Authors:
Ceyda Tuba Sengel*
Affiliations:
Ceyda Tuba Sengel-Turk, Ankara University Faculty of Pharmacy, Department of Pharmaceutical Technology 06100 Tandogan / Ankara, Turkey
Dates:
Received: 30 October, 2015; Accepted: 19 November, 2015; Published: 09 December, 2015
*Corresponding author:

Ceyda Tuba Sengel-Turk (Assistant Professor) Ankara University Faculty of Pharmacy Department of Pharmaceutical Technology 06100 Tandogan / Ankara Turkey, Contact Nos: +90 312 203 31 52; Fax Nos: +90 312 213 10 81; E-mail: @

Citation:
Sengel CT (2015) Delivery of Nanoparticles for the Treatment of Cardiovascular Diseases. Glob J Obes Diabetes Metab Syndr 2(1): 018-021.10.17352/2455-8583.000010
Copyright:
© 2015 Sengel CT. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Keywords:
Nano; Stents; Biomedical implants; Cardiovascular implants

Cardiovascular diseases are still one of the major causes of death for the people in the world. Biomedical implantable devices are the basic approach on the treatment of cardiovascular diseases. However, unexpected and serious complications can be observed in the case of their usage. Nanotechnology gives a promising perspective to overcome these drawbacks. Nanoparticulate drug delivery systems have develop superior medical solutions and offer better prospects to patients. This review comprehensively summarizes the recent situation, the benefits and the role of nanocarriers in cardiovascular implant technology.

Introduction

Nanotechnology is a promising and emerging field which uses nanoparticles to facilitate the treatment and/or diagnosis of various diseases such as cancer [1,2], diabetes [3], osteoarthritis [4], brain and retinal diseases [5], cardiovascular diseases [6,7] and bacterial infections [8,9]. Nanoparticles are defined as colloidal particulate dispersions or particles ranging from 10 to 1000 nm in size [10,11]. A similar definition was made by Buzea et al. They described the nanoparticles as “particles with at least one dimension smaller than 1 micron and potentially as small as atomic and molecular length scale” [12]. Different types of nanostructure systems are designed for drug delivery and also the manipulation and the fabrication of biomedical implantable devices have been extensively investigated over the past decades. For this reason, the utilization of the nanoparticulate drug delivery systems in the field of biomedical is predicted to spread rapidly in recent years. [13] Biomedical implants obviously provide a wide range of medical cures for many of the disorders, such as cardiovascular diseases. Vascular grafts, defibrillators, heart valves, pacemarkers and stents are the most common cardiovascular implantable devices used in the medical field. However, the present implant technology is facing a major difficulty of being perceived by the human body as foreign substances. Nanotechnology provides a medical solution to revolutionize the biomedical implant technology exactly by modifying and designing their structures thereby to overcome these problems [14]. The topic of this review article is to designate the role and the importance of nanocarriers in cardiovascular implant technology.

