by helix cutter atherectomy: Detection of FK506-binding protein 12 upregulation.

Circulation 2001;103:1396–402. doi:10.1161/01.CIR.103.10.1396.

[67] Suzuki T, Kopia G, Hayashi S, Bailey LR, Llanos G, Wilensky R, et al. Stent-based delivery of sirolimus reduces neointimal formation in a porcine coronary model. Circulation

2001;104:1188–93. doi:10.1161/hc3601.093987.

[68] Hooks MA. Tacrolimus, a new immunosuppressant--a review of the literature. Ann Pharmacother n.d.;28:501–11.

[69] Rowinsky EK, Donehower RC. Paclitaxel (taxol). N Engl J Med 1995;332:1004–14.

[70] Dobesh PP, Stacy Z a, Ansara AJ, Enders JM. Drug-eluting stents: a mechanical and pharmacologic approach to coronary artery disease. Pharmacotherapy 2004;24:1554–77.

doi:10.1592/phco.24.16.1554.50955.

[71] Windecker S, Remondino A, Eberli FR, Jüni P, Räber L, Wenaweser P, et al. Sirolimus-eluting and paclitaxel-eluting stents for coronary revascularization. N Engl J Med 2005;353:653–62.

[72] Fattori R, Piva T. Drug-eluting stents in vascular interventions. Lancet 2003;361:247–9.

[73] Nakazawa G, Finn A V., John MC, Kolodgie FD, Virmani R. The Significance of Preclinical Evaluation of Sirolimus-, Paclitaxel-, and Zotarolimus-Eluting Stents. Am J Cardiol 2007;100:S36–44. doi:10.1016/j.amjcard.2007.08.020.

[74] Camenzind E, Wijns W, Mauri L, Kurowski V, Parikh K, Gao R, et al. Stent thrombosis and major clinical events at 3 years after zotarolimus-eluting or sirolimus-eluting coronary stent implantation: A randomised, multicentre, open-label, controlled trial. Lancet 2012;380:1396–

405. doi:10.1016/S0140-6736(12)61336-1.

[76] Mehilli J, Massberg S. Revisiting the BIOSCIENCE of drug-eluting stent technology. Lancet 2014;384:2086–8.

[77] Natsuaki M, Kozuma K, Morimoto T, Kadota K, Muramatsu T, Nakagawa Y, et al.

Biodegradable polymer biolimus-eluting stent versus durable polymer everolimus-eluting stent: A randomized, controlled, noninferiority trial. J Am Coll Cardiol 2013;62:181–90.

doi:10.1016/j.jacc.2013.04.045.

[78] Serruys PW, Ruygrok P, Neuzner J, Piek JJ, Seth A, Schofer JJ, et al. A randomised comparison of an everolimus-eluting coronary stent with a paclitaxel-eluting coronary stent:the SPIRIT II trial. EuroIntervention 2006;2:286–94.

[79] Serruys PW, Silber S, Garg S, van Geuns RJ, Richardt G, Buszman PE, et al. Comparison of zotarolimus-eluting and everolimus-eluting coronary stents. N Engl J Med 2010;363:136–46.

[80] Kandzari DE, Leon MB, Popma JJ, Fitzgerald PJ, O’Shaughnessy C, Ball MW, et al. Comparison of Zotarolimus-Eluting and Sirolimus-Eluting Stents in Patients With Native Coronary Artery Disease. J Am Coll Cardiol 2006;48:2440–7. doi:10.1016/j.jacc.2006.08.035.

[81] Park DW, Kim YH, Yun SC, Kang SJ, Lee SW, Lee CW, et al. Comparison of zotarolimus-eluting stents with sirolimus- and paclitaxel-eluting stents for coronary revascularization: The ZEST (Comparison of the Efficacy and Safety of Zotarolimus-Eluting Stent with Sirolimus-Eluting and PacliTaxel-Eluting Stent for Cor. J Am Coll Cardiol 2010;56:1187–95.

doi:10.1016/j.jacc.2010.03.086.

[82] Sousa JE, Serruys PW, Costa MA. New frontiers in cardiology: drug-eluting stents: Part I.

Circulation 2003;107:2274–9.

[83] Kamath KR, Barry JJ, Miller KM. The Taxus drug-eluting stent: A new paradigm in controlled drug delivery. Adv Drug Deliv Rev 2006;58:412–36. doi:10.1016/j.addr.2006.01.023.

[84] Moses JW, Leon MB, Popma JJ, Fitzgerald PJ, Holmes DR, O’Shaughnessy C, et al. Sirolimus-Eluting Stents versus Standard Stents in Patients with Stenosis in a Native Coronary Artery. N Engl J Med 2003;349:1315–23. doi:10.1056/NEJMoa035071.

[85] Leon MB, Mauri L, Popma JJ, Cutlip DE, Nikolsky E, O’Shaughnessy C, et al. A Randomized Comparison of the Endeavor Zotarolimus-Eluting Stent Versus the TAXUS Paclitaxel-Eluting Stent in De Novo Native Coronary Lesions. 12-Month Outcomes From the ENDEAVOR IV Trial.

J Am Coll Cardiol 2010;55:543–54. doi:10.1016/j.jacc.2009.08.067.

[86] Ding N, Pacetti SD, Tang FW, Gada M, Roorda W. XIENCE V (TM) Stent Design and Rationale. J Interv Cardiol 2009;22:S18–27. doi:DOI 10.1111/j.1540-8183.2009.00450.x.

[87] García-García HM, Vaina S, Tsuchida K, Serruys PW. Drug-eluting stents. Arch Cardiol Mex 2006;76:297–319.

[88] Raval A, Parikh J, Engineer C. Mechanism of controlled release kinetics from medical devices.

Brazilian J Chem Eng 2010;27:211–25.

[89] Moyer P, Ornato JP, Brady WJ, Davis LL, Ghaemmaghami C a., Gibler WB, et al. Development

[89] Moyer P, Ornato JP, Brady WJ, Davis LL, Ghaemmaghami C a., Gibler WB, et al. Development