Sei sulla pagina 1di 6

Available online at www.sciencedirect.

com

Pharmaceutical nanocrystals
George Duo Wang1, Franck P Mallet2, Francois Ricard2 and Jerry YY Heng1
Development of poorly soluble and/or permeable drug molecules using nanocrystal formulations has proven to be highly successful due to the greater surface/volume ratio, resulting in improvements in dissolution and bioavailability as well as enhanced permeability. These submicron nanoparticles have similar benets to conventional nanoparticles which are less than 100 nm but are scalable and can be applied in all existing manufacturing processes and dosage forms. This opinion focuses on recent developments in the preparation of nanocrystals and the role of surface properties on optimizing dissolution and targeted delivery. Some key technical challenges and environmental impact are also considered.
Addresses 1 Surfaces and Particle Engineering Laboratory, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom 2 Particle Generation, Control & Engineering, Product Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom Corresponding author: Heng, Jerry YY (jerry.heng@imperial.ac.uk)

electronics, optics, consumer products and materials science, nanotechnology has been readily embraced in product development and with great effect [10,11]. Nanoparticles are also playing a role in synergizing chemical engineering with other physical and life sciences [12,13]. A more pressing issue from which nanonization can be applied is in conventional pharmaceutical development. Solubility and permeability as described by the biopharmaceutical classication system (BCS) are key drivers for conventional drug development [14]. Classes II and IV which are both poorly water soluble, often require manipulation of the physical form, that is, the crystalline polymorph in order to achieve adequate dissolution [15]. Currently, focus has been made on effective solid-state screening and salt selection [16]. Formation of crystalline nanoparticles or nanocrystals (in this case, submicron, 1000 nm or less) can improve both solubility and permeability and ultimately bioavailability. Increase in surface area to volume ratio will result in increased cellular uptake due to the 100-fold size reduction. Animal studies have shown that oral bioavailability is greater for solid lipid nanoparticles compared to nanocrystals, the nanocrystal formulation presents a signicant improvement which is both simpler and scalable using various existing technologies [17,18]. Many drugs developed for neurological diseases exhibit poor permeability across the bloodbrain barrier, where endothelial cells prevent diffusion of large hydrophilic drug molecules into the cerebrospinal uid [19]. Nanocrystals present an opportunity to exploit other cellular uptake processes such as endocytosis and also surface functionalization to utilize the potential for targeted drug delivery [20].

Current Opinion in Chemical Engineering 2012, 1:102107 This review comes from a themed issue on Nanotechnology Edited by Hua Chun Zeng Available online 26th December 2011 2211-3398/$ see front matter Crown Copyright # 2011 Published by Elsevier Ltd. All rights reserved. DOI 10.1016/j.coche.2011.12.001

Introduction
Nanoparticles have shown great promise in terms of biomedical applications, ranging from novel drug delivery platforms and drug targeting to diagnostics and tissue engineering [1,25]. By denition a nanoparticle is a particle having one or more dimensions of the order of 100 nm or less and considered to have novel properties that differentiate nanoparticles from the bulk material typically develop at a critical length scale of under 100 nm [6]. Over the past two decades, there has been continued interest and success in utilizing nanoparticles in the development of pharmaceuticals. Cost and regulatory approval have been the main limiting factors, with less than 30 marketed formulations since Ambisome1 (liposomal amphotericin B) was rst approved in 1990 [7,8,9]. By comparison, in other elds such as
Current Opinion in Chemical Engineering 2012, 1:102107

Preparation of nanocrystals
The majority of pharmaceuticals are developed in the most stable crystalline solid form. This, however, can present issues in terms of poor solubility and bioavailability [21]. Without altering the chemistry of the drug molecule, the amorphous form can be prepared but this often conversely lacks the required stability for manufacturing and shelf life. Nanocrystals are intermediates between the crystalline and amorphous states, beneting from enhanced dissolution due to higher surface energy and surface area [2224]. Their preparation therefore cannot be conducted using conventional crystallization approaches. Stabilization, ltration, and characterization also become more difcult as a result of the reduced length scale [25].
www.sciencedirect.com

Pharmaceutical nanocrystals Wang et al. 103

Figure 1

Bottom Up (e.g. precipitation)

Feed Solution

Nanosuspension (with stabilizer)

Top Down (e.g. attrition)


Current Opinion in Chemical Engineering

Bottom up or top down approach to preparing nanocrystals.