Nanotechnology and Cardiovascular Implantable Devices

Among the various diseases, cardiovascular diseases are still the major cause of mortality in the developed countries. Nanotechnology-based systems have promised new opportunities to diagnose and treatment cardiovascular diseases. Stents are the first option for the cardiovascular therapy [15]. A stent is a tubular device used to support a segment of a blood vessel or any other anatomical lumen so as to preserve or regain its patency. Balloon angioplasty was the common choice to open the blocked vascular vessels before the introduction of coronary stents as bare metallic stents [16]. However, both stent implantation and balloon angioplasty are still cause of various complications, such as elastic recoil, vascular smooth muscle cell migration and proliferation, platelet aggregation, and at last thrombus formation [17,18]. These complications were eventually overcome by the modifications in the stent technology and could be ameliorated by the introduction of therapeutic compounds. In this context, drug eluting stents have been developed through coating the surface of the stents by a matrix polymer, bearing therapeutic agents that regulate the cell division and prevent thrombosis. Sirolimus-, biolimus-, everolimus- and paclitaxel-polymer combinations have been the major surface modifiers which are used for the surface coating and the manufacturing of the drug eluting stents [19,20]. Among them, paclitaxel-eluting TAXUS® and sirolimus-eluting CYPHER® are the most under-researched implanted stents in this field. The conducted randomized clinical studies have demonstrated that these systems had indicated their therapeutic efficiencies by reducing the risk of in-stent restenosis [21,22]. Even though the surface coating of the stents by drug-polymer combination has solved the problem of in-stent restonosis, a new critical problem surfaces after then –late stent thrombosis. This trouble is partly due to the introduction of the stent surface as a foreign substance by human body and also partly due to the incorporation of the polymers, which even they have biodegradable structure, causes inflammation and increases the sensitivity to thrombosis [23,24]. In fact, the majority of the drug eluting stents over bare metallic stents is still contradictive for these reasons. At this point, the researchers have found the solution in nanotechnology. The combination of nanotechnology with cardiovascular device provides a key to the solution by inducing the endothelial cells proliferation, while suppressing the vascular smooth muscle cell proliferation at the same time. From this perspective, nanostructured stents seem more effective in the treatment of cardiovascular disease compared to nanocarrier based stent coatings. When nanoparticles are associated with stents, the particles leaves from the stent surface, penetrate to the injured epithelium and are taken up by the arterial tissues [25]. The major benefits of the nanoparticulate drug delivery systems are tabulated in (Table 1).

  1. avatar

    Table 1:

    The superiorities of nanoparticles on cardiovascular implantable devices [14, 26, 27].

Nanoparticles Coated Biomedical Devices

The first study on this field belongs to Labhasetwar and his group. In 1997, they investigated the potency of developed polymeric nanoparticulate drug delivery systems on the treatment of restenosis by using an ex-vivo arterial model. They prepared poly(lactide-co-glycolide) (PLGA) polymeric nanoparticles and modified their surfaces by a cationic agent. The ex-vivo model study demonstrated that the arterial uptake of surface coated nanoparticles was 10-fold higher than that of the non-coated nanoparticles [28]. Although this research did not contain an in-vivo experiment, it had been a major driving force in providing a basis to incorporate nanocarriers on stent surfaces for restenosis treatment. As a continuation of this study, the same researchers designed another experiment and produced anti-restetonic drugs encapsulated polymeric nanoparticulate formulations. They evaluated the uptake ability of these systems in an ex-vivo model utilizing dog carotid arter. This study demonstrated that the arterial uptake is size-dependent and the particles of ca.100 nm diameter penetrated to the artery wall better than those of 200 nm. It was also reported that when the artery was not washed, approximately 26 percent of the nanoparticles had been retained, whereas if the nanoparticles contanct was followed by washing with Ringer’s solution, the retention dropped to 6 percent. This result indicated the possibility to wash away the coated nanoparticles from the stent surface through vascular flow [29]. At the same year, they investigated the arterial retention of the polymeric nanoparticulate systems into porcine coronary arterials through an in vivo model. However, they found a reduction on the uptake capacity of nanoparticles, compared to ex-vivo model findings [30]. In 2002, Labhasetwar and his research team evaluated the uptake potency and the localization of the nanoparticles through endothelial cells. They reported that the nanoparticles preferentially localized in the cytoplasma of the cells and the nanoparticles of about 70 nm particle size showed a 27 fold higher uptake than that of the particles which had aproximately 200 nm diameter [31,32]. Similar results were reported by different researchers. Luderer et al. prepared sirolimus loaded poly(D,L-lactide) nanoparticles with a diameter of ca. 250 nm. They concluded that the developed nanoparticles seemed more effective than the free drug in inhibiting smooth endothelial cell proliferation without affecting the endothelial cell multiplication [33].

