Rapamycin-loaded solid lipid nanoparticles: Morphology and impact of the drug loading on the phase transition between lipid polymorphs

https://doi.org/10.1016/j.colsurfa.2016.05.017Get rights and content

Highlights

  • Cold high-pressure homogenization and ultrasound-assisted emulsion were used.

  • The stability and drug content varied with regard to the method applied.

  • Modeling of the elliptical plate and flat ellipsoid SLN shapes.

  • The reduction of the phase transition point in the lipid matrix by the drug.

Abstract

In recent decades solid lipid nanoparticles (SLN) have become a well-performing tool for the site-targeted delivery of water-insoluble drugs. In this study, Compritol® 888 ATO-based SLN, coated with polysorbate 80, were loaded with rapamycin (Rap), a lipophilic immunomodulator, broadly-used in therapies of cancer and neurodegenerative diseases. Rap-SLN were formulated using cold high-pressure homogenization and ultrasound-assisted emulsion. The exploitation of these methods yielded the nanoparticles of various values of zeta-potential (from −1 mV to −20 mV) and efficacies of the Rap entrapment (from 37.5 ± 2.3% to 77.0 ± 5.4%). The SEM and AFM imaging and shape-modeling by the combined DLS-SANS analysis revealed that the Rap-SLN of the hydrodynamic radius of ∼46 nm preserve the platelet-like or flat ellipsoidal structure with a thickness as large as 8–9 nm. These dimensions correspond to a single lipid bilayer, organized in a triclinic Lβ polymorph, and covered with a 1–2-nm shell of the surfactant. Consistently, FT-IR spectra acquired in the range 52–75 °C, showed that the Rap incorporation within the lipid matrix decreases the point of the gel-liquid crystalline (Lβ-Lα) phase transition. These outcomes imply a thermodynamically-driven mechanism of the Rap release from SLN.

Introduction

In recent decades great attention has been drawn to the preparation of solid lipid nanoparticles (SLN) as a drug delivery system. SLN are formulated from a lipid matrix that preserves the solid state at room and physiological temperatures. Particularly, SLN refer to colloidal dispersions made of various lipids (waxes, fatty acids, glycerides), stabilized with surfactant on the surface, and are usually produced by using high pressure homogenization, microencapsulation and emulsions methods [1]. In comparison to widely-used polymeric nanovehicles, SLN display a series of advantages, such as: improvement of biocompatibility and bioavailability of lipophilic drugs, or fast, inexpensive and scalable manufacture [2]. Specifically, SLN based on Compritol® 888 ATO, which constitutes a physical mixture of mono-, di-, and triglycerides of behenic acid, have been reported to facilitate both transportation and therapeutic activity of a number of water-insoluble or lipophilic drugs, dedicated to various diseases of affluence, targeted for instance to central nervous system [3], cancer [4], inflammation [5], or gene therapy [6]. Such formulations can be delivered to the body in a form of oral [7], topical [8], parenteral [9], ocular [10] and intestinal lymphatic [11] administration. Up to now, these nanocarriers have been extensively studied in terms of the drug incorporation influence on the lipid matrix, thermal stability, cytotoxicity, biodistribution and biodegradation in both in vitro and in vivo models.

Compritol® 888 ATO (thereafter named Compritol) displays an evident hydrophobic nature, expressed by a hydrophilic-lipophilic balance value of 2 and relatively high melting temperature in the range of 64–74 °C [12], [13]. Compritol is soluble in chloroform and dichloromethane, when heated, slightly soluble in hot (but not cold) 96% ethanol, and insoluble in hexane, mineral oils and water. The feasibility of the industrial application of this non-toxic lipid composite, belonging to GRAS specimens and FDA Inactive Ingredients Database, has been widely applied to cosmetics, foods and pharmaceuticals, formulated as oral capsules, tablets and suspensions. In terms of structural composition, Compritol consists of monobehenin (∼20%), dibehenin (∼50%) and tribehenin (∼30%). Upon the dissolution, each of these ingredients preserves an individual pattern of recrystallization [12], [13]. During this process, followed by storage, the Compritol-based SLN initially crystallize in the non-stable hexagonal Lα-form and in the metastable orthorhombic perpendicular Lβ′ or stable triclinic parallel Lβ forms. The prevalence of one particular lamellar structure over the others depends on the size, shape and the physical state of the SLN formulation, considering that the rate of phase transition is known to be more sensitive for colloidal dispersion than for the bulk materials [12]. The polymorphism of Compritol originates from the various lateral packing of fatty acid chains: the Lα and Lβ′ polymorphs are remarkable for a mixture of long-chained 1,2-diglycerides, whilst the Lβ polymorph is most suitable for monoacid 1,3-di- and triglycerides [12].

