Thesis - Archive ouverte UNIGE
Transcription
Thesis - Archive ouverte UNIGE
Thesis Novel micellar systems for the formulation of poorly water soluble drugs : biocompatibility aspects and pharmaceutical applications DUMONTET MONDON, Karine Abstract Amongst the large number of novel drugs, 95% are lipophilic and poorly water soluble. Particularly, this renders their aqueous formulation very difficult. In this regard this thesis focused on polymeric micelles based on novel MPEG-hexPLA copolymers forming a hydrophilic shell and a very hydrophobic core that favors the incorporation of poorly water soluble drugs. Although the drug hydrophobicity and water solubility are the main parameters in respect to their incorporation efficiency, structural parameters showed an important influence, too. The biocompatibility of the novel MPEG-hexPLA micelles could be proven and promising pharmaceutical applications can be envisioned. An intravenous application for ovarian cancer diagnostics was demonstrated in vitro and in vivo in rats with the administration of a micelle formulation containing the fluorescent compound hypericin. A topical dermatology application with various antifungal compounds and possible use for oral delivery were also evidenced. Reference DUMONTET MONDON, Karine. Novel micellar systems for the formulation of poorly water soluble drugs : biocompatibility aspects and pharmaceutical applications. Thèse de doctorat : Univ. Genève, 2010, no. Sc. 4258 URN : urn:nbn:ch:unige-171669 Available at: http://archive-ouverte.unige.ch/unige:17166 Disclaimer: layout of this document may differ from the published version. [ Downloaded 25/10/2016 at 04:54:18 ] UNIVERSITÉ DE GENÈVE FACULTÉ DES SCIENCES Section des sciences pharmaceutiques Pharmacie galénique Professeur Robert Gurny Docteur Michael Möller Novel Micellar Systems for the Formulation of Poorly Water Soluble Drugs : Biocompatibility Aspects and Pharmaceutical Applications THÈSE présentée à la Faculté des sciences de l’Université de Genève pour obtenir le grade de Docteur ès sciences, mention interdisciplinaire par Karine Dumontet Mondon de Lyon (FRANCE) Thèse N°:4258 Genève Atelier de reproduction Repromail 2011 A Maëlou, Léo et Bertrand A ma famille, belle-famille et amis Remerciements Mes premiers remerciements s’adressent au jury qui a accepté d’évaluer mon travail de thèse : Professeur Jean Marie Devoisselle (Université de Montpellier I, Montpellier, France), Dr. Barbara Lueckel (F.Hoffman-La Roche AG, Bâle, Suisse) et Dr. Khadija Schwach (Novozymes, Copenhague, Danemark). Je tiens ensuite à remercier toutes les personnes qui ont contribué à cette recherche : Le Professeur Robert Gurny et le Docteur Michael Möller pour avoir proposé un sujet aussi passionant et touchant à de nombreux aspects pharmaceutiques que j’ai découvert au fur et à mesure de la thèse. Merci Robert pour votre confiance, votre disponibilité et les conditions de travail particulièrement favorables dont j’ai pu bénéficier. Merci Michael pour ton soutien sans faille, tes idées, et aussi tes critiques. Le Professeur Borkovec et le Docteur Andrea Vaccaro (Université de Genève, Départment de Chimie Minérale, Analytique et Appliquée) pour la disponibilité et l’utilisation du goniomètre. Le Docteur Christoph Bauer (Université de Genève, Département de Biologie moléculaire, Bioimaging Platform) pour les analyses en Microscopie à Transmission Electronique. Le Professeur Carrupt, et les Docteur Sophie Martel et Bruno Bard (Université de Genève, Pharmacochimie) pour la disponibilité du logiciel ACD/Chemsketch ainsi que pour leur aide scientifique dans la détermination de la solubilité dans l’eau de certaines drogues. Mes acolytes et ex-acolytes du labo 468, Thomas, Leila, Delia, Benjamin, Sema, Claudia, Lutz et Sarra. Merci Claudia pour ton soutien, pour les innombrables petits services que tu as faits pour moi qui m’ont énormément facilité la vie, merci aussi pour ton oreille attentive et pour ton amitié. Merci Lutz pour ton humour, ta joie de vivre et ta serviabilité. Merci Sarra pour tes corrections d’anglais, mais surtout merci pour nos petites pauses « papotage » qui permettaient de souffler un peu pendant l’écriture du manuscrit. Merci à tous. Cela a été un vrai plaisir de travailler à vos côtés, vous allez tous me manquer. Mes collègues et ex-collègues, je pense particulièrement à Magali, Marie, Dany et Adriana. Merci Mag de m’avoir initié au CAM, à la culture cellulaire et aux essais in vivo, qui ont fortement contribué au « succès » de ma thèse. Merci également pour ton soutien (même à distance) et aussi pour cette amitié que nous avons liée après de longues heures d’extraction d’organes et de TP info … Merci Marie pour nos discussions sur comment allier vie familiale et l’accomplissement d’une thèse, merci aussi d’organiser des petites sorties entre « mamans » qui font tellement de bien au moral. Merci Dany aussi pour les discussions mais plutôt «travaux » et « comment gérer nos filles respectives ! ». Merci Adriana pour ton soutien et ton oreille attentive. J’ai beaucoup apprécié de pouvoir échanger nos sentiments de futures mamans, et de mamans au cours de ces années de recherche. Je pense aussi au Docteur Florence Delie pour sa disponibilité à répondre à mes questions d’ordre pharmaceutique mais aussi pour tous les bons plans qu’elle a pu me conseiller. Merci Flo. Je tiens aussi à remercier Emilie, pour les expériences de fluorescence avec le Nile Red. Merci pour ta disponibilité et ton sourire, ainsi que les quelques heures de basket que nous avons pu faire ensemble. Je n’oublie pas non plus le Docteur Norbert Lange pour avoir son humour sarcastique pendant les TP info des étudiants. Je pense aussi au groupe PDT, toujours au summum dans l’ambiance du labo et pour organiser des évènements; le groupe d’Archamps, toujours présent malgré la distance géographique ; et enfin les nouveaux arrivants avec qui j’ai eu beaucoup de plaisir à discuter pendant les derniers mois de la thèse (Flo, Julie, Bénédicte, Amandine…). L’équipe technique et administrative, Myrtha, Florence, Brigitte et Marco. Merci Myrtha pour ton support administratif et moral, encore plus appréciable en fin de thèse, merci aussi pour les chocolats ou autres gourmandises que tu nous fais partager et qui sont toujours les bienvenues à la cafet’. Merci Florence pour toujours avoir répondu à mes questions administratives. Merci Brigitte pour ta serviabilité et ton aide précieuse dans la commande de produits, la connaissance des fournisseurs, mais surtout pour les agréables moments à discuter avec toi dans le labo ou à la cafet’. Enfin un énorme merci à Marco pour son assistance technique sans limite, merci d’avoir toujours trouvé le temps de réparer la HPLC même si ton emploi du temps était chargé. Finalement, je tiens aussi à remercier mes amis, particulièrement Nath et Caro, ma bellefamille, ma sœur et mes parents pour leur présence, leur soutien et leurs encouragements tout au long de ces 5-6 années. Enfin, un spécial merci à Bertrand pour son amour qui nous a permis de construire une vie de famille bien remplie avec la présence de nos deux magnifiques petits bambins. Table of contents Table of contents INTRODUCTION………………………………………………………………………………… 1 CHAPTER I Colloidal Drug Delivery Systems – Recent Advances with Polymeric Micelles…………….... 3 CHAPTER II Formulation of Poorly Water Soluble Drugs with MPEG-hexPLA Micelles: Investigations on the Effects of Drug Physical-Chemical Parameters on the Incorporation…………………………………………………………………………………… 33 CHAPTER III Novel Cyclosporin A Formulations Using MPEG-Hexyl Substituted Polylactide Micelles: A Suitability Study…………………………………………………………………………….. 61 CHAPTER IV MPEG-hexPLA Micelles as Novel Carriers for Hypericin, a Fluorescent Marker for Use in Cancer Diagnostics………………………………………………………………………...... 91 CHAPTER V Novel Micellar Formulations to Increase Cutaneous Bioavailability of Azole Antifungals…………………………………………………………………………………..... 117 CHAPTER VI MPEG-hexPLA Micelle Formulations for Oral Delivery of Poorly Water Soluble Drugs: Investigations with Cinnarizine as a Model Drug ………………………………………….…. 139 CONCLUSIONS AND PERSPECTIVES………………………………………………………....... 149 SUMMARY (IN FRENCH)……………………………………………………………………… 153 ABBREVIATIONS……………………………………………………………………………… 155 1 1 1 1 1 1234567831529 9 9 1 Introduction 1 Introduction Technological advances in drug discovery such as high-throughput screening (HTS) have considerably increased the number of “lead” compounds for a specific biological activity. The majority of these compounds are poorly soluble in water, limiting their success in the clinic because low solubility is often the cause of low bioavailability. Formulating such entities is therefore a real challenge since stability problems are often encountered. Several strategies have been developed to overcome this solubility issue, one of which is the incorporation of the drug into polymeric micelles. The poorly soluble drug is loaded into the carrier core which has a certain affinity with the drug and around a protective shell which prevents drug precipitation and exposure to the immune system. To obtain such specific structures, amphiphilic copolymers with a hydrophobic core forming block and a hydrophilic shell forming block are used. Amongst a variety of suitable polymers, biocompatible and entirely biodegradable ones are favoured. Poly(ethylene glycol) (PEG) is often the preferred hydrophilic block because of its biocompatibility and non-immunogenic properties. For the hydrophobic block, polylactides (PLA) and their derivatives are often used. However, for PLA based copolymers, the incorporation of some poorly soluble drugs is limited 1. Alkyl substituted polylactides were therefore developed to overcome this problem 2. By more hydrophobic alkyl groups on the PLA backbone, the hydrophobicity of the resulting polymer increases while preserving the biodegradability properties of PLA. Due to their physicochemical properties, two hexyl- substituted PLA (hexPLA): monohexyl- (monohex) and dihexyl- (dihex) substituted PLA were selected for the here presented pharmaceutical investigations. Copolymers of these new PLA derivatives with methoxypolyethylene glycol (MPEG) were synthesised and able to form polymeric micelles 3;4. In this thesis, such MPEG- Introduction 2 hexPLA copolymer micelles were investigated towards their biocompatibility aspects and possible pharmaceutical applications. Recent advances with colloidal drug delivery systems will be reviewed in chapter I. The incorporation of a poorly water soluble model drug into MPEG-hexPLA micelles is presented as a proof of concept in this chapter. Chapter II reports on the incorporation of several poorly soluble drugs into MPEG-hexPLA micelles and describes the relationship between incorporation results and the physico-chemical properties of the drugs, such as partition coefficient log P, water solubility, molecular weight, the number of H bond- donor or acceptor groups, etc. As novel materials for the pharmaceutical industry, the biocompatibility aspects of MPEG-hexPLA as unimers and as micelles were investigated in chapter III in terms of in vitro and in vivo toxicity and hemolysis activity. A possible intravenous application of cyclosporin A is presented as an example. In chapter IV, intravenously injected MPEG-hexPLA micelles were studied in vivo in rats as a novel ovarian cancer diagnosis tool. A promising application of these novel micelle formulations for dermatology applications will be presented in chapter V. At the end the possible oral delivery of poorly water soluble drugs with MPEG-hexPLA micelles and the initial proof of concept studies will be presented in chapter VI. References (1) Ma, L. L.; Jie, P.; Venkatraman, S. S., Block Copolymer Stealth Nanoparticles for Chemotherapy: Interactions with Blood Cells In Vitro, Adv.Funct.Mat. 2008, 18, 716-725. (2) Trimaille, T.; Gurny, R.; Möller, M., Synthesis and ring-opening polymerization of new monoalkyl-substituted lactides, J.Polym.Sci.Part A: Polym.Chem. 2004, 42, 4379-4391. (3) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. (4) Nottelet, B.; Di Tommaso, C.; Mondon, K.; Gurny, R.; Möller, M., Fully biodegradable polymeric micelles based on hydrophobic- and hydrophilic-functionalized poly(lactide) block copolymers, J.Polym.Sci.Part A: Polym.Chem. 2010, 48, 3244-3254. 1234567892AB 2CB D2236EB F783B 2656B 35B 7AB 2C8AB B 7B D56B 26E439B 4966B F98B 893B D9C9B 2366B 835853B 7AB B 7B D32499AB 7DD327B 82B D3D73B 7525B C234567892AB2CB7578B2AA837892ABF98B22B879698EB 26B !"#BD26E439B4966BF98B98B9A37BE32D29B23B73B!D8B 82B 9A23D2378B 93B 7425A8B 2CB E32D29B 35B 87AB 8B 87A73B "#B 1 1 1 1 1234567898 1 8 1 Chapitre I 3 Colloidal Drug Delivery Systems – Recent Advances with Polymeric Micelles K. Mondon, R. Gurny, and M. Möller. School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland. Published in: Chimia, 62 (10), 832-840 (2008) With the emergence of nanotechnology in drug delivery, colloidal systems and particularly polymeric micelles have attracted great attention. Polymeric micelles formed by the selfassembly of amphiphilic copolymers adopt a core-shell structure, which can be loaded with drugs and used as drug delivery systems for various medical applications. The most interesting aspects involve extended blood circulation times and stability upon dilution, which enable polymeric micelles to accumulate in tumour or inflammation sites due to the enhanced permeation and retention effect (EPR). In the first part of this paper polymeric micelles with different morphologies and different circulating-, active- and passive targeting, and stimuli responsive properties will be reviewed. Furthermore amphiphilic block-copolymers of different compositions for pharmaceutical micelle formulations will be discussed. The hydrophilic block is often composed of the biocompatible polyethylene glycol (PEG), whereas diverse polymers are used for the hydrophobic block. The biodegradable and biocompatible polylactide (PLA) is one of the most tested core-forming blocks for micelles, in part because of approval by the FDA for numerous drug products for use in humans. However, PLA has limitations with respect to the incorporation of poorly water soluble drugs. Considering this we will present in the second part of this paper briefly our strategy and contribution to overcome these limitations and recent results for the envisioned application of these micelles in the field of cancer treatment. In order to increase the hydrophobicity of PLA methyl groups are substituted by more hydrophobic hexyl groups. The hexyl-substituted polylactides in combination with PEG form the amphiphilic block copolymers PEG-hexPLA, which self-assemble in aqueous solution into stable spherical, homogenous micelles with a diameter of 20-45nm. The increased hydrophobicity of the hex-PLA micelle core leads to Recent Advances with Polymeric Micelles 4 higher incorporation rates of hydrophobic drugs, like the poorly water soluble photosensitizer, meso-tetra(p-hydroxyphenyl)porphine (THPP). THPP water solubility is increased 200-fold using these micelles. Their application in Photodynamic therapy (PDT), coupled with the increased accumulation of the photosensitizer in a tumour, followed by irradiation at a special wavelength, should lead to tumour death. The encouraging drug incorporation results reveal the potential of hexyl-substituted polylactide micelles as drug carriers for PDT applications. Keywords: Biodegradable Polymers; Block Copolymers; Drug Delivery; Polymeric Micelles; Substituted Polylactides 5 Chapitre I 1. Overview on polymeric micelles as drug delivery systems In the first part of this article we will give a brief overview on polymeric micelles as drug delivery systems. Specific properties of polymeric micelles will be outlined with an emphasis on the different compositions of widely used amphiphilic copolymers. In a second part, recent developments from our own research on hydrophobic substituted PLA-based copolymers and their possible use as micellar carriers of poorly water soluble drugs for cancer treatment will be presented. One very important aspect of pharmaceutical research is the development of novel drug carriers, which can deliver drugs to the right pathological site, at the right time, in the right dose, without affecting surrounding healthy tissues. In order to achieve this, nanotechnology has gotten into the spotlight with the exploration of various colloidal drug delivery systems like nanospheres, nanocapsules, micelles, liposomes, nanoemulsions, nanogels, lipid based nanoparticles, dendrimer nanocomposites, and others 1. Among these systems, micelles have gained increased interest in the last 15 years. The number of patents and publications is increasing exponentially. The number of published papers in 1990 dealing with “polymeric micelles” was about one hundred, whereas in 2007 it was 8 times higher 2 (Figure 1). Numerous excellent reviews are available in the literature describing micellar carrier systems for contrast agents in the imaging and diagnostic field 3, as drug carriers 4;5 for cancer 6-8, and for gene and photodynamic therapy 9. Number of publications and patents per year Recent Advances with Polymeric Micelles 900 800 700 600 500 400 300 200 100 0 1980 1985 1990 1995 2000 6 2005 Year Figure 1. Number of yearly publications and patents from 1980 to 2007 using the keyword “polymeric micelles” (source: Scifinder Scholar, February 2008). Polymeric micelles for pharmaceutical applications can be formed from amphiphilic copolymers with different architectures (Figure 2). Composed of a hydrophilic A block and a hydrophobic B block, these copolymers can either be of the linear AB diblock type, ABA or BAB triblock- or multiblock type-, or a nonlinear composition having more complex architectures like star or branched types. Consequently these copolymers form micelles of different structures. The self-assembly process is driven by the force to reach the lowest free-energy of the system10. In solution the amphiphilic copolymers exist first as unimers until their concentration exceeds the critical micellar concentration (CMC), above which they spontaneously self-assemble into micelles to form colloidal systems of the nano scale. For a given polymer concentration micelles form, when the solution temperature reaches the critical micellar temperature (CMT). The lower critical solution temperature (LCST) is the minimum temperature beyond which the polymers self-assemble into micelles. Thus both the CMC and LCST are critical parameters for the existence of stable micelles in the body. The micelle morphology is controlled mainly by 3 factors: 1) stretching of the core-forming blocks (free energy of the core), 2) the surface tension between the core-forming block and the solvent (free energy of the interface), and 3) repulsion interactions of the corona-forming blocks (free energy of the corona) 10. 7 Chapitre I Different morphologies are possible depending on the characteristics of the amphiphilic polymers (molecular weight, physical state and composition) and on the solution parameters (solvent, polymer concentration, pH, ionic strength, solvent/co-solvent ratio and others). Amphiphilic diblock copolymers self assemble into basic shapes like spheres (the predominant form), rods, and wormlike or vesicle structures. When the molecular weight of the hydrophilic block represents more than 50% of the total polymer molecular weight, spontaneous spherical micelles are formed, whereas with a hydrophilic block slightly below 50%, the micelles take on a wormlike structure. This latter morphology can also be generated by sonication from a copolymer forming initially spherical micelles, as it was shown for poly(ethylene glycol)-b-poly(-caprolactone) (PEG 5000g/mol -PCL 6500g/mol ) 11 . In degradation studies, wormlike micelles transformed into spherical micelles, when the PCL blocks hydrolysed and the percentage of hydrophobic blocks dropped to less than 50% of the overall polymer weight. The physical state of the core forming block also influences the micelle morphology. PEG-poly(-caprolactone-D,L lactide) (PEG-P(CL-DLLA)) with the amorphous poly(D,L lactide) block self-assembled into spheres, whereas cylindrical structures were formed with the crystalline poly(L,L-lactide) block (PEG-P(CL-LLA)) 12 . A strong competition between the energy of the crystalline core and the chain stretching corona may explain the formation of the cylindrical shape. Moreover the influence of the composition and the block length has been demonstrated in the following examples; PEG 2000g/mol -PCL n formed a variety of different morphologies with the increase of PCL block length from sphere, rod, wormlike to lamellae, whereas PEG 5000g/mol -PCL n only self-assembled into spherical micelles. One polymer, PEG 5000g/mol -PCL 232 , formed a mixture of spheres and lamellae 13. PCL also has been used to study the effect of the polymer- block-type and -architectures in polymeric micelles (Figure 2). Classical PEG 5000g/mol -PCL n diblock 13;14 or PCL-PEG-PCL triblock 15 copolymers self-assemble into the common spherical core-shell structure (Figure 2a and 2b, respectively). BACAB type multiblock copolymers, where C is a less hydrophobic block than B as in PCL n -(PEG-PPO-PEG) 1900g/mol -PCL n , leads to the formation of a double layer shellcore structure (Figure 2c) 16. Star-shaped copolymers like the 4 arm (PEG 5000g/mol -PCL) 4 17 or Tetronic®-PCL 18 assemble into spherical micelles, wherein the hydrophobic blocks form the core and the hydrophilic PEG chains arrange to form the shell (Figure 2d). Graft copolymers like PAsp-g-PCL can form spherical micelles with a shell, in which hydrophilic chains overlap, and entangle (Figure 2e) 19. PCL 7000g/mol –PDMA 8000g/mol brush copolymers with their 8 Recent Advances with Polymeric Micelles hydrophobic PCL-backbone form a kind a core-shell-“crosslinked” micelles (Figure 2f) 20. All of the above mentioned micelles are characterised by sizes below 100nm. Yet, not all micelle types have proved to be suitable drug carriers. Diblock copolymers Hydrophilic block Hydrophilic shell (a) Hydrophobic core Hydrophobic block Triblock copolymers (b) Multiblock copolymers (c) Less hydrophobic block 1st Hydrophilic shell 2nd Shell More hydrophobic block Double layer micelles Star shaped copolymers (d) Graft copolymers (e) Brush copolymers (f) Figure 2. Different amphiphilic copolymer types and architectures leading to different micelle structures by self-assembly. Chapitre I 9 The influence of the solvent/co-solvent ratio on micelle shape has been explored by Barghava et al. 21 . Using 2 different solvent systems, DMF/water and DMF/acetonitrile, PEG- polystyrene (PEG-PS) copolymers self-assembled first into spheres and with the decrease of DMF content into cylinders, followed by wormlike, and finally vesicle structures. Mixed morphologies (spheres and cylinders, cylinders and wormlike structures) were visible at intermediate DMF concentrations. In aqueous media, micelles have a core-shell structure with a hydrophobic dense inner core and a hydrophilic flexible outer shell. It has been shown, that many poorly water soluble drugs can be incorporated efficiently into the hydrophobic core, and thus within the polymeric micelles to become water soluble, facilitated by the hydrophilic micelle shell, which is composed mainly of polyethylene glycol (PEG). Drug loading also influences the micelle morphology. For example a change from spheres to cylinders occurred, when indomethacin loading was increased in PEG-poly[2-(diisopropylamino)ethyl methacrylate] (PEG-PDPA) micelles 22 . Further, wormlike micelles from poly(ethylene glycol)-b-poly(-caprolactone) (PEG 5000g/mol -PCL 6500g/mol ) possessed the same diameter as the corresponding spherical ones, but could incorporate twice as much paclitaxel, which is of great interest for an efficient drug loading of the micellar carrier system 11. Moreover, the stability and cytotoxicity did not differ between the two morphologies. The presence of PEG, a hydrophilic, water soluble, nontoxic and non-immunogenic polymer, at the surface of micelles enables nanocarriers to escape from renal exclusion and from opsonisation by the mononuclear phagocytic system (MPS). Therefore micelles can protect the incorporated drug from the biological fluids until it can reach the target site. With a prolonged circulation time in the blood stream they can accumulate at the inflammation site or in tumour tissues through the enhanced permeability and retention effect (EPR), which resembles a passive targeting 23 . Due to their small size (below 100nm), polymeric micelles can reach the very small vascular vessels and can be internalised into cells more efficiently unlike bigger carriers such as microparticles 24;25 . The release of the incorporated drug at the pathological site can occur by simple diffusion, destabilisation of the structure of the micelles by degradation, change in pH or by other stimuli, which will be discussed in more detail below. There are several other advantages of polymeric micelles in drug delivery. With their low CMC in the micromolar range, polymeric micelles generally have good stability upon 10 Recent Advances with Polymeric Micelles dilution. Thus the amount of the polymer surfactant can be reduced in comparison to other classical surfactants in order to formulate a stable drug carrier system. This higher stability of polymeric surfactant based formulations results in a better shelf-life. The clear appearance and the good injectability of micellar solutions are also advantages for medical treatments. The protection of drugs in the micelle core decreases the adverse side effects observed with free drugs, such as pain and inflammation at the injection site, and systemic side effects, when administered parenterally 26. Propofol is an example of the improvement of a pharmaceutical formulation with polymeric micelles. It is a water insoluble anesthetic agent, that was found to be more stable in poly(N-vinyl-2 pyrrolidone)-block-poly(D,L-lactide) (PVP-PDLLA) micelle solution than in the currently used water-in-oil emulsion. After reconstitution, the micelle formulation is stable for 4 days, whereas the standard emulsion can only be used for 6h 27 . As a second example for advantages of polymeric micelles, a formulation with the potent anticancer drug paclitaxel can be mentioned. Paclitaxel, a chemotherapeutic agent that is almost insoluble in water (0.3g/mL), is currently marketed as Taxol® and solubilised therein with Cremophor® EL, a polyoxyethylated castor oil. To replace this surfactant, which is known for its toxicity, albumin bound paclitaxel nanoparticles (Abraxane®) 28 poly(D,L-lactic acid) polymeric micelles (Genexol® PM) have been developed and PEG29 . These formulations lead to a reduction of severe side effects, thus permitting to increase doses of the chemotherapy. In vivo studies on human ovarian and breast cancer cells in nude mice demonstrated, that tumour growth was reduced 48h and 14 days, respectively after administration of Genexol® PM at its maximum tolerated dose (MTD), which is 3 times higher than in the treatment with Taxol®. Moreover, tumour regrowth which was observed for Taxol® 30 is not reported for the Genexol® PM treatment. After one month of treatment, this polymeric micelle formulation showed complete tumour regression. As mentioned before, drug targeting is a special challenge, since the delivery of a drug to the right site of action should be both selective and quantitative. Passive targeting by the EPR effect results from the long–circulation property of micelles. In order to achieve a higher drug dose at the target site, substantial effort has been put into active targeting research. Active targeting can be accomplished by modifying the micelle surface with site-specific ligands, or by creating “immunomicelles” by attaching monoclonal antibodies. One strategy for drug targeting uses folate functionalization. Receptors recognising the vitamin folic acid are over 11 Chapitre I expressed on many human cancer cells (breast, ovarian, brain, kidney and lung), consequently folate conjugated micelles are preferably recognised and bound on the tumour cells, and can then be internalised by active tumour mechanisms. This mechanism was for example. observed with adriamycin loaded folated polymeric micelles 31. The presence of these ligands on the micelle surface increased the in vitro cytotoxicity by increasing the cellular uptake and the intracellular concentration. The in vivo studies suggested that a defined number of folate groups on the micelles surface showed an optimized carrier-receptor interaction. Tumour treatment with these folated polymeric micelles was more efficient than with the non folate functionalized micelles or even the free drug. Also, other potent drugs like paclitaxel doxorubicin 35 , and tamoxifen 36 and a multidrug resistance modulator 37 32-34 , have been formulated with folate functionalized micelles and yielded improved results. A recent paper by Beduneau et al. reviews the different active targeting strategies available for treating brain tumours 38. One major obstacle for efficient drug delivery to the brain is the blood brain barrier. Different modifications of the surface of nanocarriers (liposomes, micelles or nanoparticles) by grafting endogenous and chimeric ligands, or by directly conjugating proteins and peptides through a covalent or non covalent linkage leads to internalization of the nanocarrier by brain capillary endothelial cells. Other examples of surface functionalization of micelles for drug delivery can be found in the review of Mahmud et al. 39. Stimuli responsive micelles are also an approach of controlled drug release. When nanocarriers have reached the pathological site, the drug can be released due to destabilisation by different stimulations. This can be achieved with external sources like light, ultrasound, hypo- or hyperthermia, or with internal stimuli like pH change or enzymes. pH-sensitive micelles have been widely tested in drug delivery, because of increased acidity of tumour and inflammatory tissues compared to healthy tissues. A change in the pH leads to a demicellization process and the drug is released. It has been reported that doxorubicin loaded pH-sensitive micelles formed from PEG-poly(-amino esters) disassembled at pH 6.4 leading to rapid doxorubicin release, whereas at physiological pH they remained stable 40 . A higher drug concentration at the pathological site was achieved, resulting in higher anti-tumour efficacy and a higher survival rate compared to treatment with free drug. In other examples, pH-sensitive polymeric micelles were loaded with adriamycin 41, paclitaxel 36 or tamoxifen 36. 12 Recent Advances with Polymeric Micelles Other poorly soluble drugs like triclosan fenofibrate 44, and progesterone 44 42 , candersartan cilexetil 43 , indomethacin 44 , could be successfully incorporated in similar micelles and resulted in pharmacological improvements. Thermoresponsive micelles can be used as controlled release systems. This is particularly the case for poly(N-isopropylacrylamide) (PNIPAAm)-based micelles with a LCST of about 40°C 45 . Below this LCST, AB type copolymers with PNIPAAm as the hydrophilic block form polymeric micelles, which are sterically stabilised by the water soluble flexible chains of the PNIPAAm block 46. The lipophilic drug is incorporated in the hydrophobic core and can be stored as a stable formulation. Above the LCST, PNIPAAm becomes insoluble and thus deforms the micelle structure, which results in a rapid diffusion of the drug from the micelles. In a cancer treatment study PNIPAAm-based polymeric micelles are accumulated in a solid tumour site by passive targeting, and upon local heating slightly above the LCST the encapsulated drug was released 47;48. Ultrasound-sensitive polymeric micelles based on Pluronic® P-105 were developed by the groups of Pitt and Rapoport in the late 90’s 49. Over time Rapoport and co-workers optimised these micelles by mixing them with PEG-distearoylphosphatidylethanolamine (PEG-DSPE). Mixed micelles demonstrated enhanced stability upon dilution compared to the standard Pluronic® P-105 micelles. Indeed ruboxil, a paramagnetic labelled anthracyclin, showed complete release in very diluted concentrations of fetal bovine serum when incorporated in Pluronic® P105 micelles, whereas 65% of the drug was retained in the mixed polymeric micelles 50. In addition, the treatment of tumours with doxorubicin as the drug was found to be more efficient as indicated by a higher uptake and therefore a decrease of the tumour size, when an ultrasound stimulus was applied to micelles in comparison to non-stimulated micelles. The efficiency of the tumour treatment is even more pronounced when compared to the treatment with the free drug 50 . In summary, polymeric micelles with their specific structure and long circulation in the blood stream are promising carriers for passive and active targeting, especially for cancer treatment. 