+ All Categories
Home > Documents > Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

Date post: 02-Jun-2018
Category:
Upload: esteban-araya
View: 218 times
Download: 0 times
Share this document with a friend

of 9

Transcript
  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    1/9

    International Journal of Pharmaceutics 447 (2013) 115123

    Contents lists available at SciVerse ScienceDirect

    InternationalJournal ofPharmaceutics

    journal homepage: www.elsevier .com/ locate / i jpharm

    Pharmaceutical Nanotechnology

    Systemic heparin delivery by the pulmonary route using chitosan and glycolchitosan nanoparticles

    Adriana Trapani a,1, Sante Di Gioia b,1, Nicoletta Ditaranto c, Nicola Cioffic, Francisco M. Goycoolead,Annalucia Carboneb, Marcos Garcia-Fuentes e, Massimo Coneseb, MariaJose Alonso e,

    a Department of Pharmacy-Drug Sciences,University of Bari AldoMoro, ViaOrabona, 4, 70125 Bari, Italyb Department ofMedical and Surgical Sciences, University of Foggia, Viale L. Pinto 1, 71122 Foggia, Italyc Department of Chemistry, University of Bari AldoMoro,Via Orabona 4, 70125 Bari, Italyd Institute of Plant Biology and Biotechnology, Westphalian Wilhelms UniversityMnster, Schlossgarten 3, D-48149Mnster, Germanye Centerfor Research in MolecularMedicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela(IDIS), Department of Pharmacy and Pharmaceutical

    Technology, School of Pharmacy,University of Santiago de Compostela, 15782 CampusVida, Santiago de Compostela, Spain

    a r t i c l e i n f o

    Article history:

    Received 28 December 2012

    Received in revised form 12 February 2013

    Accepted 13 February 2013

    Available online 20 February 2013

    Keywords:

    Low molecular weight heparin

    Lipoid

    Chitosan-and glycolchitosan-nanoparticles

    Lung delivery

    a b s t r a c t

    The aim ofthis study was to evaluate the performance ofchitosan (CS) and glycol chitosan (GCS) nanopar-

    ticles containing the surfactant Lipoid S100 for the systemic delivery of low molecular weight heparin

    (LMWH) upon pulmonary administration. These nanoparticles were prepared in acidic and neutral con-

    ditions using the ionotropic gelation technique. The size andzeta potential ofthe NPs were affected by the

    pHand also the type ofpolysaccharide (CS or GCS). The size (between 156 and 385 nm) was smaller and

    the zeta potential (from +11 mV to +30 mV) higher for CS nanoparticles prepared in acidic conditions. The

    encapsulation efficiency ofLMWH varied between 100% and 43% for the nanoparticles obtained in acidic

    and neutral conditions, respectively. X-ray photoelectron spectroscopy studies indicated that the surfac-

    tant Lipoid S100 was localized on the nanoparticles surface irrespective ofthe formulation conditions.

    In vivo studies showed that systems prepared in acidic conditions did not increase coagulation times

    when administered to mice by the pulmonary route. In contrast, Lipoid S100-LMWH GCS NPs prepared

    in neutral conditions showed a pharmacological efficacy. Overall, these results illustrate some promisingfeatures ofCS-based nanocarriers for pulmonary delivery ofLMWH.

    2013 Elsevier B.V. All rights reserved.

    1. Introduction

    Low molecular weight heparin (LMWH) is a linear anionic

    polysaccharide used as an anticoagulant for the prevention and

    treatment of deep veinthrombosis,pulmonary embolismand other

    thromboembolic disorders (Schulman, 2000; Yang et al., 2004).