In this study, Compritol SLN were loaded with rapamycin (Rap), an antifungal agent, naturally produced by Streptomyces hygroscopicus and collected for the first time in 1965 [14]. The activity of Rap comprises the specific inhibition of the mammalian target of rapamycin (mTOR) kinase [15]. Upon the activation by extracellular stimulation, mediated by nutrients, growth factors or stress conditions, the mTOR signaling pathway responds via the regulation of cellular growth, proliferation, metabolism and survival [16]. Ongoing studies indicate that the Rap-triggered inhibition of mTOR plays a pivotal role in the reduction of causes and effects of neurodegeneration [17], cardiovascular diseases [18], tumorigenesis or cancer [19], inflammatory diseases [20] and immunological response [21]. Nevertheless, given that Rap is a macrolydic immunosuppressive agent that possesses no ionizable groups in the pH range 1–10, displays poor solubility [22], high hydrolysis-potency in aqueous solution [23], and high affinity to red blood cells and other blood components [24], the oral bioavailability and the clinical applicability of Rap are extremely limited. Therefore, much effort has been made to improve the Rap delivery into human tissues. These are mainly: (i) enhancement of Rap solubility by hydrotropy strategy [22], or (ii) incorporation of drug molecules inside various nanocarriers, mostly based on phospholipid [25] or polymeric micelles [26], nanoparticles [27] and liposomes [28]. The exceptional compliance of SLN consigned to the brain-targeted delivery of Rap, is expected to attenuate the severe immunosuppressive effect of the drug in an organism, observed upon the intravenous administration, and to prolong the therapeutic, mTOR-specific activity in central nervous system after the topical administration or following the blood-brain barrier crossing.

The detailed investigation of the surface quality of Rap-SLN, stabilized with polysorbate 80 (PS80), was achieved by means of electron and atomic force microscopy (SEM, AFM). Additionally, the morphology was studied using light and small-angle neutron scattering (DLS, SANS), since only several lipid-based drug delivery systems have been investigated by SANS, until now [29], [30]. Such combined analysis evidenced that Rap-SLN shape is platelet-like rather than spheroidal. The stability of the nanoformulation was evaluated by measurements of mean hydrodynamic diameter, zeta-potential and polydispersity indexes, whereas the Rap content in SLN was analyzed both quantitatively and qualitatively by means of UV spectrophotometry and Raman spectroscopy, respectively. Eventually, the effect of the Rap loading on the phase transition in lipid matrix was investigated by recording FT-IR spectra in the temperature range of 52–75 °C. Taken together, these results suggest considerable contribution of the thermodynamically-driven processes leading to the Rap release from SLN.

Section snippets

Materials

Rapamycin (Rap, Sirolimus, CAS No.: 53123-88-9, purity ≥98%) was obtained from Ningbo Heyreal Import&Export Co., Ltd, China. Compritol® 888 ATO (Compritol, glyceryl behenate, 99%) was acquired from Gattefosse, Milan, Italy. Polysorbate 80 (PS80, Tween® 80, polyoxyethylene sorbitan monooleate), deuterium oxide (D2O, heavy water, 99.98%), chloroform (CHCl3, 99.5%) and dimethyl sulfoxide (DMSO, 99.9%) were from Sigma Aldrich (Poznań, Poland and Milan, Italy). No additional purification was