13 Chapitre I For the use of amphiphilic polymers in pharmaceutical applications in general, important requirements such as non-toxicity and biocompatibility need to be fulfilled. This constrains the number of available hydrophobic and hydrophilic natural or synthetic polymers. For the hydrophilic shell-forming block polyethylene glycol (PEG) is the polymer of choice, because of its superior properties, and its approval by the FDA. Despite its non degradability PEG is non-toxic and can easily be removed from the body through the excretion pathways, as long as the molecular weight is less than 15kDa. Thus PEG with molecular weights between 2 and 15kDa are suitable in polymeric micelles for drug delivery. One of the main advantages resides in the efficient protection of incorporated drugs in pegylated nanocarriers 51 . Other polymers like poly(N-vinyl-2-pyrrolidone) (PVP) 52;53, polyvinyl alcohol (PVA) and its derivatives 54 , or recently studied poly(ethyl ethylene phosphate) (PEEP) 55 could be alternatives to PEG. PVP a non ionic, biocompatible and water soluble synthetic polymer is often preferred to PEG for freeze drying formulations, because of its better cryoprotectant properties 56. Polyphosphates were recently investigated as polymers for micellar systems 55, because of their biocompatibility, degradability and pendant chain functionality, but further work is needed for a proof of concept. For the hydrophobic core-forming block, a large number of polymers meet the requirements for pharmaceutical applications. Here ionic or non-ionic, degradable or non degradable polymers can be considered. In polyion complex micelles (PICM) a charged core-forming block can more efficiently incorporate negatively charge drugs like plasmid DNA, oligodeoxyribonucleotides, some polysaccharides, enzymes or photosensitizers. The typical positively charged polymers are poly(ethylenimines) (PEI) lysine) 60 57 58;59 , polyacrylamides or poly(2-(N,N-dimethylamino)ethyl methacrylate) (PDMAEMA) , poly(L- 61 . In contrast, polyanionic polymers, like poly(methacrylic acid) (PMAA) 47 or poly(aspartic acid) (PAsp) 62, are used for the incorporation of polycationic peptides or lipids 63. Various non-ionic hydrophobic core-building degradable and non-degradable polymers have been investigated. Here we will focus on the possible polymers from three different classes including polyethers, poly(L-amino acids) and polyesters. Certainly there are other important biodegradable polymers, like polyanhydrides or polyurethanes, but to date they are used more as implants, microspheres, discs or other matrices for localised drug delivery 64;65 . A few 14 Recent Advances with Polymeric Micelles examples of polymeric micelles made from oligoanhydrides and PEG have been reported by Najafi et al., who demonstrated the ability to incorporate a hydrophilic model drug, calcein 66. The most interesting polyethers for micellar drug delivery systems are the triblock copolymers (ABA type) of PEG and poly(propylene oxide) (PPO). They are marketed and commonly known as Pluronics® (BASF) 67 or as Poloxamers, their non-proprietary name 68 . Depending on the compositions and block-lengths PEG-PPO multiblock copolymers show interesting material properties, including the formation of micellar structures or temperature dependent gels. As for example Pluronic®-based micelles were studied by Gao et al. Exner et al. 69 50 and for cancer therapy. In these studies, doxorubicin and carboplatin, respectively were successfully incorporated into the micelles. For doxorubicin loaded Pluronic® micelles high drug uptake could be achieved in multidrug resistant cell lines through ultrasonic irradiation of the tumour 50. An efficient tumour decrease was observed using this method. Deriving from natural L-amino acids, poly(L-amino acids) are very interesting biocompatible polymers and were studied as possible hydrophobic core-forming blocks 70 . Several poly(L- amino acid)-based micelles have been investigated as pH-sensitive polymeric drug carriers. At acidic pH in the tumour, the protonation of free amine and carboxyl groups in the polymer chains lead to a destabilization of the micelle structure and resulted in the controlled release of drug 71 . PEG-poly(L-histidine) micelles loaded with doxorubicin generated a decrease of human ovarian carcinoma subcutaneously xenografted in mice 72. As another example PEGpoly(L-lysine) and PEG-poly(L-ornithin) micelles were studied for DNA delivery 73. A higher condensation of DNA and a higher DNA transfection of two mammalian cell lines in vitro were achieved with these micelle carriers compared to the parent homopolymers, poly(Llysine) or poly(L-ornithine). Disadvantageously the higher haemolytic activity could limit the intravenous use of these novel vesicles. As an example for non-pH sensitive poly(L-amino acids) polymeric micelles, benzyl ester of PEG-poly(aspartic acid) (PEG-P(Asp)) micelles yielded a higher drug loading of the waterinsoluble anticancer agent camptothecin and better stability compared to other esterified copolymers and to free drug 74 . In these studies mice, transplanted with colon solid tumour cells, and treated with benzylester-PEG-P(Asp) micelles, had a longer circulation time, an increased accumulation in the tumour, and thus a more efficient antitumour activity 23. Several Chapitre I 15 hydrophobic core-forming poly(L-amino acids) could improve micelle stability and showed promising results for cancer or gene therapy applications. Poly(hydroxyalkanoic acids) are the most commonly used polymers in PEG-polyester micelles for drug delivery, because of their outstanding biodegradability and biocompatibility. Due to the vast number of published articles on PEG-polyester micelles we can only present some selected examples of recent achievements, and would like to refer the readers to other more detailed review articles on polymeric micelles 5;6;75;76. Poly(butyrolactone) (PBL) is preferentially used in its form, and is derived from the natural production in microorganisms. -PBL is characterized by a higher hydrophobicity and crystallinity compared to the synthetic -PBL. Despite a good stability, a low CMC and small size, the potential of PEG-PBL-PEG triblock micelles (ABA type) was limited due to the slow biodegradation of the PBL blocks in vivo 77 . Contrary the PBL-PEG-PBL copolymers, BAB type, showed a faster biodegradation rate, while having similar sizes and CMC values 78 . However, the incorporation of hydrophobic drugs needs to confirm their feasibility for drug delivery, even if pyrene as a model compound was incorporated with success. Different studies on the semi-crystalline poly(valerolactone) (PVL) as the hydrophobic coreforming block were reported 79-81. As one example, PEG 2000g/mol -PVL 2000g/mol was used for the formulation of paclitaxel 79 . With these micelles the water solubility was increased to 9mg/mL. When compared to the actual paclitaxel solubilisation of 6mg/mL in the mixture Cremophor® EL/dehydrated alcohol in Taxol®, the result suggests that a lower amount of PEG-PVL surfactant would be needed to achieve the same dose. In vivo studies need to show that due to the slow degradation of PVL, and the expected longer elimination time from the body will not be a problem. Poly(caprolactone) (PCL) is another very suitable polymer for the core-forming block, because of its proven biocompatible properties, therefore PEG-PCL diblock copolymers have commonly been investigated for drug delivery. Recently, protoporphyrin IX, a hydrophobic photosensitizer, was incorporated into PEG 5000g/mol -PCL 4100g/mol micelles, and a higher cellular uptake and higher photocytotoxicity was obtained compared to the free photosensitizer 82. In another example reported by Aliabadi et al., cyclosporine A, an immunosuppressive agent 16 Recent Advances with Polymeric Micelles was encapsulated in PEG 5000g/mol -PCL 13000g/mol micelles 83 . Here the drug solubility was increased by 80 fold compared to its water solubility 84. High drug loadings in these micelles envision the possible replacement of other surfactants, like Cremophor® EL in the current formulation Sandimmun®, which shows severe side effects. In studies for cancer treatment PEG-PCL polymeric micelles were loaded with chemotherapeutic agents like cisplatin doxorubicin 85;86 and curcumin , paclitaxel 87 and recently tested newer compounds such as -lapachone 20 , 86 88;89 . Other approaches focus on tumour specific targeting PEG-PCL carriers functionalized with folic acid, heparin or epidermal growth factor (EGF) for the release of a chemotherapeutics like paclitaxel 32, of both indomethacin and basic fibroblast growth factor (bFGF) 90 or ellipticine 91. Higher internalisation and higher cytotoxicity were observed, and thus improved significantly the tumour treatment compared to the non-functionalized micelles. Among the various poly(hydroxyalkanoic acids), poly(lactic acid) (PLA) and poly(lactic-coglycolic acid) (PLGA) copolymers are the most outstanding polyesters in drug delivery applications. Environmental friendliness, synthesis from renewable resources, biodegradability into non-toxic lactic acid, and excellent biocompatibility have made it the polymer of first choice for many medical applications. Available stereochemically different polylactides are characterized by different physical properties. The racemic D,L-PLA is amorphous, whereas the enantiomeric pure L- or D-PLA (PLLA and PDLA, respectively) are crystalline materials, resulting in different biodegradation times. Poly(lactide-glycolic acid) (PLGA), which is a PLA functionalized with glycolic acid units or blocks in the polymer chain, degrades faster than pure PLA and is often used to tailor the degradation time of PLA drug carrier systems. Therefore depending on the polymer used these PLA-based micelles have significantly different properties. PEG n -P(CL-DLLA) 8121g/mol had a lower CMC than PEG n -P(CL-LLA) 6614g/mol , and both micelles adopted different morphologies 12. Mixing PEGPLLA and PEG-PDLA copolymer led to a stereocomplex formation of the PLA chains in the micelle core, which had better micelle stability with a lower CMC, smaller size, and higher incorporation efficacy of rifambin, than PEG-PLLA or PEG-PDLA micelles 93. In Genexol® PM, PEG-PDLLA micelles are used as the drug carrier for paclitaxel 30. A multicenter Phase II trial for the treatment of advanced non-small-cell lung cancer in combination with cisplatin 29 has been successfully finished, and the same formulation enters into a Phase II study for advanced pancreatic cancer treatment 94. Other drugs like 5-fluoroacil 95, doxorubicin 86;96;97 , 17 Chapitre I -lapachone 86;98, campthothecin 99 and amphotericin B 100 have been incorporated into PEGPLA micelles, and different polymer architectures like linear or star branched, diblock or triblock polymers, homopolymer or mixed polymer micelles were investigated. Star branched polymeric micelles showed no difference in comparison to linear polymeric micelles for 5fluoroacil release, whereas paclitaxel was released more rapidly and completely 95. In mixed micelles with doxorubicin loaded poly(N-isopropylacrylamide-co-methacrylicacid)-graftpoly(D,L-lactide) (P(NIPAAm-co-MAAc)-g-PDLLA) the PEG-PLA did not affect the pHand thermoresponsive properties, but advantageously prevented the adsorption of albumin, increasing the circulation time in the blood stream 97 . PLA-PEG-PLA triblock copolymers with acryl end groups self-assembled into micelles, which were transformed into nanogels by UV irradiation 99 . Loaded with campthothecin these nanogels showed better stability and a sustained release for at least 20 days in contrast to complete release for non-irradiated micelles after few hours. Hydrophilic shell-forming blocks other than PEG have been explored for PLA- or PLGA- based copolymer micelles. Recently PLA-poly(L-glutamic acid) micelles were developed for magnetic resonance imaging (MRI) diagnostic systems 101 . Gadolinium (Gd) ions chelated with DTBA were incorporated into these micelles as an MRI probe. They were stable upon dilution and showed a two times higher relaxivity compared to the free DTBA-Gd complex. Further in vivo studies should evaluate the feasibility of these micellar diagnostic systems for their application in patients. Poly(L-lysine) could be another alternative polymer to PEG as the shell-building block. Grafted on PLGA, the resulting amphiphilic polymer self-assembled into micelles, which showed lower cytotoxicity and higher transfection efficiency than poly(L-lysine) itself, and could be a possible carrier for gene delivery 102. In comparison to PEG, these poly(L-amino acid) based micelles are entirely biodegradable. Stimuli responsive micelles based on PEG- poly(L-histidine) (PEG-PHis) copolymer micelles were also investigated. Upon a change of pH in tumour tissue, the imidazole side groups along the poly(L-histidine) backbone are ionized (pH<pK b ), transforming the hydrophobic PHis-block into an hydrophilic water soluble one. This is accompanied by a swelling of the micelles and a concurrent drug release 103 . Loaded with doxorubicin PLA-PEG-PHis micelles showed an effective treatment of human breast tumours96. Thermoresponsive micelles made of PLA-based polymers were studied with (PNIPAAm-co-DMAAm)-PLGA copolymers for the delivery of chemotherapeutic agents like paclitaxel 104 and doxorubicin 59. These micelles showed a higher drug release upon thermo- stimulation above the LCST (39.5°C) and a higher cytotoxicity on tumour cells compared to Recent Advances with Polymeric Micelles 18 free drug. As an example for tumour targeting micelles, folate conjugated PEG-PLGA micelles were studied by Yoo et al. 105 and Zhao et al. 35 . A higher uptake, higher cytotoxicity, and higher apoptosis were obtained with these folate-functionalised micelles compared to the non-folated micelles and the free drug, respectively 35. PEG–polyester micelles are potent drug carriers compared to many classical surfactant based micelles. A higher stability in diluted solutions due to their lower CMC, PEG copolymer micelles have long circulation properties and can be functionalized as targeting or as stimuli responsive drug release carrier systems. An increased accumulation, a higher drug uptake by the tumour or inflammation cells accompanied with a more effective tumour treatment can often be achieved with polymeric micelles. Regarding this, albeit an incomplete list, PCL, PLA and PLGA are the most promising polyesters. Because of their approval by the Food and Drug Administration (FDA) for drug products in human use, new formulations with these pegylated excipients might lead easier and faster to new applicable therapeutics. 2. Recent developments of hydrophobic substituted PLA based polymeric micelles Despite the outstanding applications of PLA in medical applications, limitations for the incorporation of hydrophobic drugs for pharmaceutical formulations are often encountered. Since a more hydrophobic core is desirable for a better incorporation into PLA-based polymeric micelles, we focused in our own research efforts on the controlled functionalization of PLA with hydrophobic substituents. Therefore methyl groups along the PLA polymer backbone were substituted by hexyl groups leading to the more hydrophobic hexyl-substituted poly(lactides) (hexPLA) 106 . PEG-hexPLA copolymers formed micelles in aqueous solutions and incorporated hydrophobic drugs, e.g. griseofulvin, more efficiently than comparable PEG-PLA micelles 107 . Recently we investigated the potential of these novel PEG-hexPLA micelles for possible use in photodynamic therapy (PDT) in cancer treatment. Initial results in 19 Chapitre I incorporating the hydrophobic photosensitizer meso-tetra(p-hydroxyphenyl)porphine (THPP) (Figure 3), and stability studies of THPP loaded PEG-hexPLA micelles will be presented. OH HO N NH HN N HO OH Figure 3. Molecular structure of meso-tetra(p-hydroxyphenyl)porphine (THPP). In order to compare PEG-monohexPLA and PEG-dihexPLA micelles 107 for their increased hydrophobicity and a possible higher incorporation rate of THPP, a comparable PEG“standard” poly(D,L-lactide) (PEG-PDLLA) was used as a control. All three copolymers were of comparable molecular weights of around 5200g/mol and a comparable polydispersity of 1.2. The PEG-(hex)PLA polymeric micelles were prepared by the co-solvent evaporation technique. Both the drug and the copolymer were dissolved in the water-miscible organic solvent acetone or THF/acetone (1:1). After the drop wise addition of the organic solution into water under sonication, accompanied by the self-assembly to micelles, the organic solvent was slowly evaporated. After equilibrium overnight of the remaining aqueous solution the micelles were characterized by their sizes, morphologies and drug incorporation. Unloaded PEG-(hex)PLA micelles had sizes between 18-27nm, whereas THPP loaded micelles were only slightly larger with 21-45nm. Thus the loaded micelle sizes remained below 50nm, a preferred “sub-100nm” size, facilitating a good internalisation into tumour cells by the EPR effect. Dynamic light scattering at different detection angles confirmed a unimodal size distribution for loaded and unloaded PEG-hexPLA and PEG-PLA micelles. The micelles sizes and morphologies were confirmed by TEM measurements. As shown in Figure 4 unloaded and loaded PLA-based polymeric micelles had comparable sizes with spherical shapes. Recent Advances with Polymeric Micelles (a) 20 (b) 100 nm (c) 100 nm (d) 100 nm 100 nm Figure 4. TEM images of unloaded (a) and THPP loaded PEG-monohexPLA (b) micelles, and for comparison unloaded PEG-dihexPLA (c) and PEG-PLA (d) micelles A study of polymeric micelle solutions stored at room temperature for 10 months proved their stability and the long shelf-life of this formulation. The amount of THPP in the diverse hexPLA- and PLA-based micelles was assessed by UV spectroscopy. The results for THPP incorporation into these micelles are presented in Figure 5. 21 Chapitre I 100% efficiency Incorporated drug loading (mgTHPP/gcopolymer) 200 80%efficiency 150 100 50 0 0 25 50 75 100 125 150 175 200 225 250 275 300 Intended drug loading (mgTHPP/gcopolymer) Figure 5: Incorporation of THPP in PEG-PLA (○,●), PEG-monohexPLA (□,■) and PEG-dihexPLA (,) micelles in function of the intended drug loading for the two preparation methods, with acetone (empty symbol) and THF/acetone (plain symbol), respectively Shown is the actual achieved loading in dependence of the intended loading, which is based on the drug concentration of the organic solution used for the micelle preparation. Furthermore, the two lines indicate a theoretical 80 and 100% incorporation, demonstrating the efficiency of the THPP incorporation. THPP incorporation of micelles prepared with acetone and with THF/acetone, respectively as solvent systems are presented. For intended small loadings up to 50mg THPP/g copolymer no significant differences were observed between the three different polymers and the organic solvents used. Incorporation rates were at least 80%. For a desired higher drug loading of 100mg THPP/g copolymer and 300mg THPP/g copolymer, respectively better incorporation rates were obtained when using a polymer/drug solution in the THF/acetone (1:1) mixture, which facilitates drug solubility. It is to point out, that in both cases the incorporation was higher and much more efficient with the novel PEG-hexPLA micelles than with the standard PEG-PLA micelles. The highest obtained drug loading was 123mg THPP/g PEG-dihexPLA, although with less efficiency (41%) compared to 96 and 88 mg THPP for the dihexPLA and the monohexPLA, respectively, which achieved around 90% efficiency for the intended lower drug loading. Since the molecular weights and ratios of PEG/hexPLA are not optimized, it is expected, that the loading capacity for THPP could be higher. Nevertheless the actual results already show a Recent Advances with Polymeric Micelles 22 prominent increased water solubility of THPP in PEG-hexPLA micelles. Within the dihexPLA micelles 480mg THPP/L water could be dissolved compared to only 2.2mg/L in pure water, which corresponds to an increase of 218 fold! As mentioned previously, this result still stands for non-optimized conditions, thus further developments in PEG-hexPLA micellar formulations for THPP delivery could lead to a promising alternative for PDT applications in cancer treatment. The increased THPP drug loadings within the hexyl-substituted PLA-based micelles in comparison to standard PLA validate our strategy of improving hydrophobic drug formulations with “hydrophobized” PLA. As outlined in our previous papers the hexPLA polymers degrade by hydrolysis to non-toxic lactic acid and 2-hydroxyoctanoic acid, which has already been approved for topical applications, and which is an important aspect for the feasibility of this polymer for medical applications. Therefore, on-going studies focus on the toxicity issues of these novel PEG-hexPLA micelles. Initial cell culture- and human blood toxicity tests show positive results and will soon be reported. 3. Summary Polymeric micelles as colloidal drug delivery systems have attracted great interest in the last 15 years due to their favorable properties. Formed by the self-assembly of amphiphilic copolymers, polymeric micelles have a core-shell structure, which can adopt several morphologies. The protective hydrophilic shell gives the micelles long circulation times in the blood stream, and a low CMC a high stability upon dilution. This can enable micelles to accumulate in tumours by passive targeting and the EPR effect. Active targeting to the site of action can be achieved by binding specific ligands or antibodies on micelle surfaces. Using stimulus responsive polymeric micelles can enhance the controlled drug release. For drug delivery, PEG is the most outstanding and used hydrophilic polymer for the micelle shell. Considerations regarding toxicity and biocompatibility limit the choice of numerous hydrophobic polymers for the core-forming block. Next to several polyethers, poly (L-amino acids), polyesters, particularly PLA has distinguished properties for medical applications. Nevertheless, the incorporation of hydrophobic drugs into PLA micelles often has limitations. In order to increase the hydrophobicity along the PLA backbone for better incorporation of Chapitre I 23 hydrophobic drugs we followed the strategy to substitute the methyl with more hydrophobic hexyl side groups. Hexyl-substituted lactides copolymerized with PEG lead to amphiphilic copolymers, which self-assemble in aqueous solution into homogeneous spherical micelles ranging in sizes from 20 to 45nm. These PEG-hexPLA micelle formulations showed 10 months of stability at room temperature. The incorporation of a poorly water soluble drug, like the photosensitizer THPP, did not affect the micelle size or the micelle morphology. However, by incorporation into PEG-hexPLA micelles the water solubility was increased more than 200 times compared to the water solubility of the free drug. These initial results suggest that the use of PEG-hexyl-substituted polylactides micelles as drug delivery carriers for photodynamic therapy is feasible. References (1) Koo, O. M.; Rubinstein, I.; Onyuksel, H., Role of nanotechnology in targeted drug delivery and imaging: a concise review, Nanomedicine: NBM 2005, 1, 193-212. (2) Search with Scifinder with keywords "polymeric micelles" after refinement by Document Type "Journal, Review and Patent" and analysis by Publication Year. 2008. (3) Torchilin, V. P., PEG-based micelles as carriers of contrast agents for different imaging modalities, Adv.Drug Deliv.Rev. 2002, 54, 235-252. (4) Torchilin, V., Micellar Nanocarriers: Pharmaceutical Perspectives, Pharm.Res. 2007, 24, 1-16. (5) Gaucher, G.; Dufresne, M. H.; Sant, V. P.; Kang, N.; Maysinger, D.; Leroux, J. C., Block copolymer micelles: preparation, characterization and application in drug delivery, J.Control.Release 2005, 109, 169-188. (6) Sutton, D.; Nasongkla, N.; Blanco, E.; Gao, J., Functionalized Micellar Systems for Cancer Targeted Drug Delivery, Pharm.Res. 2007, 24, 1029-1046. (7) Le Garrec, D.; Ranger, M.; Leroux, J. C., Micelles in Anticancer Drug Delivery, Am.J.Drug Deliv. 2004, 2, 15-42. (8) Rapoport, N., Physical stimuli-responsive polymeric micelles for anti-cancer drug delivery, Prog.Polym.Sci. 2007, 32, 962-990. Recent Advances with Polymeric Micelles 24 (9) Nishiyama, N.; Kataoka, K., Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery, Pharmacol.Ther. 2006, 112, 630-648. (10) Zhang, L.; Eisenberg, A., Formation of crew-cut aggregates of various morphologies from amphiphilic block copolymers in solution., Polym.Advan.Technol. 1998, 9, 677699. (11) Cai, S.; Vijayan, K.; Cheng, D.; Lima, E.; Discher, D., Micelles of Different Morphologies-Advantages of Worm-like Filomicelles of PEO-PCL in Paclitaxel Delivery, Pharm.Res. 2007, 24, 2099-2109. (12) Zhang, J.; Wang, L. Q.; Wang, H.; Tu, K., Micellization Phenomena of Amphiphilic Block Copolymers Based on Methoxy Poly(ethylene glycol) and Either Crystalline or Amorphous Poly(caprolactone-b-lactide), Biomacromolecules 2006, 7, 2492-2500. (13) Du, Z. X.; Xu, J. T.; Fan, Z. Q., Micellar Morphologies of Poly(e-caprolactone)-bpoly(ethylene oxide) Block Copolymers in Water with a Crystalline Core, Macromolecules 2007, 40, 7633-7637. (14) Gyun Shin, I. L.; Yeon Kim, S.; Moo Lee, Y.; Soo Cho, C.; Yong, K. S., Methoxy poly(ethylene glycol)/[epsilon]-caprolactone amphiphilic block copolymeric micelle containing indomethacin.: I. Preparation and characterization, J.Control.Release 1998, 51, 1-11. (15) Piao, L.; Dai, Z.; Deng, M.; Chen, X.; Jing, X., Synthesis and characterization of PCL/PEG/PCL triblock copolymers by using calcium catalyst, Polymer 2003, 44, 2025-2031. (16) Liu, C.; Gong, C.; Pan, Y.; Zhang, Y.; Wang, J.; Huang, M.; Wang, Y.; Wang, K.; Gou, M.; Tu, M.; Wei, Y.; Qian, Z., Synthesis and characterization of a thermosensitive hydrogel based on biodegradable amphiphilic PCL-Pluronic (L35)PCL block copolymers, Colloid.Surface A 2007, 302, 430-438. (17) Lu, C.; Liu, L.; Guo, S. R.; Zhang, Y.; Li, Z.; Gu, J., Micellization and gelation of aqueous solutions of star-shaped PEG-PCL block copolymers consisting of branched 4-arm poly(ethylene glycol) and polycaprolactone blocks, Eur.Polym.J. 2007, 43, 1857-1865. (18) Lee, J. S.; Go, D. H.; Bae, J. W.; Lee, S. J.; Park, K. D., Heparin conjugated polymeric micelle for long-term delivery of basic fibroblast growth factor, J.Control.Release 2007, 117, 204-209. (19) Jeong, J. H.; Kang, H. S.; Yang, S. R.; Park, K.; Kim, J. D., Biodegradable poly(asparagine) grafted with poly(caprolactone) and the effect of substitution on selfaggregation, Colloid.Surface A 2005, 264, 187-194. (20) Xu, P.; Van Kirk, E. A.; Li, S.; Murdoch, W. J.; Ren, J.; Hussain, M. D.; Radosz, M.; Shen, Y., Highly stable core-surface-crosslinked nanoparticles as cisplatin carriers for cancer chemotherapy, Colloid.Surface B 2006, 48, 50-57. Chapitre I 25 (21) Bhargava, P.; Zheng, J. X.; Li, P.; Quirk, R. P.; Harris, F. W.; Cheng, S. Z. D., SelfAssembled Polystyrene-block-poly(ethylene oxide) Micelle Morphologies in Solution, Macromolecules 2006, 39, 4880-4888. (22) Giacomelli, C.; Schmidt, V.; Borsali, R., Specific Interactions Improve the Loading Capacity of Block Copolymer Micelles in Aqueous Media, Langmuir 2007, 23, 69476955. (23) Kawano, K.; Watanabe, M.; Yamamoto, T.; Yokoyama, M.; Opanasopit, P.; Okano, T.; Maitani, Y., Enhanced antitumor effect of camptothecin loaded in long-circulating polymeric micelles, J.Control.Release 2006, 112, 329-332. (24) McClean, S.; Prosser, E.; Meehan, E.; O'Malley, D.; Clarke, N.; Ramtoola, Z.; Brayden, D., Binding and uptake of biodegradable poly--lactide micro- and nanoparticles in intestinal epithelia, Eur.J.Pharm.Sci. 1998, 6, 153-163. (25) Desai, M. P.; Labhasetwar, V.; Walter, E.; Levy, R. J.; Amidon, G. L., The Mechanism of Uptake of Biodegradable Microparticles in Caco-2 Cells Is Size Dependent, Pharm.Res. 1997, 14, 1568-1573. (26) Picard, P.; Tramer, M. R., Prevention of Pain on Injection with Propofol: A Quantitative Systematic Review, Anesth. Analg. 2000, 90, 963-969. (27) Ravenelle, F.; Gori, S.; Le Garrec, D.; Lessard, D.; Luo, L.; Palusova, D.; Sneyd, J.; Smith, D., Novel Lipid and Preservative-free Propofol Formulation: Properties and Pharmacodynamics, Pharm.Res. 2008, 101, 186-193. (28) Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.; Beals, B.; Dykes, D.; Noker, P.; Yao, R.; Labao, E.; Hawkins, M.; Soon-Shiong, P., Increased Antitumor Activity, Intratumor Paclitaxel Concentrations, and Endothelial Cell Transport of Cremophor-Free, Albumin-Bound Paclitaxel, ABI-007, Compared with Cremophor-Based Paclitaxel, Clin. Cancer Res. 2006, 12, 1317-1324. (29) Kim, D. W.; Kim, S. Y.; Kim, H. K.; Kim, S. W.; Shin, S. W.; Kim, J. S.; Park, K.; Lee, M. Y.; Heo, D. S., Multicenter phase II trial of Genexol-PM, a novel Cremophorfree, polymeric micelle formulation of paclitaxel, with cisplatin in patients with advanced non-small-cell lung cancer, Ann.Oncol. 2007, 18, 2009-2014. (30) Kim, S. C.; Kim, D. W.; Shim, Y. H.; Bang, J. S.; Oh, H. S.; Kim, S. W.; Seo, M. H., In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and efficacy, J.Control.Release 2001, 72, 191-202. (31) Bae, Y.; Nishiyama, N.; Kataoka, K., In Vivo Antitumor Activity of the FolateConjugated pH-Sensitive Polymeric Micelle Selectively Releasing Adriamycin in the Intracellular Acidic Compartments, Bioconjugate.Chem. 2007, 18, 1131-1139. (32) Park, E. K.; Kim, S. Y.; Lee, S. B.; Lee, Y. M., Folate-conjugated methoxy poly(ethylene glycol)/poly([var epsilon]-caprolactone) amphiphilic block copolymeric micelles for tumor-targeted drug delivery, J.Control.Release 2005, 109, 158-168. Recent Advances with Polymeric Micelles 26 (33) Seow, W. Y.; Xue, J. M.; Yang, Y. Y., Targeted and intracellular delivery of paclitaxel using multi-functional polymeric micelles, Biomaterials 2007, 28, 17301740. (34) Wang, Y.; Yu, L.; Han, L.; Sha, X.; Fang, X., Difunctional Pluronic copolymer micelles for paclitaxel delivery: Synergistic effect of folate-mediated targeting and Pluronic-mediated overcoming multidrug resistance in tumor cell lines, Int.J.Pharm. 2007, 337, 63-73. (35) Zhao, H.; Yung, L. Y. L., Selectivity of folate conjugated polymer micelles against different tumor cells, Int.J.Pharm. 2008, 349, 256-268. (36) Licciardi, M.; Giammona, G.; Du, J.; Armes, S. P.; Tang, Y.; Lewis, A. L., New folate-functionalized biocompatible block copolymer micelles as potential anti-cancer drug delivery systems, Polymer 2006, 47, 2946-2955. (37) Yang, X.; Deng, W.; Fu, L.; Blanco, E.; Gao, J.; Quan, D.; Shuai, X., Folatefunctionalized polymeric micelles for tumor targeted delivery of a potent multidrugresistance modulator FG020326, J.Biomed.Mater.Res.A 2007, 86A, 48-60. (38) Beduneau, A.; Saulnier, P.; Benoit, J. P., Active targeting of brain tumors using nanocarriers, Biomaterials 2007, 28, 4947-4967. (39) Mahmud, A.; Xiong, X. B.; Aliabadi, H. M.; Lavasanifar, A., Polymeric micelles for drug targeting, J.Drug Target. 2007, 15, 553-584. (40) Ko, J.; Park, K.; Kim, Y. S.; Kim, M. S.; Han, J. K.; Kim, K.; Park, R. W.; Kim, I. S.; Song, H. K.; Lee, D. S.; Kwon, I. C., Tumoral acidic extracellular pH targeting of pHresponsive MPEG-poly([beta]-amino ester) block copolymer micelles for cancer therapy, J.Control.Release 2007, 123, 109-115. (41) Bae, Y.; Nishiyama, N.; Fukushima, S.; Koyama, H.; Yasuhiro, M.; Kataoka, K., Preparation and Biological Characterization of Polymeric Micelle Drug Carriers with Intracellular pH-Triggered Drug Release Property: Tumor Permeability, Controlled Subcellular Drug Distribution, and Enhanced in Vivo Antitumor Efficacy, Bioconjugate.Chem. 2005, 16, 122-130. (42) Chiappetta, D. A.; Degrossi, J.; Teves, S.; D'Aquino, M.; Bregni, C.; Sosnik, A., Triclosan-loaded poloxamine micelles for enhanced topical antibacterial activity against biofilm, Eur.J.Pharm.Biopharm. 2008, 69, 535-545. (43) Satturwar, P.; Eddine, M. N.; Ravenelle, F.; Leroux, J. C., pH-responsive polymeric micelles of poly(ethylene glycol)-b-poly(alkyl(meth)acrylate-co-methacrylic acid): Influence of the copolymer composition on self-assembling properties and release of candesartan cilexetil, Eur.J.Pharm.Biopharm. 2007, 65, 379-387. (44) Sant, V. P.; Smith, D.; Leroux, J. C., Novel pH-sensitive supramolecular assemblies for oral delivery of poorly water soluble drugs: preparation and characterization, J.Control.Release 2004, 97, 301-312. Chapitre I 27 (45) Liu, B.; Yang, M.; Li, R.; Ding, Y.; Qian, X.; Yu, L.; Jiang, X., The antitumor effect of novel docetaxel-loaded thermosensitive micelles, Eur.J.Pharm.Biopharm. 2008, 69, 527-534. (46) Wei, H.; Zhang, X. Z.; Cheng, H.; Chen, W. Q.; Cheng, S. X.; Zhuo, R. X., Selfassembled thermo- and pH responsive micelles of poly(10-undecenoic acid-b-Nisopropylacrylamide) for drug delivery, J.Control.Release 2006, 116, 266-274. (47) Wei, H.; Zhang, X. Z.; Zhou, Y.; Cheng, S. X.; Zhuo, R. X., Self-assembled thermoresponsive micelles of poly(N-isopropylacrylamide-b-methyl methacrylate), Biomaterials 2006, 27, 2028-2034. (48) Chung, J. E.; Yokoyama, M.; Yamato, M.; Aoyagi, T.; Sakurai, Y.; Okano, T., Thermo-responsive drug delivery from polymeric micelles constructed using block copolymers of poly(N-isopropylacrylamide) and poly(butylmethacrylate), J.Control.Release 1999, 62, 115-127. (49) Munshi, N.; Rapoport, N.; Pitt, W. G., Ultrasonic activated drug delivery from Pluronic P-105 micelles, Cancer Lett. 1997, 118, 13-19. (50) Gao, Z. G.; Fain, H. D.; Rapoport, N., Controlled and targeted tumor chemotherapy by micellar-encapsulated drug and ultrasound, J.Control.Release 2005, 102, 203-222. (51) Otsuka, H.; Nagasaki, Y.; Kataoka, K., PEGylated nanoparticles for biological and pharmaceutical applications, Adv. Drug Deliv. Rev. 2003, 55, 403-419. (52) Le Garrec, D.; Gori, S.; Luo, L.; Lessard, D.; Smith, D. C.; Yessine, M. A.; Ranger, M.; Leroux, J. C., Poly(N-vinylpyrrolidone)-block-poly(D,L-lactide) as a new polymeric solubilizer for hydrophobic anticancer drugs: in vitro and in vivo evaluation, J.Control.Release 2004, 99, 83-101. (53) Chung, T. W.; Cho, K. Y.; Lee, H. C.; Nah, J. W.; Yeo, J. H.; Akaike, T.; Cho, C. S., Novel micelle-forming block copolymer composed of poly ([var epsilon]caprolactone) and poly(vinyl pyrrolidone), Polymer 2004, 45, 1591-1597. (54) Zuccari, G.; Carosio, R.; Fini, A.; Montaldo, P. G.; Orienti, I., Modified polyvinylalcohol for encapsulation of all-trans-retinoic acid in polymeric micelles, J.Control.Release 2005, 103, 369-380. (55) Wang, Y. C.; Tang, L. Y.; Sun, T. M.; Li, C. H.; Xiong, M. H.; Wang, J., SelfAssembled Micelles of Biodegradable Triblock Copolymers Based on Poly(ethyl ethylene phosphate) and Poly(e-caprolactone) as Drug Carriers, Biomacromolecules 2008, 9, 388-395. (56) Hubalek, Z., Protectants used in the cryopreservation of microorganisms, Cryobiology 2003, 46, 205-229. (57) Nam, Y. S.; Kang, H. S.; Park, J. Y.; Park, T. G.; Han, S. H.; Chang, I. S., New micelle-like polymer aggregates made from PEI-PLGA diblock copolymers: micellar characteristics and cellular uptake, Biomaterials 2003, 24, 2053-2059. Recent Advances with Polymeric Micelles 28 (58) Li, Y. Y.; Zhang, X. Z.; Kim, G. C.; Cheng, H.; Cheng, S. X.; Zhuo, R. X., Thermosensitive Y-Shaped Micelles of Poly(oleic acid-Y-N-isopropylacrylamide) for Drug Delivery, Small 2006, 2, 917-923. (59) Liu, S. Q.; Tong, Y. W.; Yang, Y. Y., Incorporation and in vitro release of doxorubicin in thermally sensitive micelles made from poly(N-isopropylacrylamideco-N,N-dimethylacrylamide)-b-poly(d,l-lactide-co-glycolide) with varying compositions, Biomaterials 2005, 26, 5064-5074. (60) Jang, W. D.; Nakagishi, Y.; Nishiyama, N.; Kawauchi, S.; Morimoto, Y.; Kikuchi, M.; Kataoka, K., Polyion complex micelles for photodynamic therapy: Incorporation of dendritic photosensitizer excitable at long wavelength relevant to improved tissuepenetrating property, J.Control.Release 2006, 113, 73-79. (61) Zhang, L.; Nguyen, T. L. U.; Bernard, J.; Davis, T. P.; Barner-Kowollik, C.; Stenzel, M. H., Shell-Cross-Linked Micelles Containing Cationic Polymers Synthesized via the RAFT Process: Toward a More Biocompatible Gene Delivery System, Biomacromolecules 2007, 8, 2890-2901. (62) Jiang, T. Y.; Wang, Z. Y.; Chen, C.; Mo, F. K.; Xu, Y. L.; Tang, L. X.; Liang, J. j., Poly(aspartic-acid) derivatives as polymeric micelle drug delivery systems, J.Appl.Polym.Sci. 2006, 101, 2878. (63) Kim, S. H.; Jeong, J. H.; Mok, H.; Lee, S. H.; Kim, S. W.; Park, T. G., Folate Receptor Targeted Delivery of Polyelectrolyte Complex Micelles Prepared from ODN-PEG-Folate Conjugate and Cationic Lipids, Biotechnol.Prog. 2007, 23, 232237. (64) Santerre, J. P.; Woodhouse, K.; Laroche, G.; Labow, R. S., Understanding the biodegradation of polyurethanes: From classical implants to tissue engineering materials, Biomaterials 2005, 26, 7457-7470. (65) Lee, K.; Choi, H. S.; Kim, J. Y.; Ahn, I. S., Distribution of phenanthrene between soil and an aqueous phase in the presence of anionic micelle-like amphiphilic polyurethane particles, J.Hazard.Mater. 2003, 105, 179-197. (66) Najafi, F.; Sarbolouki, M. N., Biodegradable micelles/polymersomes from fumaric/sebacic acids and poly(ethylene glycol), Biomaterials 2003, 24, 1175-1182. (67) Kabanov, A. V.; Batrakova, E. V.; Alakhov, V. Y., Pluronic(R) block copolymers as novel polymer therapeutics for drug and gene delivery, J.Control.Release 2002, 82, 189-212. (68) Chiappetta, D. A.; Sosnik, A., Poly(ethylene oxide)-poly(propylene oxide) block copolymer micelles as drug delivery agents: Improved hydrosolubility, stability and bioavailability of drugs, Eur.J.Pharm.Biopharm. 2007, 66, 303-317. (69) Exner, A. A.; Krupka, T. M.; Scherrer, K.; Teets, J. M., Enhancement of carboplatin toxicity by Pluronic block copolymers, J.Control.Release 2005, 106, 188-197. Chapitre I 29 (70) Lavasanifar, A.; Samuel, J.; Kwon, G. S., Poly(ethylene oxide)-block-poly(-amino acid) micelles for drug delivery, Adv.Drug Deliv.Rev. 2002, 54, 169-190. (71) Yin, H.; Lee, E. S.; Kim, D.; Lee, K. H.; Oh, K. T.; Bae, Y. H., Physicochemical characteristics of pH-sensitive poly(l-Histidine)-b-poly(ethylene glycol)/poly(lLactide)-b-poly(ethylene glycol) mixed micelles, J.Control.Release 2008, 126, 130138. (72) Gao, Z. G.; Lee, D. H.; Kim, D. I.; Bae, Y. H., Doxorubicin loaded pH-sensitive micelle targeting acidic extracellular pH of human ovarian A2780 tumor in mice, J.Drug Target. 2005, 13, 391-397. (73) Brown, M. D.; Schatzlein, A.; Brownlie, A.; Jack, V.; Wang, W.; Tetley, L.; Gray, A. I.; Uchegbu, I. F., Preliminary Characterization of Novel Amino Acid Based Polymeric Vesicles as Gene and Drug Delivery Agents, Bioconjugate.Chem. 2000, 11, 880-891. (74) Opanasopit, P.; Yokoyama, M.; Watanabe, M.; Kawano, K.; Maitani, Y.; Okano, T., Block Copolymer Design for Camptothecin Incorporation into Polymeric Micelles for Passive Tumor Targeting, Pharm.Res. 2004, 21, 2001-2008. (75) Croy, S. R.; Kwon, G. S., Polymeric Micelles for Drug Delivery, Curr.Pharm.Design 2006, 12, 4669-4684. (76) Park, J. H.; Lee, S.; Kim, J. H.; Park, K.; Kim, K.; Kwon, I. C., Polymeric nanomedicine for cancer therapy, Prog.Polym.Sci. 2008, 33, 113-137. (77) Li, X.; Mya, K. Y.; Ni, X.; He, C.; Leong, K. W.; Li, J., Dynamic and Static Light Scattering Studies on Self-Aggregation Behavior of Biodegradable Amphiphilic Poly(ethylene oxide)-Poly[(R)-3-hydroxybutyrate]-Poly(ethylene oxide) Triblock Copolymers in Aqueous Solution, J.Phys.Chem.B 2006, 110, 5920-5926. (78) Chen, C.; Yu, C. H.; Cheng, Y. C.; Yu, P. H. F.; Cheung, M. K., Biodegradable nanoparticles of amphiphilic triblock copolymers based on poly(3-hydroxybutyrate) and poly(ethylene glycol) as drug carriers, Biomaterials 2006, 27, 4804-4814. (79) Lee, H.; Zeng, F.; Dunne, M.; Allen, C., Methoxy Poly(ethylen glycol)-block-Poly(dvalerolactone) Copolymer Micelles for Formulation of Hydrophobic Drugs, Biomacromolecules 2005, 6, 3119-3128. (80) Lin, W. J.; Wang, C. L.; Juang, L. W., Characterization and comparison of diblock and triblock amphiphilic copolymers of poly(e-valerolactone), J.Appl.Polym.Sci. 2006, 100, 1836-1841. (81) Zeng, F.; Lee, H.; Chidiac, M.; Allen, C., Synthesis and Characterization of Six-Arm Star Poly(e-valerolactone)-block-Methoxy Poly(ethylene glycol) Copolymers, Biomacromolecules 2005, 6, 2140-2149. (82) Li, B. H.; Moriyama, E. H.; Li, F. G.; Jarvi, M. T.; Allen, C.; Wilson, B. C., Diblock copolymer micelles deliver hydrophobic protoporphyrin IX for photodynamic therapy, Photochem.Photobiol. 2007, 83, 1505-1512. Recent Advances with Polymeric Micelles 30 (83) Aliabadi, H. M.; Mahmud, A.; Sharifabadi, A. D.; Lavasanifar, A., Micelles of methoxy poly(ethylene oxide)-b-poly([var epsilon]-caprolactone) as vehicles for the solubilization and controlled delivery of cyclosporine A, J.Control.Release 2005, 104, 301-311. (84) Aliabadi, H. M.; Elhasi, S.; Mahmud, A.; Gulamhusein, R.; Mahdipoor, P.; Lavasanifar, A., Encapsulation of hydrophobic drugs in polymeric micelles through co-solvent evaporation: The effect of solvent composition on micellar properties and drug loading, Int.J.Pharm. 2007, 329, 158-165. (85) Shuai, X.; Ai, H.; Nasongkla, N.; Kim, S.; Gao, J., Micellar carriers based on block copolymers of poly([var epsilon]-caprolactone) and poly(ethylene glycol) for doxorubicin delivery, J.Control.Release 2004, 98, 415-426. (86) Sutton, D.; Wang, S. H.; Nasongkla, N.; Gao, J. M.; Dormidontova, E. E., Doxorubicin and beta-lapachone release and interaction with micellar core materials: Experiment and modeling, Exp.Biol.Med. 2007, 232, 1090-1099. (87) Geng, Y.; Discher, D. E., Visualization of degradable worm micelle breakdown in relation to drug release, Polymer 2006, 47, 2519-2525. (88) Letchford, K.; Liggins, R.; Burt, H., Solubilization of hydrophobic drugs by methoxy poly(ethylene glycol)-block-polycaprolactone diblock copolymer micelles: Theoretical and experimental data and correlations., J.Pharm.Sci. 2008, 97, 1179-1190. (89) Ma, Z.; Haddadi, A.; Molavi, O.; Lavasanifar, A.; Lai, R.; Samuel, J., Micelles of poly(ethylene oxide)-b-poly(-caprolactone) as vehicles for the solubilization, stabilization, and controlled delivery of curcumin, J.Biomed.Mater.Res.A 2007, in Press, doi:10.1002/jbm.a.31584. (90) Lee, J. S.; Bae, J. W.; Joung, Y. K.; Lee, S. J.; Han, D. K.; Park, K. D., Controlled dual release of basic fibroblast growth factor and indomethacin from heparinconjugated polymeric micelle, Int.J.Pharm. 