Nanocrystals can be obtained directly through bottom up by modied crystallization/antisolvent precipitation (but this approach is less common [26]) or indirectly top down by mechanical breakage/attrition of a crystalline powder (see Figure 1). The production of nanosized particle by direct crystallization/precipitation can be carried out using extreme supersaturation conditions in order to favor nucleation over growth, but kinetic phase(s) will be promoted. This technique can be suitable for preparing large quantities of material which has no polymorph issue, or if a metastable polymorph is required for formulation. The stability upon agglomeration during rapid crystallization, needs to be assessed for development feasibility [27]. Another way to generate nanocrystals is to limit the amount of material available for crystallization by reducing the working volume (conned crystallization). Microuidics set-up or emulsion crystallization can be the method of choice to generate microcrystalline material, but the throughput of these techniques is a major limiting factor, as for microuidics it will be restrained by the quantity of parallel set-up available or for emulsion crystallization, the operating volume is critical for mixing scaling-up. Spray drying or spray freeze drying uses the same approach by crystallizing/precipitating particles within or on the surface of the droplet by evaporation of solvent [28]. The feed and the drying kinetics (operating conditions) will determine the output material properties, but use of these equipments at larger scale can be envisaged, and powder obtained from spray drying can be handled and used for subsequent conventional formulation [28]. Spray freeze drying combines both current spray drying and freeze drying technology,
www.sciencedirect.com

whereby the atomized feed solution is frozen and dried to produce monodispersed submicron spherical solute particles. This reduces the time and cost associated with conventional lyophilization. Solubility can be further controlled by the addition of polymers for encapsulation and solid dispersions [29]. Porosity and crystallinity can also be controlled by altering processing parameters [30]. These existing methods are further optimized by the use of supercritical uids (SCF) which utilize the unique properties of SCF [31]. In order to be comprehensive in this review, microuidics can also be considered in the preparation of nanocrystals as they reduce the scale of process(s) to improve efciency and reduce waste, in addition presenting a physical environment not achievable at full scale. It enables nanocrystallization and parallel scale-out in order to meet industrial needs [32]. Mechanical milling can be carried out on dry powders by using high-energy ball milling, cryo-milling [33] or jet milling (micronization). In these examples, the process is highly energetic, thus the likelihood of amorphous formation is not negligible, therefore monitoring will be needed as well as prevention of contamination from milling equipment. On the other hand, milling of slurries reduces the likelihood of generating amorphous material. Other common techniques to achieve nanosized material include wet bead milling, microuidization, and piston gap homogenization. As these techniques involve the use of a solvent, the generation of amorphous material is minimized, but these techniques necessitate that there are no hydrate or solvate known for the operating solvent. Wet bead milling and high-pressure homogenization are the most widely used
Current Opinion in Chemical Engineering 2012, 1:102107

104 Nanotechnology

Figure 2

Ball Milling Micronization

Scalability/control

Wet Bead Milling

High Pressure Homogenization

Freeze Spray Drying

Spray Drying

Microfluidics

Precipitation/ Crystallization

Particle size distribution


Current Opinion in Chemical Engineering

Preparation of nanocrystals depending on scalability/control and particle size distribution requirements.

methods employed by industry and have been patented (Elan Nanocrystal1 and SkyePharma Dissocubes1, respectively) [27,34,35]. One of the main properties of nanoparticles is their high surface energy but this induces a tendency of those particles to aggregate, therefore nanosuspensions need to be stabilized to maintain dispersion, preventing aggregation or growth which may occur via Ostward ripening. Stabilizing agents used need to be pharmaceutically acceptable, typical excipients include polymers (cellulose based, povidone, pluronics) and surfactants (polysorbate, anionic). These can then be used directly in the liquid form with minor processing in oral, parenteral or topical dosage forms. Drying of the nanosuspensions can be carried out using existing equipment, for example, uidized bed or spray drying or more optimized novel approaches such as spray freeze drying, freeze drying, microuidics, and supercritical processing [36]. Holland et al. have presented the use of a combination of wet bead milling and spray drying at plant scale to prepare nanosized pharmaceutical particles [37]. Filtration of nanosuspension presents more of a challenge as the cake will be compressible, which lead to longer ltration time, and the dry powder will be highly subject to agglomeration [38]. Dried API can then be further formulated by conventional means. An extensive review of current ller et al. [39,40] strategies is presented by Mu (Figure 2).
Current Opinion in Chemical Engineering 2012, 1:102107