Influence of the preparation method on drug content and short-term stability

The Compritol-based aqueous dispersions of blank SLN and Rap-SLN, stabilized with PS80 surfactant (Fig. 1), were prepared using two methods: cold high-pressure homogenization (CHPH) and ultrasound-assisted emulsion (UAE). The formulation procedures followed the well-established protocols, described in details elsewhere [7], [31]. Table 1. presents the methodologies, preliminary characteristics and the efficacy of the Rap encapsulation in SLN. The CHPH method employed a 5-cycle long

Conclusions

Rapamycin-loaded Compritol-based SLN were prepared for the first time by using a cold high-pressure homogenization and ultrasound-assisted emulsion methods. Spectroscopic analysis proved the efficient drug loading within SLN. Further investigation of the chemical interactions between Rap and glyceryl behenate molecules is required to comprehend the accurate mechanism of the drug entrapment. FT-IR spectra, recorded in the range of 52–75 °C, showed that the Rap incorporation between the fatty acid

Conflict of interest

The authors declare no conflicts of interests.

Acknowledgements

This project was supported by the International PhD Project Program, Foundation for Polish Science operated within the Innovative Economy Operational Programme 2007–2013 within European Regional Development Fund, and the grant PBS1/A9/13/2012 of the National Centre for Research and Development. The work was also supported by Fondazione Cassa di Risparmio di Perugia (Grant no. 20140222.021).

References (75)

  • K. Hallensleben et al.

    Identification of a new metabolite of macrolide immunosuppressant like rapamycin and SDZ RAD, using high performance liquid chromatography and electrospray tandem mass spectrometry

    J. Am. Soc. Mass Spectrom.

    (2000)
  • Y.C. Chen et al.

    Rapamycin encapsulated in dual-responsive micelles for cancer therapy

    Biomaterials

    (2013)
  • H. Bunjes et al.

    Characterization of lipid nanoparticles by differential scanning calorimetry, X-ray and neutron scattering

    Adv. Drug Deliv. Rev.

    (2007)
  • A. Kovacevic et al.

    Polyhydroxy surfactants for the formulation of lipid nanoparticles (SLN and NLC): effects on size, physical stability and particle matrix structure

    Int. J. Pharm.

    (2011)
  • P. Blasi et al.

    Lipid nanoparticles for brain targeting I. Formulation optimization

    Int. J. Pharm.

    (2011)
  • P. Blasi et al.

    Lipid nanoparticles for brain targeting III. Long-term stability and in vivo toxicity

    Int. J. Pharm.

    (2013)
  • D.J. Trepanier et al.

    Rapamycin: distribution, pharmacokinetics and therapeutic range investigations: an update

    Clin. Biochem.

    (1998)
  • M. Ricciutelli et al.

    Evaluation of rapamycin chemical stability in volatile-organic solvents by HPLC

    J. Pharm. Biomed. Anal.

    (2006)
  • M. Kamberi et al.

    UV-visible spectroscopy as an alternative to liquid chromatography for determination of everolimus in surfactant-containing dissolution media: a useful approach based on solid-phase extraction

    J. Pharm. Biomed. Anal.

    (2012)
  • A. Noack et al.

    Physicochemical characterization of curcuminoid-loaded solid lipid nanoparticles

    Int. J. Pharm.

    (2012)
  • A. Belu et al.

    Chemical imaging of drug eluting coatings: combining surface analysis and confocal Raman microscopy

    J. Controlled Release

    (2008)
  • Y.C. Kuo et al.

    Entrapment and release of saquinavir using novel cationic solid lipid nanoparticles

    Int. J. Pharm.

    (2009)
  • N. Passerini et al.

    Solid lipid microparticles produced by spray congealing: influence of the atomizer on microparticle characteristics and mathematical modeling of the drug release

    J. Pharm. Sci.

    (2010)
  • S. Das et al.

    Formulation design, preparation and physicochemical characterizations of solid lipid nanoparticles containing a hydrophobic drug: effects of process variables

    Colloids Surf. B

    (2011)
  • J. You et al.

    Preparation and characteristic of vinorelbine bitartrate-loaded solid lipid nanoparticles

    Int. J. Pharm.

    (2007)
  • P. Shahgaldian et al.