2008, 346, 57-63. (91) Lee, H.; Hu, M.; Reilly, R. M.; Allen, C., Apoptotic Epidermal Growth Factor (EGF)Conjugated Block Copolymer Micelles as a Nanotechnology Platform for Targeted Combination Therapy, Mol.Pharm. 2007, 4, 769-781. (92) Chen, L.; Xie, Z.; Hu, J.; Chen, X.; Jing, X., Enantiomeric PLA–PEG block copolymers and their stereocomplex micelles used as rifampin delivery, J.Nanopart.Res. 2007, 9, 777-785. (93) From the website of the US National Institutes of Health, http://clinicaltrials.gov with the keywords "genexol", 2008. (94) Jie, P.; Venkatraman, S. S.; Min, F.; Freddy, B. Y. C.; Huat, G. L., Micelle-like nanoparticles of star-branched PEO-PLA copolymers as chemotherapeutic carrier, J.Control.Release 2005, 110, 20-33. Chapitre I 31 (95) Lee, E. S.; Oh, K. T.; Kim, D.; Youn, Y. S.; Bae, Y. H., Tumor pH-responsive flowerlike micelles of poly(l-lactic acid)-b-poly(ethylene glycol)-b-poly(l-histidine), J.Control.Release 2007, 123, 19-26. (96) Lo, C. L.; Huang, C. K.; Lin, K. M.; Hsiue, G. H., Mixed micelles formed from graft and diblock copolymers for application in intracellular drug delivery, Biomaterials 2007, 28, 1225-1235. (97) Blanco, E.; Bey, E. A.; Dong, Y.; Weinberg, B. D.; Sutton, D. M.; Boothman, D. A.; Gao, J., [beta]-Lapachone-containing PEG-PLA polymer micelles as novel nanotherapeutics against NQO1-overexpressing tumor cells, J.Control.Release 2007, 122, 365-374. (98) Lee, W. C.; Li, Y. C.; Chu, I. M., Amphiphilic Poly(D,L-lactic acid)/Poly(ethylene glycol)/Poly(D,L-lactic acid) Nanogels for Controlled Release of Hydrophobic Drugs, Macromol.Biosci. 2008, 6, 846-854. (99) Yang, Z. L.; Li, X. R.; Yang, K. W.; Liu, Y., Amphotericin B-loaded poly(ethylene glycol)-poly(lactide) micelles: Preparation, freeze-drying, and in vitro release, J. Biomed. Mater. Res. 2008,85,539-46 (100) Zhang, G.; Zhang, R.; Wen, X.; Li, L.; Li, C., Micelles Based on Biodegradable Poly(L-glutamic acid)-b-Polylactide with Paramagnetic Gd Ions Chelated to the Shell Layer as a Potential Nanoscale MRI-Visible Delivery System, Biomacromolecules 2008, 9, 36-42. (101) Jeong, J. H.; Park, T. G., Poly(-lysine)-g-poly(-lactic-co-glycolic acid) micelles for low cytotoxic biodegradable gene delivery carriers, J.Control.Release 2002, 82, 159166. (102) Lee, E. S.; Shin, H. J.; Na, K.; Bae, Y. H., Poly(-histidine)-PEG block copolymer micelles and pH-induced destabilization, J.Control.Release 2003, 90, 363-374. (103) Liu, S. Q.; Tong, Y. W.; Yang, Y. Y., Thermally sensitive micelles self-assembled from poly(N-isopropylacrylamide-co-N,N-dimethylacrylamide)-b-poly(d,l-lactide-coglycolide) for controlled delivery of paclitaxel, Molecular BioSystems 2005, 1, 158165. (104) Yoo, H. S.; Park, T. G., Folate receptor targeted biodegradable polymeric doxorubicin micelles, J.Control.Release 2004, 96, 273-283. (105) Trimaille, T.; Gurny, R.; Möller, M., Synthesis and ring-opening polymerization of new monoalkyl-substituted lactides, J.Polym.Sci.Part A: Polym.Chem. 2004, 42, 4379-4391. (106) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. Recent Advances with Polymeric Micelles 32 123456789A89BC58648D4C234A889EF4BC3948EE3483E4941456C84234573EE34 786D5934C234C9BC3F4 1 4 123489342F98A28574C234573EE347893!4C2343CC394C23456789A89BC58648D4B42F98A285749"#4 4 83$39!4C2542FA8C23546334C8434$395D534C84B6394C234%3C586&4 1 '4C23478673AC4BAAE57BE34C84BEE423(A889EF4BC3948EE3494D89EBC586)#4 1 1 1 12345678998 8 8 1 Chapter II 33 Note Formulation of Poorly Water Soluble Drugs with MPEG-hexPLA Micelles: Investigations on the Effects of Drug Physical-Chemical Parameters on the Incorporation K. Mondon, R.Gurny, and Michael Möller. School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland Today, over 9 out of 10 pharmaceutical lead products are lipophilic and poorly water soluble, which render their formulation difficult and limits their clinical use. In this regard, solubilisation of such drugs into micelles made of methoxypoly(ethylene glycol) hexylsubstituted poly(lactide) (MPEG-hexPLA) was investigated as an alternative pharmaceutical formulation. The incorporation efficiency of 18 different drugs into MPEG-hexPLA micelles was studied. Relationships and trends between the known physical-chemical properties of the 18 drugs and their incorporation capacity were examined. All drug loaded micelles had a size below 200 nm. The incorporation of these drugs was generally higher in MPEG-hexPLA than in the comparable MPEG-polylactide (MPEG-PLA) micelles. For some drugs the aqueous solubility could be improved up to 100 times. The physical-chemical parameters, water solubility, log P, number of H bond acceptor (Ha) and H bond donor (Hd) were found to strongly influence drug incorporation into the micelles. Concerning the aqueous drug solubility and the aqueous solubility increase with micelles, the drug water solubility was the most influencing parameter followed by Hd, Ha, and log P. Hd was the most predominant parameter for the drug/copolymer ratio in the micelles, followed by log P, Ha, and the drug water solubility. The drug molecular weight and the surface tension were found as minor Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 34 influencing parameters. A qualitative trend was observed for drugs with low water solubility (< 0.055 mM). They showed a high increase of their aqueous solubility when incorporated into MPEG-dihexPLA micelles. Amongst the variety of investigated drugs, some drugs do not fit the Lipinski’s “rule of 5” but have suitable physico-chemical parameters for an efficient incorporation into MPEG-hexPLA micelles. This envisions the use of micelle solutions as an alternative formulation approach for these poorly water soluble drugs. Keywords: polymeric micelles; prediction; poorly water soluble drugs; solubilisation, substituted polylactides. Chapter II 35 1. Introduction In the past two decades drug discovery has been accelerated by the use of high throughput screening (HTS) and combinatorial chemistry methodologies. Amongst the large library of novel active compounds 95% are lipophilic and poorly water soluble 1, which is often the critical point to their success in the clinics 2;3. Different strategies to improve solubilisation of poorly water soluble drugs have been developed. Co-solvency is one approach to enhance water solubility 4 with the advantage of the protection of the drug from hydrolysis. However, unstable formulations upon dilution in the blood can lead to local drug precipitation after administration, inducing serious side effects like pain, inflammation or haemolysis 5;6 . Alternatively a complexation with carriers like cyclodextrins can be used, but its application is limited by high toxicity at high concentrations 7 and by the susceptibility of the complex dissociation upon dilution. Oil-in-water emulsion is another approach for very lipophilic drugs, however the use of low molecular weight surfactants could lead to some toxicity issues like it is the case for Cremophor®EL 8;9 . Polymer based drug delivery systems are a further alternative choice for the formulation of poorly water soluble drugs. Amongst the variety of applied biocompatible and biodegradable, respectively polymers 10 (poly(ethers), poly(esters), poly(L-amino acids etc.) with different structure (linear, dendrons, multi block, etc.), amphiphilic diblock copolymers have the particularity to self-assemble in aqueous media into a specific micelle core-shell structure. Such nanocarriers can incorporate a drug in their core, preventing its precipitation in an aqueous environment. The nanosize and pegylation of studied carrier systems facilitate long-circulation, and ability to cross certain membranes and passively targeting diseased tissues. The hydrophilic corona most often composed of the biocompatible poly(ethylene glycol) (PEG) limits protein adsorption at the surface and therefore warranties long circulating properties 11 . In addition polymeric micelles with sizes below 200 nm can accumulate at sites of leaky vasculature like inflamed or cancer tissues 12;13, which make them interesting for a passive targeting 14 . Concerning the hydrophobic core, usually poly(ethers), poly(L-amino acids) and poly(esters) are used 15. The micelle core forms a reservoir into which a drug can be incorporated and consequently the micellar system enhances significantly its aqueous solubility. The amount of drug that can be incorporated within the core depends principally on the compatibility of the drug with the Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 36 polymer of the core forming block, as outlined for PEG-poly(caprolactone) (PCL) 16-21, PEGPCL and PEG-poly(lactide) (PLA) vinyl pyrrolidone-styrene) 16-21 16-21 , poly(ethylene oxide-propylene oxide) and poly(N- . In our own work, cyclosporin A was incorporated into MPEG-hexPLA micelles with different degrees of hydrophobicity of the core forming block 21 . The results revealed that the more hydrophobic the core, the higher the drug incorporation. Better affinity between the core and the hydrophobic drug was created with the presence and increasing number of hexyl groups in the PLA core forming block, as shown for the di-hexyl substituted PLA (dihexPLA) compared to the mono-hexyl substituted PLA (monohexPLA) and PLA. The affinity of a drug to the core forming polymer can be estimated with the theoretical Flory-Huggins parameters (χsp), via the calculation of the theoretical Hildebrand or Hansen solubility parameters of both the drug and the polymer 22 . The difference obtained between the solubilisate (the drug) and the solvent (the polymer) indicates the miscibility of both components. The closer χsp is to 0, the better is the miscibility reflecting a higher affinity of the drug and solubility in the given polymer, here resembling its incorporation efficiency into the micelle core. To evaluate a possible prediction or estimation of the drug incorporation into MPEG-hexPLA micelles, 18 poorly soluble drugs were tested. The theory of Flory-Huggins was first applied to each drug/copolymer system to evaluate the different affinities. Relationships and trends between the known physical-chemical parameters of the drugs and the experimentally obtained incorporation results (i.e. drug content in micelle solution, drug aqueous solubility in micelle solution, aqueous solubility increase) are presented. 2. Materials and methods 2.1. Materials Chemicals Tetrahydrofuran (THF) was supplied by SDS (Toulouse, France), and dried by distillation. Methoxy-poly(ethylene glycol) with a molecular weight of 2000 g/mol (MPEG2000g/mol) was a gift from BASF (Ludwigshafen, Germany). D,L-lactide, tin(II) 2-ethylhexanoate (Sn(Oct)2), and acetone p.a. were purchased from Purac Biochem (Gorinchem, The Netherlands), Aldrich (Buchs, Switzerland), and Fluka (Buchs, Switzerland), respectively and used as received. The Chapter II 37 monomers, mono-hexyl-substituted lactide (monohexLA) and di-hexyl-substituted lactide (dihexLA) were synthesized as described previously 23. Dichloromethane (DCM) (HPLC grade) and toluene were supplied by Fluka (Buchs, Switzerland). Methanol (MeOH) and acetonitrile (ACN) (HPLC grade) were purchased from VWR (Nyon, Switzerland). Ammonium dihydrogen phosphate, di-potassium hydrogen phosphate, sodium chloride, acetic acid, pyrophosphoric acid, phosphoric acid, formic acid were supplied by Fluka, (Buch, Switzerland). Diethylamine was purchased from Acros organics (Geel, Belgium) Drugs Clotrimazole (CLZ), Danazol (DAN), Diclofenac sodium (DICLO), Econazole nitrate (ECZ), Etoposide (ETO), Fluconazole (FLZ), Griseofulvin (GF), Ketoconazole (KETO), Piroxicam (PIRO), Quercetin dehydrate (QUER) and Naproxen sodium (NAPRO) were all supplied by Sigma (Buchs, Switzerland). Cyclosporin A (CsA), Docetaxel (DTX), Hydrocortisone (HYDRO), Hypericin (HY), Paclitaxel (PTX), and meso-tetra(p-hydroxyphenyl)porphine (THPP) were purchased from Fluka, AlfaChem (Kings Point, NY, US), from Aldrich, from Alexis Corporation (Lausen, Switzerland), Oskar Tropitzsch (Marktredwitz, Germany), and Frontier Scientific (Carnforth, UK), respectively. Verteporfin (VER) was extracted from Visudyne (Novartis Pharma, Bern, Switzerland) purchased from a local pharmacy. 2.2. Synthesis and characterization of MPEG-(hex)PLA copolymers The MPEG-(hex)PLA copolymers with a molecular weight of around 5000g/mol were synthesized by ring opening polymerization (ROP) with MPEG 2000 as initiator as described previously 21;24 , see Scheme 1 for their chemical structures. The copolymers were characterized by their molecular weight (Mn) and polydispersity index (P.I.) using gel permeation chromatography (GPC). The GPC setup was composed of a Waters system with Waters Styragel HR1-3 columns and a Waters 410 differential refractometer (Waters, Milford, USA). The analysis was carried out using polystyrene (PS) of different molecular weights as calibration standards (PSS, Mainz, Germany). The degree of polymerization of MPEG-(hex)PLA was calculated from 1H NMR (CDCl3, Bruker 300 MHz). Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 38 Please note that in the following text, “MPEG-(hex)PLA” refers to both MPEG-PLA and MPEG-hexPLA copolymers. O O H3C 45 R1, R2 = CH3 1 R 2 O O R 1 O H m O : MPEG2000-PLA 2 R =C6H13, R =CH3 : MPEG2000-monohexPLA R1, R2 =C6H13 : MPEG2000-dihexPLA Scheme 1. Structure of MPEG-hexPLA and MPEG-PLA block copolymers 2.3. Preparation of drug loaded MPEG-(hex)PLA micelles The preparation and incorporation of drugs into MPEG-(hex)PLA micelles followed the cosolvent evaporation method. Six milligrams of drug and 20 mg of copolymer were dissolved in 2 mL acetone or THF. The organic solution was added dropwise under stirring into 4 mL of ultra pure water using a peristaltic pump. The organic solvent was removed under stirring by evaporation at 200 PSI in a desiccator for 2h under vacuum at 200 mbar. Final micelle concentrations were adjusted to 5 mgcopolymer/mL by adding ultra pure water. The solutions were left to equilibrate overnight and then centrifuged at 9500 x g for 15 min to remove nonincorporated drug. Afterwards, drug loaded MPEG-(hex)PLA micelles were analyzed for their size and their drug content. 2.4. Size determination of drug loaded MPEG-(hex)PLA micelles The hydrodynamic- (Zav) and number-weight (dn) diameters of drug loaded MPEG-(hex)PLA micelles were measured by dynamic light scattering (DLS) using a Zetasizer HS 3000 (Malvern, Worcestershire, UK). Analyzes were performed at an angle of 90° at 25°C. For each sample, the mean diameters were obtained in triplicate. Chapter II 39 2.5. Drug loading determination of drug loaded MPEG-(hex)PLA micelles The drug loading of MPEG-(hex)PLA micelles were determined after centrifugation of micelle formulations. The supernatant was diluted in acetonitrile at a ratio 1:10 to break up the micelles and to release the drug for quantitative analysis. The drug loading of MPEG-(hex)PLA was mainly performed by HPLC for the majority of the micelles (see Supplementary Material for the different HPLC conditions). For VER and THPP loaded micelles UV absorbance was used to quantify the drug content. The drug content (DC), the drug solubility in micelles ([Drug]micelles) and the “solubility increase factor” were calculated using the following equations: Drug content [mgdrug /g copolymer ] = [Drug]micelles[mM] = mass of drug incorporated in micelles [mg] mass of copolymer used [g] DC [mgdrug /g copolymer ] × concentration of copolymer [g/L] Molecular weight of the drug [g/mol] "Solubility increase factor" = [Drug]micelles [mM] Drug water solubility [mM] (1) (2) (3) 2.6. Determination of the Flory-Huggins parameter χsp The Flory-Huggins parameter reflects the compatibility between the drug and the (hex)PLA micelle core. It was calculated by the following equation: χ sp = (δ drug −δ copolymer )2 × V RT (4) δdrug and δpolymer are the solubility parameters for the drug, and for the core-forming block of the copolymer, respectively. R is the gas constant, T the temperature in Kelvin, and V the molar volume of the drug. Values for δdrug and δpolymer were calculated by the additive group contribution method as described by Van Krevelen 25 using the Material Studio Software (Accelrys Inc., USA). The molar volume V of the drug was calculated by the group contribution method (GCM) according to Fedors 26 using the same software. The temperature was set at 298 K (25°C). Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 40 3. Results and Discussion Due to the large number of potent drugs with low water solubility and high lipophilicity, and the resulting limitations for pharmaceutical formulations, polymeric micelles have attracted a lot of attention as possible drug delivery systems 19 . With their specific core-shell structure, micelles can incorporate a drug in their core, while the hydrophilic corona renders aqueous solubility. Considering that the affinity of a drug and the core-forming block is affecting the incorporation of a drug into micelles, novel MPEG-hexPLA based micelles with an increased hydrophobicity of the micelle core were developed 24. These MPEG-hexPLA micelles showed higher drug loadings, lower critical micellar concentrations and higher stabilities compared to the standard MPEG-PLA micelles. As an example, the immunosuppressant drug cyclosporin A (CsA) was 4 times more incorporated into MPEG-dihexPLA than into MPEGPLA micelles, leading to an aqueous solubility of 1.4 mg/mL 21 . MPEG-dihexPLA micelles showed also a superior stability at body temperature, both in water and in PBS pH 7.4 27 , in comparison to the less hydrophobic MPEG-monohexPLA micelles and the standard MPEGPLA micelles. To further generally examine the incorporation efficiency of MPEG-hexPLA and MPEG-PLA micelles, respectively, 18 poorly water soluble drugs with different physicochemical properties were tested. The experimental results were then correlated with the FloryHuggins parameter and the physico-chemical parameters of the drug regarding the obtained drug content (drug/copolymer ratio) in micelle solution, the drug solubility in micelle solution and the aqueous solubility increase (ratio between drug aqueous solubility obtained in micelles over drug water solubility). The used copolymers, MPEG-monohexPLA and MPEG-dihexPLA, and the “standard” MPEG-PLA were synthesized in a controlled manner by ROP with a defined molecular weight of around 5000 g/mol (± 1 monomer unit) (Table 1), thus allowing a good comparison of the influence of the hexyl substituents on the drug incorporation. Chapter II 41 Table 1. Characteristics of MPEG-(hex)PLA copolymers Copolymer Mn a (g/mol) P.I. b # hexyl groups MPEG-PLA 5050 1.17 0 MPEG-monohexPLA 5040 1.13 14 MPEG-dihexPLA 5250 1.15 23 a determined by 1H NMR; b determined by GPC. The investigated drugs (Figure 1) were selected to cover a wide range of lipophilicity values (log P), and of different physical-chemical characteristics (Table 2). They are all known to be poorly water soluble. The chosen drugs are used in various pharmaceutical applications such as cancer-, transplantation rejection-, dermatology-, or ophthalmology treatments. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 42 O N N N N HO O N N N N O N Cinnarizine CIN O H Clotrimazole CLZ H 3C O O H N N O N O O Cl O H N H N H 3C HO N N O CH O Danazol DAN Cyclosporine A CsA HO HO + NH O Na - O NH O OH O O Cl O O Cl H H O Cl N H O O N O OH O Cl O HO OO N OH O O O O O H O O HO O Cl HO Docetaxel DTX Diclofenac sodium DICLO Econazole nitrate ECZ Etoposide ETO O O Cl Cl OH N N Cl N MeO HO HO O N N F OMe N O O HO O HO OMeO O N F OH O OH N O Hypericin HY Hydrocortisone HYDRO Griseofulvin GF Fluconazole FLZ H3C Ketoconazole KETO OH HO O O O OH OH O O OH H3C O N H3C HO OH O H NH O H N NH NH N O O OH N O OO O S HN N O OH O O OH HO Piroxicam PIRO Paclitaxel PTX THPP MeO OMe H3C OH OH O O HO O HO OH OH + Na O Quercetin dihydrate QUER O O CH3 .2H 2 O O O - Naproxen sodium NAPRO Figure 1. Structure of all tested drugs O N H N N H N Verteporfin VER O N Chapter II 43 Table 2. Characteristics of the investigated drugs Drug Mw [g/mol] Exp. logP CLZ 344.80 6.1 a Exp. water solubility [mM] 0.0880 b CsA 1202.60 8.2 c 0.0100 c 23.389 5 17 31.6 DAN 337.5 3.8 a 0.0017 e 20.77 1 2 55.0 DICLO 318.1 4.5 d 0.0560 f 23.21 1 0 57.9 DTX 807.9 2.4 a 0.0068 g 23.66 5 10 66.2 - 1 5 45.4 c δdrug Van Krevelen [MPa 1/2] Hd bond donor Ha bond acceptor Surface tension [dyne/cm] 20.80 0 2 - ECZ 444.7 - 1.9316 ETO 588.9 1a 0.1593 h 24.52 3 10 76.4 FLZ 306.3 0.5 b 0.0033 i 24.16 1 8 55.4 GF 352.8 3.18 a 0.0600 c 21.08 0 6 52.5 HYDRO 362.5 1.72 a 0.0003 j 25.11 3 2 58.8 HY 504.4 8.78 a 0.0500 c 33.40 6 2 150.2 KETO 531.5 4.35 d 0.0320 k 21.08 0 8 52.1 PTX 853.9 3a 0.0004 l 23.38 4 11 68.4 PIRO 331.2 3.06 d 0.0695 d 24.11 2 5 79.5 QUER 338.3 1.48 d 0.1774 d 32.73 4 2 114.8 THPP 678.5 - 0.0030 c 25.94 6 2 75.5 VER 718.8 3.74 a 0.0110 c 21.29 3 9 70.1 NAPRO 230.3 2.8 0.0691 c - 1 1 - With Mw the molecular weight; Exp. log P, the experimental partition coefficient between octanol and water; δdrug the solubility parameter calculated with the software Material Studio according to the Van Krevelen group contribution method; a data taken from http://www.drugbank.ca/drugs; b OSPAR Commission, 2005. http://www.ospar.org/documents/dbase/publications/p00199_BD%20on%20clotrimazole.pdf; c values obtained experimentally by Carrupt et al. (University of Geneva, Pharmacochemistry, Geneva, Switzerland, unpublished data); d data taken from ChemIDplus Advanced http://chem.sis.nlm.nih.gov/chemidplus/. e Erlich et al., Int. J. Pharm., 1999, 179, 49-53; f Llinas et al., J. Med. Chem., 2007, 50, 979-983; g Ali et al., J. Med. Chem., 1997, 40, 236-241; h Shah et al., Int. J. Pharm., 1995, 113, 103-111; i Loftsson T, Hreinsdóttir D. Determination of Aqueous Solubility by Heating and Equilibration: A Technical Note. AAPS PharmSciTech. 2006; 7(1): E1-E4. j Ould-ouali et al., Pharm. Res., 2004, 21, 1581-1590; k BioAqueous Solubilization Services, Dowpharma. http://www.dow.com/webapps/lit/litorder.asp?filepath=pharma/pdfs/noreg/715-00011.pdf&pdf=true; l Soga et al., J.Control. Release, 2005, 103, 341-353; m Soga et al., J.Control. Release, 2005, 103, 341-353 Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 44 All drugs were formulated and incorporated into MPEG-(hex)PLA micelles by a co-solvent evaporation standard procedure. It has to be pointed out that neither the preparation procedure nor the polymer molecular weight and polymer block-ratio have been optimized for the specific drugs. It is expected that a systematic optimization could significantly improve the incorporation results. In general the micelle size obtained for all formulations were similar, with hydrodynamic diameters (Zav) varying from 70 to 200 nm and number weight diameters (dn) from 17 to 150 nm (Table 3). This small carrier size could be of importance for a possible use for passive drug targeting to inflamed or diseased tissues 12;14 . The majority of the drugs was much more efficient and higher incorporated into MPEG-dihexPLA than into MPEGmonohexPLA micelles. In contrast, the drug incorporation results i.e. the drug content (mgdrug/gcopolymer), the drug solubility in micelle solution (mM), as well as the “solubility increase factor” (between drug solubility in micelles and water solubility) were significant different for the tested formulations. Various observations are remarkable when comparing drugs with similar log P values or similar water solubility values. For example, GF (log P =3.18) incorporated at 16.6 mg/g shows a more than 4 times higher drug incorporation in MPEG-dihexPLA micelles than PIRO (log P=3.06). For drugs with a significantly different log P, FLZ (log P=0.5) and CLZ (log P=6.1), a higher drug incorporation in MPEG-dihexPLA micelles was surprisingly obtained for FLZ with an achieved drug content of 268 mg/g, corresponding to a 4 times higher drug content than for CLZ (59 mg/g). As another example, PTX, with an experimental water solubility of 0.0004 mM compared to HYDRO with a similar water solubility of 0.0003mM, could be 5 times less incorporated in MPEG-dihexPLA micelles. In consequence, it is not applicable to draw on only one physical-chemical parameter like log P to predict the incorporation of a selected drug into polymeric micelles. Therefore, the influence of the various physical-chemical parameters needs to be evaluated in more detail for a possible prediction or estimation of the drug incorporation ability. Chapter II 45 Table 3. Incorporation results, size characteristics and Flory-Huggins parameter of all the investigated drug loaded MPEG-(hex)PLA micelle formulations. (a) MPEG-PLA micelles Micelle size Drug CLZ χsp Drug content Van Krevelen [mgdrug/gcopolymer] 1/2 [MPa ] 0.49 129.5 [Drug]micelles [mM] 1.877 Solubility increase Zav P.I. dn [%]dn factor 21.3 56 0.42 25 99.9 CsA 1.14 66.2 0.275 27.5 89 0.39 28 99.9 DAN 15.02 3.5 0.051 30.0 118 0.43 29 99.9 DICLO 1.18 76.8 1.183 21.1 179 0.16 156 100.0 DTX 2.68 4.2 0.026 3.8 194 0.69 28 100.0 ECZ - 177.1 1.991 1.0 70 0.29 29 100.0 ETO 8.46 229.0 1.937 12.2 154 0.53 26 95.7 FLZ 3.82 250.2 4.051 1240.7 92 0.44 20 99.2 GF 1.34 18.7 0.265 4.4 HYDRO 6.82 47.4 0.649 2163.3 HY 32.42 6.7 0.066 1.3 126 0.52 17 100.0 KETO 1.94 99.1 0.923 28.8 129 PTX 9.09 1.8 0.011 27.5 208 0.69 28 100.0 PIRO 3.87 1.4 0.021 0.3 184 0.33 76 QUER 19.07 9.4 0.139 0.8 100 0.52 23 100.0 THPP 13.68 4.6 0.033 11.0 107 0.50 35 100.0 VER 3.01 11.9 0.151 13.7 111 0.46 26 100.0 - 14.6 0.064 0.9 161 0.24 83 NAPRO 114 0.43 25 100.0 66 0.24 23 99.9 25 100.0 99.9 99.8 With [Drug]micelles, the concentration of drug in micelles; Zav the hydrodynamic diameter, dn the number weight diameter and [%]dn the percentage of micelles with a given dn. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 46 (b) MPEG-monohexPLA micelles Micelle size Drug χsp Solubility Drug content [Drug]micelles Van Krevelen increase [mgdrug/gcopolymer] [mM] [MPa 1/2] factor Zav P.I. dn [%]dn CLZ 1.378 34.7 0.503 5.7 33 0.02 19 100.0 CsA 16.711 213.7 0.907 90.7 82 0.54 24 100.0 DAN 1.424 8.1 0.120 70.6 188 0.25 140 96.5 DICLO 2.996 149.9 2.269 40.5 147 0.34 19 99.8 DTX 10.638 11.6 0.072 10.6 178 0.41 45 97.9 ECZ - 252.7 2.841 1.5 145 0.41 130 97.7 ETO 9.252 275.8 2.375 14.9 183 0.44 24 100.0 FLZ 4.201 271.7 4.452 1363.5 112 0.51 18 100.0 GF 1.598 13.7 0.195 3.3 145 0.51 20 100.0 HYDRO 7.408 83.2 1.143 3810.0 159 0.52 22 100.0 HY 33.732 84.0 0.809 16.2 211 0.25 217 100.0 KETO 2.313 46.6 0.443 13.8 183 0.36 22 PTX 10.121 5.1 0.030 75.0 205 0.48 103 74.7 PIRO 4.252 1.6 0.025 0.4 149 0.56 22 100.0 QUER 19.880 28.3 0.418 2.4 172 0.40 187 100.0 THPP 14.737 11.7 0.086 28.7 1179 0.06 406 66.5 VER 3.548 45.7 0.318 28.9 1243 0.23 112 97.6 - 25.9 0.112 1.6 149 NAPRO 97.1 0.41 25 100.0 With [Drug]micelles, the concentration of drug in micelles; Zav the hydrodynamic diameter, dn the number weight diameter and [%]dn the percentage of micelles with a given dn. Chapter II 47 (c) MPEG-dihexPLA micelles Micelle size Drug χsp Solubility Drug content [Drug]micelles Van Krevelen increase Zav [mgdrug/gcopolymer] [mM] [MPa 1/2] factor P.I. dn [%]dn CLZ 1.49 59.16 0.858 9.7 75 0.26 30 99.8 CsA 17.45 320.7 1.100 110.0 86 0.27 68 89.9 DAN 1.54 17.5 0.255 150.0 85 0.45 26 100.0 DICLO 3.13 119.5 1.839 32.8 99 0.59 25 100.0 DTX 11.09 32.7 0.201 29.6 135 0.34 35 99.5 ECZ - 295.1 3.318 1.7 95 0.30 29 100.0 ETO 9.60 67.8 0.609 3.8 183 0.44 23 99.9 FLZ 4.37 268.3 4.381 1341.8 71 0.40 27 98.2 GF 1.71 16.6 0.196 4.4 163 0.37 31 99.9 HYDRO 7.66 78.6 1.067 3556.7 205 0.47 65 94.0 HY 34.29 112 1.122 22.4 101 0.16 17 99.9 KETO 2.48 55.4 0.494 15.4 109 0.26 34 99.7 PTX 10.57 14.1 0.082 205.0 50 0.52 23 98.9 PIRO 4.42 3.4 0.051 0.7 113 0.26 29 99.8 QUER 20.22 12.5 0.182 1.0 205 0.26 110 100.0 THPP 15.19 12.2 0.088 29.3 177 0.17 126 100.0 VER 3.79 85.0 0.591 53.8 193 0.45 29 100.0 NAPRO - 26.9 0.117 1.7 104 0.27 29 97.2 With [Drug]micelles, the concentration of drug in micelles; Zav the hydrodynamic diameter, dn the number weight diameter and [%]dn the percentage of micelles with a given dn. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 48 3.1. Examination of Flory-Huggins parameter and incorporation data It would be desirable to have a fast access to a prediction or estimation of the incorporation of any drug into the polymeric micelle carrier system by simply calculating this from known physical-chemical parameters of the drug. As demonstrated above a solely characterization of the drug lipophilicity and affinity with the hydrophobic micelle core via the log P is not possible. A generally applied method for the determination of the solubility of a compound in a solute is given by the Flory-Huggins theory. Here, the Flory-Huggins parameter χsp of a drug would reflect its solubility and affinity, respectively in the hydrophobic micelle core. It is assumed that first the drug is only solubilized by the core forming block copolymer and second that the core of the micelles can be represented as a bulk polymer. The first condition is a hypothesis since one cannot exclude drug dispersion in the micelle core. The second condition is met for the hexPLA block as e.g. CsA was found to be dissolved in the MPEGdihexPLA micelle core 28 , and e.g. haloperidol could be completely dissolved in bulk hexPLA 29. Based on this the theory of Flory-Huggins ought to be applicable here. χsp was calculated for all drug-copolymer systems using equation 4. The results are presented in Table 3. The closer χsp is to zero, the better is the miscibility between the drug and the copolymer. The 18 investigated drugs showed significant different χsp-affinities with the 3 different coreforming polymers. With a slightly lower δpolymer (δdihexPLA= 17.19 MPa δmonohexPLA=17.33 and δPLA=17.64 MPa 1/2 1/2 ) compared to , respectively, dihexPLA micelles should have a slightly lower affinity/compatibility with those drugs which have a corresponding higher δ values. However, this is not the case. The majority of the tested drugs e.g. CsA, THPP, VER, HY, PIRO, NAPRO, PTX, DTX, VER, DAN are better incorporated in MPEG-dihexPLA micelles rather than in MPEG-PLA micelles. Only KETO and CLZ were found to be better incorporated in MPEG-PLA micelles. Hy and CsA with a high χsp of 34.29 and 17.45 MPa1/2 with MPEG-dihexPLA, respectively were expected to be less incorporated than drugs with lower χsp. However, both drugs were among the 5 best incorporated drugs for MPEGdihexPLA micelles (Table 3c), whereas THPP with a χsp of 15.19 yielded one of the lowest drug contents with only 12.2 mg/g. Hence, no clear relationship between χsp and the drug content in MPEG-(hex)PLA micelles was found. Thus the Flory-Huggins theory does not seem to be applicable to predict drug incorporation into MPEG-hexPLA based polymeric micelles. As mentioned above so far only non-optimized polymers towards the drugs were used for this study. Letchford et al. reported for MPEG-PCL copolymers with varying MPEG and PCL Chapter II 49 block lengths after incorporation of 5 drugs with different χsp 11;18 that (1) drugs with low χsp were solubilised in higher extent than those with high χsp, indicating a better affinity and thus a better incorporation, and (2) that copolymers with higher molecular weights of the PCL block (higher block length) yielded higher drug incorporations. These findings envision the possibility of improving drug incorporation into MPEG-hexPLA micelles by adjusting the molecular weight of the core forming block. 3.2. Differentiated views on the drug water solubility and log P and experimental incorporation results As an alternative approach the here tested 18 different drugs were classified related to the Biopharmaceutics Classification System (BCS) modified by the addition of high/low log P and high/low water solubility as parameters (Figure 2). The set limit for high and low log P was inspired here by the Lipinski’s “rule of 5” 2;14 . Concerning the solubility in water, the limit was set for an intermediate value of 0.055 mM. The results of the drug incorporation into MPEG-dihexPLA micelles (drug content and “solubility increase factor”) are summarized in Figure 2. Water solubility >0.055 mM <0.055 mM Low log P (log P<5) Drug QUER PIRO NAPRO GF ETO DICLO KETO DTX VER DAN PTX FLZ HYDRO High log P (log P>5) Drug Solubility Drug Solubility content increase Drug content increase [mg/g] [× times] [mg/g] [× times] 12.5 (×1) 3.4 (×1) 26.9 (×1.7) CLZ 59.2 (×10) 18.7 (×3) 68.0 (×4) 119.5 (×33) 55.4 (×15) 32.7 (×30) 85.0 (×54) (×22) HY 112.0 17.5 (×150) CsA 321.0 (×110) 14.1 (×205) 268.0 (×1342) 78.6 (×3559) Figure 2. Classification of the tested drugs by high/low log P and high/low water solubility. (Log P of ECZ and THPP are not available) Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 50 As the drug content differed from 4 to 295 mg/g for drugs with a considered high water solubility and from 12 to 321 mg/g for those with a considered low water solubility, no tendency for the drug incorporation could be found. However, a trend for the “solubility increase factor” was observed. For drugs with a considered low water solubility, drug aqueous solubility (solubilised by micelles) could be increased from 15 to 3559 times, when incorporated into MPEG-dihexPLA micelles. For drugs with a considered high water solubility, the increase solubility factor varied only from 1 to 10 (Figure 3). These results confirm the findings of Letchford et al., who also reported for the drug with the low aqueous solubility the highest solubility increase 18. 0.055 mM Solubility increase factor (F) 4000 3000 2000 1000 500 300 100 60 50 40 30 20 10 0 0.00 F>10 F<10 0.01 0.1 0.2 1.5 Drug water solubility [mM] 2.5 Figure 3. Influence of drug water solubility of the 18 tested drugs on the “solubility increase factor”. For drugs with a similar log P (2.4 <log P< 3.8) in the group of “low solubility and low log P”, the drug content increased with the water solubility of 4 selected drugs (VER, DTX, DAN, and PTX) for the 3 investigated MPEG-(hex)PLA micelle carrier systems (Figure 4). Chapter II 51 VER 90 Drug content [mg/g] 80 70 60 50 DTX 40 30 DAN 20 PTX 10 0 0 0.002 0.004 0.006 0.008 0.01 0.012 S water [mM] Figure 4. Influence of the drug water solubility (Swater) in drug content obtained in MPEG-PLA (), MPEH-monohexPLA () and MPEG-dihexPLA () micelles for drugs with log P between 2.4 and 3.8: PTX (paclitaxel), DAN (danazol), DTX (docetaxel), and VER (verteporfin). The more hydrophobic MPEG-dihexPLA micelles incorporate considerably higher quantities of those poorly water soluble drugs in comparison to MPEG-monohexPLA and MPEG-PLA micelles. This superior incorporation of poorly water soluble drugs into MPEG-hexPLA micelles compared to MPEG-PLA micelles was also observed for the majority of the tested drugs (Figure 5). 350 CsA ECZ Drug content (mg/g) 300 QUER FLZ 250 200 DICLO 150 CLZ VER 100 HYDRO 50 KETO HY DTX GF DAN THPP PTX 0 0.000 0.005 0.010 0.03 NAPRO ETO PIRO 0.05 0.07 0.09 0.1 1.1 Swater (mM) 2.1 Figure 5. Drug contents of all tested drugs in MPEG-PLA (○) MPEG-monohexPLA () and MPEGdihexPLA () micelles in function of the water solubility (Swater) of the investigated drugs. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 52 Looking closer at two drugs with similar χsp and chemical structures, as it is the case for PTX and DTX, significant different incorporation results were obtained. A drug content of 14.12 mgPTX/g and 32.7 mgDTX/g was found for MPEG-dihexPLA micelles. However, this corresponds to a “solubility increase factor” of 205 for PTX and only 30 for DTX. The structure of both drugs differs only in two chemical groups, at the same C positions. PTX has a benzamid goup at the secondary amine, and a methoxy group, whereas DTX bears a tertbutoxy group at the secondary amine, and has a free hydroxyl group (Figure 1). PTX has a slightly higher log P (log P=3) compared to DTX (log P=2.4). Interestingly here PTX with its higher log P yielded a lower drug content, but the higher “solubility increase factor”. This high increase of aqueous solubility of PTX in micelles might be due to its very low water solubility value of 0.0004 mM, which is 17 times lower than that of DTX (0.0068 mM). As another example one can compare the photosensitizer drugs THPP and VER which also have similar chemical structures. In this case THPP with the lower water solubility of 0.003 mM has a “solubility increase factor” of 29 whereas VER with its higher water solubility of 0.011 mM leads to a “solubility increase factor” of 54. These results show clearly the importance of the drug water solubility as such on the “solubility increase factor”. The, to some extent, contradictory results highlight that the incorporation efficiency under same preparation conditions can not be predicted by a simplistic look at only one physicalchemical parameter of the drug, especially log P, which is often used in the literature as a major value to describe the hydrophobic character of a drug and does not reflect consistently the obtained experimental data. 3.3. Examination of other physical-chemical parameters Since the incorporation data obtained for the 18 drugs with MPEG-hexPLA could not be explained via the log P or the water solubility parameter only, further parameters like the molecular weight (Mw) of the drug, the number of hydrogen bond donors (Hd) and hydrogen bond acceptors (Ha) in the drug structure were taken into consideration, as well as the surface tension of the drug, which was calculated with the software ACD/ChemSketch software (Advanced Chemistry Development, Inc., Canada). Because simultaneous interpretation with 6 chemical-physical parameters (Mw, Hd, Ha, drug water solubility, log P, and surface tension) becomes difficult to handle manually, the incorporation results (the drug content, the solubility in micelles, and the “solubility increase factor”) were analyzed by a linear multiple Chapter II 53 regression in order to find the most influencing parameters. The corresponding regression coefficients of each chemical-physical parameter were obtained by using the Data Analysis tool of Microsoft Office Excel 2003. The higher is the absolute value of the coefficient, the higher is the influence of the parameter on the specific response. The classification of the influence of the different tested parameters regarding the obtained regression coefficients is given in brackets in Table 4. Please note that these analyses were done only with those drugs for which all the defined parameters were available. Table 4. Classification of the influence of the different chemical physical parameters of the tested drugs on the incorporation responses in MPEG-dihexPLA micelles after a linear multiple regression. In brackets the ranking of importance is given, whereby 1 is high and 6 is low importance. Responses “Solubility increase factor” -6190.6 (1) Drug water solubility [mM] Solubility in micelles [mM] -6.06 (1) Hd 0.56 (2) 668.2 (2) 65.3 (1) Ha 0.34 (3) 39.8 (4) 30.2 (3) Log P 0.19 (4) -49.0 (3) 34.4 (2) Surface tension [dyne/cm] 0.02 (5) -31.4 (5) 3.4 (5) Mw [g/mol] 0.01 (6) -5.4 (6) 0.8 (6) Chemical – physical parameters Drug content [mg/g] 29.5 (4) The results reveal that the drug content is mainly influenced by Hd, log P, Ha and the drug water solubility, Mw and the surface tension have less influence. Similarly these parameters affect the drug solubility in micelles. Here the drug’s water solubility appeared to be the most influencing parameter, followed by Hd and Ha, and then log P. Like for drug content, Mw and the surface tension of the drug had only a slight influence. The parameter ranking of “the solubility increase factor” response is similar to the one of the solubility in micelles, confirming the observation that for a drug with lower water solubility (< 0.055 mM) a much higher increase of its aqueous solubility can be achieved when incorporated in MPEGhexPLA micelles. The influence of Ha and Hd on the incorporation results in MPEG-hexPLA micelles may explain the difference found and discussed above for the similar drugs PTX and DTX, wherein the latter DTX has 1 more Hd group than PTX. The obtained results show in Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 54 general that Hd is the more influencing parameter for the drug content, and indeed for DTX a higher drug content in MPEG-hexPLA micelles was obtained. Thus the presence of chemical groups with Ha and Hd functions also influences the drug incorporation in MPEG-hexPLA micelles. Indeed, the drug/copolymer affinity was found to be induced by the formation of Hbonds between the multiple H-bond site of the PCL copolymer and single H-bond site of two cucurbitacin drugs 30 . Drugs with multiple H bond donors and acceptors have favourable χsp values and are better solubilised 31 . In consequence, the prediction or estimation of drug incorporation in MPEG-hexPLA micelles needs also to take structural parameters into account. It becomes clear that a quantitatively relationship between a drug via its physicalchemical parameters and the experimentally obtained incorporation results is difficult to draw. From a practical point of view towards suitable pharmaceutical formulations, we applied the criteria of Lipinski’s “rule of 5” for of the here tested drugs (Figure 6). “Rule of 5” PIRO NAPRO Mw<500 g/mol QUER KETO Ha<10 Hd <5 Log P<5 GF PTX DICLO VER ETO ECZ DTX CsA DAN THPP HY HYDRO FLU CLZ Inverse “rule of 5” Mw>500 g/mol Ha>10 Hd >5 Log P>5 Figure 6. The Lipinski’s “rule of 5” applied on the investigated drugs As shown half of the drugs do not fit the rules (Mw<500 g/mol; Ha<10; Hd<5 and log P<5), and have in majority a considered low solubility, which would limit their success in clinics. Amongst those drugs, CsA is a drug that in theory would have only little chances of bioavailability by all the Lipinski’s criteria. However, as demonstrated here CsA would be a preferable drug for incorporation in MPEG-dihexPLA micelles with its high number of H bond donor and acceptor groups, its high log P (8.2) and its very low water solubility (0.01 mM). Indeed, the successful CsA incorporation into MPEG-hexPLA micelles was presented previously 21. Chapter II 55 4. Conclusions Eighteen different poorly water soluble drugs, chosen to cover a wide range of lipophilicity and of water solubility, were incorporated into MPEG-hexPLA micelles. The calculation of the Flory-Huggins parameter χsp does not confirm the general better affinity of the hydrophobic drugs with the most hydrophobic micelle-core forming block (dihexPLA). The value of log P did not consistently correlate the experimental incorporation data either. Therefore the influence of other drug physical-chemical parameters on the incorporation results in micelles was studied. The molecular weight and the surface tension of the drug show only minor contribution on the drug incorporation data, whereas the log P, the drug water solubility and the number of hydrogen bond donor Hd– groups and hydrogen acceptor Ha- groups were found to be major influencing parameters. Drugs with a low water solubility (< 0.055 mM) showed a high increase of their aqueous solubility when incorporated into MPEG-dihexPLA micelles. Drugs with a high number of Hd yielded higher drug contents in MPEG-hexPLA micelles. Therefore the prediction or estimation of drug incorporation into MPEG-hexPLA micelles depends also on the structural characteristics of the drugs. In order to take the maximum of factors into consideration, physical-chemical and structural, molecular dynamics (MD) simulation of the presented results of MPEG-hexPLA incorporation are currently under investigation. It has to be pointed out that drugs which do not fit the Lipinski’s “rule of 5” indicating a low oral bioavailability, can efficiently be incorporated and formulated in MPEG-hexPLA micelles. Thus MPEG-hexPLA micelles can be envisioned as alternative pharmaceutical formulations for poorly water soluble drugs. Acknowledgments The authors thank the Swiss National Science Foundation (SNF) for financial support (SNF 200020-103752). The authors thank also Aliya Kasimova (University of Geneva, Geneva, Switzerland) for the determination of the solubility parameters with the Material Studio software. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 56 References (1) Cheng, W. P.; Thompson, No.2010/0,029,544. C., Composition, U.S.Patent 2010, Application (2) Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J., Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings, Adv.Drug Delivery Rev. 1997, 23, 3-25. (3) Bittner, B.; Mountfield, R. J., Intravenous administration of poorly soluble new drug entities in early drug discovery: The potential impact of formulation on pharmacokinetic parameters, Curr.Opin.Drug Discov.Devel. 2002, 5, 59-71. (4) Yalkowsky S. H.; Roseman, T. J. Techniques of Solubilization of Drugs.; Dekker, New York ed.; 1981; Chapter 3. (5) Johnson, J. L. H.; He, Y.; Yalkowsky, S. H., Prediction of precipitation-induced phlebitis: A statistical validation of an in vitro model, J.Pharm.Sci. 2003, 92, 15741581. (6) Yalkowsky, S. H.; Krzyaniak, J. F.; Ward, G. H., Formulation-related problems associated with intravenous drug delivery, J.Pharm.Sci. 1998, 87, 787-796. (7) Uchenna Agu, R.; Jorissen, M.; Willems, T.; Van den Mooter, G.; Kinget, R.; Verbeke, N.; Augustijns, P., Safety assessment of selected cyclodextrins -- effect on ciliary activity using a human cell suspension culture model exhibiting in vitro ciliogenesis, Int.J.Pharm. 2000, 193, 219-226. (8) Thiel, G., Nephrotoxicity of ciclosporin, Trends Pharmacol.Sci. 1986, 7, 169. (9) Tibell, A.; Norrlind, B., Cyclosporin A in fat emulsion carriers: studies on the immunosuppressive potential, using the heterotopic heart transplant model in rats, Transplant Int. 1994, 7, 438-441. (10) Jones, M. C.; Leroux, J. C., Polymeric micelles - a new generation of colloidal drug carriers, Eur.J.Pharm.Biopharm. 1999, 48, 101-111. (11) Gref, R.; Lück, M.; Quellec, P.; Marchand, M.; Dellacherie, E.; Harnisch, S.; Blunk, T.; Müller, R. H., Stealth corona-core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption, Colloid.Surface B 2000, 18, 301-313. (12) Park, J. H.; Lee, S.; Kim, J. H.; Park, K.; Kim, K.; Kwon, I. C., Polymeric nanomedicine for cancer therapy, Prog.Polym.Sci. 2008, 33, 113-137. (13) Kohane, D. S., Microparticles and nanoparticles for drug delivery, Biotechnol.Bioeng. 2007, 96, 203-209. Chapter II 57 (14) Singh, R.; Lillard, J., Nanoparticle-based targeted drug delivery, Exp.Molec.Pathol. 2009, 86, 215-223. (15) Croy, S. R.; Kwon, G. S., Polymeric Micelles for Drug Delivery, Curr.Pharm.Design 2006, 12, 4669-4684. (16) Nagarajan, R.; Barry, M.; Ruckenstein, E., Unusual selectivity in solubilization by block copolymer micelles, Langmuir 1986, 2, 210-215. (17) Liu, J. B.; Xiao, Y. H.; Allen, C., Polymer-drug compatibility: A guide to the development of delivery systems for the anticancer agent, ellipticine, J.Pharm.Sci. 2004, 93, 132-143. (18) Letchford, K.; Liggins, R.; Burt, H., Solubilization of hydrophobic drugs by methoxy poly(ethylene glycol)-block-polycaprolactone diblock copolymer micelles: Theoretical and experimental data and correlations., J.Pharm.Sci. 2008, 97, 1179-1190. (19) Gaucher, G.; Dufresne, M. H.; Sant, V. P.; Kang, N.; Maysinger, D.; Leroux, J. C., Block copolymer micelles: preparation, characterization and application in drug delivery, J.Control.Release 2005, 109, 169-188. (20) Allen, C.; Maysinger, D.; Eisenberg, A., Nano-engineering block copolymer aggregates for drug delivery, Colloid.Surface B 1999, 16, 3-27. (21) Mondon, K.; Zeisser-Labouèbe, M.; Gurny, R.; Möller, M., Novel Cyclosporin A formulations using MPEG-hexyl substituted polylactide micelles: a suitability study, Eur.J.Pharm.Biopharm. 2011, 77,56-65. (22) Barton, A. F. M. Handbook of solubility parameters and other cohesion parameters,CRC Press, 2nd edition; 1991. (23) Trimaille, T.; Gurny, R.; Möller, M., Synthesis and ring-opening polymerization of new monoalkyl-substituted lactides, J.Polym.Sci.Part A: Polym.Chem. 2004, 42, 4379-4391. (24) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. (25) Van Krevelen, D. W. Properties of Polymers- their correlations with chemical structure, their numerical estimation and prediction from additive group contribution, 3rd edition; 1997. (26) Fedors, R. F., A method for estimating both the solubility parameters and molar volumes of liquids, Polym.Eng.Sci. 1974, 14, 147-154. (27) Nottelet, B.; Di Tommaso, C.; Mondon, K.; Gurny, R.; Möller, M., Fully biodegradable polymeric micelles based on hydrophobic- and hydrophilic-functionalized poly(lactide) block copolymers, J.Polym.Sci.Part A: Polym.Chem. 2010, 48, 3244-3254. (28) Di Tommaso, C.; Como, C.; Gurny, R.; M÷ller, M., Investigations on the lyophilisation of MPEG-hexPLA micelle based pharmaceutical formulations, Eur. J. Pharm. Sci. 2010, 40, 38-47. Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 58 (29) Asmus, L. R.; Gurny, R.; Möller, M., Solutions for Lipophilic Drugs - A biodegradable polymer acting as solvent, matrix and carrier to "solve" drug delivery issues, Int. J. Artificial. Organs 2010, Submitted . (30) Patel, S. K.; Lavasanifar, A.; Choi, P., Roles of Nonpolar and Polar Intermolecular Interactions in the Improvement of the Drug Loading Capacity of PEO-b-PCL with Increasing PCL Content for Two Hydrophobic Cucurbitacin Drugs, Biomacromolecules 2009, 10, 2584-2591. (31) Patel, S. K.; Lavasanifar, A.; Choi, P., Molecular dynamics study of the encapsulation capability of a PCL-PEO based block copolymer for hydrophobic drugs with different spatial distributions of hydrogen bond donors and acceptors, Biomaterials 2010, 31, 1780-1786. Supplementary material HPLC conditions developed for the determination of the drug content of each tested drug Drug Type of Column T [°] Mobile Phase Flow rate (mL/min) UV detection Calibration Standards (µg/mL) CLZ C18 a 35°C MeOH:0.05 M K2HPO4 (75:25) 1.5 214 nm 5.5 – 220 CsA C18 b 65°C ACN:Water (75:25) 1.2 210 nm 0.15 - 200 DAN C18 b 25°C ACN:Water (70:30) 1.0 283 nm 2.1 – 213 DICLO C18 b 30°C ACN: 20mM K2HPO4 (40:60) 1.0 275 nm 2.0 – 200 (pH = 3.1 adjusted with PA) (pH = 3.5 adjusted with PA) DTX C18 b 25°C ACN:Water (50:50) 1.0 227 nm 1.6 – 307 ECZ C18 a 35°C ACN:0.05 M K2HPO4 (60:40) 1.5 214 nm 2.0 – 200 ETO C18 b 25°C ACN:Water:AA (34:65:1) 1.0 230 nm 10.0 – 200 FLZ C18 a 350C MeOH:0.1 M NH4H2PO4 (50:50) 1.5 214 nm 5.0 – 200 Chapter II GF C18 a 25°C ACN:20 mM KH2PO4 (45:55) 59 1.0 293 nm 2.0 – 200 (pH = 3.0 adjusted with PPA) HY C-8 c 25°C MeOH:Water (90:10) + 1% FA 1.0 590 nm 2.0 – 200 HYDRO C18 a 35°C ACN:0.03% PA 1.0 243 nm 2.0 – 200 KETO C18 a 25°C MeOH:Water:DEA (74:26:0.1) 1.0 240 nm 2.1 – 207 PIRO C18 a 30°C ACN: 20mM K2HPO4 (40:60) (pH = 3.5 adjusted with PA) 1.0 355 nm 1.9 – 190 PTX C18 a 25°C ACN:Water(50:50) 1.0 227 nm 1.0– 200 QUER C18 a 25°C ACN:1% AA (45:55) 1.0 230 nm 2.0– 200 NAPRO C18 a 30°C ACN: 20mM K2HPO4 (40:60) 1.0 331 nm 2.0– 200 (pH = 3.5 adjusted with PA) AA, acetic acid; DEA, diethylamine; FA, formic acid; K2HPO4, potassium biphosphate buffer; NH4H2PO4, ammonium phosphate monobasic; KH2PO4, potassium dihydrogen phosphate; PA, phosphoric acid; PPA, pyrophosphoric acid. a Lichrospher® RP-18 column (124 mm × 4 mm) b Nucleosil 100-5 C18 column (250 mm × 4 mm) c Zorbax- Eclips XDB-C8 column (150 mm× 4.6 mm) Incorporation of Poorly Water Soluble Drugs into MPEG-hexPLA Micelles 60 1234562678693A738B63456CADE23FB4865234DD5386532DE6395E23A8B33BAB37E653D33 D793 FF648D53 793 9D7D53 5DE423 F73 63 C653 6AFA6783 D5E8693 A8B3 B6 13 AF6623 793 A7F562A73 2E2878A3 8B6A53 8653 2DEAA8!3 "73 8B63 DDA73 3 D963 1 D5E8AD73D3FFD24D5A7313238628693D53A823676534B5F6E8AF32EA8AA8345D4658A62!3 1 1 1 1 123456789998 8 1 Chapter III 61 Novel Cyclosporin A Formulations Using MPEG-Hexyl Substituted Polylactide Micelles: A Suitability Study K. Mondon, M. Zeisser-Labouèbe, R. Gurny, and M. Möller School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland Submitted to: European Journal of Pharmaceutics and Biopharmaceutics The immunosuppressive agent Cyclosporin A (CsA) has very poor solubility in water and, in consequence, non-aqueous formulations have been developed for its intravenous administration to treat patients with transplant rejection. In this article, aqueous micelle solutions of novel amphiphilic copolymers based on methoxy-poly(ethylene glycol) (MPEG) and hexyl-substituted poly(lactides) (hexPLA) were studied for possible incorporation and formulation of CsA, and for their biocompatibility towards novel pharmaceutical applications. Above the critical micellar concentration (CMC), MPEG-hexPLA block-copolymers selfassemble into unimodal micelles with diameters of around 30 nm, either unloaded or drugloaded. The best shelf-life stability of these formulations was observed when stored at 4°C with a drug loss inferior to 7% after 1 year. The polymer and micelle toxicities were evaluated in vitro for three different cell lines and in vivo using the chick embryo chorioallantoic membrane (CAM) model. The hemolytic property was assessed using human blood samples. As the studies revealed, MPEG-hexPLAs are non-toxic and do not show hemolysis; the same was found for the comparable MPEG-PLAs, both as unimers below their CMC and as polymeric micelles up to copolymer concentrations of 20mg/mL. At this concentration, CsA was efficiently incorporated into MPEG-hexPLA micelles up to 6 mg/mL, which corresponds to a 500-fold increase of its water solubility. The current recommended clinical concentration administered per infusion (0.5-2.5 mg/mL) can be easily achieved and requires four times less copolymer than with the often-used Cremophor ®EL surfactant. In this regard, MPEGhexPLA micelle formulations can be an applicable formulation in transplant rejection treatments as an injectable CsA carrier system. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 62 Keywords: biocompatibility; CAM model; Cyclosporin A; drug delivery; polymeric micelles; substituted polylactides. Chapter III 63 1. Introduction To deliver drugs to the right pathological site, at the right time, in the right dose, and without altering surrounding tissues is one of the important aims of drug delivery systems. For this purpose, nanocarriers, such as nanoparticles 1;2 , liposomes 3 and micelles 4-6 , have been studied in pharmaceutical research. Due to their small size of less than 1 μm, nanosized vehicles can improve the therapeutic and pharmacological properties of drugs by escaping the typical response of the immune system and therefore reaching and accumulating into pathological sites, such as tumors, due to the enhanced permeability and retention effect (EPR) 7. For other pathological sites, the nanosize of micelles allows for better internalization in cells and increases the therapeutic effect of the drug compared to larger carriers 8-10. Polymeric micelles of amphiphilic copolymers, which are composed of hydrophobic and hydrophilic blocks, are characterized by self-assembled core-shell structures in aqueous media. For pharmaceutical use, polymeric micelles with such a structure are of interest due to their ability to incorporate and protect hydrophobic drugs within their hydrophobic core, while the hydrophilic corona stabilizes and solubilizes the micelles in the aqueous environment. Usually the hydrophilic block is formed by the water-soluble, biocompatible and non-immunogenic poly(ethylene glycol) (PEG), which is known for its ability to reduce opsonization and recognition by the mononuclear phagocytic system (MPS). This consequently provides the long circulation properties of, for example, pegylated nanoparticles 11. The hydrophobic block can be composed of various polymers, which must be non-toxic and biocompatible for pharmaceutical applications, i.e., polyethers acids 13 , or polyesters 14 12 , polyamino . Amongst the hydrolysable polyesters, polycaprolactone (PCL) and polylactides (PLA) are the polymers of highest interest due to their known biodegradability, biocompatibility and their FDA status of “Generally Regarded as Safe” (GRAS). Due to its faster degradation time, PLA is often preferred to PCL in medical applications. However, PLA has limited applicability and performance as a carrier of hydrophobic drugs; the ability of micelles to solubilize drugs within their core depends on the compatibility of the core forming block and the drug itself 15-17 . Therefore, novel hydrophobic and biocompatible polymers based on the biodegradable PLA backbone were developed by T. Trimaille et al., who synthesized different alkyl substituted polylactides by ring opening polymerization (ROP) 18. Due to their interesting physico-chemical properties, viscous and injectable hexyl- CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study substituted polylactides (hexPLA) were investigated for pharmaceutical applications 64 19 . HexPLA polymers of the two different structures, that is to say the mono- and di- hexylsubstituted polylactides (replacement of every second methyl group and all methyl groups by hexyl groups, respectively along the polymer backbone), are named here as monohexPLA and dihexPLA, respectively (Scheme 1). The presence of hexyl groups results in the highest hydrophobicity for dihexPLA, followed by monohexPLA, and the smallest for PLA. When hexPLAs are copolymerized with methoxy-poly(ethylene glycol) (MPEG), amphiphilic copolymers that self-assemble in aqueous media into polymeric micelles are formed and they incorporate hydrophobic drugs like griseofulvin hydroxyphenyl)porphine (THPP) 21 20 or the photosensitizer meso-tetra(p- much more efficiently than comparable “standard” MPEG-PLA micelles. In this work, the very hydrophobic drug Cyclosporin A (CsA), which has a log P of 8.2, was formulated for the first time with MPEG-hexPLA micelles. We also investigated their biocompatibilities to determine the feasibility of these novel drug nanocarriers as conceivable pharmaceutical formulations. The neutral, cyclic, undecapeptide CsA is a potent immunosuppressive agent used to reduce graft rejection after organ transplantation 22;23 . With its rigid structure and its poor water solubility (0.012mg/mL, determined experimentally at 25°C), CsA must be solubilized in non-aqueous media with surfactants like CremophorEL, as is done for the Sandimmune formulation (Novartis Pharma). Here, MPEG-hexPLA micelles can facilitate an aqueous formulation for the solubilization and delivery of CsA for intravenous applications. Due to this newly-developed drug carrier system, the biocompatibility issues of the MPEG-hexPLA micelles needed to be addressed. Therefore, we first characterized both unloaded and CsA-loaded micelle formulations and studied their drug incorporations and shelf-life stabilities. Secondly, the biocompatibilities of the unloaded micelles and their unimers (possible compounds from disassembled micelles below their CMC that can occur under the dilution conditions in intravenous applications) were studied. These studies included (a) the in vitro toxicity on three different cell-lines, which differed in their mammalian origins (murine or human) and their biological origins (intestinal or cancerous), (b) the in vivo toxicity using the chick embryo chorioallantoic membrane (CAM) model, and (c) the hemolytic property using human blood. The obtained favorable and encouraging results are presented here. Chapter III 65 2. Materials and methods 2.1 Materials Triton® X-100 was purchased from AppliChem (Gatersleben, Germany). Tetrahydrofuran (THF) was supplied by SDS (Toulouse, France), and distilled over sodium to make it anhydrous when needed. Methoxy-poly(ethylene glycol) with a molecular weight of 2000 g/mol (MPEG2000g/mol) was a gift from BASF (Ludwigshafen, Germany). D,L-lactide, tin(II) 2-ethylhexanoate (Sn(Oct)2), and acetone p.a. were purchased from Purac Biochem (Gorinchem, The Netherlands), Aldrich (Buchs, Switzerland), and Fluka (Buchs, Switzerland), respectively and used as received. The monomers, mono-hexyl-substituted lactide (monohexLA) and di-hexyl-substituted lactide (dihexLA) were synthesized as described previously 18. CremophorEL was supplied by BASF and Sandimmune (Novartis Pharma, Bern, CH) was purchased from a local pharmacy. Cyclosporine A was supplied by Fluka (Buchs, Switzerland). 2.2 Synthesis and characterization of MPEG-(hex)PLA copolymers Please note that in the following text, “MPEG-(hex)PLA” refers to both MPEG-PLA and MPEG-hexPLA polymers. MPEG-hexPLA and MPEG-PLA copolymers of 5000 g/mol were synthesized by ring opening polymerization (ROP). Briefly, 3.0 g of the corresponding lactide monomer were polymerized in bulk at 100°C with the required amount of the initiator MPEG2000g/mol to reach the targeted molecular weight. The catalyst Sn(Oct)2 was added in an equimolar ratio to the initiator. The copolymerization reactions were stopped after 1.5 h by adding 5 mL of THF (1% water) into the reaction mixture. After the removal of THF by evaporation the resulting copolymers were precipitated dropwise into 120 mL of cold methanol, then filtrated and dried under vacuum. Finally they were analyzed by 1H NMR (Brüker, 300 MHz) to detect any residual organic solvent traces. The copolymers were characterized to determine their molecular weight (Mn) and polydispersity index (P.I.) using gel permeation chromatography (GPC). The GPC setup was composed of a Waters system with Waters Styragel HR1-3 columns and a Waters 410 differential refractometer (Waters, Milford, USA). The analysis was carried out using CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 66 polystyrene (PS) of different molecular weights as calibration standards (PSS, Mainz, Germany). 2.3 Preparation and size characterization of unloaded MPEG-(hex)PLA micelles A MPEG-(hex)PLA copolymer solution was dissolved at 100 mg/mL in acetone (2 mL) and was added dropwise under sonication into 4 mL of isotonic saline solution. The organic solvent was slowly removed by evaporation at 15 mbar. Final micelle concentrations were adjusted to 50 mgcopolymer/mL by adding isotonic saline solution. Afterwards, MPEG(hex)PLA micelles were analyzed for their size. The hydrodynamic- (Zav) and numberweighted (dn) diameters were measured using dynamic light scattering (DLS) with a Zetasizer HS 3000 (Malvern, Worcestershire, UK). The analyzes were performed at an angle of 90° at 25°C. For each sample, the mean diameters were obtained after three runs of ten measurements. 2.4 Incorporation of CsA into MPEG-hexPLA micelles The preparation and incorporation of CsA into MPEG-(hex)PLA micelles followed the similar co-solvent evaporation method as described for unloaded micelles. Here, CsA and 20 mg MPEG-(hex)PLA were dissolved in acetone (2 mL), and the final micelle concentrations were adjusted to 5 mgcopolymer/mL by adding ultra-pure water or isotonic saline solution. When needed, the isotonicity of micelles solution prepared in water was obtained by addition of the corresponding amounts of sodium chloride. The solutions were left to equilibrate overnight and then centrifuged at 9500 x g for 15 min to remove non-incorporated, non-soluble CsA. For drug-loading determination, the supernatant was diluted in acetonitrile at a ratio 1:10 to break up the micelles and to release the CsA for quantitative analysis. For more highly-concentrated CsA loadings in MPEG-dihexPLA micelles, the copolymer concentration was increased up to 20 mg/mL with a targeted drug loading set at 400 mgCsA/gcopolymer. To ensure complete micelle destruction before CsA quantification, a 1:100 dilution in acetonitrile was made. The CsA concentration was quantified by HPLC using a C-18 column (250 mm × 4.6 mm) heated at 65°C. The flow rate was 1.2 mL/min and the mobile phase was a mixture of acetonitrile/water (75:25) with a pH of 3.1 after the addition of phosphoric acid. Samples of Chapter III 67 20 µL were injected with a running time fixed at 6 min. The CsA peak was detected by UV at λ = 210 nm and appeared at 4.1 min. The CsA calibration standards from 0.15 to 200 µg/mL were prepared and the resulting calibration curves were obtained with a regression coefficient superior to 0.99. All samples were measured in triplicates. 2.5 Preparation of CremophorEL micelles CremophorEL micelles were prepared by adding 400 mg of the surfactant into 4 mL isotonic saline solution with stirring to reach a final concentration of 100 mg/mL. They have been characterized by size as previously mentioned for unloaded MPEG-(hex)PLA micelles. 2.6 Cell culture A Caco-2 cell line, a human epithelial colorectal adenocarcinoma cell line was maintained in Dulbecco's Modified Eagle's medium (DMEM) (Gibco Life Technologies, Carlsbad, USA) supplemented with 10% fetal bovine serum (FBS) (Brunschwig, Amsterdam, The Netherlands), 1% of non-essential amino acids and 1% of penicillin and streptomycin 24. The cells were grown for 2 weeks to allow for monolayer formation and cell differentiation 25. A NuTu-19 cell line, a poorly differentiated Fischer 344 rat-derivative epithelial ovarian cancer cell line 26, was kindly provided by Dr. A Major (Geneva University Hospital, Geneva, Switzerland). The cells were cultured in DMEM culture medium supplemented with 10% FBS and 100 U/mL penicillin–streptomycin (Gibco Life Technologies). A SKOV-3 (HTB-77) cell line, a human ovarian carcinoma cell line, was purchased from American Tissue Culture Collection (ATCC, Manassas, USA). The cells were grown in Roswell Park Memorial Institute medium (RPMI 1640) (Gibco Life Technologies) and supplemented with 10% FBS, 1% l-glutamine, and 1% penicillin/streptomycin. All cell lines were cultured at 37°C in a humidified atmosphere containing 5% CO2. 2.7 In vitro toxicity The in vitro toxicity of unloaded micelles was determined by standard MTT tests on Caco-2, NuTu-19, and SKOV-3 cells. Briefly, the cells were seeded in a 96-well plate at a density of 1.5×104 cells per well in 100 μL of culture medium and incubated in a humidified atmosphere with 5% CO2 at 37°C for 24 h. After removal of the culture media and washing with PBS, the CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 68 cells were incubated with 100 μL MPEG-(hex)PLA and CremophorEL micelle solutions or isotonic saline solution. After 24 h incubation, 50 μL of a 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide (MTT) solution of 1 mg/mL in PBS were added to each well and the plate was incubated for 3 h to allow the soluble yellow MTT to be reduced into the dark-blue, insoluble formazan crystals by the metabolically active cells. Afterwards, the formazan crystals were dissolved by addition of 200 μL dimethyl sulfoxide (DMSO) in the incubator at 37°C for 1 h. The UV absorbance of individual wells was measured at 595 nm with a microplate reader (Model 550, Bio-RAD, Hercules, USA). Formulation concentrations below the CMC (0.01mgcopolymer/mL) were studied to determine the toxicity of MPEG-(hex)PLA as unimers, whereas concentrations above the CMC yielded the toxicity of MPEG-(hex)PLA as micellar structures. Different copolymer concentrations were obtained by dilution in culture media from the most concentrated MPEG-(hex)PLA formulation (50 mgcopolymer/mL). The same procedure was carried out for CremophorEL solutions, which have a CMC of 0.9 mg/mL. The culture media and a 0.5% TritonX-100 solution in 1 N NaOH were used as positive (100% survival) and negative (0% survival) controls, respectively. The cell viability was determined by the following formula: Cell viability (%) = A sample − A 0% A100% − A 0% × 100 where Asample, A0% and A100% are the respective absorbances of the sample, the negative and positive control. 2.8 In vivo toxicity by the CAM model The in vivo toxicity of the MPEG-(hex)PLA micelles was assessed by using the chick embryo chorioallantoic membrane (CAM) model adapted from Lange et al. 27. The egg incubation procedure followed the one described by Vargas et al. 28 . Briefly, fertilized hen eggs, kindly provided by the Animalerie Universitaire of the University of Geneva (Geneva, Switzerland) were placed with the narrow apex down into an incubator Savimat MG 200 at 37°C with a relative humidity of 65%. The eggs were rotated twice a day until the embryo development day 3 (EDD3). On EDD4, a 3-mm hole was drilled into the eggshell and the narrow apex was covered by an adhesive tape. The eggs were incubated in a static mode until EDD12. After the removal of the tape, a bigger hole of 2-3 cm was drilled, thus allowing the visualization of the CAM vasculature. Portions of 20 µL of unloaded MPEG-(hex)PLA formulations were Chapter III 69 injected via the main vessel. The eggs were returned to the incubator and the survival rate was evaluated at 24 h post-injection. The unloaded MPEG-(hex)PLA micelles at 0.0001, 5, 10, and 50 mgcopolymer/mL from five chick egg embryos were investigated. The highest concentrated micelle solution of 50 mgcopolymer/mL was used as prepared and the other three were obtained by dilution with isotonic saline solution. The isotonic saline solution itself was used as the 100% survival control. 2.9 Hemolysis test Blood samples were freshly collected from healthy human volunteers in acid citrate dextrosecoated (ACD) tubes at the Blood Transfusion Center (Geneva University Hospital, Geneva, Switzerland). The formulation samples were incubated with blood at a 3:1 sample/blood ratio at 37°C for 24 h. After centrifugation at 770 x g for 10 min, the supernatant was collected in a 96-well plate and the release of hemoglobin was measured by UV absorbance at 575 nm with four points of measurement per well using a microplate reader (Safire, Tecan, Männedorf, Switzerland). Various concentrations (below and above the CMC) of unloaded MPEG-(hex)PLA, CsAloaded MPEG-dihexPLA micelles and Sandimmune were tested. Samples of different concentrations were prepared by dilution of the highest copolymer or surfactant concentration with isotonic saline solution. Both the concentrated CsA-loaded MPEG-dihexPLA formulations and the concentrated Sandimmune were diluted in order to obtain final CsA concentrations of 0.5 mg/mL and of 2.5 mg/mL. An isotonic saline solution and 1% TritonX-100 solution were tested as the 0% lysis control and 100% lysis control, respectively. The percentage of hemolysis was calculated as follows: Hemolysis (%) = A sample − A 0% A100% − A 0% × 100 where Asample, A0%, A100% represent the absorbances of the sample, the negative and positive control, respectively. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 70 2.10 Shelf-life stability The size stability of unloaded and CsA-loaded micelles in ultra-pure water was investigated at 25°C over one year. The shelf-life of CsA-loaded micelles in isotonic saline solution in terms of drug content and size was studied at 3 different temperatures (4°C, 25°C and 37°C) over 3 months and one year. The micelle size was determined by DLS at multiple detection angles with a goniometer ALV/CGS-5 (ALV-GmbH, Langen, Germany) and a power of 0.2 W. Briefly, 100 µL of unloaded or CsA-loaded MPEG-(hex)PLA micelle formulations were diluted in 2 mL of isotonic saline solution or ultra-pure water in a clean, clear and capped tube resulting in a copolymer concentration of 0.24 mg/mL. After initial size measurements, the samples were stored at their respective storage conditions. At the desired time points, they were allowed to equilibrate to room temperature for 2 h before being analyzed directly in the sample tubes. A similar procedure was applied for the formulation stability tests. After the desired storage time, the CsA-loaded MPEG-(hex)PLA formulation samples were allowed to equilibrate to room temperature and then centrifuged at 9500 g for 15 min to remove any non-entrapped CsA. The CsA drug content in micelles was quantified by the HPLC method described above. 3. Results 3.1 Synthesis of MPEG-(hex)PLA copolymers The MPEG-monohexPLA and MPEG-dihexPLA copolymers were prepared by ring opening polymerization (ROP) in bulk using Sn(Oct)2 as the catalyst and MPEG2000g/mol as the initiator. These molecular structures are presented in Scheme 1. The MPEG-PLA was synthesized by the same method and was used as the “reference”. Copolymers with molecular weights of 5000 g/mol (± 1 monomer unit) were obtained with low polydispersity indices (P.I.) smaller than to 1.1 (Table 1). The MPEG-hexPLAs of the same molecular weight have half and double number of hydrophobic hexyl side groups, respectively, thus permitting us to investigate the influence of hydrophobicity on the micellization, stability, drug loading and toxicity of their resulting micelles. Chapter III 71 O O H3C 45 R O O R R1, R2 = CH3 1 2 1 O H m O : MPEG2000g/mol-PLA3000g/mol for m=21 2 R =C6H13, R =CH3 : MPEG2000g/mol-monohexPLA3000g/mol for m=14 R1, R2 =C6H13 : MPEG2000g/mol-dihexPLA3000g/mol for m=11 Scheme 1. Structure of MPEG-hexPLA and MPEG-PLA block copolymers. 3.2 Unloaded MPEG-hexPLA micelles Unloaded MPEG-hexPLA micelles were prepared in isotonic saline solution by the co-solvent evaporation method and had the number weighted (dn) sizes between 18-27 nm with a population of 99-100% (Table 1). The values obtained for the hydrodynamic diameter (Zav) showed sizes of 70-90 nm with a polydispersity between 0.22-0.35. This higher polydispersity is related to a minor population of some larger micelles, which could be filtered off. Table 1. Characteristics of synthesised MPEG-(hex)PLA copolymers and their corresponding unloaded micelles prepared in isotonic saline solution. a Micelle size [%]dn Zav [nm] P.I. 18 100.0 75 0.35 1.03 26 99.1 70 0.22 1.05 27 99.6 87 0.31 Mwa [g/mol] P.I.a dn [nm] MPEG-PLA 5100 1.07 MPEG-monohexPLA 5000 MPEG-dihexPLA 5300 Copolymer determined by GPC using PS standards CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 72 3.3 CsA incorporation in MPEG-hexPLA micelles Formulations with a copolymer concentration of 5 mg/mL and different targeted drug loadings from 10 to 1000 mgCsA/gcopolymer were prepared. The amount of incorporated CsA was assessed by HPLC after micelle disruption by the addition of acetonitrile. The results are summarized in Fig. 1, where the incorporation efficiency of 100% and 80% are drawn in plain and dashed lines, respectively. 