In 2004, Westedt and co-workers firstly designed the nanoparticle coated catheter balloons which releases nanocarriers locally and shows a biphasic drug release. Westedt and his team evaluated the arterial uptake ability of the fluorescent labeled nanoparticles which were released from a microporous balon catheter. In conclusion, they observed a higher uptake of nanoparticules [34]. This viewpoint brought another perspective and a new approach of “coating the nano-sized carriers on the stent surface instead of coating of nanoparticles on ballons” has emerged. In 2009, Nakano et al. [35] firstly reported the development of nanoparticle-eluting stents. They prepared nanoparticle formulations and coated their surfaces with chitosan to achieve electrodeposition. Then, the nanoparticles were coated on a metallic stent surface by a cationic electro-deposition coating technique. Chitosan coating also gave a cationic surface charge to the nanoparticles, thus this cationicity further helps intracellular uptake because of interaction with negatively charged cellular membranes. In vivo study results performed on the porcine model indicated that fluorescein isothiocyanate (FITC)-encapsulated nanoparticles coated stents showed fluorescence in neo-intima and media layers compared to solely FITC-polymer coated stents which did not have any fluorescence.

Joo et al., [36] also developed a novel simple coating process to coat of nanoparticles on the stents surfaces. They called this technology as “Ring Shaped Surface Tension Method”. It relies on the principle that a liquid is held between two very closely spaced surfaces in the form of a meniscus, as a result of capillarity. A specially designed ring trails along the immobilized stent surface held along its axis, just like a ring slides over a finger. The nanoparticle suspension was injected between the stent surface and the ring. The deposition occurred at the wedge where the meniscus met the surface when the ring was moved up or down [16]. Uniform deposition of the nanoparticles on the stent surface was observed through the scanning electron microscopy images. The major benefit of this coating process is that the amount of drug on the stent surface can be modified by various ways [36].

In 2013, concercium of Concept Medical Research Private Ltd., Envision Scientific Private Ltd. and Professor Lemos’s working team have reported a new cardiovascular implantable stent. This system consists of a polymer-free novel phospholipid based sirolimus encapsulated nanocarrier system coated on stand-alone balloon catheters and on stents with precrimped balloons [37]. Calcium-phosphorous based components rarely settled at the nanoparticle surface and lead to the release of the encapsulated drug through the pH changes. However, precilinical studies were performed solely on the nanosystem caoted ballons. The clinical phase trials of these systems are still going on to initiate its clinical use.

In summary, the current coating technologies of nanoparticles on cardioavscular implantable stents is an up and coming treatment strategy for the therapy of cardiovascular diseases. Recent developments in this field will further boost the transition of these strategies to the market.

Nanostructured Biomedical Devices

In recent years, a great amount of research is carried out on the nanostructured biomedical implants which designed by the way of surface topography. The first study in this field belonged to Reed et al. in 1998 [38]. They evaluated the integration of microstructure technology with vascular coronary stents that enables delivery of antirestenotic drugs into coronary arteries by piercing through the plaque. However, this system was not able to transport the drugs into the arterial layers on the contrary of nanoparticles. A solution for this problem came from Wang et al. [39]. Wang and co-workers stressed on a polymer free stent coating by opting for a composite coating of carbon nanotubes and magnetic mesoporous silica nanoparticles. Iakovou et al. [40] developed an alternative approach - create a nanotopology – instead of coating nanotubes on to stents. This approach involved the design of a metallic stent surface to create a nanostructure. With carving out a polymer-free surface, the risk of polymer-induced thrombosis disappeared and thus, the major complication of the currently marketed stents will be overcomed. Iakovou et al., also emphasized with this study that the risk and the rate of the formation of thrombosis were significantly lower in bare metallic stents than drug-eluting stents [40]. This in itself puts into question the practice of forming a drug-polymer matrix on the stent surface; it does pave a way to effectively provide a long term drug concentration locally but stems an altogether new problem of thrombosis [41].