    AFM imaging of calixarene based solid lipid nanoparticles in gel matrices

    Eur. J. Pharm. Biopharm.

    (2003)
  • C. Vitorino et al.

    The size of solid lipid nanoparticles: an interpretation from experimental design

    Colloids Surf. B

    (2011)
  • S. Kumar et al.

    Preparation and characterization of Paliperidone loaded solid lipid nanoparticles

    Colloids Surf. B

    (2013)
  • N. Seetapan et al.

    Rheological and morphological characterizations on physical stability of gamma-oryzanol-loaded solid lipid nanoparticles (SLNs)

    Micron

    (2010)
  • A. Illing et al.

    Investigation on the flow behavior of dispersions of solid triglyceride nanoparticles

    Int. J. Pharm.

    (2004)
  • K. Gao et al.

    Influence of particle size on transport of methotrexate across blood brain barrier by polysorbate 80-coated polybutylcyanoacrylate nanoparticles

    Int. J. Pharm.

    (2006)
  • E. Banachowicz et al.

    Solution structure of biopolymers: a new method of constructing a bead model

    Biophys. J.

    (2000)
  • S.M. Martins et al.

    Brain targeting effect of camptothecin-loaded solid lipid nanoparticles in rat after intravenous administration

    Eur. J. Pharm. Biopharm.

    (2013)
  • R.N. Lewis et al.

    Membrane lipid phase transitions and phase organization studied by Fourier transform infrared spectroscopy

    Biochim. Biophys. Acta

    (2013)
  • R.N. Lewis et al.

    Studies of highly asymmetric mixed-chain diacyl phosphatidylcholines that form mixed-interdigitated gel phases: Fourier transform infrared and 2H NMR spectroscopic studies of hydrocarbon chain conformation and orientational order in the liquid-crystalline state

    Biophys. J.

    (1994)
  • K. Priyanka et al.

    Preparation and evaluation of montelukast sodium loaded solid lipid nanoparticles

    J. Young Pharm.

    (2012)
  • Z. Rahman et al.

    Non-destructive methods of characterization of risperidone solid lipid nanoparticles

    Eur. J. Pharm. Biopharm.

    (2010)
  • Cited by (26)

    • Chitosan hydrogel for controlled crystallization of loaded drug: Role of interplay of assembly processes

      2022, Colloids and Surfaces A: Physicochemical and Engineering Aspects
      Citation Excerpt :

      In order to minimize the risk of phase transition, it is desirable to choose the most stable form of crystalline phase in the initial stages of drug development. However, using stable form may result in low solubility, dissolution rate and bioavailability [5]. In that case, the metastable form or their amorphous counterparts with stable kinetics at room or lower temperature are usually considered to be ideal to market [6].

    • Streptomycin sulphate loaded solid lipid nanoparticles show enhanced uptake in macrophage, lower MIC in Mycobacterium and improved oral bioavailability

      2021, European Journal of Pharmaceutics and Biopharmaceutics
      Citation Excerpt :

      The conformational disordering of an all-trans polymethylene chain is accompanied by an upward shift in the sνCH2 IR band maxima by almost 8 cm−1 in STRS-SLNs. This reflects a probable hydrocarbon chain conformational disorder [44]. The 2918 and 2850 cm−1 IR absorption bands indicate hydrocarbon phase transitions, but the 2850 cm−1 band is invariably used to define such transitions, because the asνCH2 (corresponding to 2918 cm−1) band also contains contributions from CH3 groups.

    • Immunogenicity evaluating of the SLNs-alginate conjugate against Pseudomonas aeruginosa

      2021, Journal of Immunological Methods
      Citation Excerpt :

      The chemical structures of SLNs and Alg-SLN conjugate were characterized using a Fourier-transform infrared (FTIR) spectrometer (Model: Tensor27, Manufacturer: BRUKER, Country: Germany). All FTIR spectra were obtained in the spectral region of 500–4000 cm−1(Mazuryk et al., 2016; Singh et al., 2016). The mice were divided into 4 groups of 25 each.

    View all citing articles on Scopus
    View full text