100% efficiency Actual drug loading [mgCsA/gcopolymer] 500 80% efficiency 400 300 200 100 0 0 100 200 300 400 Targeted drug loading [mgCsA /gcopolymer] 500 600 Figure 1. Incorporation of CsA in MPEG-PLA (○), MPEG-monohexPLA (□), and MPEG-dihexPLA () micelles in function of the targeted drug loading. As seen in the figure, the CsA loading increased with the targeted loadings until a plateau at 80 mgCsA/gcopolymer for MPEG-PLA, 230 mgCsA/gcopolymer for MPEG-monohexPLA and 300 mgCsA/gcopolymer for MPEG-dihexPLA copolymer was reached. At the latter angular points, loadings with efficiencies of 80% and higher could be achieved. For MPEG-monohexPLA micelles, the highest and most efficient drug loading of 230 mgCsA/gcopolymer was obtained from the targeted loading of 300 mg/g, with an incorporation efficiency of 77%. For the same target loading, MPEG-dihexPLA incorporated 265 mgCsA/gcopolymer with an efficiency of 88%. For the higher targeted loading of 500 mg/g, the highest drug loading of 320 mg/g for a MPEG-hexPLA formulation could be achieved, still with an efficiency of 65%. Any nonincorporated drug could be removed by centrifugation of the formulation. Chapter III 73 Micelle sizes, Zav and dn of CsA-loaded MPEG-(hex)PLA micelles were determined by DLS at a detection angle of 90°. The results for different drug loadings are presented in Fig. 2, where the sizes are given on the primary Y-axis and the [%]dn on the secondary axis. The MPEG-(hex)PLA micelles had a number weighted diameter between 16 and 30 nm for all tested formulations. The sizes were identical to the unloaded micelle solutions (Table 1). It is notable that the average hydrodynamic diameter Zav increased with the incorporation amount of CsA in MPEG-hexPLA micelles, which was due to the presence of a small number of larger micelles in contrast to unloaded micelle formulations. Still, 98-99% of all the micelles 100 90 80 70 60 50 40 30 20 10 0 [%]dn 90 80 70 60 50 40 30 20 10 0 9 19 35 39 67 77 86 10 27 37 68 100 229 234 9 26 33 68 100 265 327 Size [nm] had sizes dn smaller than 30 nm (Fig. 2). MPEG-PLA micelles MPEGmonohexPLA micelles MPEG-dihexPLA micelles Drug loading [mg/g] Figure 2. Size characteristics of CsA loaded PEG-(hex)PLA micelles in dependence of the drug loading with the hydrodynamic diameter Zav ( ), the number weighted diameter dn ( ) and the percentage of micelles with a given dn ([%]dn (●)). For the formulations envisioned for intravenous application, isotonic saline solutions were prepared by first generating MPEG-hexPLA polymeric micelles in water and then adjusting the solution isotonicity with the addition of NaCl. The average hydrodynamic diameter, Zav, increased with the isotonicity change in the medium (Table 2), whereas the diameter dn remained at 25 nm. The CsA solubility in polymeric micelles was found to be a little bit lower with 1.4 mg/mL, as compared to 1.5 mg/mL in water. The more hydrophobic MPEGdihexPLA micelles incorporated higher amounts of CsA than MPEG-monohexPLA and MPEG-PLA-based formulations. More highly-concentrated micelle solutions of 6 mg/mL could be prepared by increasing the MPEG-dihexPLA copolymer concentration in the medium while keeping the same drug to copolymer ratio. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 74 Table 2. Characteristics of CsA loaded MPEG-dihexPLA micelles with different copolymer concentrations in different aqueous media. MPEG-dihexPLA concentration [mg/mL] Aqueous Medium CsA loading [mgCsA/gcopolymer] CsA concentration [mgCsA/mLmicelles] Size dn [nm] [%]dn Size Zav [nm] P.I. 5 Water 307 ± 1 1.51 ± 0.01 26 100.0 40 0.24 5 Isoton. Saline Sol. 286 ± 11 1.43 ± 0.06 25 99.7 83 0.50 10 Water 318 ± 4 3.23 ± 0.05 26 100.0 40 0.24 20 Water 295 ± 5 5.97 ± 0.11 27 99.7 41 0.28 Chapter III 75 3.4 In vitro toxicity of MPEG-hexPLA solutions To evaluate the in vitro toxicity of the unloaded MPEG-(hex)PLA micelles, three different cell lines were tested: human intestinal epithelial Caco-2 cells, murine NuTu-19 and human SKOV-3 ovarian cancer cells. Various copolymer concentrations were investigated, both below the CMC for studying the toxicity of MPEG-(hex)PLA as unimers, and above the CMC for studying the micelle solutions. The isotonic saline solution and CremophorEL solutions were tested for comparison (Fig. 3). Triton X-100 and culture media were used as 0% survival and 100% survival controls, respectively. After 24 h incubation at 37°C on Caco-2 cells (Fig. 3a), MPEG-(hex)PLA polymeric micelles were non-toxic with a cell survival above 80% for copolymer concentrations ranging from 0.0001 to 25 mgcopolymer/mL, whereas CremophorEL micelles showed no toxicity only for solutions with low concentrations (<1 mgcopolymer/mL). Negative cell survivals were observed for higher concentrations, which can be explained by a washing-out of the cells from the test plates. On NuTu-19 cells, MPEG-(hex)PLA micelles were non-toxic for concentrations up to a 10 mgcopolymer/mL for MPEG-dihexPLA and MPEG-PLA solutions, and up to 1 mgcopolymer/mL for the comparable MPEG-monohexPLA solutions (Fig. 3b). CremophorEL micelles showed no toxicity for low concentrations (<0.5 mg/mL). On the human SKOV-3 ovarian cancer cells, the three MPEG-(hex)PLA micelle solutions were non-toxic up to concentrations of 10 mgcopolymer/mL, whereas CremophorEL micelles started to show a toxic effect for a concentration of 1 mg/mL (Fig. 3c). At higher concentrations MPEG-hexPLA micelles showed a slightly better cell viability than compared to MPEG-PLA micelles. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 76 Figure 3. In vitro toxicity on (a) Caco-2, (b) Nutu-19 and (c) SKOV-3 cells of MPEG-PLA ( ), MPEG-monohexPLA ( different concentrations. ), MPEG-dihexPLA ( ), CremophorEL ( ) and NaCl 0.9% () for Chapter III 77 3.5 In vivo toxicity of MPEG-hexPLA micelle solutions In vivo toxicity of MPEG-(hex)PLA micelles was evaluated using the CAM model. The chick embryo survival was determined 24 h after the intravascular injection of 20 µL micelle solution for four different copolymer concentrations of 0.0001, 5, 10 and 50 mg/mL. All tested concentrations of MPEG-(hex)PLA formulations were non-toxic, since 100% of the tested chick embryos were still alive at this crucial time point. 3.6 Hemolysis test of MPEG-hexPLA micelle solutions The hemolytic property of MPEG-(hex)PLA solutions was assessed on fresh human blood after incubation at 37°C for 24 h. The method was adapted from a hemolysis test protocol described earlier by Mottu et al. 29. The percentage of hemolysis was determined for various copolymer concentrations (Table 3). As compared to the control of an isotonic saline solution, the unloaded MPEG-(hex)PLA micelle solutions showed less than 1% of red blood cell lysis up to a copolymer concentration of 20 mgcopolymer/mL. Likewise, under much diluted conditions that were below the CMC, the non hemolytic activity of MPEG-(hex)PLA unimers was proven (Table 3a). For the highest concentration of 50 mgcopolymer/mL, which is much higher than the formulation concentration that is considered to be practical, a hemolysis of 2.0% for MPEG-dihexPLA, 9.2% for MPEG-monohexPLA and 9.7% for MPEG-PLA were observed. The incorporation of CsA into MPEG-dihexPLA micelles did not induce any red blood cell lysis (Table 3b). A formulation of CsA in MPEG-dihexPLA (20 mg/mL) with a CsA concentration of 6 mg/mL affected less than 1% lysis (Entry 1). Also, the formulations with lower concentrations that are similar to those used in current clinical CsA doses (Entry 2 and 3) do not show hemolysis. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 78 Table 3. Haemocompatibility of (a) unloaded and (b) CsA loaded MPEG-hexPLA micelles of different copolymer concentrations after 24h incubation with human blood. (a) % lysis Copolymer concentration [mg/mL] 50 0.01 to 20 0.0001 to 0.005 a a MPEG-dihexPLA MPEG-monohexPLA MPEG-PLA 2.0% ± 0.9% <1% <1% 9.2% ± 3.6% <1% <1% 9.7% ± 2.3% <1% <1% Concentrations below CMC (b) Entry 1 2 3 4 a CremophorEL CsA Surfactant concentration concentration % lysis [mg/mL] [mg/mL] 6 78 <1% 2.5 32.5 <1% 0.5 6.5 <1% <0.09 a <1% MPEG-dihexPLA Copolymer concentration % lysis [mg/mL] 20 <1% 8.3 <1% 1.7 <1% <0.008 a <1% Concentrations below CMC 3.7 Shelf-life stability The sizes and possible size changes of MPEG-hexPLA micelles were monitored using DLS at different detection angles over a period of one year. At small detection angles of 30-40°, large particles scatter more light than small particles, whereas at larger detection angles of 50-140°, mainly the small particles are detectable. Therefore, it is important to measure micelle sizes at the full range of detection angles in order to observe a possible micelle enlargement or aggregation with time. The results obtained for MPEG-hexPLA micelles prepared in ultrapure water and stored at 25°C for one year are given in Fig. 4a. Directly after preparation, the unloaded MPEG-hexPLA micelles had an average hydrodynamic diameter (observation between 50°-140°) of around 30 nm for MPEG-monohexPLA and of around 40 nm for MPEG-dihexPLA micelles, respectively. Only a few micelles larger than 50 nm were visible at the small angles of 30°-40°. After one year of storage, the size curves of the initial and stored solutions were similar, except at the low detection angle of 30°, where some larger micelles in the stored solutions were observable. For all other detection angles, size changes Chapter III 79 of only 10 nm for MPEG-dihexPLA and of 17 nm for MPEG-monohexPLA were observed. The formation of larger micelles at small angles was also found for CsA-loaded micelles (Fig. 4b). This was more pronounced for MPEG-monohexPLA micelles, for which a substantial increase of the diameter occurred at detection angles between 30° to 60°, showing the existence of 1-2% of larger micelles in the stored solutions. For all other detection angles the difference in diameter values after one year at 25°C varied in the same range as for the unloaded micelles. The results from intermediate time points (data not shown) revealed that micelle size enlargement or aggregation started only after five months for CsA-loaded MPEG-monohexPLA and MPEG-PLA micelles. Hydrodynamic diameter [nm] (a) 250 200 150 100 50 0 30 50 70 90 110 130 110 130 Detection angle [°] Hydrodynamic diameter [nm] (b) 250 200 150 100 50 0 30 50 70 90 Detection angle [°] Figure 4. Hydrodynamic diameters Zav at different detection angles of (a) unloaded- and (b) CsA loaded- MPEG-monohexPLA (,) and MPEG-dihexPLA (,) micelles in ultra pure water directly after preparation (filled symbols) and after 1 year (unfilled symbols) at 25°C. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 80 MPEG-(hex)PLA micelle formulations in isotonic saline solution were assessed for their shelf-life stability, including a size and drug content evaluation. Hydrodynamic diameter [nm] (a) 1000 750 500 425 375 325 300 200 100 0 30 50 70 90 110 130 110 130 110 130 Detection angle [°] Hydrodynamic diameter [nm] (b) 1000 750 500 425 375 325 300 200 100 0 30 50 70 90 Detection angle [°] Hydrodynamic diameter [nm] (c) 1000 750 500 425 375 325 300 200 100 0 30 50 70 90 Detection angle [°] Figure 5. Hydrodynamic diameters Zav at different detection angles of CsA loaded MPEG-PLA (●,○), MPEG-monohexPLA (,) and MPEG-dihexPLA (,) polymeric micelles in isotonic saline solution directly after preparation (filled symbols) and after 3 months (unfilled symbols) at (a) 4°C, (b) 25°C and (c) 37°C. Chapter III CsA concentration [µg/mLmicelles] (a) 81 1200 1000 800 600 400 200 0 CsA concentration [µg/mLmicelles] (b) 0 2 4 0 2 4 0 2 4 6 8 Time [months] 10 12 14 10 12 14 10 12 14 1200 1000 800 600 400 200 (c) 1200 CsA concentration [µg/mLmicelles] 0 1000 6 8 Time [months] 800 600 400 200 0 6 8 Time [months] Figure 6. Formulation stability of CsA incorporated in MPEG-PLA (), MPEG-monohexPLA () and MPEG-dihexPLA () micelles in isotonic saline solution (a) at 4°C, (b) 25°C and (c) 37°C (n=3, standard deviation for most time points smaller than symbols). CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 82 Fig. 5 shows the micelle hydrodynamic diameters of the three micelle solutions at (a) 4°C, (b) 25°C and (c) 37°C. Here the formation of larger micelles was visible at detection angles between 30° to 60° after three months of storage time. The size change in these formulations was influenced by the storage temperature. Larger micelles were observed at 37°C (200-450 nm) than at 25°C (100-750 nm) and at 4°C (150-220 nm). The drug content of CsA-loaded MPEG-(hex)PLA formulations in isotonic saline solution at the same storage temperatures was followed up over time (Fig. 6). The CsA content in micelles remained most stable at 4°C, with a drug loss of less than 7% for all tested formulations after one year. At 25°C, the drug loss increased to 26%, 82% and 100% for MPEG-dihexPLA, -monohexPLA, and MPEGPLA micelles, respectively. At 37°C, the entire incorporated amount of CsA was released after three months in MPEG-PLA micelles, after seven months in MPEG-monohexPLA and 13 months in MPEG-dihexPLA micelles. 4. Discussion Polymeric micelles based on MPEG-hexPLA are characterized by an increased hydrophobicity of the micelle core in comparison to the standard MPEG-PLA. It has been demonstrated that this enables higher drug loadings of poorly water-soluble drugs 21 . In this paper, we investigated the biocompatibility of these novel micelles towards formulations of the poorly water-soluble drug Cyclosporin (CsA) for intravenous applications. The novel copolymers, MPEG-monohexPLA and MPEG-dihexPLA, and the “reference” standard MPEG-PLA were synthesized in a controlled manner by ROP with a defined molecular weight of 5000 g/mol (± 1 monomer unit), thus allowing a good comparison of the influence of the introduced hexyl substituents along the PLA backbone. The increase of hydrophobic interactions between copolymer chains favors micellization at lower critical micellar concentrations (CMC). Indeed, the CMCs of MPEG-hexPLA micelles decrease with the increase of hexyl groups in the core-forming block, leading to a value of 1.6×10-6 M and 1.7×10-6 M for MPEG-dihexPLA and -monohexPLA compared to a value of 2.0×10-6 M for MPEG-PLA micelles 20 . These values in the micro molar range are in agreement with data reported in the literature 5;30. Compared to MPEG-PLA and to other typical surfactants, which Chapter III 83 have CMCs with 10 to 1000 times higher concentrations 4, the low CMC of MPEG-hexPLA micelles facilitates better stability upon dilution, which can improve these formulations for envisioned intravenous applications. Moreover, MPEG-hexPLA micelles are very small and truly nanocarriers. Around 99% of MPEG-hexPLA micelles had number weighted diameters (dn) between 18-30 nm, confirming a unimodal size distribution in the unloaded (Table 1) or CsA-loaded state (Fig. 2). The incorporation of CsA into the micelles did not influence the micelle size, dn. As a nanosized drug delivery system, MPEG-hexPLA micelles should have the same advantages as described for other polymeric micelles, in particular the ability to escape the mononuclear phagocyte system (MPS) uptake and renal clearance, allowing a long circulation in the body, which should result in a higher probability of reaching the target 30. The potential of MPEG-hexPLA micelles to solubilize the hydrophobic drug CsA was herein demonstrated in comparison to MPEG-PLA micelles and the CremophorEL surfactant. Comparing the obtained CsA loadings for MPEG-hexPLA with the corresponding MPEGPLA formulations, it becomes obvious that the increased hydrophobicity of the new hexPLAbased micelles improve the loading and the solubility capacity. In aqueous MPEG-hexPLA formulations, CsA was solubilized very efficiently up to the concentration of 1.5 mg/mL with 5.0 mg/mL MPEG-dihexPLA excipient (Table 2). Increasing the concentration of the MPEGhexPLA copolymer while keeping the drug/copolymer ratio constant leads to higher concentrated formulations. At comparable polymer concentrations of 10 mg/mL, MPEGhexPLA could incorporate twice the amount of CsA as MPEG-polycaprolactone (MPEG5000g/mol–PCL13000g/mol) micelles (~1.3 mg/mL) as reported by Aliabadi et al. 31 . Formulations with 20 mg/mL MPEG-hexPLA facilitated a CsA concentration of 6 mg/mL, corresponding to an increase in water solubility of CsA by a factor of 500. For comparable CsA concentrations, MPEG-dihexPLA formulations would require four times less excipient than a currently marketed formulation with CremophorEL as the surfactant. Toxicity studies carried out on three different cell lines showed that MPEG-hexPLA micelles had a lower toxicity than formulations with CremophorEL micelles on human ovarian cancer SKOV-3 cells. On the two other cell lines, CremophorEL formulations at higher concentrations than 0.5 mg/mL washed out cells from the test plates, and thus cell viabilities could not be determined (Fig. 3). This was not the case for MPEG-hexPLA micelle CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 84 formulations, for which non-toxicity could be found for concentrations at least up to 10 mg/mL. For possible intravenous pharmaceutical applications, isotonic micelle solutions of MPEGhexPLA micelles were prepared first in water and then adjusted by the addition of NaCl crystals before use. By this procedure, CsA formulations with a concentration of 1.4 mg/mL could be more efficiently prepared and the loss of drug observed in other procedures could be significantly reduced. For example, by direct preparation in isotonic saline solution, MPEGhexPLA micelles could solubilize around 1 mg/mL CsA, which corresponds to an incorporation of 48% (results not shown) as opposed to 73% when prepared in water and adjusted afterwards to isotonicity. This was also observed for other copolymers like MPEG5000g/mol-PCL13000g/mol micelles, which incorporated CsA with 28% or 37% efficiency when prepared in isotonic solution, whereas in water a 37% or 64% efficiency could be achieved, depending on the initial applied drug loading 32 . The choice of the medium is not only of importance for the drug loading efficiency, but it is also an important factor regarding the stability of the micelle formulations. In isotonic saline solutions, the formation of larger micelles was observable after three months, whereas in water, no size increase could be detected for MPEG-dihexPLA formulations after one year of storage (Fig. 4). A micelle size enlargement after addition of salt has also been observed by Jain et al. and has been explained by the dehydratation effects on the PEG units of the micelle shell 33 . For the investigated formulations, the increase in micelle diameter was dependent on the storage temperature and the copolymer. A storage temperature of 4°C showed the best formulation shelf-life stability, whereas in the isotonic saline solution only a very few larger MPEG-hexPLA micelles were observed after three months (Fig. 5), still with diameters below 200 nm, which is the maximum size for particles with long-circulating properties 34 . To avoid their potential consequences, larger micelles or aggregates could be simply filtered off with common sterile filters after extended storage times. Regarding the CsA concentrations in the MPEG-hexPLA formulations, the CsA amounts remained very constant over one year when stored at 4°C, with a final drug loss of only 7% (Fig. 6). In contrast, at 25°C and 37°C, the initial drug content in the micelles decreased more rapidly. At 37°C, a complete CsA release was observed at earlier time points; the MPEG-hexPLA micelles showed a higher stability than the MPEG-PLA micelles. A recent paper of Nottelet et al. have also demonstrated the superior stability of MPEG-hexPLA micelles in PBS modeling intravenous conditions (pH Chapter III 85 7.4 at 37°C) compared to MPEG-PLA micelles 35. These results could be associated with the degradation of the polymers under these conditions. Previous studies on hexPLA polymers have shown that, despite a similar degradation profile, the homopolymer PLA had a faster molecular weight decrease and weight loss than hexPLA in PBS at pH 7.4 after 50 days 19 . Thus, the slower polymer degradation and the lower CMC increase the excipient stability in the formulations and stabilize the solubilized CsA drug content. Nevertheless, the data obtained here for the higher temperatures indicate that the carrier system has the properties of a certain shelf-life stability next to its ultimate degradation and drug release, when applied in the human body. The biocompatibility aspects of both MPEG-hexPLA unimers and micelles were investigated by studying their toxicity in vitro and in vivo and by assessing their hemolytic activity on red blood cells. Practically, the biocompatibility of unimers was carried out by modeling the dilution effect after intravenous application and thus the disassembly of micelles. Unloaded MPEG-hexPLA micelles were tested on three cell lines with different mammalian origins and from different sources: human epithelial intestinal cells (Caco-2), murine (NuTu-19) and human ovarian cancer cells (SKOV-3). Non-toxicity was found for MPEG-dihexPLA unimers and for their polymeric micelles at least up to 10 mgcopolymer/mL on all tested cell lines and was comparable to the results obtained for MPEG-PLA copolymers as controls (Fig. 3). In addition to the MTT tests, the in vivo toxicity of MPEG-hexPLA micelles was studied on the CAM model. This model is an alternative to mammalian models and has been proven helpful for testing intravenous formulations due to its well-developed vasculature network 36 . Injection volumes of 20 µL into the vasculature of the chick embryos were found to be welltolerated 28 and were therefore chosen for the toxicity studies of the MPEG-hexPLA micelle solutions. The survival rate of chick embryos was assessed 24 h after injection, since longer times usually give identical results. The CAM model results showed neither a toxicity for MPEG-hexPLA unimers nor for the MPEG-hexPLA micelle formulations of the three tested concentrations. All chick embryos survived the injection of the formulations with the novel excipient. It has to be pointed out that MPEG-hexPLA micelles were non-toxic up to a concentration of 50 mgcopolymer/mL, the same as was found for the control MPEG-PLA micelle solution, whose biocompatibility has been reported in the literature 37. To add to these toxicity studies, the compatibility with human blood was assessed. The hemolytic property of MPEGhexPLA unimers and unloaded micelles was investigated at different concentrations and CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 86 showed no hemolytic activity (<1% lysis) up to a copolymer concentration of 20 mg/mL. A slight hemolytic activity was observed for the three different MPEG-(hex)PLA micelles for the “extreme” copolymer concentration of 50 mgcopolymer/mL, which is far above the concentrations possibly needed for an envisioned intravenous application. Also, under these conditions, the novel hexPLA based micelles did not differ in their hemolytic activity from the standard PLA based micelles. Considering the practical application of CsA MPEG-hexPLA formulations, the corresponding drug concentrations of 0.5 mg/mL up to 2.5 mg/mL, as used in the current CremophorEL based products, were found to not induce hemolysis. The high drug loading capacity of MPEG-hexPLA micelles allows a significant reduction of polymeric excipient for formulating the same amounts of CsA. This increases the potential intravenous use of such micelle formulations, which may improve the maximum tolerated dose (MTD) in the treatment. In a previous study, MPEG-PLA micelles could increase four times the MTD of paclitaxel in nude mice compared to the current treatment with Taxol, which is also formulated with CremophorEL 38 . In addition, regarding the influence of the physical state of the hydrophobic core forming block on the micelle stability, MPEG-hexPLA micelles are expected to retain the drug more efficiently than CremophorEL micelles 31;37. 5. Summary and Conclusions In this paper, we demonstrated the biocompatibility and non-toxicity of polymeric micelles based on MPEG-hexPLA. The amphiphilic MPEG-hexPLA copolymers self-assembled in aqueous media into polymeric micelles of 30 nm. The resulting micelle formulations show no toxicity and no hemolytic activity in MTT tests, the CAM model and on human blood tests. All results are comparably good or better than those obtained for the controls of standard MPEG-PLA. By increasing the MPEG-hexPLA copolymer concentration while keeping the drug:copolymer ratio constant, the immunosuppressive and hydrophobic drug Cyclosporin A could be solubilized and formulated with concentrations up to 6 mg/mL, which is equivalent to a 500-fold increase of the drug’s water solubility. The current clinically-used CsA Chapter III 87 concentration administrated intravenously with Sandimmune can be prepared in a MPEGhexPLA micelle formulation by a simple procedure and would need four times less copolymer than is used with the surfactant CremophorEL. MPEG-hexPLA micelles have the potential to be very interesting non-toxic injectable nanosized drug carriers for improved formulations of poorly soluble drugs. Acknowledgments The authors thank the Swiss National Science Foundation (SNF) for financial support (SNF 200020-103752). The authors are grateful to Professor Michal Borkovec and Dr. Andrea Vaccaro (University of Geneva, Geneva, CH) for the availability and use of the goniometer ALV/CGS-5. References (1) Byrne, J. D.; Betancourt, T.; Brannon-Peppas, L., Active targeting schemes for nanoparticle systems in cancer therapeutics, Adv.Drug Deliv.Rev. 2008, 60, 1615-1626. (2) Chatterjee, D. K.; Fong, L. S.; Zhang, Y., Nanoparticles in photodynamic therapy: An emerging paradigm, Adv.Drug Deliv.Rev. 2008, 60, 1627-1637. (3) Torchilin, V. P., Recent advances with liposomes as pharmaceutical carriers, Nat.Rev.Drug Discov. 2005, 4, 145-160. (4) Croy, S. R.; Kwon, G. S., Polymeric Micelles for Drug Delivery, Curr.Pharm.Design 2006, 12, 4669-4684. (5) Kwon, G. S., Polymeric micelles for delivery of poorly water-soluble compounds, Crit. Rev. Ther. Drug Carrier Syst. 2003, 20, 357-403. (6) Torchilin, V., Micellar Nanocarriers: Pharmaceutical Perspectives, Pharm.Res. 2007, 24, 1-16. (7) Maeda, H.; Bharate, G. Y.; Daruwalla, J., Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect, Eur.J.Pharm.Biopharm. 2009, 71, 409-419. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 88 (8) Desai, M. P.; Labhasetwar, V.; Walter, E.; Levy, R. J.; Amidon, G. L., The Mechanism of Uptake of Biodegradable Microparticles in Caco-2 Cells Is Size Dependent, Pharm.Res. 1997, 14, 1568-1573. (9) Jain, R. K., Delivery of molecular medicine to solid tumors: lessons from in vivo imaging of gene expression and function, J.Control.Release 2001, 74, 7-25. (10) McClean, S.; Prosser, E.; Meehan, E.; O'Malley, D.; Clarke, N.; Ramtoola, Z.; Brayden, D., Binding and uptake of biodegradable poly--lactide micro- and nanoparticles in intestinal epithelia, Eur.J.Pharm.Sci. 1998, 6, 153-163. (11) Gref, R.; Lück, M.; Quellec, P.; Marchand, M.; Dellacherie, E.; Harnisch, S.; Blunk, T.; Müller, R. H., Stealth corona-core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption, Colloid.Surface B 2000, 18, 301-313. (12) Kabanov, A. V.; Batrakova, E. V.; Alakhov, V. Y., Pluronic(R) block copolymers as novel polymer therapeutics for drug and gene delivery, J.Control.Release 2002, 82, 189212. (13) Lavasanifar, A.; Samuel, J.; Kwon, G. S., Poly(ethylene oxide)-block-poly(-amino acid) micelles for drug delivery, Adv.Drug Deliv.Rev. 2002, 54, 169-190. (14) Liggins, R. T.; Burt, H. M., Polyether-polyester diblock copolymers for the preparation of paclitaxel loaded polymeric micelle formulations, Adv.Drug Deliv.Rev. 2002, 54, 191-202. (15) Allen, C.; Maysinger, D.; Eisenberg, A., Nano-engineering block copolymer aggregates for drug delivery, Colloid.Surface B 1999, 16, 3-27. (16) Liu, J. B.; Xiao, Y. H.; Allen, C., Polymer-drug compatibility: A guide to the development of delivery systems for the anticancer agent, ellipticine, J.Pharm.Sci. 2004, 93, 132-143. (17) Nagarajan, R.; Barry, M.; Ruckenstein, E., Unusual selectivity in solubilization by block copolymer micelles, Langmuir 1986, 2, 210-215. (18) Trimaille, T.; Gurny, R.; Möller, M., Synthesis and ring-opening polymerization of new monoalkyl-substituted lactides, J.Polym.Sci.Part A: Polym.Chem. 2004, 42, 4379-4391. (19) Trimaille, T.; Gurny, R.; Möller, M., Poly(hexyl-substituted lactides): Novel injectable hydrophobic drug delivery systems, J.Biomed.Mater.Res.A 2007, 80A, 55-65. (20) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. (21) Mondon, K.; Gurny, R.; Möller, M., Colloidal Drug Delivery Systems Recent Advances With Polymeric Micelles, Chimia 2008, 62, 832-840. Chapter III 89 (22) Czogalla, A., Oral cyclosporine A - the current picture of its liposomal and other delivery systems, Mol.Biol.Lett. 2008, 14, 139-152. (23) Schumacher, A.; Nordheim, A., Progress towards a molecular understanding of cyclosporin A-mediated immunosuppression., Clin.Invest. 1992, 70, 773-779. (24) Pietzonka, P.; Rothen-Rutishauser, B.; Langguth, P.; Wunderli-Allenspach, H.; Walter, E.; Merkle, H. P., Transfer of Lipophilic Markers from PLGA and Polystyrene Nanoparticles to Caco-2 Monolayers Mimics Particle Uptake, Pharm.Res. 2002, 19, 595-601. (25) Gaumet, M.; Vargas, A.; Gurny, R.; Delie, F., Nanoparticles for drug delivery: The need for precision in reporting particle size parameters, Eur.J.Pharm.Biopharm. 2008, 69, 19. (26) Rose, G. S.; Tocco, L. M.; Granger, G. A.; DiSaia, P. J.; Hamilton, C. T.; Santin, A. D.; Hiserodt, J. C., Development and characterization of a clinically useful animal model of epithelial ovarian cancer in the Fischer 344 rat, Am.J.Obstet.Gynecol. 1996, 175, 593599. (27) Lange, N.; Ballini, J. P.; Wagnieres, G.; van den Bergh, H., A New Drug-Screening Procedure for Photosensitizing Agents Used in Photodynamic Therapy for CNV, Invest.Ophthalmol.Vis.Sci. 2001, 42, 38-46. (28) Vargas, A.; Pegaz, B.; Debefve, E.; Konan-Kouakou, Y.; Lange, N.; Ballini, J. P.; van den Bergh, H.; Gurny, R.; Delie, F., Improved photodynamic activity of porphyrin loaded into nanoparticles: an in vivo evaluation using chick embryos, Int.J.Pharm. 2004, 286, 131-145. (29) Mottu, F.; Stelling, M. J.; Rüfenacht, D. A.; Doelker, E., Comparative hemolytic activity of undiluted organic water-miscible solvents for intravenous and intra-arterial injection, PDA J.Pharm.Sci.Technol. 2001, 55, 16-23. (30) Torchilin, V. P., Structure and design of polymeric surfactant-based drug delivery systems, J.Control.Release 2001, 73, 137-172. (31) Aliabadi, H. M.; Brocks, D. R.; Lavasanifar, A., Polymeric micelles for the solubilization and delivery of cyclosporine A: pharmacokinetics and biodistribution, Biomaterials 2005, 26, 7251-7259. (32) Aliabadi, H. M.; Mahmud, A.; Sharifabadi, A. D.; Lavasanifar, A., Micelles of methoxy poly(ethylene oxide)-b-poly([epsilon]-caprolactone) as vehicles for the solubilization and controlled delivery of cyclosporine A, J.Control.Release 2005, 104, 301-311. (33) Jain, N. J.; Aswal, V. K.; Goyal, P. S.; Bahadur, P., Salt induced micellization and micelle structures of' PEO/PPO/PEO block copolymers in aqueous solution, Colloid.Surface A 2000, 173, 85-94. (34) Moghimi, S. M.; Hunter, A. C.; Murray, J. C., Long-Circulating and Target-Specific Nanoparticles: Theory to Practice, Pharmacol.Rev. 2001, 53, 283-318. CsA/ MPEG-hexPLA Micelle Formulations : A Suitability Study 90 (35) Nottelet, B.; Di Tommaso, C.; Mondon, K.; Gurny, R.; Möller, M., Fully biodegradable polymeric micelles based on hydrophobic- and hydrophilic-functionalized poly(lactide) block copolymers, J.Polym.Sci.Part A: Polym.Chem. 2010, 48, 3244-3254. (36) Vargas, A.; Zeisser-Labouèbe, M.; Lange, N.; Gurny, R.; Delie, F., The chick embryo and its chorioallantoic membrane (CAM) for the in vivo evaluation of drug delivery systems, Adv.Drug Deliv.Rev. 2007, 59, 1162-1176. (37) Burt, H. M.; Zhang, X. C.; Toleikis, P.; Embree, L.; Hunter, W. L., Development of copolymers of poly(D,L-lactide) and methoxypolyethylene glycol as micellar carriers of paclitaxel, Colloid.Surface B 1999, 16, 161-171. (38) Kim, S. C.; Kim, D. W.; Shim, Y. H.; Bang, J. S.; Oh, H. S.; Kim, S. W.; Seo, M. H., In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and efficacy, J.Control.Release 2001, 72, 191-202. 1234563746489AB93CDE69EF6CEFC93CDE6379367416C4AA469546EDE3DC69EF6FD6ED36 7D6 74DAC6 93C8C36 9E6 CE35984EDB6 9AC93CDE6 D26 7416 C4AA46 C376 3746 1 2ABD544E36 745CCE6 95456 96 3BFC4F6 96 96 E46 FC9ED3C6 2D5BA93CDE6 2D56 D895C9E6 9E4569EF643939C6F4343CDE 66 1 1 1 1 123456789A8 8 1 Chapter IV 91 MPEG-hexPLA Micelles as Novel Carriers for Hypericin, a Fluorescent Marker for Use in Cancer Diagnostics K. Mondon, M. Zeisser-Labouèbe, R. Gurny, and M. Möller School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30 quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland. Submitted to: Photochemistry and Photobiology Ovarian cancer is the most common gynecological cancer diagnosed in Western countries. Detection of micrometastases at an early stage of the disease could lead to a cure rate of 90% by limiting the spread of the disease outside the ovaries. In this article, hypericin (Hy), a hydrophobic photosensitizer used for the photodynamic diagnosis of ovarian cancer, was efficiently incorporated into a core of micelles made from methoxy-poly(ethylene glycol) (MPEG) and hexyl-substituted poly(lactides) (hexPLA) copolymers. The fate of these micelles following intravenous injection was studied in vivo in two ovarian tumor-bearing animal models. In the chick embryo chorioallantoic membrane (CAM) model, 17 times more Hy accumulated in tumor nodules when Hy was delivered with micelles than when Hy was delivered as an ethanol solution. Studies of the biodistribution of Hy in Fisher rats revealed escape of these nanosized micelles (<32 nm) from the mononuclear phagocyte system. Hyloaded micelles showed maximal accumulation in tumors and demonstrated the best tumor/muscle contrast visible 3h after injection in the rat model. The rapid and highly selective accumulation of Hy in tumors that we demonstrated in this study suggests that these micelle formulations could be used for the photodynamic diagnosis of ovarian cancer in the future. Keywords: diagnosis; drug delivery; hypericin; imaging; ovarian cancer; polymeric micelles Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 92 Chapter IV 93 1. Introduction In the current era, cancer is overtaking cardiovascular disease and becoming the leading cause of death worldwide. According to the World Health Organization, cancer-related deaths will reach 12 million in 2030 as compared to 7.4 million in 2004. Early detection of many types of cancers can reduce fatality rates by as much as 30%. Additionally, a precise localization of tumor tissues is essential before surgery, radiotherapy or chemotherapy is considered 1. Despite the progress that has been made in imaging technology (ultrasound, endoscopy and radiography), ovarian cancer is still the leading cause of death from gynecological malignancies in Western countries 2. Due to the absence of symptoms during the early stages of the disease, the majority of affected patients have already developed metastases when their cancer is diagnosed. As a consequence, the 5-year survival rate very much depends on the stage of the disease at the time of diagnosis. For stage I (localized) disease, the 5-year survival rate is 93%, whereas for stage IV (distant) disease, it decreases drastically to 31% 2. Early surgery combined with taxane- or platinum-based chemotherapy has improved the overall survival rate of ovarian cancer patients, but survival rates are still unacceptably low for patients with advanced disease. Half of these patients will relapse within five years after surgery 3, mainly due to non-detected residual metastases in the peritoneal cavity 4. A newly developed selective photodynamic diagnosis (PD) imaging technology has recently been shown to efficiently detect bladder cancer 5 . This technique consists of administrating a photosensitizer (PS) or a precursor for the in situ formation of a PS, which preferentially accumulates in diseased tissues 6. Due to its fluorescence properties, the PS is easily located when excited at the appropriate wavelength, enabling easy detection of malignant tissues. Of the PSs that have been studied, 5-aminolevulinic acid (5-ALA)-mediated protoporphyrin IX (PP IX), Photofrin®, Temoporfin (Foscan®) and Metvix® have been successfully used for the detection of various cancers 7, such as bladder 5, esophageal 8, skin 9 and head and neck cancer 10. In ovarian cancer, 5-ALA-mediated PP IX has shown feasibility and promise in detecting metastases in both animal models 11 and humans 4. Another PS that has been investigated for use in cancer detection is hypericin (Hy). Hy is an interesting PD compound due to its low photobleaching 12 and its natural origin as well as its photoactivity in several cancer cell lines 13-15 and rodent models 13;14;16 . Recently, Hy has been found to be effective at detecting oral cancer 17 as well as ovarian tumors in rats 22 challenge due to its high degree of hydrophobicity (log P=8.78 23 and bladder tumors in humans 18-21 . Formulating Hy is a ). Polylactide (PLA) Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 94 nanoparticles (Nps) loaded with Hy have recently been evaluated and have been shown to specifically reach tumor nodules in rats following intravenous (i.v.) administration. Because these polymeric Nps had a longer half-life as compared to the drug in solution as well as a small size (~200 nm), they were able to extravasate from the neovascularature and accumulate in diseased tissues through the enhanced permeation retention (EPR) effect 22;24 . Their small size, long circulation time and specific accumulation and transport of the drug into tumor tissues are the reasons that there has been increasing interest in nanocarrier systems in cancer research 25-27. Amongst the different nanocarrier systems that have been studied, polymeric micelles have gained special interest because of their nanoscale small size, in vivo stability, high drug-loading capacity and good biocompatibility 28 . Polymeric micelles have a specific core-shell structure formed by the self-assembly of amphiphilic copolymers. The outer shell, which is composed of a hydrophilic polymer, most often poly(ethylene glycol) (PEG), reduces interactions with blood proteins, facilitating a long circulation time. The inner core, which is formed by the hydrophobic block, can successfully be loaded with lipophilic drugs, such as PSs 29-33 . Biocompatible and biodegradable polymers like PLA are preferred for use in the hydrophobic core polymer, but some hydrophobic drugs can only be incorporated into PLA micelles in a limited fashion 34. Thus, it would be ideal to identify and create polymers with an increased degree of hydrophobicity that would allow more efficient drug loading, which, as a result, could lead to better clinical results. Our group recently demonstrated the efficiency with which hydrophobic drugs, such as the PS meso-tetra(p-hydroxyphenyl)porphine (THPP) could be incorporated into (hexPLA)-based micelles. These micelles were shown to load this hydrophobic drug more efficiently than standard PLA. Successful loading of 123 mgTHPP/gcopolymer was achieved with MPEG-hexPLA, as compared to 62 mgTHPP/gcopolymer that was achieved with MPEG-PLA micelles 35 . To the best of our knowledge, there are no previous reports in the literature examining the potential pharmaceutical applications of polymeric micelle-incorporated Hy. In this paper, we investigated the potential utility of Hy formulated in MPEG-hexPLA micelle solutions in ovarian cancer diagnostics. Hy-loaded MPEG-hexPLA micelle formulations with a high drug-loading capacity were prepared and investigated in two different ovarian tumor-bearing animal models, the chick embryo chorioallantoic-membrane (CAM) and the female Fisher rat F-344. Chapter IV 95 2. Materials and methods 2.1. Materials Hypericin (Hy) was purchased from Alexis Corporation (Lausen, Switzerland). Tetrahydrofuran (THF) was supplied by SDS (Toulouse, France). Methoxy-poly(ethylene glycol) (MPEG) with a molecular weight of 2000 g/mol was a gift from Union Carbide Corporation (USA). D,L-lactide, tin(II) 2-ethylhexanoate (Sn(Oct)2) and acetone p.a. were purchased from Purac Biochem (Gorinchem, The Netherlands), Aldrich (Buchs, Switzerland) and Fluka (Buchs, Switzerland), respectively, and used as received. The monomers mono-hexyl-substituted lactide (monohexLA) and di-hexyl-substituted lactide (dihexLA) were synthesized as described in a previous publication 36. 