Various research groups currently work on the manufacturing technologies of fully metallic wires with nanostructured surface features. High temperature chemical vapor deposition is a well-known technique for chemically etching surfaces that results in desired surface properties. Recently, Loya et al. explored radio frequency plasma for the creation of radially emanating metallic nanopillar structures on stent surfaces creating a dense and porous texture capable of affecting vascular cells [42,43]. Such metallic nanostructures on stent surfaces may provide an opportunity to improve the safety of stent usage, with a polymer-free approach which reduced the risk of thrombus formation in patients. The stent applications which composed of nanostructuring metallic surfaces may possibly lead to reutilization of bare metallic stents [44].

An important perspective on the design and engineering of the stents is their interaction with the endothelial cells and their effects on endothelialization process. Jia et al., investigated the effectiveness of the nanostructured stainless steel stents on endothelialization mechanism of the cells [45]. Paclitaxel loaded nanostructured stainless steel stents was successfully prepared in this concept. In vivo studies, which were performed on a porcine model, demonstrated that (i) the prepared stents lead to rapid re-endothelialization, (ii) they promoted vessel healing with less deposition of fibrin, and (iii) they reduced inflammatory responses when compared to a polymer based sirolimus stent and a bare metallic stent.

In summary, the technology to chisel a metallic stent surface with a nanoscale positively alters the vascular cell responses to specifically boost the adhesion and proliferation of the required cell type, especially endothelial cells leading to re-endothelialization and recede that of the vascular smooth muscle cells that lead to an adverse response. These are expected not only to eliminate the risk of stent related thrombosis, but also to contribute to the restoration of physiological function of treated vessels.

Conclusion and Future Perspectives

Cardiovascular disesases are still remained overly significant in human life. Over the years, cardiovascular biomedical implantation technology has made gradual progress. Utilization of nanoparticulate drug carriers on implantable stent technology is a promising approach for the treatment of cardiovascular diseases. For this reason, future aspects in this field should focus to minimize the toxic effects of drugs, find out new strategies to deliver the nanoparticles by using non-polymeric materials, and move the coating technologies to an endustrial scales. These recent developments on the nanoscale biomedical implants described in this review will allow us to reach these technologies to the market in the near future.