In terms of health and safety, the handling of dry nanoparticles will undoubtedly require special attention regarding dust explosivity as the quantity of nes which can be suspended within the equipment atmosphere will not be negligible. Therefore, working in an inert environment may be obligatory. The protection of the people inside and outside the facility from ultrane particle will need to be guaranteed by high performance lters [41].

Physical properties of nanocrystals


The therapeutic advantages of nanocrystals are predominantly due to reduction in particle size (less than 1 mm) [42]. Suspensions of nanocrystals with 100% drug loading are much simpler and easier to prepare and scale-up than carrier based nanoparticles, nanocapsules or nanospheres, with only the need for stabilization. Nanocrystals will also have similar gains in bioavailability and the relative monodispersity minimizes the likelihood of Ostwald ripening [17,38,43,44]. Improved solubility and dissolution is due to higher saturation solubility and dissolution velocity as a result of the increase in surface area [4547], and the reduction in particle radius (Kelvin equation). Enhancement in bioavailability can also be attributed to partial or full conversion to the amorphous form upon processing, with the metastable form or nanocrystalline phase having higher surface energy [48,49]. Also it has been shown that increased adhesion may lead to improved oral absorption [50]. Surface functionalization can alter and further enhance properties of nanocrystals
www.sciencedirect.com

Pharmaceutical nanocrystals Wang et al. 105

Figure 3

Targeting Ligand Adhesion Ligand

Fluorescence Ligand

Uptake Ligand Image Contrast Ligand Magnetic Ligand

Surfactant Nanocrystal Core

Controlled Release Polymer Layer


Current Opinion in Chemical Engineering

Surface functionalization of nanocrystals.

depending on the needs for release prole and targeted drug delivery [51,52,53] (Figure 3). As well as the usual characterization techniques to determine physical properties, that is, dynamic light scattering (DLS) and scanning electron microscopy, novel techniques such as nanoparticle tracking analysis (NTA), dual polarization interferometry (DPI) have emerged. NTA is based on DLS but actually tracks the Brownian motion of nanoparticles quantitatively which enables the study of nanocrystal and stabilizer interactions, whereas DPI is a much more powerful technique based on interference through waveguides which can be used to monitor, for example, the nucleation of nanocrystals at a molecular level [54,55].

particles in a dry powder inhalation whereby surface properties can be further optimized by ne particles [61].

Conclusions
Nanocrystals and nanosuspensions are fast growing alternative formulation routes being readily used by industry to deal with poor soluble drug compounds during development. Although not traditionally considered as nanoparticles, they are somewhat analogous and provide some of their unique properties as well being practically feasible for large-scale manufacturing. Further research is required to optimize production of nanocrystals and fully utilize their novel surface properties.

References and recommended reading Role of surface properties


Surface properties play an important role throughout development of nanocrystals, for example during preparation, in the coating of beads for wet milling to maximize attrition and minimize erosion [39]. Stabilization occurs when the interaction(s) between nanocrystals and excipient(s) are optimized which is a function of surface chemistry and has been shown to be anisotropic for crystalline pharmaceutical powders [43,56]. Surface energy also plays a major role in stabilization which was found to be heterogenous for pharmaceutical powders and highly dependent on processing history [5759]. A recent development based on nite dilution inverse gas chromatography at different surface coverages, enables the degree of surface energy heterogeneity to be determined. In addition the surface energy heterogeneity can be modeled [60]. This approach overcomes a range of drawbacks and limitations to conventional powder bed approaches [57]. Post-milling, nanocrystals can also be delivered through the inhaled route by using carrier
www.sciencedirect.com Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest

1. Farokhzad OC, Langer R: Impact of nanotechnology on drug  delivery. ACS Nano 2009, 3:16-20. This is an excellent concise overview of the use of nanotechnology in pharmaceutical development by one of the foremost experts in drug delivery and pharmaceutical nanotechnology. 2. 3. Irvine DJ: Drug delivery: one nanoparticle, one kill. Nat Mater 2011, 10:342-343. Cormode DP, Skajaa T, Fayad ZA, Mulder WJM: Nanotechnology in medical imaging probe design and applications. Arterioscler Thromb Vasc Biol 2009, 29:992-1000. Shi J, Votruba AR, Farokhzad OC, Langer R: Nanotechnology in drug delivery and tissue engineering: from discovery to applications. Nano Lett 2010, 10:3223-3230. Joachim C: To be nano or not to be nano? Nat Mater 2005, 4:107-109. BSI: Nanoparticles. Vocabulary. United Kingdom: British Standards Institution; 2011:. 36. Current Opinion in Chemical Engineering 2012, 1:102107

4.

5. 6.

106 Nanotechnology

7. Allen TM, Cullis PR: Drug delivery systems: entering the  mainstream. Science 2004, 303:1818-1822. This review is slightly dated but provides a fantastic detailed history of the application of nanoparticles in pharmaceuticals. 8. Dusastre V: Nanomedicine: a matter of rhetoric? Nat Mater  2006, 5:243. This short editorial summarizes the economic impact of nanotechnology on current pharmaceutical development and the growing investment from government and private sector. 9. Eier AC, Thaxton CS: Nanoparticle therapeutics: FDA approval, clinical trials, regulatory pathways, and case study. In Biomedical Nanotechnology, vol 726. Edited by Hurst SJ. Humana Press; 2011:325-338.

and stabilization methods. J Pharm Pharmacol 2010, 62: 1569-1579. 28. Vehring R: Pharmaceutical particle engineering via spray  drying. Pharm Res 2008, 25:999-1022. This expert review from Reinhard Vehing gives to the reader an in depth theoretical and experimental approaches to the particle engineering using spray drying. The literature review conducted by the author is complete with more than 400 references. This article needs to be studied not only read as it teaches a lot on the possibility of the technique. 29. Six K, Verreck G, Peeters J, Brewster M, Mooter GVd: Increased physical stability and improved dissolution properties of itraconazole, a class II drug, by solid dispersions that combine fast- and slow-dissolving polymers. J Pharm Sci 2004, 93:124-131. 30. Lee J, Cheng Y: Critical freezing rate in freeze drying nanocrystal dispersions. J Control Release 2006, 111:185-192. 31. Jung J, Perrut M: Particle design using supercritical uids: literature and patent survey. J Supercrit Fluids 2001, 20:179-219. 32. Panagiotou T, Mesite SV, Fisher RJ: Production of noroxacin nanosuspensions using microuidics reaction technology through solvent/antisolvent crystallization. Ind Eng Chem Res 2009, 48:1761-1771. 33. Niwa T, Nakanishi Y, Danjo K: One-step preparation of pharmaceutical nanocrystals using ultra cryo-milling technique in liquid nitrogen. Eur J Pharm Sci 2010, 41:78-85. ller RH: Drug nanocrystals of poorly soluble drugs 34. Keck CM, Mu produced by high pressure homogenisation. Eur J Pharm Biopharm 2006, 62:3-16. 35. Kesisoglou F, Panmai S, Wu Y: Nanosizing oral formulation  development and biopharmaceutical evaluation. Adv Drug Deliv Rev 2007, 59:631-644. This review discusses the need for nanosizing and includes fundamental principles, production and characterization of the nanoformulations, for both small molecule and biopharmaceuticals. 36. Beirowski J, Inghelbrecht S, Arien A, Gieseler H: Freeze-drying of nanosuspensions: 1. Freezing rate versus formulation design as critical factors to preserve the original particle size distribution. J Pharm Sci 2011, 100:1958-1968. 37. Holland SJ, Knight WA, Leonard GS: Wet milling process. US 2004/0089753 A1. United States; 2004:9. 38. Na GC, Stevens J, Yuan BO, Rajagopalan N: Physical stability of ethyl diatrizoate nanocrystalline suspension in steam sterilization. Pharm Res 1999, 16:569-574. ller RH: Nanocrystal technology, drug 39. Junghanns J-UAH, Mu  delivery and clinical applications. Int J Nanomed 2008, 3: 295-310. ller from Free University of Berlin is one of the worlds foremost Dr Mu experts on nanocrystals and nanosuspensions with over 300 publications relating to this eld, and this is one of his most recent and easily accessible reviews. This and others by him and his group are must reads in order to gain an insight to this eld of research. ller RH, Shegokar R, Gohla S, Keck CM: Nanocrystals: 40. Mu production, cellular drug delivery, current and future products. In Intracellular Delivery, vol 5. Edited by Prokop A. Netherlands: Springer; 2011:411-432. 41. Health and Safety Executive: Literature review explosion hazards associated with nanopowders. Press Release March 2004. 42. Lee J: Drug nano- and microparticles processed into solid dosage forms: physical properties. J Pharm Sci 2003, 92:20572068. 43. Choi JY, Yoo JY, Kwak HS, Nam BU, Lee J: Role of polymeric stabilizers for drug nanocrystal dispersions. Curr Appl Phys 2005, 5:472-474. 44. Patravale VB, Date AA, Kulkarni RM: Nanosuspensions: a promising drug delivery strategy. J Pharm Pharmacol 2004, 56:827-840. 45. Dressman JB, Amidon GL, Reppas C, Shah VP: Dissolution testing as a prognostic tool for oral drug absorption: www.sciencedirect.com