2.2. Synthesis and characterization of MPEG-hexPLA copolymers The synthesis of MPEG-hexPLA copolymers with a molecular weight of approximately 5000g/mol has been described previously (34-35). Briefly, MPEG-hexPLA copolymers were synthesized by ring opening polymerization (ROP) with a MPEG of 2000g/mol as the initiator and tin octanoate as the catalyst. The obtained copolymers were characterized by their molecular weight (Mn) and polydispersity index (P.I.) by gel permeation chromatography (GPC). The structures of MPEG-hexPLA are presented in Scheme 1. O H3C O 45 R O O R 1 2 O H m O R1=C6H13, R2=CH3 : MPEG2000g/mol-monohexPLA3000g/mol for m=14 R1, R2 =C6H13 : MPEG2000g/mol-dihexPLA3000g/mol Scheme 1. Structure of MPEG-hexPLA block copolymers for m=11 Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 96 2.3. Preparation of Hy-loaded MPEG-hexPLA micelles Hy-loaded MPEG-hexPLA micelles were prepared by the co-solvent evaporation method described by Mondon et al. 37 . Briefly, 100 mg/mL solutions of MPEG-hexPLA copolymers in acetone and a 10 mg/mL solution of Hy in acetone were prepared. Next, 500 µL or 200 µL of Hy solution were gently mixed with 400 µL of MPEG-dihexPLA or 800 µL of MPEG-monohexPLA solution, respectively. Acetone was added to the organic phase in order to obtain a final volume of 1 mL. The organic mixture was poured into 2 mL isotonic saline solution under sonication and then slowly removed by evaporation at 15 mbar. The final micelle concentrations were adjusted by adding isotonic saline solution to reach a copolymer concentration of 20 mgcopolymer/mL for MPEG-dihexPLA and 40 mgcopolymer/mL for MPEG-monohexPLA (see structural difference in Scheme 1). The solutions were left to equilibrate overnight and then centrifuged at 9500 x g for 15 min to remove non-incorporated Hy. Both MPEG-hexPLA micelles were then analyzed for size and drug loading. 2.4. Characterization of Hy-loaded MPEG-hexPLA micelles The hydrodynamic (Zav) and number-weighted (dn) diameters of micelles were measured by dynamic light scattering (DLS) after centrifugation. Analyzes were performed at 25 °C with a Zetasizer HS 3000 system (Malvern, Worcestershire, UK) at an angle of 90°. For each sample, mean diameters were obtained after 3 runs of 10 measurements. For drug-loading determination, the micelles were broken up by a 10-fold dilution with acetonitrile. The released Hy was quantified in triplicate by reversed-phase HPLC as previously described by Zeisser-Labouèbe et al. 38. The HPLC was calibrated with standard solutions of 2.0–200 µg/mL of Hy dissolved in acetone. The resulting calibration curves were obtained with regression coefficients greater than 0.99. The incorporation efficiency and drug loading were calculated using equation (1) and (2), respectively: Incorporation efficiency (%) = Drug loading (% w/w) = mass of drug incorporated in micelles (g) × 100 mass of drug introduced (g) mass of drug incorporated in micelles (g) × 100 mass of copolymer used (g) (1) (2) Chapter IV 97 2.5. Animals Female Fisher rats F-344 (150-200g) were purchased from Charles River Laboratories (L’Arbresle, France) and housed in a temperature-controlled room with a 12 h light/dark cycle. They were given a commercial basal diet and water ad libitum. All animal experiments and animal husbandry were carried out in compliance with national regulations and approved by the cantonal veterinary office of Geneva, Switzerland (Certificate number 31.1.1020/3065/2). 2.6. Cell culture and cell preparation for in vivo inoculation The NuTu-19 cell line, a poorly differentiated Fischer 344 rat-derivative epithelial ovarian cancer cell line 39 , was kindly provided by Dr. A. Major (Geneva University Hospital, Geneva, Switzerland). The cells were cultured in Dulbecco's Modified Eagle's medium (DMEM) (Gibco Life Technologies, Carlsbad, USA) supplemented with 10% fetal bovine serum (FBS) (Brunschwig, Amsterdam, The Netherlands) and 100 U/mL penicillin–streptomycin (Gibco Life Technologies) at 37 °C (atm. 5% CO2). Before tumor implantation onto the CAM or into rats, NuTu-19 cells were washed twice with phosphate buffered saline (Gibco Life Technologies, Carlsbad, USA), harvested using 0.5% Trypsin-EDTA (Gibco Life Technologies, Carlsbad, USA) and counted. After centrifugation, an equal mixture of complete culture medium and Matrigel matrix (BD Biosciences, Bedford, USA) was prepared. A suspension of 108 cells/mL was prepared for the CAM experiments and a suspension of 5x106 cells/mL was prepared for the rat experiments. 2.7. Chick embryo chorioallantoic membrane (CAM) study The chick embryo chorioallantoic membrane (CAM) model was adapted from the model described by Lange et al. 40. Egg incubation was performed according to the procedure described by Vargas et al. 24;41 . Briefly, fertilized hen eggs, which were kindly provided by the Geneva University Animal House (Geneva, Switzerland) were placed with the narrow apex down in an incubator (Savimat MG 200, Chauffry, France) at 37 °C with a relative humidity of 65%. The eggs were rotated twice a day until embryo development day 3 (EDD3). On EDD4, a 3-mm hole was drilled into the eggshell and the narrow apex was covered with an adhesive tape. The eggs were incubated in a static mode until EDD8, when the hole in the eggshell was enlarged by 2 or 3 mm to allow the placement of a silicon O-ring (Apple Rubber products inc., Lancaster, USA) on Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 98 the CAM. Next, 20 µL of the ovarian cancer NuTu-19 cell suspension (108 cells/mL) were inoculated topically to allow tumor growth inside the O-ring area. The open hole was then sealed with a plastic film (Parafilm, Pechiney Plastic Packaging, Chicago, USA) to avoid contamination and desiccation of the CAM. Eggs were returned to the static incubator until EDD12, the day of the test formulation administration. A total of 3 formulations were tested on 5 eggs each: (a) an Hy-loaded MPEG-monohexPLA formulation with a concentration of 0.69 mg/mL Hy; (b) an Hyloaded MPEG-dihexPLA formulation with a concentration of 1.0 mg/mL Hy; and (c) a Hy solution (1.0 mg/mL Hy in a mixture of ethanol, PEG 400 and water). These formulations were injected via the main vessel of the CAM at a dose of 2 mg/kg (i.e., 20 ng/egg). The eggs were returned to the incubator and the fluorescence of the tumor nodules was evaluated at 1 min, 1 h, 3 h and 6 h post-injection by fluorescence microscopy with an attenuation coefficient of 4 [see details of the set-up in reference 24. The relative tumor fluorescence (Frel) was calculated using equation (3): Frel = (Ft - F0 ) , F0 (3) where Ft is the tumor fluorescence intensity at the different time points and F0 is the tumor autofluorescence prior to drug injection. 2.8. Biodistribution of Hy-loaded MPEG-dihexPLA micelles in rats Ovarian tumor-bearing female Fisher rats (F-344) were used for Hy biodistribution studies. The rats were inoculated intraperitoneally with 1 mL of ovarian cancer NuTu-19 cell suspension (5x106 cells) to induce tumor growth. After five weeks, ascites were palpable and tumors had grown sufficiently to allow us to perform the study. The rats were injected intravenously in the tail vein with Hy formulations at a dose of 2 mg/kg. Three formulations were investigated: (a) Hy loaded MPEG-dihexPLA micelles; (b) Hy solution (2 mg/mL in a mixture of ethanol, PEG 400 and water); and (c) isotonic saline solution (which was administered to two rats as a control). After intravenous injection of the Hy formulations, 4 rats per formulation were sacrificed by CO2 asphyxia at 1, 3, 6 and 24 h after injection for the micelle formulations and 1 h after injection for Hy in ethanol solution. In order to visualize the tumors by fluorescence imaging, the abdominal cavity was opened and images were compared under white and blue light, respectively using the set-up described previously by Chapter IV 99 Zeisser-Labouèbe et al. 22. Blood samples were obtained by cardiac puncture with heparinized tubes. The liver, spleen, lung, tumor and muscles surrounding the tumors were excised. All samples were weighed and stored at -20 °C until analysis. Tissue samples were extracted with tetrahydrofuran (THF) complemented with a tissue homogenizer (Eurostar digital, IKA®Werke, Staufen, DE). Blood samples were prepared for analysis by THF extraction and sonication (5 times for 5 s with a sonifier S-450D®, Branson Ultrasonic S.A, Geneva, CH). After sample centrifugation, the supernatants were evaporated under nitrogen. The residues were dissolved in 0.3 mL DMSO and their Hy fluorescence was determined with a microplate reader (Safire®, Tecan, Salzburg, AT) at the excitation and emission wavelengths of 530 and 645 nm. The obtained fluorescence intensity was corrected by subtraction of the fluorescence of control samples. The respective Hy concentration was calculated from the calibration curve (1.95−25 ng/mL), which had a regression coefficient higher than 0.999. 2.9. Stability of Hy-loaded MPEG-dihexPLA micelles in blood plasma. Human blood plasma from an anonymous AB donor (Geneva University Hospital, Switzerland) was used for the study of the stability of Hy loaded MPEG-dihexPLA micelles in the presence of plasma. A volume of 187 μL Hy loaded MPEG-dihexPLA micelles was added and mixed with 12 mL plasma for 1, 3, 6 and 24h in the dark at 37°C under orbital shaking to mimic the experimental conditions of the in vivo studies. A control experiment with Hy solution (2 mg/mL in a mixture of ethanol, PEG 400 and water) was also carried out under the same conditions, except that 94 μL of the Hy solution were mixed with 6 mL plasma. After shaking, 4 mL of the plasma/micelles mixture were centrifuged for 5 min at 10 000 rpm (Beckman, AvantiTM, Fullerton, USA) in order to remove non-entrapped drug. One milliliter of the supernatant was dissolved in 2 mL THF under sonication. The resulting organic mixture was afterwards centrifuged for 5 min at 10 000 rpm to remove blood components that have precipitated with the addition of THF. The entire remaining supernatant was collected, dried, and dissolved in 400 μL acetone. The acetone solution was finally centrifuged to remove all other precipitates (5 min, 10 000 rpm), and directly analyzed (with no dilution) by HPLC for the Hy drug content. Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 100 Please note that the used volume of blood plasma was chosen to simulate the in vivo concentrations in the animal model, and was calculated from the following equation (4) for the blood volume and the body weight of the rats 42: Blood volume (mL) = 0.06 × body weight (g) + 0.77 (4) Here, the body weight corresponded to 187 g, the average body weight calculated from the 22 rats studied in the in vivo experiments. 2.10. Statistical analysis Results are expressed as means ± SDs (standard deviations). The significance of betweengroup differences was determined using Student’s t-test. All p-values <0.05 were considered to be statistically significant. 3. Results 3.1. Hypericin/MPEG-hexPLA micelle formulations Hypericin (Hy)-loaded micelle formulations were prepared for in vivo evaluation in the CAM model. Table 1 shows the size characteristics and Hy incorporation of these micelles. MPEGmonohexPLA and MPEG-dihexPLA micelles had number weighted diameters (dn) of 32 nm and of 19 nm and mean hydrodynamic diameters (Zav) of 133 nm and 85 nm, respectively. The high Hy incorporation efficiency that we observed (>70%) yielded a Hy solubility of 0.69 mg/mL for MPEG-monohexPLA micelles and 2.02 mg/mL for MPEG-dihexPLA micelles. These values correspond to a drug-loading of 1.7% and 9.8 % (%w/w), respectively. It is notable that with the MPEG-dihexPLA micelles, half of the amount of excipient was needed to create a formulation with double the Hy concentration in comparison to the less hydrophobic MPEG-monohexPLA. Chapter IV 101 Table 1. Hy loaded MPEG-monohexPLA and MPEG-dihexPLA micelle formulations and characteristics. Polymeric micelles Mn (g/mol) Mn/Mw Copolymer Hy Encapsulation concentration concentration Efficiency Drug loading (%w/w) Zav (nm) P.I. dn [%]dn MPEG-monohexPLA 5600 1.15 40 mg/mL 0.69 mg/mL 69.6% 1.7 133 0.1 32 100 MPEG-dihexPLA 5351 1.12 20 mg/mL 2.02 mg/mL 80.3% 9.8 85 0.3 19 100 P.I.: Polydispersity index Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 102 3.2. Chick embryo chorioallantoic membrane (CAM) study Four days after topical inoculation of NuTu-19 cells onto the CAM (which occurred on EDD8), visible tumor nodules with extensive neovascularization were observed at the membrane surface. Hy-loaded micelles and the Hy solution were injected at the same dose (2 mg/kg). The fluorescence intensity of the accumulated Hy in the nodules was observed and photographed 1 min, 1 h, 3 h and 6 h following the administration. The corresponding fluorescence images showed the increase in fluorescence intensity over time within the ovarian tumor nodules as compared to the non-tumoral surrounding tissues (Fig. 1). Compared to Hy in solution (upper row), both Hy-loaded MPEG-hexPLA micelle formulations (lower row) led to a much higher fluorescent signal in the nodules. Figure 1. Fluorescence images of nodules after injection of Hy in solution and Hy loaded MPEGdihexPLA micelle formulation at a dose of 2 mg/kg in chick embryo (CAM model). Chapter IV 103 The higher accumulation of Hy within the tumors was confirmed by the plot of the relative tumor fluorescence intensity versus time (Fig. 2). No statistically significant differences were found between the two micelle formulations. However, the micelle formulations led to a significantly higher fluorescence as compared to the drug in solution at all time points (p<0.05). Six hours after injection, the fluorescence intensity in the tumor nodules that was achieved by the MPEG-hexPLA micelle formulations was 13 to 17 times higher than that achieved by the Hy in ethanol solution. 15 Corrected Fluorescence intensity (AU) * * * 10 * * * 5 * * 0 0 1 2 3 Time (h) 4 5 6 Figure 2. Relative tumor fluorescence intensity over time after i.v. administration of Hy loaded MPEG-monohexPLA micelles (▲), MPEG-dihexPLA micelles (■) and Hy solution () at a dose of 2 mg/kg in chick embryos (mean ±S.D., n=5). * Significantly different from chick embryos injected with Hy solution (Student’s t test, p<0.05). Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 104 3.3. Biodistribution of Hy-loaded MPEG-dihexPLA micelles The biodistribution of Hy-loaded micelles was investigated following injection into Fisher rats at a dose of 2 mg/kg body weight. In this experiment, the Hy-MPEG-dihexPLA formulation with the higher drug-loading efficiency (at a concentration of 1 mg/mL) was chosen. Hy in a clear ethanol-PEG 400-water solution was used for comparison. No sign of precipitation was visually observed at the site of injection for both tested solutions. The plasma Hy concentration profiles observed for the Hy solution and Hy-loaded micelles are shown in Fig. 3. The results for the Hy solution concerning the 3, 6 and 24h time points were taken from the published work of ZeisserLabouèbe et al. (who performed their studies on Fisher 344 rats) 22. At all tested time points, the Hy plasma levels that were obtained were significantly higher when Hy was incorporated into micelles as compared to Hy in ethanol solution. At 1 h after injection, Hy loaded in micelles showed a plasma concentration that was 6 times higher concentration than that was achieved when the rats were injected with ethanol solution. After 3 h, the plasma concentration achieved by the Hy-loaded micelles was 4 times higher than was achieved with Hy in ethanol solution and after 6 h, it was 2 times higher. After 24 h, the plasma concentration achieved by the Hy-loaded micelles was 3 times higher than for the drug in solution, indicating that the circulation time of Hy was significantly prolonged when it was incorporated in the micelle carriers. 8.00 Hy concentration (µg/ml) * 6.00 4.00 * 2.00 * * 0.00 1 3 Time (h)6 24 Figure 3. Plasma concentration profile of Hy after i.v. injection in rats of Hy solution (white bars) and of Hy loaded MPEG-dihexPLA micelles (dashed bars) at a dose of 2 mg/kg. (Mean ±SD, n=3-4). * Significantly different from rats injected with Hy solution (Student’s t test, p<0.05). Results of Hy in solution after 3, 6 and 24h were taken from reference 22. Chapter IV 105 The distribution of both micelle formulations that were tested in rat livers, spleens and lungs is illustrated in Fig. 4. The uptake of Hy following administration of Hy in solution was higher in the spleen than in the lungs and liver. Hy delivered in micelles was found in higher concentrations in the spleen and lungs than in the liver. A total of 24 h after administration, Hy that had been loaded in micelles was eliminated by these organs at the same rate as the drug solution. Both formulation profiles were similar for the liver and led to a final Hy concentration of 0.60 and 0.43 µg/g tissue when Hy was delivered in solution and in micelles, respectively. Hy-loaded micelles exhibited a slower elimination from the spleen than Hy in ethanol solution at 1 h, 3 h and 6 h after injection, but showed the same concentration after 24 h. In the lungs, Hy loaded in micelles showed a similar profile as in the spleen, except that after 24 h, Hy that had been administrated in micelle formulations was more rapidly eliminated than the drug solution. Hy concentration (µg/g) 25.00 20.00 15.00 10.00 5.00 0.00 1h 3h 6h 24h 1h Liver 3h 6h 24h 1h Spleen 3h 6h 24h Lung Figure 4. Biodistribution of Hy in liver, spleen and lung over time after injection of Hy solution (white bars) and of Hy loaded MPEG-dihexPLA micelles (dashed bars) at a dose of 2 mg/kg in rats (Mean ±SD, n=3-4). Results of Hy in solution after 3, 6 and 24h were taken from reference 22. Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 106 In addition to examining the tissue distribution of Hy, we also determined the accumulation of Hy-loaded MPEG-dihexPLA micelle in the tumors and surrounding muscle tissue. For both tested formulations, the Hy concentration in the surrounding muscles was very low as compared to the concentration that was observed in the other investigated organs. However, a slightly higher Hy accumulation in muscles was observed for Hy-loaded micelles as compared to Hy in solution. In the tumors, a significant accumulation of Hy-loaded micelles was found for the first 6 h after injection, with a maximum level detected at 3 h (Fig. 5). After 24 h, the Hy accumulation decreased to a concentration that was similar to the concentration that was obtained with the drug in solution. 1.25 * * Hy concentration (µg/g) 1 0.75 * * 0.5 * * * 0.25 0 0 6 12 Time (h) 18 24 Figure 5. Biodistribution of Hy in tumors (open symbols) and the surrounding muscle (plain symbols) over time after injection of Hy solution (,) and of Hy loaded MPEG-dihexPLA micelles (,■) at a dose of 2 mg/kg in rats (Mean ±SD, n=3-4). * Significantly different from rats injected with Hy solution (Student’s t test, p<0.05). Results of Hy in solution after 3, 6 and 24h were taken from reference 22. Chapter IV 107 For the application of Hy-loaded micelles for tumor imaging to be successful, a high contrast between healthy tissues (muscle) and diseased tissues (tumor) is essential. The visual distinction between these tissues is possible by using endoscopy with blue light. Under white light, no difference between muscle tissues and tumor nodules could be observed (Fig. 6). However, under blue light, the red fluorescence of Hy accumulated in the tumor nodules became clearly visible, while the absence or near absence of red fluorescence in the healthy muscle indicated the absence or very low accumulation of Hy. Figure 6. Pictures of ovarian metastasis in the peritoneal cavity of rats under white light and blue light after injection of Hy loaded MPEG-dihexPLA micelles. Below the given contrast was obtained between diseased tissues and surrounding healthy muscle. Visually, the maximum contrast between healthy and diseased tissues was observed 3 h after micelle injection. To achieve more quantitative measurements, the tumor-to-muscle ratio was calculated from the extracted and analyzed Hy concentrations. This analysis also found that the highest contrast between tumor and muscle tissue was present 3 h after injection of Hyloaded micelles (Fig. 7). It is also worth noting that this time was also the point at which maximum concentration of the fluorescent marker could be detected in the tumor tissue (Fig. 5). Hy Ratio Tumour/Muscle Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 108 7.0 6.0 5.0 4.0 3.0 2.0 1.0 0.0 1 3 6 24 Time (h) Figure 7. Tumor to muscle contrast over time after injection of Hy solution (white bars) and of Hy loaded MPEG-dihexPLA micelles (dashed bars) at a dose of 2 mg/kg in rats. Results of Hy in solution after 3, 6 and 24h were taken from reference 22. 3.4. Stability of Hy-loaded MPEG-dihexPLA micelles in blood plasma The stability of Hy loaded micelles has been studied in blood plasma and has been performed under similar conditions as in the in vivo experiments, i.e. with the same dilution factor and the same dose. Hy loaded micelles retained more than 80% of the introduced Hy quantity in plasma at all the tested time points (1, 3, 6 and 24h), whereas for the control of Hy in ethanol solution only 10-15% of Hy was found in the plasma (Fig. 8). Figure 8. Percentage of Hy recuperated from human blood plasma when added as an ethanol solution () and when added within MPEG-dihexPLA micelles (■), respectively. Chapter IV 109 4. Discussion Over the last decades, nanosized polymeric micelles have become a topic of increasing interest in the field of drug delivery. Because of their specific core-shell structure, they can carry potent hydrophobic drugs within their core, while their hydrophilic shell facilitates water solubility. Their ability to accumulate at tumor sites via the EPR effect envisions localized cancer treatments 43. In the present study, the objective was to investigate novel MPEG-hexPLA micelles for their potential use in ovarian cancer diagnosis. We examined their ability to achieve efficient drug loading, a long circulation time and selective accumulation in tumor tissue in a series of in vivo experiments. As demonstrated, the relatively water insoluble fluorescent marker Hypericin was successfully incorporated into MPEG-hexPLA micelles, reaching an aqueous solubility of ~2 mg/mL, which represents a high increase in its water solubility as compared to the drug alone. In addition, incorporating Hy in MPEG-hexPLA micelles increased its circulation time in the bloodstream, which is a major advantage for passive tumor targeting. Even 24 h after injection into ovarian tumor-bearing rats, Hy loaded MPEG-dihexPLA micelles were still present in the plasma. The distribution of Hy has been proven in vitro to be due to a stable incorporation of Hy into micelles for 24h and not from a gradually release of the drug from the micelles. The loss of Hy when added within micelles in human blood plasma (<20%) observed after 1h can be explained either by the multiple steps needed for the extraction of the drug, by a possible equilibrium needed to be reached during the first hour between the micelles and the plasma components, or by both phenomena. The observed low Hy plasma concentration even after 1h can be attributed to a probably very fast decrease of the Hy loaded micelle concentration in the blood between the injection and the first studying time point. Indeed, a study with comparable MPEG-PLA micelles (molecular weight around 5000g/mol with MPEG 2000g/mol) and with paclitaxel as the incorporated drug demonstrated a biphasic nature of the plasma profile with an initial fast distribution phase completed within the first hour 44. The presence of inert hydrophilic PEG on the nanocarrier surface provided good steric hindrance, limiting blood serum protein binding and as expected increasing the carrier’s circulation time 45;46 . In addition, the increased hydrophobicity of the hexPLA core, in comparison to a PLA or PCL micelle core, allowed the novel MPEG-dihexPLA micelles to have a lower critical micelle concentration 47 , thus a higher Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 110 stability upon dilution, and a higher stability in PBS modeling intravenous conditions (pH 7.4 at 37°C) 48, consequently increasing the life time of the Hy loaded in micelles in the blood stream. For cancer diagnosis, increased accumulation of the fluorescent marker at the tumor sites enables better tumor detection. In both in vivo studies, Hy loaded micelles primarily accumulated in ovarian tumors and thereby meeting this requirement. The accumulation of free Hy is often associated with aggregation, and a possible loss of fluorescence intensity. Here, this quenching phenomenon is not likely to occur as Hy released from the micelles at the site of interest would not aggregate due to the presence of proteins like LDL, HDL or albumin49;50 to which Hy rapidly binds and does not influence the fluorescence properties. In the CAM studies, accumulated Hy-loaded micelles showed a much higher intensity of fluorescence as reported for Hy-loaded nanoparticles (Nps) 24 . A 5-fold increased accumulation of Hy in tumors was obtained with Hy-loaded MPEG-hexPLA micelles as compared to Hy-loaded Nps 3 h after injection. In ovarian tumor-bearing rats, the biodistribution study confirmed that Hy micelles rapidly accumulated in tumor tissues at high concentrations. Hy loaded in MPEG-dihexPLA micelles reached a maximum concentration and a maximum tumor-to-muscle contrast 3 h after injection. In contrast Hy-Nps showed its highest accumulation 24 h after injection 22. The differences that were observed in the tumor accumulation profiles between the two animal models (rat versus CAM) can be attributed to the absence of an elimination process in the CAM model 24 . In the rat experiments, the differences between the two Hy nanocarriers (i.e., micelles and Nps) can be explained by differences in the size of the two carrier systems. It has been reported that the accumulation effect is primarily influenced by the size of the carrier 51 . Because of their small size, nanocarriers can easily pass through the gaps in leaky endothelial walls and are efficiently taken up by tumor cells. Nagayasu et al. reported that for liposomes, a diameter of <100 nm seemed to be most suitable in order to achieve a long half-life in the blood circulation, tumorspecific drug accumulation and in vivo drug release 52. Moreover, another biological process of micelle uptake in tumor tissues has recently been described by Kawaguchi et al. 53. They discovered by histological tracking that labeled PEG-poly(aspartate) copolymer micelles accumulated more rapidly (after 1 h) via the blood vessels at the tumor periphery than via tumor blood vessels due to the EPR effect. The Hy-loaded MPEG-hexPLA micelles (Zav= 90 nm and dn< 30 nm) accumulated faster and more efficiently in tumors 3 h after injection than Hy-PLA Nps (Zav= 200-300 nm), which needed at least 24 h to reach their highest Chapter IV 111 accumulation 22. For the MPEG-hexPLA formulations, it was found that the disappearance of Hy from the bloodstream coincided with Hy uptake in tissues and, more specifically, tumor uptake. The decrease in plasma Hy concentration 6 h after injection demonstrates the elimination of Hy and Hy-loaded MPEG-hexPLA micelles from the bloodstream. Similarly, Burt et al. showed that MPEG-PLA copolymers were rapidly eliminated from the body through the urine 44. The degradation of the copolymer in acidic conditions, which are present in tumor tissues, is one plausible explanation 54;55 . The emerging lower molecular weight components are also more rapidly filtered and eliminated by the kidneys 29;56. This suggests a low risk of chronic accumulation of a micelle formulation in the human body if it were to be administered in a clinical setting. With regard to the need for an early detection of ovarian cancer and any metastases that are present that was discussed above, it can be concluded that Hy-loaded MPEG-hexPLA formulations lead to a rapid (3 h) accumulation of the fluorescent marker in rat tumors with an excellent contrast to surrounding healthy tissues. When one considers the procedures that are currently practiced in the clinical setting, it becomes apparent that laparoscopic examination using blue light following intravenous administration of Hy micelle formulations could increase the visualization of tumors and micrometastases in the pelvis. This would allow for a efficient, early diagnosis of ovarian cancer. It would also lead to more accurate staging. Both of these factors could result in improved patient prognosis due to earlier identification of the appropriate treatment regimen for each patient. Follow-up laparoscopic examinations should be possible with the same efficacy, since the very small size MPEG-hexPLA micelles should not induce the accelerated blood clearance (ABC) phenomenon that is often observed with pegylated nanoassemblies >50 nm following a second injection 57;58 . In addition to cancer diagnosis, laparoscopy under blue light could be performed during surgery, facilitating complete removal of diseased tissues. For both of these possible clinical applications, the fast and efficient micelle accumulation that can be achieved with these formulations is a great advantage, since it can reduce the time needed for a routine patient check-up or preparation prior to a necessary surgery. The proof of concept that we achieved in the CAM and rat model showed promising results. In view of their possible clinical applications, MPEG/dihexPLA molecular weights and ratios could be further tailored to modulate and optimize their circulation time, loading capacity and diagnostic properties in future studies. Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 112 Acknowledgments The authors would like to thank the Swiss National Science Foundation (SNF) for its financial support (SNF 200020-103752) References (1) Hennessy, B. T.; Coleman, R. L.; Markman, M., Ovarian cancer, Lancet 2009, 374, 1371-1382. (2) Jemal, A.; Siegel, R.; Ward, E.; Hao, Y.; Thun, M. J., Cancer Statistics, 2009, CA Cancer J.Clin. 2009, 59, 225-249. (3) Collinet, P.; Sabban, F.; Farine, M. O.; Villet, R.; Vinatier, D.; Mordon, S., Laparoscopic Photodynamic Diagnosis of Ovarian Cancer Peritoneal Micro Metastasis: An Experimental Study, Photochem.Photobiol. 2007, 83, 647-651. (4) Löning, M.; Diddens, H.; Diedrich, K.; Hüttmann, G., Laparoscopic fluorescence detection of ovarian carcinoma metastases using 5-aminolevulinic acid-induced protoporphyrin IX, Cancer 2004, 100, 1650-1656. (5) Song, X.; Ye, Z.; Zhou, S.; Yang, W.; Zhang, X.; Liu, J.; Ma, Y., The application of 5aminolevulinic acid-induced fluorescence for cystoscopic diagnosis and treatment of bladder carcinoma, Photodiagn.Photodyn.Ther. 2007, 4, 39-43. (6) Hillemanns, P.; Soergel, P.; Löning, M., Fluorescence diagnosis and photodynamic therapy for lower genital tract diseases - A review, Med.Laser Appl. 2009, 24, 10-17. (7) Allison, R. R.; Sibata, C. H., Oncologic photodynamic therapy photosensitizers: A clinical review, Photodiagn.Photodyn.Ther. 2010, 7, 61-75. (8) Moghissi, K.; Dixon, K.; Stringer, M.; Thorpe, J. A. C., Photofrin PDT for early stage oesophageal cancer: Long term results in 40 patients and literature review, Photodiagn.Photodyn.Ther. 2009, 6, 159-166. (9) Lopez, R. F. V.; Lange, N.; Guy, R.; Bentley, M. V. L. B., Photodynamic therapy of skin cancer: controlled drug delivery of 5-ALA and its esters, Adv.Drug Delivery Rev. 2004, 56, 77-94. (10) Lorenz; K.J; Maier, H., Squamous cell carcinoma of the head and neck. Photodynamic therapy with Foscan®, HNO 2008, 56, 402-409. Chapter IV 113 (11) Ascencio, M.; Delemer, M.; Farine, M. O.; Jouve, E.; Collinet, P.; Mordon, S., Evaluation of ALA-PDT of ovarian cancer in the Fisher 344 rat tumor model, Photodiagn.Photodyn.Ther. 2007, 4, 254-260. (12) Pytel, A.; Schmeller, N., New aspect of photodynamic diagnosis of bladder tumors: fluorescence cytology, Urology 2002, 59, 216-219. (13) Saw, C. L. L.; Olivo, M.; Soo, K. C.; Heng, P. W. S., Delivery of hypericin for photodynamic applications, Cancer Lett. 2006, 241, 23-30. (14) Liu, C. D.; Kwan, D.; Saxton, R. E.; McFadden, D. W., Hypericin and Photodynamic Therapy Decreases Human Pancreatic Cancer in Vitro and in Vivo, J.Surg.Res. 2000, 93, 137-143. (15) Colasanti, A.; Kisslinger, A.; Liuzzi, R.; Quarto, M.; Riccio, P.; Roberti, G.; Tramontano, D.; Villani, F., Hypericin photosensitization of tumor and metastatic cell lines of human prostate, J.Photochem.Photobiol.,B 2000, 54, 103-107. (16) Uzdensky, A.; Iani, V.; Ma, L. W.; Moan, J., On hypericin application in fluorescence diagnosis and cancer treatment: Pharmacokinetics and photosensitizing efficiency in nude mice bearing WiDr carcinoma, Med.Laser Appl. 2006, 21, 271-276. (17) Thong, P. S. P.; Olivo, M.; Chin, W. W. L.; Bhuvaneswari, R.; Mancer, K.; Soo, K. C., Clinical application of fluorescence endoscopic imaging using hypericin for the diagnosis of human oral cavity lesions, Br.J.Cancer 2009, 101, 1580-1584. (18) Kubin, A.; Meissner, P.; Wierrani, F.; Burner, U.; Bodenteich, A.; Pytel, A.; Schmeller, N., Fluorescence Diagnosis of Bladder Cancer with New Water Soluble Hypericin Bound to Polyvinylpyrrolidone: PVP-Hypericin, Photochem.Photobiol. 2008, 84, 15601563. (19) Sim, H. G.; Lau, W. K.; Olivio, M.; Tan, P. H.; Cheng, C. W., Is photodynamic diagnosis using hypericin better than white-light cystoscopy for detecting superficial bladder carcinoma?, BJU International 2010, 95, 1215-1218. (20) D'Hallewin, M. A.; Bezdetnaya, L.; Guillemin, F., Fluorescence Detection of Bladder Cancer: A Review, Eur.Urol. 2002, 42, 417-425. (21) D'Hallewin, M. A.; De Witte, P. A.; Waelkens, E.; Merlevede, W.; Baert, L., Fluorescence detection of flat bladder carcinoma in situ after intravesical instillation of Hypericin, J.Urol. 2000, 164, 349-351. (22) Zeisser-Labouèbe, M.; Delie, F.; Gurny, R.; Lange, N., Benefits of nanoencapsulation for the hypercin-mediated photodetection of ovarian micrometastases, Eur.J.Pharm.Biopharm. 2009, 71, 207-213. (23) From the website of the United States http://chem.sis.nlm.nih.gov/chemidplus/. 2010. national library of medicine, Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 114 (24) Zeisser-Labouèbe, M.; Delie, F.; Gurny, R.; Lange, N., Screening of nanoparticulate delivery systems for the photodetection of cancer in a simple and cost-effective model, Nanomedicine 2009, 4, 135-143. (25) Singh, R.; Lillard, J., Nanoparticle-based targeted drug delivery, Exp.Molec.Pathol. 2009, 86, 215-223. (26) Park, J. H.; Lee, S.; Kim, J. H.; Park, K.; Kim, K.; Kwon, I. C., Polymeric nanomedicine for cancer therapy, Prog.Polym.Sci. 2008, 33, 113-137. (27) Alexis, F.; Rhee, J. W.; Richie, J. P.; Radovic-Moreno, A. F.; Langer, R.; Farokhzad, O. C., New frontiers in nanotechnology for cancer treatment, Urol.Oncol. 2001, 26, 74-85. (28) Torchilin, V. P., Structure and design of polymeric surfactant-based drug delivery systems, J.Control.Release 2001, 73, 137-172. (29) Nishiyama, N.; Morimoto, Y.; Jang, W. D.; Kataoka, K., Design and development of dendrimer photosensitizer-incorporated polymeric micelles for enhanced photodynamic therapy, Adv.Drug Delivery Rev. 2009, 61, 327-338. (30) Rijcken, C. J. F.; Hofman, J. W.; van Zeeland, F.; Hennink, W. E.; van Nostrum, C. F., Photosensitiser-loaded biodegradable polymeric micelles: Preparation, characterisation and in vitro PDT efficacy, J.Control.Release 2007, 124, 144-153. (31) Chowdhary, R. K.; Chansarkar, N.; Sharif, I.; Hioka, N.; Dolphin, D., Formulation of Benzoporphyrin Derivatives in Pluronics-Â, Photochemistry and Photobiology 2009, 77, 299-303. (32) Sezgin, Z.; Yuksel, N.; Baykara, T., Investigation of pluronic and PEG-PE micelles as carriers of meso-tetraphenyl porphine for oral administration, Int.J.Pharm. 