  1. Sanna V, Sechi M (2012) Nanoparticle therapeutics for prostate cancer treatment. Nanomedicine 8: S31-S36.
  2. Graves RA, Ledet GA, Glotser EY, Mitchner DM, Bostanian LA, et al. (2015) Formulation and evaluation of biodegradable nanoparticles for the oral delivery of fenretinide. Eur J Pharm Sci 76: 1-9.
  3. Tisch R, McDevitt H (1996) Insulin-dependent diabetes mellitus. 85: 291–297.
  4. Zhou Y, Liu SG, Peng H, Yu L, He B, et al. (2015) In vivo anti-apoptosis activity of novel berberine-loaded chitosan nanoparticles effectively ameliorates osteoarthritis. Int Immunopharm 28: 34-43.
  5. Jo DH, Kim JH, Lee TG, Kim JH (2015) Size, surface charge, and shape determine therapeutic effects of nanoparticles on brain and retinal diseases. Nanomedicine 11: 1603-1611.
  6. Klugherz BD, Jones PL, Cui X, Chen W, Meneveau NF, et al. (2000) Gene delivery from a DNA controlled-release stent in porcine coronary arteries. Nat Biotechnol 18:1181–1184.
  7. Perlstein I, Connolly JM, Cui X, Song C, Li Q, et al. (2003) DNA delivery from an intravascular stent with denatured collagen-polylactic-polyglycolic acid-controlled release coating: mechanisms of enhanced transfection. Gene Ther 10: 1420–1428.
  8. Lecaroz C, Gamazo C, Blanco-Prieto MJ (2006) Nanocarriers with gentamicin to treat intracellular pathogens. J Nanosci Nanotechnol 6: 3296–3302.
  9. Toti US, Guru BR, Hali M, McPharlin CM, Wykes SM, et al, (2011) Targeted delivery of antibiotics to intracellular chlamydial infections using PLGA nanoparticles. Biomaterials 32: 6606-6613.
  10. Paul SD, Dewangan D (2015) Nanotechnology and Neutraceuticals. Int J Nanomater Nanotechnol Nanomed 1: 30-33.
  11. Kumar SSD, Mahadevan S, Vijayaraghavan R, Mandal AB, MacFarlane DR (2014) Curcumin loaded poly(2-hydroxyethlene methacrylate) nanoparticles from gelled ionic liquid – In vitro cytotoxicity and anti-cancer activity in SKOV-3 cells. Eur J Pharm Sci 51: 34-44.
  12. Buzea C, Pacheco II, Robbie K (2007) Nanomaterials and nanoparticles: sources and toxicity. Biointerphases 2: 17-71.
  13. Naahidi S, Jafari M, Edalat F, Raymond K, Khademhosseini A, et al. (2013) Biocompatibility of engineered nanoparticles for drug delivery. J Control Release 166: 182-194.
  14. Arsiwala AM, Raval AJ, Patravale VB (2013) Nanocoatings on implantable medical devices. Pharm Pat Anal 2: 499–512.
  15. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, et al. (2012) PLGA-based nanoparticles: An overview of biomedical applications. J Control Release 161: 505-522.
  16. Arsiwala A, Desai P, Patravale V (2014) recent advances in micro/nanoscale biomedical implants. J Control Release 189: 25-45.
  17. Pengkai Qia, Manfred F, Maitzb c, Nan Huang (2013) Surface modification of cardiovascular materials and implants. Surf Coat Technol 233: 80–90.
  18. Satzl S, Henn C, Christoph P, Kurz P, Stampfl U, et al, (2007). The efficacy of nanoscale poly [bis(trifluoroethoxy) phosphazene] (PTFEP) coatings in reducing thrombogenicity and late in-stent stenosis in a porcine coronary artery model. Investig Radiol 42: 303–311.
  19. Puranik AS, Dawson ER, Peppas NA (2013) recent advances in drug eluting stents. Int J Pharm 441: 665–679.
  20. Garg S, Serruys PW (2010) Coronary stents: current status. J Am Coll Cardiol 56: S1–S42.
  21. Silber S (2004) When are drug-eluting stents effective? A critical analysis of the presently available data. Z Kardiol 93: 649–663.
  22. Ong AT, Serruys PW (2005) Technology insight: an overview of research in drug-eluting stents. Nat Clin Pract Cardiovasc Med 2: 647–658.
  23. Luscher TF, Steffel J, Eberli FR, Joner M, Nakazawa G, et al, (2007) Drug eluting stent and coronary thrombosis: biological mechanisms and clinical implications. Circulation 115: 1051–1058.
  24. Joner M, Finn AV, Farb A, Mont EK, Kolodgie FD, et al, (2006) Pathology of drug-eluting stents in humans: delayed healing and late thrombotic risk. J Am Coll Cardiol 48: 193–202.
  25. Karagkiozaki V, Karagiannidis PG, Kalfagiannis N, Kavatzikidou P, Patsalas P, et al, (2012) Novel nanostructured biomaterials: implications for coronary stent thrombosis. Int J Nanomedicine 7: 6063–6076.