    Unfortunately, LMWH exhibits poor oral bioavailability and, con-

    sequently, has to be administered via parenteral route. Due to

    poorpatientcompliance andside effects associatedwith injections,

    alternative routes for non-invasive LMWH administration have

    been actively investigated (Motlekar and Youan, 2006). Amongthose, the pulmonary route has attracted notable interest as a

    potential strategy to deliver therapeutically useful amounts of the

    anticoagulant (Qi et al., 2004). The large alveolar surface area avail-

    able for drug absorption, the low thickness of the epithelial barrier,

    its extensive vascularization and relatively low proteolytic activity

    make pulmonary delivery of drugs of particular interest even for

    Corresponding author. Tel.: +34 881815454; fax: +34 881815403.

    E-mail address:[email protected](M.J. Alonso).1 These authors contributed equally to this work.

    chronic therapy (Hussain et al., 2003; Labiris and Dolovich, 2003;

    Craparo et al.,2011; Licciardi et al.,2012). Moreover,it hasalsobeen

    observedthat LMWHitself causes, uponpulmonaryadministration,

    a transient opening of the tight junctions in the lung epithelium,

    leading to a rapid onset of action and a Cmaxcomparable to subcu-

    taneous administration (Qietal.,2004). Nevertheless, beyond these

    positive aspects, it is believed that, without the use of penetration

    enhancers and adequate delivery vehicles, the amount of LMWH

    overcoming the pulmonary barriers and reaching the systemic cir-

    culation might be insufficient for an adequate pharmacological

    response.Among the differentpulmonary drug delivery vehicles,chitosan

    (CS)-basednanoparticles areparticularlyattractive.Indeed,besides

    promising properties including low toxicity, biocompatibility and

    high loading of hydrophilic molecules (Garcia-Fuentes and Alonso,

    2012; Ieva et al., 2009), CS shows excellent mucoadhesive charac-

    teristicsand it is capable of opening the tight junctions of epithelial

    cells, thereby improvingthe uptake of hydrophilic drugs (Mao et al.,

    2010). A CS derivative conjugated with ethylene glycol branches,

    i.e., glycol chitosan (GCS), which is water soluble at neutral and

    acidic pH values, has also been described (Siew et al., 2012). In

    addition to its adequate biocompatibility, GCShas been reported to

    0378-5173/$ seefrontmatter 2013 Elsevier B.V. All rights reserved.

    http://dx.doi.org/10.1016/j.ijpharm.2013.02.035

    http://localhost/var/www/apps/conversion/tmp/scratch_2/dx.doi.org/10.1016/j.ijpharm.2013.02.035http://localhost/var/www/apps/conversion/tmp/scratch_2/dx.doi.org/10.1016/j.ijpharm.2013.02.035http://www.sciencedirect.com/science/journal/03785173http://www.elsevier.com/locate/ijpharmmailto:[email protected]://localhost/var/www/apps/conversion/tmp/scratch_2/dx.doi.org/10.1016/j.ijpharm.2013.02.035http://localhost/var/www/apps/conversion/tmp/scratch_2/dx.doi.org/10.1016/j.ijpharm.2013.02.035mailto:[email protected]://www.elsevier.com/locate/ijpharmhttp://www.sciencedirect.com/science/journal/03785173http://localhost/var/www/apps/conversion/tmp/scratch_2/dx.doi.org/10.1016/j.ijpharm.2013.02.035
  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    2/9

    116 A. Trapani et al./ International Journal of Pharmaceutics 447 (2013) 115123

    retain the mucoadhesive properties inherent to CS (Trapani et al.,

    2009; Makhlof et al., 2010).

    There are some previous reports on the potentialof CS nanopar-

    ticles for the local and systemic delivery of macromolecules

    following pulmonary administration. For example, nanoparticles

    made of CS and hyaluronic acid (HA) have been used for local

    heparin delivery to the lungs (Oyarzun-Ampuero et al., 2009). The

    results of this work have shown that CSHA nanoparticles are able

    to enhance the inherent ability of heparin to block the degranula-

    tion of mast cells (Oyarzun-Ampuero et al., 2009). Similarly, NPs

    made of CS or GCS have been recently described for pulmonary

    delivery of DNA (Bivas-Benita et al., 2004) and therapeutic pep-

    tides such as insulin (Al-Qadi et al., 2012) or calcitonin (Makhlof

    et al., 2010).