2007, 332, 161-167. (33) Roby, A.; Erdogan, S.; Torchilin, V. P., Solubilization of poorly soluble PDT agent, meso-tetraphenylporphin, in plain or immunotargeted PEG-PE micelles results in dramatically improved cancer cell killing in vitro, Eur.J.Pharm.Biopharm. 2006, 62, 235-240. (34) Ma, L. L.; Jie, P.; Venkatraman, S. S., Block Copolymer lsquoStealthrsquo Nanoparticles for Chemotherapy: Interactions with Blood Cells In Vitro, Adv.Funct.Mat. 2008, 18, 716-725. (35) Mondon, K.; Gurny, R.; Möller, M., Colloidal Drug Delivery Systems Recent Advances With Polymeric Micelles, Chimia 2008, 62, 832-840. (36) Trimaille, T.; Gurny, R.; Möller, M., Synthesis and ring-opening polymerization of new monoalkyl-substituted lactides, J.Polym.Sci.Part A: Polym.Chem. 2004, 42, 4379-4391. (37) Mondon, K.; Zeisser-Labouèbe, M.; Gurny, R.; Möller, M., Novel Cyclosporin A formulations using MPEG-hexyl substituted polylactide micelles: a suitability study, Eur.J.Pharm.Biopharm. 2010, 77, 56-65. Chapter IV 115 (38) Zeisser-Labouèbe, M.; Lange, N.; Gurny, R.; Delie, F., Hypericin-loaded nanoparticles for the photodynamic treatment of ovarian cancer, Int.J.Pharm. 2006, 326, 174-181. (39) Rose, G. S.; Tocco, L. M.; Granger, G. A.; DiSaia, P. J.; Hamilton, C. T.; Santin, A. D.; Hiserodt, J. C., Development and characterization of a clinically useful animal model of epithelial ovarian cancer in the Fischer 344 rat, Am.J.Obstet.Gynecol. 1996, 175, 593599. (40) Lange, N.; Ballini, J. P.; Wagnieres, G.; van den Bergh, H., A New Drug-Screening Procedure for Photosensitizing Agents Used in Photodynamic Therapy for CNV, Invest.Ophthalmol.Vis.Sci. 2001, 42, 38-46. (41) Vargas, A.; Pegaz, B.; Debefve, E.; Konan-Kouakou, Y.; Lange, N.; Ballini, J. P.; van den Bergh, H.; Gurny, R.; Delie, F., Improved photodynamic activity of porphyrin loaded into nanoparticles: an in vivo evaluation using chick embryos, Int.J.Pharm. 2004, 286, 131-145. (42) Lee, H. B.; Blaufox, M. D., Blood Volume in the Rat, J Nucl Med 1985, 26, 72-76. (43) Torchilin, V., Tumor delivery of macromolecular drugs based on the EPR effect, Adv.Drug Delivery Rev. 2010, doi:10.1016/j.addr.2010.03.011. (44) Burt, H. M.; Zhang, X. C.; Toleikis, P.; Embree, L.; Hunter, W. L., Development of copolymers of poly(D,L-lactide) and methoxypolyethylene glycol as micellar carriers of paclitaxel, Colloid.Surface B 1999, 16, 161-171. (45) Owens III, D. E.; Peppas, N. A., Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles, Int.J.Pharm. 2006, 307, 93-102. (46) Li, S. D.; Huang, L., Stealth nanoparticles: High density but sheddable PEG is a key for tumor targeting, J.Control.Release 2010, 145, 178-181. (47) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. (48) Nottelet, B.; Di Tommaso, C.; Mondon, K.; Gurny, R.; Möller, M., Fully biodegradable polymeric micelles based on hydrophobic- and hydrophilic-functionalized poly(lactide) block copolymers, J.Polym.Sci.A Polym.Chem. 2010, 48, 3244-3254. (49) Kohler, M.; Gafert, J.; Friedrich, J.; Falk, H.; Meyer, J., Hole-Burning Spectroscopy of Proteins in External Fields: Human Serum Albumin Complexed with the Hypericinate Ion, J.Phys.Chem. 1996, 100, 8567-8572. (50) Crnolatac, I.; Huygens, A.; Agostinis, P.; Kamuhabwa, A. R.; Van Aerschot, A.; De Witte, P. A. M., In vitro accumulation and permeation of hypericin and lipophilic analogues in 2-D and 3-D cellular systems, Int.J.Oncol. 2007, 30, 319-324. (51) Torchilin, V. P., PEG-based micelles as carriers of contrast agents for different imaging modalities, Adv.Drug Deliv.Rev. 2002, 54, 235-252. Hy/ MPEG-hexPLA Micelle Formulations for Ovarian Cancer Diagnostics 116 (52) Nagayasu, A.; Uchiyama, K.; Kiwada, H., The size of liposomes: a factor which affects their targeting efficiency to tumors and therapeutic activity of liposomal antitumor drugs, Adv.Drug Delivery Rev. 1999, 40, 75-87. (53) Kawaguchi, T.; Honda, T.; Nishihara, M.; Yamamoto, T.; Yokoyama, M., Histological study on side effects and tumor targeting of a block copolymer micelle on rats, J.Control.Release 2009, 136, 240-246. (54) Tannock, I. F.; Rotin, D., Acid pH in Tumors and Its Potential for Therapeutic Exploitation, Cancer Res 1989, 49, 4373-4384. (55) Engin, K.; Leeper, D. B.; Cater, J. R.; Thistlethwaite, A. J.; Tupchong, L.; McFarlane, J. D., Extracellular pH distribution in human tumours, Int.J.Hyperther 1995, 11, 211-216. (56) Yamamoto, Y.; Nagasaki, Y.; Kato, Y.; Sugiyama, Y.; Kataoka, K., Long-circulating poly(ethylene glycol)-poly(-lactide) block copolymer micelles with modulated surface charge, J.Control.Release 2001, 77, 27-38. (57) Ishida, T.; Kiwada, H., Accelerated blood clearance (ABC) phenomenon upon repeated injection of PEGylated liposomes, Int.J.Pharm. 2008, 354, 56-62. (58) Koide, H.; Asai, T.; Hatanaka, K.; Urakami, T.; Ishii, T.; Kenjo, E.; Nishihara, M.; Yokoyama, M.; Ishida, T.; Kiwada, H.; Oku, N., Particle size-dependent triggering of accelerated blood clearance phenomenon, Int.J.Pharm. 2008, 362, 197-200. Next to intravenous applications of the aqueous MPEG-hexPLA formulation the versatility of the novel drug delivery systems was investigated for the topical delivery of azole antifungals. CHAPTER V Chapter V 117 Novel Micelle Formulations to Increase Cutaneous Bioavailability of Azole Antifungals Y.G. Bachhava, K. Mondona, Y.N. Kalia, R. Gurny, and M. Möller School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30 quai Ernest-Ansermet, CH-1211 Geneva 4, Switzerland. a both authors have equally contributed to this work Published in: Journal of Controlled Release (2011) Efficient topical drug administration for the treatment of superficial fungal infections would deliver the therapeutic agent to the target compartment and reduce the risk of systemic side effects. However, the physicochemical properties of the commonly used azole antifungals make their formulation a considerable challenge. The objective of the present investigation was to develop aqueous micelle solutions of clotrimazole (CLZ), econazole nitrate (ECZ) and fluconazole (FLZ) using novel amphiphilic methoxy-poly(ethylene glycol)-hexyl substituted polylactide (MPEG-hexPLA) block copolymers. The CLZ, ECZ and FLZ formulations were characterized with respect to drug loading and micelle size. The optimal drug formulation was selected for skin transport studies that were performed using full thickness porcine and human skin. Penetration pathways and micellar distribution in the skin were visualized using fluorescein-loaded micelles and confocal laser scanning microscopy. The hydrodynamic diameters of the azole loaded micelles were between 70 and 165 nm and the corresponding number weighted diameters (dn) were 30 to 40 nm. Somewhat surprisingly, the lowest loading efficiency (< 20 %) was observed for CLZ (the most hydrophobic of the three azoles tested); in contrast, under the same conditions, ECZ was incorporated with an efficiency of 98.3% in MPEG-dihexPLA micelles. Based on the characterization data and preliminary transport experiments, ECZ loaded MPEG-dihexPLA micelles (concentration 1.3 mg/mL; dn < 40 nm) were selected for further study. ECZ delivery was compared to that from Pevaryl® cream (1% w/w ECZ), a marketed liposomal formulation for topical application. ECZ deposition in porcine skin following 6 h application using the MPEG-dihexPLA micelles was ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 118 >13-fold higher than that from Pevaryl® cream (22.8 ± 3.8 and 1.7 ± 0.6 µg/cm2, respectively). A significant enhancement was also observed with human skin; the amounts of ECZ deposited were 11.3 ± 1.6 and 1.5 ± 0.4 µg/cm2, respectively (i.e., a 7.5-fold improvement in delivery). Confocal laser scanning microscopy images supported the hypothesis that the higher delivery observed in porcine skin was due to a larger contribution of the follicular penetration pathway. In conclusion, the significant increase in ECZ skin deposition achieved using the MPEG-dihexPLA micelles demonstrates their ability to improve cutaneous drug bioavailability; this may translate into improved clinical efficacy in vivo. Moreover, these micelle systems may also enable targeting of the hair follicle and this will be investigated in future studies. Keywords: Antifungal; azole; polymeric micelle; skin deposition; substituted polylactides, follicular delivery. Chapter V 119 1. Introduction The incidence of mycoses especially superficial fungal infections is increasing and according to a recent report more than 25% of the world’s population is affected 1;2; disease progression is more rapid and severity increased in patients with compromised immune function 3. Host immunity can be impaired during infancy, in old age, by pregnancy, by disease, e.g. diabetes mellitus, or through the administration of antibiotics and glucocorticoids 4. Azole antifungals such as clotrimazole (CLZ), econazole nitrate (ECZ) and fluconazole (FLZ) are the first line treatments for various fungal infections 5. Topical therapy is desirable since, in addition to targeting the site of infection, it reduces the risk of systemic side effects. In general, azole antifungals tend to be highly lipophilic (although there are exceptions (e.g., FLZ)) and they can readily partition into the lipid-rich intracellular space in the stratum corneum; the challenge is to develop a simple stable formulation that facilitates drug release into the skin 6. Given the desirable properties of aqueous formulations and the lipophilic character and poor water solubility of azoles, it was decided to investigate polymeric micelles as a drug carrier system. Due to their stability, size and ability to incorporate significant amounts of hydrophobic drugs in their core, these systems seem to be well-suited for use with azole antifungals. In previous studies, micelle formulations using two novel amphiphilic methoxypoly(ethylene glycol)- hexyl-substituted poly(lactides) (MPEG-hexPLA) block copolymers, mono- and di-hexyl-substituted (MPEG-monohexPLA and –dihexPLA, respectively) demonstrated their ability to incorporate several poorly water soluble drugs with high loading efficiencies antifungals, 7-9 . The present study investigated the micelle formulations of three azole clotrimazole, econazole nitrate and fluconazole, possessing different physicochemical properties (Table 1). The specific objectives were (i) to develop and to characterize micelle formulations for CLZ, ECZ and FLZ using the novel excipients – MPEG-monohexPLA and -dihexPLA – and to compare them with formulations using standard MPEG-polylactide (MPEG-PLA), (ii) to select the best drug candidate (as determined by incorporation efficiency and micelle properties) and to optimize the formulation for skin deposition, (iii) to quantify drug deposition in full thickness porcine and human skin and to compare delivery to that from a ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 120 commercial formulation and (iv) to visualize micellar transport pathways using fluoresceinloaded micelles and confocal laser scanning microscopy. Table 1. Physico-chemical characteristics of the three antifungal agents MWa (g/mol) log Po/w b Aqueous solubility (g/L) pKa Clotrimazole (CLZ) 344.84 5.9 10 0.030 11 5.83 12 Fluconazole (FLZ) 306.27 0.4 c 0.001 c - Econazole nitrate (ECZ) 444.70 5.2 d10 a Molecular weight b Experimental partition coefficient between octanol and water c Data taken from http://www.drugbank.ca/drugs d Data taken from econazole e Determined experimentally by the shake-flask method (24h at 25°C) 0.800 e 6.65 d 10 2. Materials and Methods 2.1 Materials Clotrimazole (CLZ), econazole nitrate (ECZ), fluconazole (FLZ), dipotassium hydrogen phosphate, monobasic ammonium phosphate, acetone, and fluorescein acid were purchased from Sigma Aldrich (Buchs, Switzerland). Methanol and acetonitrile (Chromasolv HPLC grade) and nylon membrane filters (0.22 μm) were purchased from VWR (Nyon, Switzerland). Pevaryl® cream (1% w/w ECZ ) was purchased from Janssen-Cilag (7HB5P01); it contains PEG-6 stearate, glycol stearate and PEG-32 stearate (Téfose 63), liquid parafin, a polyoxyethylated kernel oil (Labrafil M 1944 CS), benzoic acid (E 210), perfumes (essential oils of rose, jasmine, iris, sandalwood, coriander, ylang-ylang, vétyver, linalol, cinnamic alcohol, cinnamic aldehyde), butylhydroxyanisole (E 320), purified water. Chapter V 121 O H3C O 45 R O O R 1 2 O H m O for m=21 : MPEG2000g/mol-PLA3000g/mol R1, R2 =CH3 2 1 R =C6H13, R =CH3 : MPEG2000g/mol-monohexPLA3000g/mol for m=14 R1, R2 =C6H13 for m=11 : MPEG2000g/mol-dihexPLA3000g/mol Scheme 1. Structure of MPEG-(hex)PLA copolymers The block copolymers, methoxy-poly(ethylene glycol) −di-hexyl-substituted lactide (MPEGdihexPLA), −mono-hexyl-substituted lactide (MPEG-monohexPLA) and −polylactide (MPEG-PLA) were synthesized with MPEG2000g/mol as initiator as described previously 7;9 The structures of these three block copolymers are presented in Scheme 1 and their molecular weights and polydispersity indices are shown in Table 2. Table 2. Characteristics of MPEG-hexPLA and MPEG-PLA copolymers Copolymer MPEG-PLA MWa (g/mol) P.I.b 5050 1.17 MPEG-monohexPLA 5040 1.13 MPEG-dihexPLA (1) 5554 1.13 MPEG-dihexPLA (2) 4881 1.11 a Determined by 1H NMR (Bruker, 300 MHz) b Polydispersity index (P.I.) determined by GPC (1) Copolymer used only for comparison of CLZ, ECZ and FLZ micelle formulations prepared by Method 1 (2) Copolymer used for the optimization of ECZ MPEG-dihexPLA micelle formulations, skin experiments and for the fluorescein micelle formulation ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 122 Please note that in the following text, the term “MPEG-hexPLA” is used to refer collectively to the MPEG-monohexPLA and MPEG-dihexPLA polymers. 2.2 Preparation of drug loaded MPEG-hexPLA and MPEG-PLA micelles micelles by stirring (method 1) The micelles were prepared by a co-solvent evaporation method. Briefly, 6 mg of drug were dissolved in 1 mL acetone and mixed with 1 mL copolymer solution (20 mg/mL) in acetone. The organic solution was added dropwise every 5 s, using a peristaltic pump, into 4 mL of ultra-pure water under continuous stirring. Acetone was then slowly removed by evaporation (with stirring) in a desiccator under vacuum (2 h, 200 mbar). The final micelle concentration was adjusted by adding ultra-pure water in order to reach a copolymer concentration of 5 mg/mL. After overnight equilibration, the solution was centrifuged at 9500 x g for 15 min to remove non-incorporated drug. 2.3 Preparation of ECZ loaded MPEG-dihexPLA micelles by sonication (method 2) Econazole nitrate (ECZ) loaded MPEG-dihexPLA micelle formulations with copolymer concentrations of 5 and of 10 mg/mL were also prepared by the co-solvent evaporation sonication method 7. ECZ and MPEG-dihexPLA copolymer were dissolved in 2 mL acetone and added drop wise every 5 s into 4 mL ultra-pure water under sonication. The remaining acetone was removed by evaporation with a rotavapor at 15 mbar. The concentration of copolymer was adjusted and the non-incorporated drug was removed by centrifugation. 2.4 Preparation of fluorescein loaded MPEG-dihexPLA micelles Fluorescein loaded MPEG-dihexPLA micelles were prepared by method 2. Chapter V 123 2.5 Size characterization of drug loaded MPEG-(hex)PLA micelles Dynamic light scattering measurements (at 25°C at an angle of 90°) using a Zetasizer HS 3000 (Malvern Instruments Ltd; Malvern, UK) were made to determine the hydrodynamic diameter (Zav), the number-weighted diameter (dn) and the percentage of micelles having the number-weighted diameter ([%]dn). All measurements were done in triplicate. 2.6 Morphology determination of drug loaded micelles The morphology of drug loaded micelles was determined by transmission electron microscopy (TEM) (EM 410, Philips, 60kV) using the negative staining method. Briefly, 30 µL of the micellar solution were dropped onto an ionised carbon-coated copper grid (0.3 torr, 400 V for 20 s). The grid was then deposited for 1 s onto a 100 µL drop of uranyl acetate solution (400 µL of a saturated uranyl acetate solution dissolved in 600 µL of distilled water) and afterwards onto a second 100 µL drop for 30 s. Excess of the staining solution was removed and the grid was dried at room temperature prior to the measurement. 2.7 Measurement of drug content in micelles Drug loading was quantified by HPLC analysis. To ensure complete micelle destruction and release of incorporated drug, a 1:10 dilution in acetonitrile was made for micelles with a copolymer concentration of 5 mg/mL and a 1:50 dilution for those with a copolymer concentration of 10 mg/mL. The azole content was quantified using a Lichrospher® RP-18 column (124 mm × 4 mm) thermostated at 35°C. The mobile phases comprised (i) for CLZ, a 75:25 mixture of methanol/0.05M potassium biphosphate buffer , (ii) for ECZ, a 60:40 mixture of acetonitrile/0.05 M potassium biphosphate buffer, and (iii) for FLZ, a 50:50 mixture of methanol/0.1M monobasic ammonium phosphate buffer. All analyses were performed using a flow rate of 1.5 mL/min and a detection wavelength of 214 nm. All samples were measured in triplicate. Sharp peaks for ECZ, CLZ and FLZ were obtained at 4.4, 5.0 and 4.6 min, respectively. The HPLC methods were linear over the concentration range of 0.5-100 µg/mL. The limits of detection for ECZ, CLZ and FLZ were found to be 0.15, 0.43 and 0.25 µg/mL, respectively. The corresponding limits of quantification were ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 0.45, 1.32 and 0.75 µg/mL for ECZ, CLZ and FLZ, respectively. 124 The incorporation efficiency and drug content were calculated using equation (1) and (2), respectively: Incorporation efficiency (%) = Drug content mass of drug incorporated in micelles (g) × 100 mass of drug introduced (g) (1) mass of drug incorporated in micelles (mg) mass of copolymer used (g) (2) (mg drug /g copolymer ) = 2.8 Skin preparation Full thickness porcine and human skin was used to perform skin transport experiments. Porcine ears were supplied by a local abattoir (CARRE; Rolle, Switzerland) shortly after sacrifice. After cleaning under cold running water, the whole skin was removed carefully from the outer region of the ear and separated from the underlying cartilage with a scalpel. The skin samples were wrapped in ParafilmTM and maintained at −20°C and used within 2 months of harvesting. Human skin samples were obtained shortly after surgery from the Department of Plastic, Aesthetic and Reconstructive Surgery, Geneva University Hospital (Geneva, Switzerland), fatty tissue was removed and the skin was wrapped in Parafilm™ before storage at −20°C for a maximum period of 3 days. The donation was approved by the Central Committee for Ethics in Research (CER: 08-150 (NAC08-051); Geneva University Hospital). 2.9 Evaluation of ECZ deposition in the skin Porcine skin Full thickness porcine skin samples were equilibrated in 0.9% NaCl solution for 30 min and then mounted on standard Franz diffusion cells (area = 2.0 ± 0.1 cm2); silicone grease was applied to ensure a watertight seal. The receptor compartment (volume 10 mL) was filled with phosphate buffered saline (PBS, pH 7.4). After equilibration, 1 mL of micelle formulation (containing 1.3 mg ECZ) or 140 mg of Pevaryl® cream (1% w/w ECZ corresponding to 1.4 mg ECZ) was added to the donor compartment. The receptor compartment was stirred at 250 rpm at room temperature; 0.6 mL of receptor phase was sampled at the end of the experiment (after 1, 3 or 6 h). Chapter V 125 Upon completion of the permeation experiments, the diffusion cells were dismantled and the skin surface washed in running water to remove the residual formulation. The amount of ECZ deposited in the skin was extracted by cutting the skin samples into small pieces and soaking in 10 mL of acetonitrile for 4 h with continuous stirring at room temperature. The samples were filtered through 0.22 µm nylon membrane filters (VWR, Nyon, Switzerland) prior to HPLC analysis. Human skin Deposition of ECZ after application of either the micelle formulation or Pevaryl® cream in human skin was determined as before for porcine skin except that specially designed Franz diffusion cells (area = 0.66 cm2) with a receptor volume of 4 mL were used and skin extraction was achieved using 4 mL of acetonitrile as the extraction medium. 2.10 Confocal laser scanning microscopy Full thickness porcine skin samples were equilibrated in 0.9 % NaCl and mounted in standard Franz diffusion cells; 1 mL of a fluorescein loaded micelle formulation was placed in the donor compartment and kept in contact with the epidermal surface for either 6 or 24 h after which the diffusion cells were dismantled and skin samples were washed with 20% ethanol solution. The samples were then placed on a glass slide with the stratum corneum side up and covered with distilled water. A cover slip was placed on the skin sample and fluorescein fluorescence was visualized by using a Laser Scan Microscope 510 Meta (Carl Zeiss; Jena, Germany) at a power of 0.95W with a pinhole of 84µm and with a master gain of 541 for 6 h experiments and of 671 for 24 h experiments. The fluorescence excitation and emission wavelengths were set with a filter 505-530 nm, respectively using an Ar laser at an excitation lines at 488 nm. Samples of skin in contact with a solution of fluorescein in water for 6 h and 24 h (2 mg fluorescein was partly dissolved in 6 mL water, then the solution was filtered using a 0.22 μm filter) were also tested as controls. The confocal images were obtained with an Achroplan 63× objective and analyzed using Zeiss LSM Image Browser software. Each image was the average of eight repeated scans. ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 126 3. Results and Discussion 3.1 Characterization of CLZ, ECZ and FLZ micelle formulations The simpler and faster Method 1 was used to prepare the first series of formulations in order to enable a more rapid screening. The incorporation of the three azoles yielded MPEGhexPLA and MPEG-PLA micelles of homogeneous size with a number weighted diameter (dn) between 30-40 nm for more than 97% of the micelles (Table 3). The “nanosize” of the micelles was confirmed by TEM – an image of ECZ loaded MPEG-dihexPLA micelles is shown in Figure 1. The high values of the hydrodynamic diameter (Zav) from 70 to 165 nm and the relatively high polydispersity index can be attributed to the presence of a few larger micelles, which can be seen in the TEM image. Since these larger micelles scattered more light than their smaller counterparts, the mean diameter deduced from the overall intensity gives a higher mean value (Zav) than dn and consequently a high polydispersity index. Thus, the number weighted diameter dn and the corresponding percentage [%]dn were considered to be the best parameters to describe micelle size and confirmed that these micelles are true nanocarriers with diameters of 30-40 nm and a narrow distribution. Earlier studies have suggested that carrier size can influence skin deposition. Smaller liposomes with sizes of 110120 nm increased cumulative permeation of cyproterone acetate through split thickness porcine abdominal skin 13 and skin deposition of the fluorescent marker, caroboxyfluorescein in human abdominal skin 14. However, no clear size dependence was observed for delivery of liposomes containing cyclosporin A across porcine skin 15. Figure. 1. TEM picture of drug loaded micelles with econazole nitrate Chapter V 127 The MPEG-hexPLA micelles containing FLZ and ECZ showed much higher incorporation efficiencies than those with CLZ (84-98% cf. 11-19%) (Table 3 and Fig. 2). Moreover, the less hydrophobic MPEG-PLA standard (control), which usually incorporated less hydrophobic drug than the MPEG-hexPLA micelles 7-9 incorporated twice as much CLZ, although the incorporation efficiency remained relatively modest at only 36%. Possessing the highest log P of the three drugs, CLZ might have been assumed a priori to be the best candidate for incorporation into the hydrophobic core of the MPEG-hexPLA micelles. However, previous results with other drugs have shown that, in addition to log P, the aqueous solubility, the number of H-bond donor (Hd) and H-bond acceptor (Ha) groups can also affect incorporation efficiency into MPEG-hexPLA micelles 16. Multiple linear regression analysis of data from an investigation into the incorporation of eighteen different drugs in MPEG-hexPLA micelles, suggested that the incorporation efficiency was influenced in decreasing order by Hd, log P, Ha and the aqueous solubility. The molecular weight and the surface tension of the drug had less effect. The influence of Ha and Hd groups on the incorporation results in MPEG-hexPLA micelles may explain the differences in incorporation found for CLZ, FLZ and ECZ. Both FLZ and ECZ contain one Hd group; in contrast CLZ has none – the presence of Hd was found to be the most important factor governing drug incorporation into MPEG-hexPLA micelles – thus, it is consistent that FLZ and ECZ show manifold higher incorporation efficiencies than CLZ. Moreover, CLZ has fewer Ha groups (two) in comparison to ECZ (five) and FLZ (eight), again consistent with its poor incorporation efficiency. Drug/copolymer affinity was also found to be favoured by the formation of H-bonds between the multiple H-bond sites of PCL copolymer and the single Hbond site of two cucurbitacin drugs 17. Drugs with multiple H bond donors and acceptors have also been found to be better solubilised 18. Similarly, Liu et al. considered the overall system when selecting the most suitable polymer for an ellipticine micelle formulation 19. ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 128 Table 3. Characteristics of MPEG-(hex)PLA micelles loaded with clotrimazole (CLZ), fluconazole (FLZ) and econazole nitrate (ECZ), prepared by method 1. Micelle size Drug Copolymer Drug content (mgdrug/gcopolymer) Incorp. efficiency (%) (nm) MPEG-PLA 108.3±2.2 36.1 72 0.28 37 99.9 33.3±0.5 11.1 163 0.51 20 100.0 MPEG-dihexPLA 58.0±0.2 19.3 95 0.30 29 98.7 MPEG-PLA 250.2±2.7 83.4 92 0.44 20 99.2 271.7±2.7 90.7 112 0.50 18 100.0 MPEG-dihexPLA 268.3±2.7 89.4 71 0.40 27 98.2 MPEG-PLA 177.1±3.1 59.0 70 0.29 29 100.0 252.7±1.0 84.4 145 0.41 30 97.7 295.1±5.8 98.3 95 0.30 29 100.0 CLZ MPEG-monohexPLA FLZ MPEG-monohexPLA ECZ MPEG-monohexPLA MPEG-dihexPLA a Hydrodynamic diameter b Polydispersity index c Number weighted diameter d Percentage of micelles having the diameter dn Zava b P.I. dnc (nm) [%]dnd FLZ was successfully encapsulated into the different micelles, achieving drug contents between 250 and 268 mg/g and incorporation efficiencies of 83 to 91%. ECZ showed similar high drug loadings and an even higher incorporation efficiency (98%) in MPEG-dihexPLA micelles; this was an advantage since the hydrophobic dihexPLA core increases the storage stability of the micelle formulations 9;20 . Based on the loading efficiency, the ECZ MPEG- dihexPLA micelle formulation was selected for further optimization prior to skin transport studies. Chapter V 129 350 Drug loading in micelles (mg/g copolymer) 300 250 200 150 100 50 0 Clotrimazole Fluconazole Econazole nitrate Figure 2. Drug loading in MPEG-PLA (), MPEG-monohexPLA () and MPEG-dihexPLA ( ) micelles for 3 antifungal agents: clotrimazole, fluconazole and econazole nitrate (Mean ±SD, n=3) 3.2 Optimizing the preparation of ECZ MPEG-dihexPLA micelle formulations Evaluation of the preparation method As mentioned earlier, micelle size may influence skin deposition. Previous experiments showed that the evaporation step in the co-solvent evaporation method influenced both the size and size distribution of the resulting micelles [Mondon et al., unpublished data]. Here, two micelle preparation methods were tested which differed in the type of mixing during the addition and evaporation steps. Method 1 involved a simple stirring and a slow evaporation (2 h at 200 mbar), whereas Method 2 involved sonication during the addition step and a faster evaporation (10 min) at 15 rpm. MPEG-dihexPLA micelles with smaller Zav, dn, and a narrower polydispersity index were obtained by Method 2 (Table 4). In addition, ECZ incorporation was also slightly higher with Method 2, 79.0% (cf. 76.5% for Method 1). Therefore, based on these results, the sonication method was chosen to prepare the ECZ MPEG-dihexPLA micelle formulations for testing in the skin deposition experiments. ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 130 Table 4. Characteristics of MPEG-dihexPLA micelle formulations prepared by method 1 (stirring, slow evaporation) and method 2 (sonication, faster evaporation). Micelle size Batch Micelle prep. method 1 Method 1 2 Method 2 a Incorp. Zav dn [ECZ]mica (g/L) (nm) 76.5 1.12±0.02 133 0.22 50 99.9 79.0 1.20±0.03 91 0.16 42 99.8 Efficiency (%) P.I. (nm) [%]dn ECZ concentration in micelles Zav, dn and [%]dn defined in Table 3 Improving ECZ drug loading Micelle preparation using Method 2 was also used to optimize ECZ loading and to maximize the drug/copolymer ratio (mgdrug/gcopolymer). Different target drug loadings (represented by different amounts of drug dissolved in the initial acetone phase) were tested using two copolymer concentrations: 5 and 10 mg/mL. In previous studies, increasing MPEG-dihexPLA concentration in the micelle formulation yielded formulations with higher drug concentration without changing micelle size as confirmed by DLS and TEM analysis 7. However, in these studies higher ECZ drug content was observed at the lower copolymer concentration (~285 and ~182 mgECZ/gcopolymer at 5 and 10 mgcopolymer/mL, respectively) (Table 5). Incorporation efficiencies were >90% with respect to the target drug loadings of 300 and 200 mg/g for micelles with 5 and 10 mgcopolymer/mL, respectively. As seen for other drugs, incorporation efficiency decreased at higher target drug loadings 8. A higher drug / copolymer ratio of 28% (w/w) with 95% incorporation efficiency and higher shelf-life stability were achieved for micelles with 5 mgcopolymer /mL; thus, this copolymer concentration was chosen for the next experiments. Since 74 nm ≤ Zav ≤ 95 nm and 22 nm ≤ dn ≤ 38 nm for the different micelles, it is clear that the sonication method was capable of preparing truly nanosized drug carrier systems with high ECZ loading. Chapter V 131 Table 5. Characteristics of MPEG-dihexPLA micelle formulations of two copolymer concentrations (5 and 10 mg/mL) for different drug contents. Incorp. Micelle size Target ECZ content (mgECZ/gcopolymer) Actual ECZ content (mgECZ/gcopolymer) Efficiency (%) 200a 186.5±2.2 92.5 0.93±0.01 86 0.26 38 98.3 300a 285.8±6.4 94.9 1.42±0.03 80 0.27 33 99.8 400a 251.4±5.9 65.6 1.27± 0.03 74 0.27 22 90.5 500a 221.1±6.4 46.1 1.15 ±0.03 75 0.36 27 99.9 200b 181.8±8.1 92.0 1.84±0.08 95 0.31 24 98.8 300b 172.3±12.4 56.6 1.69±0.12 81 0.34 25 100.0 [ECZ]micelles (g/L) Zav (nm) PI dn (nm) [%]dn [ECZ]micelles defined in Table 4 Zav, dn and [%]dn defined in Table 3 a Micelles of 5 mgcopolymer/mL b Micelles of 10 mgcopolymer/mL 3.3 ECZ skin deposition studies using MPEG-dihexPLA micelles and Pevaryl® cream Porcine skin An ECZ MPEG-dihexPLA micelle formulation was prepared by sonication with a copolymer concentration of 5 mg/mL and an ECZ concentration of 1.3 mg/mL with Zav < 90 nm and dn < 40 nm. Skin deposition of ECZ from the MPEG-dihexPLA micelle formulation was investigated as a function of application time and compared to that from Pevaryl®, a commercially available liposomal formulation, in which the liposomes are reported to have diameters (Zav) of 160-200 nm 21. A similar dose of ECZ was applied using each formulation (1.3 mg MPEG-dihexPLA micelles and 1.4 mg for Pevaryl®, respectively). No ECZ was detected in the receiver compartment from any of the tested formulations. Skin deposition of ECZ from the MPEG-dihexPLA micelles was significantly higher than that observed with Pevaryl® after formulation application for 1, 3 and 6 h (4.7 ± 1.7, 14.7 ± 1.7 and 22.8 ± 3.8 µg/cm2, cf. 1.0 ± 0.3, 1.4 ± 0.2 and 1.7 ± 0.6 µg/cm2, respectively; Fig. 3) (Student’s t-test, α=0.05). This corresponded to 5-, 10- and 13-fold improvements in ECZ deposition using the ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 132 MPEG-dihexPLA micelle formulation at the 1, 3 and 6 h time-points, respectively. Indeed, despite the presence of permeation enhancers such as labrafil M 1944 CS, linalol, cinnamic alcohol and cinnamic aldehyde in this formulation, Pevaryl® application resulted in lower skin deposition of ECZ than with the much simpler MPEG-dihexPLA micelles (which contained only water, copolymer and drug). The increase in ECZ skin deposition may be attributed to the difference of the carrier size; the MPEG-dihexPLA micelles are approximately half the size of the liposomes and may have a larger contact area with the skin surface. In addition, their viscosity may facilitate film formation resulting in a depot at the skin surface and so increase contact time and drug deposition. Another factor that may affect delivery is the relative thermodynamic activity of ECZ in the two formulations since this will determine partitioning from the formulation into the stratum corneum. It has also been reported that the physical state of the surfactant used in the preparation of niosomes or liposomes had an influence on the deposition of finasteride into sebaceous gland region 22 . The liquid state vesicles showed higher permeation than gel state vesicles. This was attributed to a better penetration of the liquid state surfactant molecules into the stratum corneum compared than those forming a gel-state. By analogy, the physical state of the polymeric micelles is rather fluid due to the low Tg of the dihexPLA core 23 and this may also facilitate drug deposition. Skin deposition of ECZ (µg/cm2) 30 * 25 20 * 15 10 * 5 0 1 3 Time (h) 6 Figure 3. Skin deposition of ECZ from the MPEG-dihexPLA micelle formulation () and the Pevaryl® (■) after 1, 3 and 6h application on porcine skin (Mean ± SD, n=5-6). * Significantly different from Pevaryl® (Student’s t-test, α= 0.05). Chapter V 133 Human skin ECZ delivery from the MPEG-dihexPLA micelle formulation and Pevaryl® was also investigated using human skin (Fig. 4); for these experiments skin deposition of ECZ was determined only at the 6 h time-point. ECZ deposition from the micelle formulation was again almost an order of magnitude superior to that from Pevaryl® (11.3 ± 1.6 and 1.5 ± 0.4 µg/cm2, respectively). As with the porcine skin experiments, no ECZ permeation was observed across the human skin from either formulation. ECZ deposition following application of Pevaryl® was equivalent for porcine and human skin. In contrast, a 2-fold difference in ECZ deposition from the MPEG-dihexPLA micelle formulation was observed between human and porcine skin. The human skin used for these experiments was obtained following breast reduction surgery and, in contrast to porcine ear skin, was devoid of hair follicles. Thus, the higher ECZ deposition observed using the MPEGdihexPLA micelle formulation was tentatively attributed to penetration of the smaller micelles via the hair follicles. 30 Skin deposition of ECZ (µg/cm2) * 25 20 15 * 10 5 0 Pevaryl® Micelle formulation Figure 4. Comparing deposition of ECZ using the MPEG-dihexPLA micelle formulation Pevaryl® after application for 6h on porcine skin () and human skin (■) (Mean ± SD, n=5-6). * Significantly different from Pevaryl® (Student’s t-test, α= 0.05). ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 134 Visualizing micellar penetration pathways using confocal laser scanning microscopy In order to determine the role of the follicular pathway, fluorescein loaded MPEG-dihexPLA micelles (dn~28 nm) were prepared using the same method as that for the ECZ MPEGdihexPLA micelle formulation and applied to porcine skin for 6 and 24 h. A solution of fluorescein in water was applied on the skin for 6 h and 24 h, as control. The skin samples were subsequently analyzed using confocal laser scanning microscopy in order to visualize micelle deposition. In contrast to the untreated skin, (Fig. 5a) and to the skin treated with fluorescein solution (Fig. 5b and 5c), the images in the XY-plane following treatment with fluorescein loaded micelles for 6 and 24 h show considerable fluorescence (Fig. 5d and 5e). Figure 5. Confocal laser scanning microscopy images of porcine skin in the XY-plane show that in contrast to (a) the untreated porcine skin and skins treated for (b) 6 h and (c) 24 h with a fluorescein solution, considerable fluorescence was observed after contact with fluorescein loaded micelles for (d) 6 h and (e) 24 h. Chapter V 135 Similarly, no fluorescence is observed in the XZ-plane in skin treated only with a fluorescein solution (Fig. 6a and 6b). However, the images following treatment with fluorescein loaded micelles show appreciable fluorescence at depths of up to 40 µm (Fig. 6c and 6d). The image recorded in the XZ-plane at 24 h (Fig. 6d) shows greater fluorescence intensity and also shows the presence of localized fluorescence in the follicles penetrating deeper into the skin. The follicular pathway has been reported as the primary penetration route for nanoparticles 24; it is thought to be a size dependent process, where smaller nanoparticles show highest penetration 25;26 . Moreover, nanoparticles of similar sizes to the MPEG-dihexPLA micelles were shown to cross the skin barrier via hair follicles 27. Thus, the MPEG-dihexPLA micelles may target delivery to the follicles and their accumulation, depot formation, and subsequent release of a drug may be used to provide more effective, sustained treatments. Figure 6. Confocal laser scanning microscopy images of porcine skin from the XZ-plane show that in contrast to the skin treated with a fluorescein solution for (a) 6 h and (b) 24 h, fluorescence appeared to accumulate in epidermis via the hair follicles (white squares) when skin was treated with fluorescein loaded micelles for (c) 6 h and (d) 24 h . ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 136 4. Conclusion The results demonstrate that hydrophobic MPEG-dihexPLA copolymers encapsulated econazole nitrate and fluconazole with high incorporation efficiencies. The preparation method was shown to impact on micelle properties; faster evaporation and addition under sonication yielded smaller micelles. Econazole nitrate delivery using MPEG-dihexPLA micelle formulations resulted in significantly higher drug deposition in both porcine and human skin as compared to Pevaryl®, a marketed liposomal formulation. Confocal laser scanning microscopy studies using similarly-sized fluorescein loaded micelles provided support for the potential role of the follicular pathway and suggested that the MPEGdihexPLA micelles may facilitate targeted follicular delivery References (1) Ameen, M., Epidemiology of superficial fungal infections, Clin.Dermatol. 2010, 28, 197-201. (2) Hay, R., Superficial fungal infections, Medicine 2009, 37, 610-612. (3) Ramos-e-Silva, M.; Lima, C. M. O.; Schechtman, R. C.; Trope, B. M.; Carneiro, S., Superficial mycoses in immunodepressed patients (AIDS), Clin.Dermatol. 2010, 28, 217-225. (4) Davis, J. D., Superficial fungal infections of the skin: Tinea corporis, tinea pedis, and Candida intertrigo, Prim Care Update Ob Gyns. 1995, 2, 157-161. (5) Hof, H., Developments in the epidemiolgy of invasive fungal infections - implications for the empiric and targeted antifungal therapy, Mycoses 2008, 51, 1-6. (6) Naik, A.; Kalia, Y. N.; Guy, R. H., Transdermal drug delivery: overcoming the skin's barrier function, Pharm.Sci.Technol.Today 2000, 3, 318-326. (7) Mondon, K.; Zeisser-Labouèbe, M.; Gurny, R.; Möller, M., Novel Cyclosporin A formulations using MPEG-hexyl substituted polylactide micelles: a suitability study, Eur.J.Pharm.Biopharm. 2010, 77, 56-65. (8) Mondon, K.; Gurny, R.; Möller, M., Colloidal Drug Delivery Systems Recent Advances With Polymeric Micelles, Chimia 2008, 62, 832-840. Chapter V 137 (9) Trimaille, T.; Mondon, K.; Gurny, R.; Möller, M., Novel polymeric micelles for hydrophobic drug delivery based on biodegradable poly(hexyl-substituted lactides), Int.J.Pharm. 2006, 319, 147-154. (10) Vanden Bossche, H.; Marichal, P., Mode of action of anti-Candida drugs: focus on terconazole and other ergosterol biosynthesis inhibitors., Am.J.Obstet.Gynecol. 1991, 165, 1193-1199. (11) Loftsson, T.; Hreinsdóttir, D., Determination of Aqueous Solubility by Heating and Equilibration: A Technical Note, AAPS PharmSciTech. 2006, 7, E1-E4. (12) Shalaeva, M.; Kenseth, J.; Lombardo, F.; Bastin, A., Measurement of dissociation constants (pKa values) of organic compounds by multiplexed capillary electrophoresis using aqueous and cosolvent buffers, J.Pharm.Sci. 2008, 97, 2581-1606. (13) Valenta, C.; Janisch, M., Permeation of cyproterone acetate through pig skin from different vehicles with phospholipids, Int.J.Pharm. 2003, 258, 133-139. (14) Verma, D. D.; Verma, S.; Blume, G.; Fahr, A., Particle size of liposomes influences dermal delivery of substances into skin, Int.J.Pharm. 2003, 258, 141-151. (15) du Plessis, J.; Ramachandran, C.; Weiner, N.; Muller, D. G., The influence of particle size of liposomes on the deposition of drug into skin, Int.J.Pharm. 1994, 103, 277-282. (16) Mondon, K., Novel Micellar Systems for the Formulation of Poorly Water Soluble Drugs: Biocompatibility Aspects and Pharmaceutical Applications, PhD Thesis 2010, University of Geneva, Report no 4258. (17) Patel, S. K.; Lavasanifar, A.; Choi, P., Roles of Nonpolar and Polar Intermolecular Interactions in the Improvement of the Drug Loading Capacity of PEO-b-PCL with Increasing PCL Content for Two Hydrophobic Cucurbitacin Drugs, Biomacromolecules 2009, 10, 2584-2591. (18) Patel, S. K.; Lavasanifar, A.; Choi, P., Molecular dynamics study of the encapsulation capability of a PCL-PEO based block copolymer for hydrophobic drugs with different spatial distributions of hydrogen bond donors and acceptors, Biomaterials 2010, 31, 1780-1786. (19) Liu, J. B.; Xiao, Y. H.; Allen, C., Polymer-drug compatibility: A guide to the development of delivery systems for the anticancer agent, ellipticine, J.Pharm.Sci. 2004, 93, 132-143. (20) Nottelet, B.; Di Tommaso, C.; Mondon, K.; Gurny, R.; Möller, M., Fully biodegradable polymeric micelles based on hydrophobic- and hydrophilic-functionalized poly(lactide) block copolymers, J.Polym.Sci.Part A: Polym.Chem. 2010, 48, 3244-3254. (21) Naeff, R., Feasibility of topical liposome drugs produced on an industrial scale, Adv.Drug Deliver.Rev. 1996, 18, 343-347. (22) Tabbakhian, M.; Tavakoli, N.; Jaafari, M. R.; Daneshamouz, S., Enhancement of follicular delivery of finasteride by liposomes and niosomes: 1. In vitro permeation and ECZ/MPEG-hexPLA Micelle Formulations for Topical Application 138 in vivo deposition studies using hamster flank and ear models, Int.J.Pharm. 2006, 323, 1-10. (23) Trimaille, T.; Gurny, R.; Möller, M., Poly(hexyl-substituted lactides): Novel injectable hydrophobic drug delivery systems, J.Biomed.Mater.Res.A 2007, 80A, 55-65. (24) Shim, J.; Seok Kang, H.; Park, W. S.; Han, S. H.; Kim, J.; Chang, I. S., Transdermal delivery of mixnoxidil with block copolymer nanoparticles, J.Control.Release 2004, 97, 477-484. (25) Knorr, F.; Lademann, J.; Patzelt, A.; Sterry, W.; Blume-Peytavi, U.; Vogt, A., Follicular transport route - Research progress and future perspectives, Eur.J.Pharm.Biopharm. 2009, 71, 173-180. (26) Wosicka, H.; Cal, K., Targeting to the hair follicles: Current status and potential, J.Dermatol.Sci. 2010, 57, 83-89. (27) Vogt, A.; Combadiere, B.; Hadam, S.; Stieler, K. M.; Lademann, J.; Schaefer, H.; Autran, B.; Sterry, W.; Blume-Peytavi, U., 40nm, but not 750 or 1,500nm, Nanoparticles Enter Epidermal CD1a+ Cells after Transcutaneous Application on Human Skin, J.Invest.Dermatol. 2006, 126, 1316-1322. 1234567892A92BC6975DEE686D78F567C95D568DD5CD57B796D5B75CD5CB66CF59E5 C65 2345 D6 D2F5 FCD5 8935 B95 B D5 B B7CB4D935 A29AD2C6D5 675 92B5 E923BC697!5 "D2D5676C6B5C36D5C9#B25CD536CB66CF59E5D5E923BC6975B5B7592B59B4D5 1 E925B2D5A2DD7CD!5 1 1 1 1 123456789A88 8 1 8 1 Chapter VI 139 MPEG-hexPLA Micelle Formulations for Oral Delivery of Poorly Water Soluble Drugs: Investigations with Cinnarizine as a Model Drug K. Mondon, R. Gurny, and M. Möller. School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 30, Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland. 1. Introduction Oral drug delivery remains the most patient-friendly way of medication. The drug discovery with the present use of high throuput screening methods gives leads of many novel potential drug candidates, which tend to have a high lipophilicity and low water solubility 1. However, the Lipinski’s “rule of 5” 2, which suggests that an orally active drug should not have more than 5 hydrogen bond donors, not more than 10 hydrogen bond acceptors, a molecular weight below 500 daltons, and an octanol-water partition coefficient log P of less than 5, is often not matched. Next to the poor water solubility, the poor permeability of the drug is another major cause of the low bioavailability of drugs in the gastrointestinal (GI) tract 3. Different methods to improve the biopharmaceutical properties of poorly water soluble drugs have thus been studied. Amongst other drug delivery systems, polymeric micelles have been investigated for oral applications 4 . With their specific core-shell structure micelles can protect the hydrophobic drug by incorporating it within its core, while the hydrophilic shell facilitates aqueous solubility to the formulation. Regarding the range of pH encountered in the GI tract (pH of 1.2 in the empty stomach, 5-7 in the small intestine and 6-7.5 in the colon), the biocompatible micelle carriers need to be stable enough to keep the drug in their core at the low pH during the passage of the stomach before reaching or releasing the drug if the intestinal epithelium is targeted. MPEG-hexPLA Micelles for Oral Delivery 140 In this work the possible oral application of MPEG-hexPLA micelle formulations was investigated using the antihistaminic class II drug, cinnarizine (CIN). Cinnarizine is a lipophilic piperazine derivative (log P ~5.3 5) with a low water solubility of 2 mg/L at 37°C 6. CIN was chosen for these studies because it is reported as an oral drug model of choice7;8. The stability of MPEG-(hex)PLA micelles was assessed under gastric pH. Therefore the intactness of the micelle structure was studied via the fluorescence of incorporated Nile Red. A Nile Red fluorescence intensity can only be detected if this probe remains in the hydrophobic environment of the micelle core 9. The stability of CIN loaded MPEG-hexPLA micelles regarding the drug concentration, and the molecular weight of the block copolymer, was further studied towards possible oral formulations. 2. Materials and methods 2.1. Materials Cinnarizine (CIN), acetone, acetonitrile (HPLC grade), hydrochloric acid (HCl) were supplied by Sigma-Aldrich (Buch, Switzerland). THF and ammonium dihydrogen phosphate were purchased by SDS (Toulouse, France) and by Fluka (Buch, Switzerland), respectively. 2.2. Preparation of MPEG-(hex)PLA block copolymers MPEG-hexPLA block copolymers were prepared by ring opening polymerization of hexyl substituted lactide as described previously by Mondon et al. 10. Methoxypoly(ethylene glycol) (MPEG2000) of 2000 g/mol was used as initiator. Copolymers with the molecular weights of the (hex)PLA block of 4000g/mol (MPEG2000-hexPLA4000) and of 8000g/mol (MPEG2000hexPLA8000) were prepared. The overall molecular weights of the block copolymer were thus of 6000g/mol and 10000g/mol, respectively. Please note that in the following text, “MPEG-(hex)PLA” refers to both MPEG-PLA and MPEG-hexPLA block copolymers. Chapter VI 2.3. 141 Preparation of CIN-loaded MPEG-(hex)PLA polymeric micelles CIN loaded micelles were prepared by dissolving 1 mL copolymer solution (20 mg/mL in acetone) with 100 µL of a CIN solution (20 mg/mL) and 900 µL acetone. The resulting acetone solution was added drop wise every 5s into 4 mL ultra pure water under sonication. Acetone was removed by evaporation and water was added afterwards to get a final micelle solution concentration of 5 mgcopolymer/mL. After overnight equilibration, CIN loaded micelles were centrifuged at 9500 x g for 15 min to remove non-incorporated drug. 2.4. Preparation of unloaded polymeric micelles Unloaded micelles were prepared following the method described above, whereby pure acetone was used instead of the drug/acetone solution. 2.5. Determination of CIN loading by HPLC The determination of the CIN loading in micelles was performed after centrifugation of the micelle solution. One hundred microliter of the supernatant was taken and dissolved with 900 µL of acetonitrile to break the micelles and to release the drug. CIN was then quantified by HPLC using a Nucleosil 100-5 C18 column (250 mm × 4 mm). The mobile phase was a mixture of acetonitrile/ 20 mM ammonium dihydrogen phosphate buffer (50/50) eluting at a flow rate of 1.0 mL/min. The CIN signal was detected by UV at λ= 250 nm and appeared at around 8 min. Calibration curves with a regression coefficient superior to 0.99 were obtained. 2.6. Gastric-pH-tests of CIN loaded MPEG-hexPLA micelles The gastric-pH-tests were performed by adding CIN loaded polymeric micelles under slow stirring to 10 mL of 0.1N HCl (pH=1.2) at 37°C. After 30 min and 1h stirring, 500 µL of the acidic solution were removed and centrifuged at 9500 x g for 15 min. The supernatant was then diluted in acetonitrile in a 1:1 ratio and analysed by HPLC for the drug content. MPEG-hexPLA Micelles for Oral Delivery 2.7. 142 Stability studies of Nile Red loaded MPEG-(hex)PLA micelles in 0.1N HCl For the stability studies, Nile Red loaded MPEG-(hex)PLA4 micelles were prepared by incorporating 10µL Nile Red solution (6 10-4 M) into 300 µL unloaded MPEG2000(hex)PLA4000 micelles (ccopolymer= 1.66 mg/mL) under stirring for 3h. The resulting Nile Red loaded micelles were diluted in 1.5 mL 0.1N HCl at 37°C. After complete dilution the Nile Red fluorescence intensity was studied for 1h. The Nile Red fluorescence was detected at an excitation wavelength of 537 nm and an emission wavelength of 600 nm using a Fluoromax spectrofluorometer (Spex, Stanmore, UK). The Nile Red fluorescence and its intensity change was visualized by diluting 1 mL of Nile Red loaded MPEG2000-(hex)PLA4000 micelles into 5 mL 0.1N HCl under slow stirring at 37°C and taking pictures just after dilution, after 30 min and after 1h under acidic pH. A control sample with 0.1N HCl containing the same quantity of Nile Red was prepared and analysed in parallel. 2.8. Degradation tests of the block copolymers at gastric pH The copolymer degradation was assessed by determining the copolymer molecular weight and the polydispersity index by GPC (Waters, Milford, USA) before and after gastric-pH-tests (1h at 0.1N HCl). A calibration curve with a regression coefficient superior to 0.99 was obtained using 6 polystyrene standards ranging from 3460 to 277000g/mol (PSS, Mainz, Germany). The copolymer characteristics after the gastric-pH-tests were determined after evaporation of the remaining acidic solution containing CIN loaded micelles and its dilution into 1.5 mL THF. 3. Results and Discussion The effective absorption of an orally administered drug depends on the dissolution within the gastrointestinal (GI) fluids and on the permeation through the intestine 11 . Drugs with low water solubility and/or low permeability are unfavoured for oral administration. Indeed, poor drug water solubility is often the cause of an early drug precipitation into the GI tract concomitant with unreached therapeutic doses. The incorporation of such drugs into MPEG- Chapter VI 143 hexPLA polymeric micelles was shown to be an efficient strategy to increase their aqueous solubility. Promising results for the solubilisation in such MPEG-hexPLA micelles for the intravenous application of cyclosporin A 10 and hypericin (Mondon et al. see Chapter IV), or for the topical applications of econazole nitrate (Mondon et al. see Chapter V) were demonstrated. In the current study, the possible use of MPEG-hexPLA micelles in oral delivery was evaluated incorporing the model drug cinnarizine (CIN). 3.1. CIN loaded MPEG-(hex)PLA micelle characteristics Two series of MPEG-(hex)PLA micelles with block copolymers of 2 different molecular weights (Mw) were investigated. The used block copolymers, MPEG2000-(hex)PLA4000 of 6000g/mol and MPEG2000-(hex)PLA8000 of 10000 g/mol, were synthesized in a controlled manner by ring opening polymerization as described previously 10;12 . All block copolymers were obtained with a narrow polydispersity index inferior to 1.2. The resulting CIN loaded micelles obtained a relative narrow size distribution with a polydispersity index below 0.3 (Table 1). Table 1. Characteristics of CIN loaded MPEG-(hex)PLA micelle formulations Micelle size Copolymer MPEG2000–PLA4000 MPEG2000-monohexPLA4000 MPEG2000-dihexPLA4000 MPEG2000-PLA8000 MPEG2000-monohexPLA8000 MPEG2000-dihexPLA8000 Drug content [drug]micelles [mgCIN/gcopolymer] [mg/L] 87.0 43.09 89.5 44.89 90.6 45.32 57.53 28.69 104.50 52.28 109.00 55.67 Zav P.I. 30 51 61 69 74 82 0.07 0.22 0.30 0.035 0.16 0.28 dn [%]dn 25 34 32 54 39 44 100.0 100.0 99.5 100.0 100.0 99.9 With [drug]micelles, the concentration of the drug in micelles; Zav, the hydrodynamic diameter; P.I., the polydispersity index; dn the number weighed diameter, and. [%]dn the percentage of micelles with a given dn The micelle size described by the hydrodynamic diameter Zav and the number-weight diameter dn was slightly higher for copolymers with higher Mw. This has also been observed with MPEG-poly(caprolactone) polymeric micelles 13 . By increasing the Mw of the core forming block, the hydrophilic/hydrophobic ratio of the block copolymer decreases and thus favors the structure of micelles towards bigger structures like i.e. wormlike micelles. MPEG-hexPLA Micelles for Oral Delivery 144 However, for the here tested formulation of the high Mw micelles a small amount of larger spherical micelles in the formulation are assumed for the slight increase in size. Only 1% of micelles have a number weighed diameter superior to 40-55 nm for micelles of MPEG2000(hex)PLA8000, and to 25-35 nm for those with MPEG2000-(hex)PLA4000. In general, all the tested MPEG-(hex)PLA micelles had a hydrodynamic diameter below or around 80 nm, the range of the size generally observed for polymeric micelles 14 , and are thus real nanosized drug carriers. CIN was successfully incorporated into MPEG-(hex)PLA micelles. For the 3 tested copolymers the maximum drug loading was achieved with efficiencies superior to 80% for a targeted drug loading of 100 mgCIN/gcopolymer. A superior incorporation of CIN in the more hydrophobic hexyl-substituted PLA micelle core compared to the standard PLA was revealed. For example, with copolymers of 6000g/mol MPEG-dihexPLA micelles a CIN concentration of 45.3 mg/L compared to 44.9 mg/L for the less hydrophobic MPEG-monohexPLA micelles and 43.1 mg/L for the even less hydrophobic standard MPEG-PLA micelles was obtained. When using MPEG-hexPLA copolymers with the higher Mw of 10000g/mol, corresponding to the increased hexPLA block, the incorporation of CIN was increased by more than 15%. A CIN aqueous concentration of 55.6 mg/L in MPEG2000-dihexPLA8000 micelles in comparison to 45.3 mg/L in MPEG2000-dihexPLA4000 micelles (Table 1) was reached. This shows that an optimization of the polymer molecular weight and hydrophilic/hydrophobic block ratio can significantly improve the incorporation results. 3.2. Stability of MPEG-(hex)PLA micelle formulations under gastric pH As mentioned before, an oral administrated formulation should be stable enough in gastric pH to pass to the intestine. This is even more true for the oral formulation of CIN which has its aqueous solubility decreasing with the pH increase of the GI tract 6. In consequence, the MPEG-hexPLA micelle formulations are required to be stable at least 30 min at gastric pH. This stability means that the intactness of the micelle core-shell structure and the CIN drug content should be preserved during this time. MPEG-(hex)PLA micelle formulations were therefore assessed for their stability under gastric pH (pH=1.2) for 1h. The fluorescent Nile Red probe was incorporated into the MPEG2000-(hex)PLA4000 micelles to evidence the Chapter VI 145 intactness of the micelle core-shell structure. A Nile Red fluorescence intensity can only be detected if this probe remains in the hydrophobic environment of the micelle core 15 . The obtained constant fluorescence intensity confirms that the core-shell structure was almost unaffected for 1h at pH 1.2. In addition this result also proves that the hydrophobic dye does not diffuse out of the micelles under these conditions (Figure 1). Nile Red Fluorescence Intensity 1.2E+07 1.0E+07 8.0E+06 6.0E+06 4.0E+06 2.0E+06 0.0E+00 0 0.5 Time in HCl [h] 1 Figure 1. Nile Red fluorescence in function of time after dilution of Nile Red loaded MPEG2000dihexPLA4000 (), MPEG2000-monohexPLA4000 () and MPEG2000-PLA4000 (ο) micelle formulations in HCl 0.1N (pH=1.2). The stability of the formulations within this time in gastric pH was also visualized by photos, which show the stable violet colouring of the Nile Red micelle formulations (Figure 2). 1 (a) 2 3 4 1 (b) 2 3 4 1 2 3 4 (c) Figure 2. Fluorescence observations of Nile Red loaded MPEG2000-dihexPLA4000 (1), MPEG2000monohexPLA4000 (2), and MPEG2000-PLA4000 (3) micelle formulation diluted in 0.1N HCl at t =0 min (a), 30 min (b) and 1h (c). The same Nile Red quantity was added in 0.1N HCl as control (4). MPEG-hexPLA Micelles for Oral Delivery 146 The stability of the CIN loaded MPEG-hexPLA micelle formulations under gastric pH was investigated by following the CIN loading over time after dilution of CIN loaded MPEG(hex)PLA micelles in 0.1N HCl solution (pH=1.2). After 30 min the drug content was the same as the initial loading for the two tested series of micelle formulations. However, a drug loss of maximum 12% was observed after 1h (Figure 3). In contrast no drug loss was observed for Nile Red. This can be attributed to the difference of the CIN and Nile Red, respectively water solubilities. CIN with a higher water solubility at gastric pH than the Nile Red might easier diffuse out of the micelles into the aqueous environment. Moreover the degradation study of the MPEG2000-(hex)PLA4000 block copolymer revealed that the (hex)PLA micelle core decreased by 2-3 monomer units after 1h in gastric pH (Figure 4), possibly slightly decreasing the micelle stability and increasing drug diffusion. Nevertheless, the block copolymer degradation did not affect the micelle core-shell structure as such, as evidenced by the observed constant Nile Red fluorescence intensity (Figure 1). As the result MPEG-hexPLA micelles could be considered for the oral delivery of CIN as only a minimal drug loss and no change in the core-shell micelle structure were observed at gastric pH after 1h. a) MPEG2000-(hex)PLA4000 b) MPEG2000-(hex)PLA8000 100 100 %CIN loading in micelles 120 % CIN loading in micelles 120 80 60 40 20 80 60 40 20 0 0 0 0.5 Time in 0.1N HCl [h] 1 0 0.5 Time in 0.1N HCl [h] 1 Figure 3. Percentage of drug loading in (a) MPEG2000-(hex)PLA4000 and (b) MPEG2000- (hex)PLA8000 micelle formulations in function of the time at gastric pH for MPEG-dihexPLA (), MPEG-monohexPLA () and MPEG-PLA (○) micelles. Chapter VI P.I. : 1.26 ; 1.31 1.17 ; 1.21 147 1.15 ; 1.16 Figure 4. Molecular weight and polydispersity index (P.I.) of MPEG2000-(hex)PLA4000 before (white bars) and after 1h under gastric pH (0.1N HCl) (black bars) 4. Conclusion It could be demonstrated that the novel MPEG-hexPLA micelles successfully incorporate the poorly water soluble cinnarizine. The stability study of these micelle formulations at gastric pH (pH=1.2) revealed that MPEG-hexPLA micelles showed no drug loss at 30 min and only 12% loss after 1h, which could be due to the slight degradation by 2-3 (hex)PLA monomer units of the block copolymer and a micelle destabilization, and the relative higher CIN water solubility at gastric pH. In conclusion, these initial in vitro investigations prove the stability of the MPEG-hexPLA micelle formulations for bypassing the stomach into the intestine for a possible oral application. MPEG-hexPLA Micelles for Oral Delivery 148 References (1) Cheng, W. P.; Thompson, No.2010/0,029,544. C., Composition, U.S.Patent 2010, Application (2) Lipinski, C. A., Lead- and drug-like compounds: the rule-of-five revolution, Drug Discovery Today: Technologies 2004, 1, 337-341. (3) Helen Chan, O.; Stewart, B. H., Physicochemical and drug-delivery considerations for oral drug bioavailability, Drug Discovery Today 1996, 1, 461-473. (4) Gaucher, G.; Satturwar, P.; Jones, M. C.; Furtos, A.; Leroux, J. C., Polymeric micelles for oral drug delivery, Eur.J.Pharm.Biopharm. 2010, 76, 147-158 (5) Data taken from http://www.drugbank.ca/drugs. 2010. (6) Stella, V. J., Why Compounds are Insoluble and What We Can Do About It!, AAPS 45th Annual Pharmaceutical Technologies Arden Conference: Formulation Strategies for Poorly Soluble Drugs 2010, West Point, NY, USA. (7) Ogata, H.; Aoyagi, N.; Kaniwa, N.; Ejima, A.; Kitaura, T.; Ohki, T.; Kitamura, K., Evaluation of beagle dogs as an animal model for bioavailability testing of cinnarizine capsules, Int.J.Pharm. 1986, 29, 121-126. (8) Boyd, B. J.; Khoo, S. M.; Whittaker, D. V.; Davey, G.; Porter, C. J. H., A lipid-based liquid crystalline matrix that provides sustained release and enhanced oral bioavailability for a model poorly water soluble drug in rats, Int.J.Pharm. 2007, 340, 5260. (9) Davis, M. M.; Helzer, H. B., Titrimetric and Equilibrium Studies Using Indicators Related to Nile Blue A, Anal. Chem. 1966, 38, 451-461. (10) Mondon, K.; Zeisser-Labouèbe, M.; Gurny, R.; Möller, M., Novel Cyclosporin A formulations using MPEG-hexyl substituted polylactide micelles: a suitability study, Eur.J.Pharm.Biopharm. 2010, 77, 56-65. (11) Lennernäs, H., Human intestinal permeability, J.Pharm.Sci. 1998, 87, 403-410. (12) Mondon, K.; Gurny, R.; Möller, M., Colloidal Drug Delivery Systems Recent Advances With Polymeric Micelles, Chimia 2008, 62, 832-840. (13) Du, Z. X.; Xu, J. T.; Fan, Z. Q., Micellar Morphologies of Poly(e-caprolactone)-bpoly(ethylene oxide) Block Copolymers in Water with a Crystalline Core, Macromolecules 2007, 40, 7633-7637. (14) Kwon, G. S., Polymeric micelles for delivery of poorly water-soluble compounds., Crit.Rev.Ther.Drug Carrier Syst. 2003, 20, 357-403. (15) Davis, M. M.; Helzer, H. B., Titrimetric and Equilibrium Studies Using Indicators Related to Nile Blue A, Anal. Chem. 1966, 38, 451-461. 1 1 1 1 1 1231456723688 93A8BCD6BC1E7FC68 1 Conclusions and Perspectives 149 Conclusions and Perspectives Amongst the large number of potent drugs for pharmaceutical applications, 95% are lipophilic and poorly water soluble. However, these special properties render the aqueous formulation of those drugs very difficult. Early drug precipitation is often encountered due to poor stability and the therapeutic dose is rarely reached. Therefore, there is an unmet need to find stable and sufficient drug loading formulations for such drugs. One strategy to overcome this formulation issue is the incorporation of poorly water soluble drugs into colloidal carriers such as liposomes, nanoparticles, and micelles. This thesis work focused on novel polymeric micelles as promising delivery systems for poorly water soluble drugs. The presented novel polymeric micelles are composed of MPEG-hexPLA copolymers. The high number of hexyl groups in the core forming block polymer compared to standard MPEG-PLA micelles, was expected to increase the hydrophobicity of the micelle core, thus the incorporation of hydrophobic drugs. In a first step, the incorporation of the hydrophobic model drug, THPP, was presented as a proof of concept. A higher incorporation was achieved with the most hydrophobic MPEGdihexPLA micelles followed by MPEG-monohexPLA and MPEG-PLA micelles. This trend was generally observed for drugs with a low water solubility (< 0.055 mM). However, the drug hydrophobicity and the drug water solubility were not the only parameters controlling the incorporation efficiency into MPEG-hexPLA micelles. Structural parameters such as the presence of H-bond donor or -acceptor groups in the drug structure showed an influence, too. Interestingly, poorly water soluble drugs which do not fit the Lipinski’s “rule of 5”, indicating poor bioavailability and limited applications in clinics, showed to have suitable physicochemical parameters for an efficient incorporation into MPEG-hexPLA micelles. This envisions the use of these novel micellar carriers as an alternative formulation approach, and was demonstrated for some selected poorly water soluble drugs, e.g. cyclosporin A, THPP, hypericin, and econazole nitrate, for very different pharmaceutical applications. Conclusions and Perspectives 150 As a novel material for a possible clinical use, MPEG-hexPLA micelles were investigated for their biocompatibility. No toxicity in vitro and in vivo or hemolytic activity on human blood was found for both, unimers (below CMC) and polymeric micelles, up to concentrations of 20 mg/mL. In consequence, MPEG-hexPLA micelle formulations could be envisioned as suitable for pharmaceutical applications. One considerable pharmaceutical application is the possible use in intravenous administrations. The micelle formulation of cyclosporin A did not show any hemolytic activity, and efficiently achieved therapeutic doses with 4 times less excipient than the surfactant in the marketed formulation. Obviously, the in vivo efficiency of such a delivery system should be evaluated in animal models to confirm its potential for pharmaceutical applications. The use of MPEG-hexPLA micelle formulations for ovarian cancer diagnostics was studied. Intravenous administration to ovarian tumor bearing rats showed a high and fast accumulation of the incorporated poorly water soluble fluorescent hypericin, in tumour nodules by passive targeting. For future applications it could be very interesting to combine the cancer diagnosis with a direct treatment by making use of the photosensitizer properties of hypericin. To improve the carrier system an active tumor targeting by conjugating MPEG-hexPLA copolymers with ligands such as folic acid could be envisioned. A promising topical application of MPEG-hexPLA micelles for antifungal treatment was evidenced in this thesis. The superior delivery of ECZ into human and porcine skin with micelles to the commercially available formulation was demonstrated. Intriguingly, the deposition into the skin may be attributed to a follicular penetration pathway. The presented findings may help to formulate and deliver various potent hydrophobic drugs into the skin. Furthermore, the oral delivery of drug loaded MPEG-hexPLA micelles could be a possible pharmaceutical application. After 1h at gastric pH, MPEG-hexPLA micelles conserved 90% of the drug content of the model class II drug, cinnarizine, and maintained an intact core-shell structure with only a slight degradation of the copolymer. This could facilitate a drug transport through the stomach into the intestine. Conclusions and Perspectives 151 In summary, these novel biocompatible MPEG-hexPLA micelles are promising for pharmaceutical formulations for intravenous, topical and oral applications of poorly water soluble drugs. Still, drug incorporation into MPEG-hexPLA micelles and biologic activity could be improved by adjusting the copolymer properties, e.g. by changing MPEG/hexPLA ratios and molecular weights, or attaching specific ligands relevant to the desired application. Conclusions and Perspectives 152 1 1 1 1 1 1233456789A7B5CADEF7 7 1 Summary (In French) 153 Résumé et Conclusions Parmi les substances bioactives les plus prometteuses pour des applications pharmaceutiques à faible poids moléculaire, 95% sont lipophiles et faiblement solubles dans l’eau, rendant leur formulation difficile et leur application clinique limitée. Après application chez l’homme, une précipitation précoce est souvent observée due à la faible solubilité dans l’eau, par conséquent les concentrations thérapeutiques sont rarement atteintes. Une des stratégies utilisée pour pallier ces problèmes de formulation est l’incorporation de ces substances dans des micelles. De part leur structure particulière « cœur-couronne », les micelles polymériques peuvent incorporer des médicaments lipophiles au sein de leur cœur hydrophobe, tout en étant solubles dans l’eau grâce à leur couronne hydrophile. Le travail de cette thèse s’est orienté sur l’étude de nouvelles micelles polymériques : les micelles à base de copolymères de methoxypolyethylène glycol (MPEG) et de polylactides (PLA) substitués par des groupes hexyls (hex), nommés MPEG-hexPLA. La présence et le nombre élevé de groupes hexyls dans les micelles de MPEG-hexPLA, comparés aux micelles de MPEG-PLA standard, augmentent l’hydrophobicité du cœur et donc l’incorporation de produits hydrophobes. L’incorporation du composé modèle hydrophobe THPP a été testée et a validé cette stratégie. Une plus grande incorporation a été mise en évidence avec les micelles les plus hydrophobes, celles composées de copolymères di-substitués avec des groupes hexyls (MPEG-dihexPLA), suivent ensuite les micelles composées de copolymères mono substitués de groupes hexyls (MPEG-monohexPLA) et enfin les micelles de MPEG-PLA. Cette tendance a été principalement observée pour les substances dont la solubilité dans l’eau dépassait 0.055mM. Cependant la solubilité dans l’eau des drogues et leur log P ne sont pas les seuls paramètres influençant leur incorporation dans les micelles. Des paramètres structuraux tels que le nombre de groupes donneurs et d’accepteurs de liaisons hydrogènes dans la structure chimique de la drogue ont aussi démontré leur influence. De façon intéressante, les drogues de faible solubilité dans l’eau qui ne suivent pas la « règle des 5 » de Lipinski, souvent synonyme de faible biodisponibilité et d’applications limitées en clinique, possèdent des propriétés physico-chimiques appropriées pour une incorporation efficace dans les micelles de Summary (In French) 154 MPEG-hexPLA. Ceci suggère l’utilisation de ces systèmes micellaires comme formulation alternative pour des substances faiblement solubles dans l’eau. Pour de possibles utilisations pharmaceutiques, les micelles de MPEG-hexPLA ont été tout d’abord étudiées pour leur biocompatibilité. Aucune toxicité in vitro et in vivo ou activité hémolytique sur le sang humain n’a été trouvée, à la fois pour les unimères et pour les micelles polymériques jusqu’à une concentration de 20 mg/mL. Par conséquent, ces nouvelles formulations micellaires s’avèrent intéressantes pour des applications pharmaceutiques. L’application intraveineuse a été une des applications envisagées. En effet, les formulations micellaires de cyclosporine A n’ont montré aucune activité hémolytique et ont pu atteindre des doses thérapeutiques avec 4 fois moins d’excipient que le surfactant utilisé dans la formulation commerciale, Sandimmune ®. De plus, l’injection intraveineuse de formulations micellaires d’hypericine a présenté chez les rats atteints de tumeurs ovariennes une accumulation rapide et élevée dans les nodules tumoraux par un ciblage passif. Ainsi les micelles de MPEG-hexPLA peuvent être envisagées pour le diagnostique du cancer de l’ovaire. Une application topique pour des traitements antifungiques a également été évaluée dans cette thèse. En effet, les micelles de MPEG-hexPLA ont montré leur supériorité lors de la pénétration du nitrate d’éconazole à l’intérieur de la peau porcine et humaine par rapport à la formulation commerciale. Ce dépôt a été curieusement attribué à une pénétration des micelles par les follicules. Les micelles de MPEG-hexPLA peuvent être considérées comme vecteurs colloidaux potentiels pour la libération d’autres produits hydrophobes dans la peau. Une possible application par voie orale des micelles a aussi été étudiée. Après 1h à pH gastrique, les micelles de MPEG-hexPLA ont conservé environ 90% de leur contenu en cinnarizine et ont gardé leur structure de « cœur-couronne » intacte malgré une légère dégradation du copolymère. Les micelles s’avère donc suffisamment stables pour passer l’estomac et atteindre l’intestin, pour une application orale. En résumé, les micelles de MPEG-hexPLA présentent de prometteuses formulations pharmaceutiques biocompatibles pour des applications intraveineuses, topiques, et orales pour les substances faiblement solubles dans l’eau. ABBREVIATIONS Abbreviations Abbreviations ACN Acetonitrile BCS Biopharmaceutics classification system bFGF Basic fibroblast growth factor CAM Chick chorioallantoic membrane CDCl3 Chloroform-d CIN Cinnarizine CL ε-Caprolactone CLZ Clotrimazole CMC Critical micellar concentration CMT Critical micellar temperature CsA Cyclosporine A DAN Danazol DC Drug content DCM Dichloromethane DICLO Diclofenac sodium dihexPLA di-hexyl-substituted polylactides DLLA D,L-lactide D-PLA Poly(D-lactide) D,L-PLA Poly(D,L-lactide) DLS Dynamic light scattering DMAAm N,N-Dimethylacrylamide DMF Dimethylformamide dn Number- weighted diameter [%]dn Percentage of micelles with a given dn DSPE Distearoylphosphatidylethanolamine DTBA p-Aminobenzyldiethylenetriaminepenta(acetic acid) DTX Docetaxel ECZ Econazole nitrate EDD Embryo development day 155 Abbreviations 156 EGF Epidermal growth factor EPR Enhanced permeation and retention effect ETO Etoposide FDA Food and drug administration FLZ Fluconazole Gd Gadolinium GF Griseofulvin GI Gastro-intestinal GRAS “Generally Regarded As Safe” Ha H bond acceptor HCl Hydrochloric acid Hd H bond donnor HexPLA Hexyl-substituted poly(lactide) (hex)PLA Hexyl-substituted poly(lactide) and poly(lactide), respectively HTS High throughput screening HY Hypericin IE Incorporation efficiency KETO Ketoconazole LCST Lower critical solution temperature LLA L,L-lactide LogP Partition coefficient between octanol and water L-PLA Poly(L-lactide) MAAc Methacrylic acid MeOH Methanol MD Molecular dynamics monohexPLA mono-hexyl-substituted poly(lactide) MPEG Methoxy-poly(ethylene glycol) MPS Mononuclear phagocytic system MRI Magnetic resonance imaging MTD Maximum tolerated dose MTT 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide) Mw Molecular weight NaCl Sodium chloride Abbreviations NAPRO Naproxen sodium NR Nile Red PAsp Poly(aspartic acid) PBL Poly(butyrolactone) PBS Phosphate buffer saline PCL Poly(ε-caprolactone) PDLA Poly(D-lactide) PDLLA Poly(D,L-lactide) PDMA Poly[2-(N,N-dimethylamino)ethyl methacrylate] PDMAEMA Poly(2-(N,N-dimethylamino)ethyl methacrylate) PDPA Poly[2-(diisopropylamino)ethyl methacrylate] PDT Photodynamic therapy PEEP Poly(ethyl ethylene phosphate) PEG Poly(ethylene glycol) PEI Poly(ethylene imines) PG Poly(L-glutamic acid) PHis Poly(L-histidine) P.I. Polydispersity index PICM Polyion complex micelles PIRO Piroxicam PLA Poly(lactic acid) or poly(lactide) PLGA Poly(lactic-co-glycolic acid) PLLA Poly(L-lactide) PMMA Poly(methacrylic acid) PNIPAAm Poly(N-isopropylacrylamide) PPO Poly(propylene oxide) PS Poly(styrene) PTX Paclitaxel PVA Poly(vinyl alcohol) PVL Poly(valerolactone) PVP Poly(N-vinyl-2-pyrrolidone) QUER Quercetine dihydrate ROP Ring opening polymerization 157 Abbreviations SC Stratum corneum Sn(Oct)2 Tin octanoate THF Tetrahydrofuran THPP meso-Tetra(p-hydroxyphenyl)porphine VER Verteporfin Zav Hydrodynamic diameter 158