10. Korkin A, Kristic PS, Wells JC: Nanotechnology for Electronics, Photonics, and Renewable Energy. Springer; 2010. 11. Nalwa HS: Handbook of Nanostructured Materials and Nanotechnology. Academic Press; 2000. 12. Sotiriou GA, Pratsinis SE: Engineering nanosilver as an antibacterial, biosensor and bioimaging material. Curr Opin Chem Eng 2011, 1:3-10. 13. Hua Chun Z: Preparation and integration of nanostructured titanium dioxide. Curr Opin Chem Eng 2011, 1:11-17. s H, Shah VP, Crison JR: A theoretical basis 14. Amidon GL, Lennerna for a biopharmaceutic drug classication: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res 1995, 12:413-420. 15. Blagden N, de Matas M, Gavan PT, York P: Crystal engineering  of active pharmaceutical ingredients to improve solubility and dissolution rates. Adv Drug Deliv Rev 2007, 59:617-630. An excellent review discussing various crystal engineering approaches, including theory and concept, benets and disadvantages of the different methods, for poorly soluble drugs. 16. Huang L-F, Tong W-Q: Impact of solid state properties on developability assessment of drug candidates. Adv Drug Deliv Rev 2004, 56:321-334. ller RH, Runge S, Ravelli V, Mehnert W, Thu nemann AF, 17. Mu Souto EB: Oral bioavailability of cyclosporine: solid lipid W nanoparticles (SLN ) versus drug nanocrystals. Int J Pharm 2006, 317:82-89. 18. Muller RH, Keck CM: Challenges and solutions for the delivery of biotech drugs a review of drug nanocrystal technology and lipid nanoparticles. J Biotechnol 2004, 113:151-170. 19. Tamai I, Tsuji A: Transporter-mediated permeation of drugs across the bloodbrain barrier. J Pharm Sci 2000, 89:1371-1388. e J-M, Port M: Recent advances in iron 20. Corot C, Robert P, Ide oxide nanocrystal technology for medical imaging. Adv Drug Deliv Rev 2006, 58:1471-1504. 21. Lipinski C: Poor aqueous solubility an industry wide problem in drug discovery. Am Pharm Rev 2002, 5:82-85. 22. Merisko-Liversidge E, Sarpotdar P, Bruno J, Hajj S, Wei L, Peltier N, Rake J, Shaw JM, Pugh S, Polin L et al.: Formulation and antitumor activity evaluation of nanocrystalline suspensions of poorly soluble anticancer drugs. Pharm Res 1996, 13:272-278. ller RH, Peters K: Nanosuspensions for the formulation of 23. Mu poorly soluble drugs: I. Preparation by a size-reduction technique. Int J Pharm 1998, 160:229-237. 24. Rabinow BE: Nanosuspensions in drug delivery. Nat Rev Drug Discov 2004, 3:785-796. 25. Gao L, Zhang D, Chen M: Drug nanocrystals for the formulation of poorly soluble drugs and its application as a potential drug delivery system. J Nanopart Res 2008, 10:845-862. 26. Hu J, Johnston KP, Williams RO: Nanoparticle engineering processes for enhancing the dissolution rates of poorly water soluble drugs. Drug Dev Ind Pharm 2004, 30:233-245. 27. Peltonen L, Hirvonen J: Pharmaceutical nanocrystals by nanomilling: critical process parameters, particle fracturing Current Opinion in Chemical Engineering 2012, 1:102107