  26. Mei L, Sun H, Jin X, Zhu D, Sun R, et al, (2007) Modified paclitaxel-loaded nanoparticles for inhibition of hyperplasia in a rabbit arterial balloon injury model. Pharm Res 24: 955–962.
  27. Catto V, Farè S, Freddi G, Tanzi MC (2014) Vascular tissue engineering: recent advances in small diameter blood vessel regeneration. ISRN Vascular Med, Article ID 923030; 1-27.
  28. Labhasetwar V, Song C, Humphrey W, Shebuski R, Levy RJ (1998) Arterial uptake of biodegradable nanoparticles: effect of surface modification. J Pharm Sci 87: 1229-1234.
  29. Song CX, Labhsetwar V, Murphy H, Qu X, Humphrey WR, et al, (1997) Formulation and characterization of biodegradable nanoparticles for intravascular local drug delivery. J Control Release 43: 197–212.
  30. Humphrey WR, Erickson LA, Simmons CA, Northrup JL, Wishka DGet al, (1997) The effect of intramural delivery of polymeric nanoparticles loadedwith antiproliferative 2-aminochrome U-86983 on neointimal hyperplasia development in balloon-injured porcine coronary artery. Adv Drug Deliv Rev 24: 87–108.
  31. Prabha S, Zhou WZ, Panyam J, Labhasetwar V (2002) Size-dependency of nanoparticle mediated gene transfection: studies with fractionated nanoparticles. Int J Pharm 244: 105–115.
  32. Davda J, Labhsetwar V (2002) Characterization of nanoparticle uptake by endothelial cells. Int J Pharm 233: 51–59.
  33. Luderer F, Lobler M, Rohm HW, Gocke G, Kunna K, et al, (2011) Biodegradable sirolimus-loaded poly(lactide) nanoparticles as drug delivery system for the prevention of in-stent restenosis in coronary stent application. J Biomater Appl 25: 851–875.
  34. Westedt U, Barbu-Tudoran L, Schaper AK, Kalinowski M, Alfke H, et al, (2004) Effects of different application parameters on penetration characteristics and arterial vessel wall integrity after local nanoparticle delivery using a porous balloon catheter. Eur J Pharm Biopharm 58: 161–168.
  35. Nakano K, Egashira K, Masuda S, Funakoshi K, Zhao G, et al, (2009) Formulation of nanoparticle-eluting stents by a cationic electrodeposition coating technology. JACC Cardiovasc Interv 2: 277–283.
  36. Joo J, Nam HY, Nam SH, Baek I, Park JS (2009) A novel deposition method of PLGA nanoparticles on coronary stents. Bull Korean Chem Soc 30: 1085–1087.
  37. Lemos PA, Farooq V, Takimura CK, Gutierrez PS, Virmani R, et al, (2013) Emerging technologies: polymer-free phospholipid encapsulated sirolimus nanocarriers for the controlled release of drug from a stent-plus balloon or a stand-alone balloon catheter. Euro Intervention 9: 148–156.
  38. Reed ML, Wu C, Kneller J, Watkins S, Vorp DA, et al, (1998) Micromechanical devices for intravascular drug delivery. J Pharm Sci 87: 1387–1394.
  39. Wang Y, Zhang W, Zhang J, Sun W, Zhang R, et al, (2013) Fabrication of a novel polymer-free nanostructured drug-eluting coating for cardiovascular stents. ACS Appl Mater Interfaces 5: 10337–10345.
  40. Iakovou I, Schmidt T, Bonizzoni E, Ge L, Sangiorgi GM, et al, (2005) Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. J Am Med Assoc 293: 2126–2130.
  41. Shuchman M (2006) Trading restenosis for thrombosis? New questions about drug eluting stents. N Engl J Med 355: 1949–1952.
  42. Oh S, Daraio C, Chen L, Pisanic T, Finones R, et al, (2006) significantly accelerated osteoblast cell growth on aligned TiO2 nanotubes. J Biomed Mater Res a 78: 97–103.
  43. Loya MC, Park E, Chen LH, Brammer KS, Jin S (2010) Radially arrayed nanopillar formation on metallic stent wire surface via radio-frequency plasma. Acta Biomater 6: 1671–1677.
  44. Loya MC, Brammer KS, Choi C, Chen LH, Jin S (2010) Plasma-induced nanopillars on bare metallic coronary stent surface for enhanced endothelialization. Acta Biomater, 6: 4589–4595.
  45. Jia H, Liu H, Kong J, Hou J, Wu J, et al, (2011) a novel polymer-free paclitaxel-eluting stent with a nanoporous surface for rapid endothelialization and inhibition of intimal hyperplasia: comparison with a polymer-based sirolimus-eluting stent and bare metal stent in a porcine model. J Biomed Mater Res a 98: 629–637.

Follow us on Academia.edu