    Taking this into account, the aim of the present work was to

    develop CS and GCS nanoparticles containing LMWH and to eval-

    uate their performance following pulmonary administration. In

    addition, we found it critical to incorporate the non ionic surfac-

    tant Lipoid S100 to the nanoparticles structure. Our hypothesis

    was that the co-encapsulation of LMWH and the penetration

    enhancer Lipoid S100 in CS and GCS nanoparticles would favour

    for the pulmonary absorption of macromolecules (Hussain et al.,

    2003). Moreover, Lipoid S100, being a mixture of natural phos-

    pholipids, was thought to improve the biocompatibility of thenanosystems in contact with the alveolar surface. Ultimately, we

    hypothesized that, by combining the mucoadhesive characteristics

    of the polymers with the nanoscale dimensions and the absorp-

    tion enhancing propertiesof the surfactant and polymers, we could

    significantly enhance the pulmonary absorption of LMWH. In addi-

    tion, nanoencapsulation was also conceived as a way to protect the

    anticoagulant drug from possible enzymatic degradation.

    2. Materials and methods

    2.1. Materials

    The following chemicals were used as received. Chitosanhydrochloride salt(Protasan, UP CL 113,Mw 110kDa, deacetylation

    degree 86%, viscosity = 13mPa/s according to manufacturer data

    sheet) was purchased from Pronova Biopolymer (Norway). Lipoid

    S100 was kindlydonated by Lipoid KG (Germany). LMWH (average

    MW 18kDa, 177 UI/mg), glycolchitosan (MW400 kDaaccording to

    supplier instructions), mucine fromporcine stomach(type II,bound

    sialic acid, 1%), glycerol, and pentasodium tripolyphospate (TPP)

    werepurchased from SigmaAldrich (Milan, Italy). Ultrapurewater

    was used throughout the study. All other standard chemicals were

    of reagent grade.

    2.2. Analytical determination of low molecular weight heparin

    The heparin amounts were directly measured using two col-orimetric assays (i.e., Stachrom Heparin, Diagnostica Stago, Italy

    (Oyarzun-Ampuero et al., 2009) and Azure A colorimetric method

    (Ramadan et al., 2011). These assays were performed according to

    the manufacturers instructions and linearity was checked in the

    140g/mL to 1.0101g/mL concentration range. For Azure Acolorimetric assay, the limit of quantification (LOQ) of LMWH was

    0.9101 g/mL.

    2.3. Preparation of nanoparticles

    LMWH loaded CS or GCS-based NPs were prepared according

    to the ionic gelation technique, as previously reported (Ieva et al.,

    2009; Mao et al., 2010; Calvo et al., 1997).

    2.4. Unloaded CS (and GCS) NPs

    CS NPs were spontaneously formed by adding1 mLof TPP (0.1%,

    w/v, in NaCl aqueous solution (87 mM)) to 3 mL of CS solution

    (0.20% (w/v) in NaCl aqueous solution (87mM) under magnetic

    stirring (VWR, VMS C-4, Italy). Such concentration of NaCl was pre-

    viously found to allow the formation of small size CS-based NPs

    (Goycoolea et al., 2009). The final CS/TPP mass ratio was 6/1. GCS

    NPs were obtained by mixing 3 mLof TPP aqueous solution (0.07%,

    w/v) to 3mLof GCS (0.2%, w/v) dissolved in acetic acid (0.1%, v/v).

    ThefinalGCS/TPP ratio was2.9/1(Ganet al., 2005). For Lipoid S100-

    containing CS (and GCS) NPs, the surfactant was dispersed to give a

    final concentration of 0.1% (w/v) in 87mM NaCl aqueous solution.