Pharmaceutical nanocrystals Wang et al. 107

immediate release dosage forms. Pharm Res 1998, 15:11-22. 46. Hecq J, Deleers M, Fanara D, Vranckx H, Amighi K: Preparation and characterization of nanocrystals for solubility and dissolution rate enhancement of nifedipine. Int J Pharm 2005, 299:167-177. ller RH, Keck CM: Kinetic solubility and 47. Mauludin R, Mu dissolution velocity of rutin nanocrystals. Eur J Pharm Sci 2009, 36:502-510. 48. Hancock BC, Parks M: What is the true solubility advantage for amorphous pharmaceuticals? Pharm Res 2000, 17: 397-404. 49. Hancock BC, Zogra G: Characteristics and signicance of the amorphous state in pharmaceutical systems. J Pharm Sci 1997, 86:1-12. ne D, Ponchel G: Bioadhesion of solid oral dosage forms, 50. Duche why and how? Eur J Pharm Biopharm 1997, 44:15-23. 51. Otsuka H, Nagasaki Y, Kataoka K: PEGylated nanoparticles for biological and pharmaceutical applications. Adv Drug Deliv Rev 2003, 55:403-419. 52. Merisko-Liversidge EM, Liversidge GG: Drug nanoparticles: formulating poorly water-soluble compounds. Toxicol Pathol 2008, 36:43-48. ller RH, Jacobs C, Kayser O: Nanosuspensions as particulate 53. Mu  drug formulations in therapy: rationale for development and what we can expect for the future. Adv Drug Deliv Rev 2001, 47:3-19. ller from Free University of Another very comprehensive review by Dr Mu Berlin on nanocrystals and nanosuspensions. Like Ref. [39], this and others by him and his group are must reads in order to gain an insight to this eld of research.

54. Filipe V, Hawe A, Jiskoot W: Critical evaluation of nanoparticle tracking analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates. Pharm Res 2010, 27:796-810. 55. Swann MJ, Peel LL, Carrington S, Freeman NJ: Dual-polarization interferometry: an analytical technique to measure changes in protein structure in real time, to determine the stoichiometry of binding events, and to differentiate between specic and nonspecic interactions. Anal Biochem 2004, 329:190-198. 56. Heng JYY, Bismarck A, Lee AF, Wilson K, Williams DR: Anisotropic surface energetics and wettability of macroscopic form I paracetamol crystals. Langmuir 2006, 22:2760-2769. 57. Thielmann F, Burnett DJ, Heng JYY: Determination of the surface energy distributions of different processed lactose. Drug Dev Ind Pharm 2007, 33:1240-1253. 58. Ho R, Wilson DA, Heng JYY: Crystal habits and the variation in surface energy heterogeneity. Cryst Growth Des 2009, 9: 4907-4911. 59. Ho R, Hinder SJ, Watts JF, Dilworth SE, Williams DR, Heng JYY: Determination of surface heterogeneity of D-mannitol by sessile drop contact angle and nite concentration inverse gas chromatography. Int J Pharm 2010, 387:79-86. 60. Jefferson AE, Williams DR, Heng JYY: Computing the surface energy distributions of heterogeneous crystalline powders. J Adhes Sci Technol 2011, 25:339-355. 61. Ho R, Muresan AS, Hebbink GA, Heng JYY: Inuence of nes on  the surface energy heterogeneity of lactose for pulmonary drug delivery. Int J Pharm 2010, 388:88-94. All books cited are appropriate starting points into understanding the vast eld of nanotechnology, nanoparticles and more specically nanocrystals and nanosuspensions with relation to development of pharmaceuticals.

www.sciencedirect.com

Current Opinion in Chemical Engineering 2012, 1:102107

Potrebbero piacerti anche