    Lipoid S100 CS NPs were prepared by mixing 0.5 mLof TPP (0.2%

    (w/v) in 87mM NaCl aqueous solution) with an aliquot of 0.2 mL

    of the dispersion of Lipoid S100. The resulting mixture was used to

    crosslink 3 mLof CS solution (0.20% (w/v) in 87mM NaCl aqueous

    solution) under stirring, and thus NPs were formed.

    For GCS based NPs (i.e., Lipoid S100 GCS pH 4.3), 3mLof TPP

    aqueous solution (0.07%, w/v) were mixed with an aliquot of 0.1mL

    of the dispersion of Lipoid S100. The resulting mixture was used to

    cross-link 3 mLof GCS solution (0.20%, w/v) previously dissolvedin

    acetic acid (0.1%, v/v).

    2.5. LMWH loaded NPs

    Lipoid S100-LMWH CS NPs were formulated starting from the

    anionic bearing species consisting of 0.2mLof Lipoid S100, 0.2 mL

    of LMWH solution and 0.5 mLof TPP (0.2%, w/v, in 87mM NaCl

    aqueous solution). Such mixture was used to cross-link 3 mL of CS

    solution (0.20%, w/v, in 87mM NaCl aqueous solution).

    Lipoid S100-LMWH GCS NPs were formulated starting from

    the anionic bearing species consisting of 0.4mL of Lipoid S100,

    0.2mL of LMWH solution and 1.8 mL of TPP (0.07%, w/v). The

    resulting mixture was used to cross-link 3mL of GCS solution

    (0.20%).

    It should be noted that all the nanosuspensions prepared as

    above displayed acidic pH values (i.e., 3.8 and 4.3 for Lipoid S100-

    LMWH CS and Lipoid S100-LMWH GCS, respectively).We have also

    prepared Lipoid S100-LMWH GCS NPs uponneutral conditions. The

    correspondingLipoid S100-LMWHCS NPs were notformulated due

    to the poor water solubility of CS at pH> 6.5. Lipoid S100-LMWH

    GCS NPs were formed as follows. Briefly, 1.5mLof TPP aqueous

    solution (0.07%, w/v) were mixed with an aliquot of 0.52mLof the

    dispersion of Lipoid S100 andan aliquot of 0.52 mLof LMWH aque-

    ous solution. The resulting mixture was used to cross-link 8.0 mL

    of GCS solution (0.20%, w/v) previously dissolved in Tris (10 mM,

    pH 7).

    NPs were isolated by ultracentrifugation (16,000g, 45min,

    Eppendorf 5415D, Germany) using a glycerol bed in order

    to facilitate their resuspension in ultrapure water by manual

    shaking.

    2.6. Physicochemical characterization of nanoparticles

    For all tested NPs, the mean particle size and distribution were

    determined in double distilled water by photon correlation spec-

    troscopy (PCS) using a Zetasizer NanoZS (ZEN 3600, Malvern,

    Herrenberg, Germany). Measurements were performed in trip-

    licate after dilution 1:1 (v/v) in double distilled water and at

    25 C. The autocorrelation functions were analyzed using the DTS

    v. 5.1 software provided by Malvern. For the determination of

    zeta-potential values, a laser Doppler velocimetry was adopted

    by using Zetasizer NanoZS after dilution with 1mM KCl (pH 7.0)

    (Montenegro et al., 2012; De Giglio et al., 2012).

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    3/9

    A. Trapani et al. / International Journal of Pharmaceutics447 (2013) 115123 117

    2.7. Atomic force microscopy (AFM)

    AFM studies were performed to characterize the morphology

    of LMWH loaded CS NPs. The AFM images were obtained using

    a Ntegra Prima AFM, on air in semi-contact mode with standard

    Si cantilever with 316kHz resonant frequency. Scan speed was

    between 2 and 3 Hz, (mean 23 lines per second). All images are

    512512 data points. For AFM visualization, a drop of nanoparti-

    cle suspension was spin-coated onto a flat gold thin film deposited

    on silicon substrate, then the sample was processed directly on the

    AFM scanner. To achieve high-resolution images of nanoparticles

    deposited on the substrate, NSG03 silicon cantilevers with a length

    of 135m were used with resonance frequency of about 90kHzand a nominal force constant around 1.8 N/m. The tip amplitude

    oscillations were kept constant in attractive forces regimen to pre-

    vent thedamage of theNPs. Thescan speed was proportional to the

    scan size with a typical scanning frequency of 23 Hz.

    2.8. Association efficiency of NPs

    The association efficiency (AE) of LMWH to the particles was

    determined after isolation of NPs by centrifugation as described in

    Section 2.3.

    AE was calculated as follows:

    %AE = 100total LMWH free LMWH

    total LMWH

    where the total amount of LMWH and the unbound amount of

    LMWH in the supernatant was determined using the colorimetric

    assays above mentioned. The association efficiency was deter-

    mined in triplicate.

    2.9. XPS analysis

    The surface chemical composition was studied by means of X-

    ray Photoelectron Spectroscopy (XPS). Prior to their analysis, the

    NPs formulations (Lipoid S100 LMWH GCS NPs and Lipoid S100

    LMWH CS NPs) were converted into powder using freeze-drying.XPS analyses were performed with a Theta Probe VG Scientific

    spectrometer equipped with a micro spot monochromatised Al Ksource (1486.6eV; spot size: 300m). Wide scans and detailedspectra were acquired in constant analyser energy (CAE) mode

    with a pass energy of 150eV and 100eV, respectively. For non-

    conductive samples, a low energy flood gun (1eV) was used

    for charge compensation. Calibration of the binding energy (BE)

    scale was performed by fixing the C C component at BE values

    of 284.80.1eV. Data were averaged over at least three points

    per sample. Acquisition and data processing were performed using

    Avantage software v. 5.74.

    2.10. Stability study of LMWH loaded NPs in different media

    Freshly prepared LMWH loaded CS and GCS NPs were resu-

    pended at 0.1% (w/v) concentration in normal saline (NaCl, 0.9%,

    pH 7; 300mOsm/kg). To evaluate the stability of the NPs under

    differentincubation conditions, the systems wereincubated in nor-

    mal saline for 12 weeks at 4 C, for 1 week at 25 C and for 4 h at

    37 C. Particle size of the samples was determined at scheduled

    time intervals.

    2.11. Mucoadhesion tests

    The mucoadhesive interaction between LMWH loaded NPs and

    mucin was determined recording the variations of zeta poten-

    tial according to the method reported in the literature (Takeuchi

    et al., 2005; Jintapattanakit et al., 2009; Das Neves et al., 2011).

    Briefly, commercially available porcin mucin was firstly suspended,

    at room temperature,in PBS(pH 7.4) at 1%(w/v) concentration.One

    milliliter of this suspension was mixed with different volumes of

    the NP suspension tested at 1 mg/ml. The resulting mixtures were

    stirred (100rpm) at 37C for 15 m in and then the zeta potential

    was measured.

    2.12. In vitro release studies

    In vitro release studies from Lipoid S100-LMWH GCS NP for-

    mulated in neutral conditions were carried out up to 24 h in

    simulated lung fluid (SLF) (Moss, 1979; Davies and Feddah, 2003).

    The composition of SLF is: 5.0mEq/L CaCl2, 1.0mEq/L MgCl2,

    1.0mEq/L MgSO4 , 1.0mEq/L K2HPO4, 1.0 mEq/L KHCO3, 1.0 mEq/L

    KCl, 7.0mEq/L sodiumacetate, 102.0mEq/L NaCl, 1.0mEq/L sodium

    citrate, 31.0mEq/L NaHCO3, and 1.0 mEq/L Na2HPO4. This medium

    had pH 7.4 and an osmolarity of 300mOsm/kg. Briefly, NPs were

    freshly prepared and then centrifuged in presence of 10L of glyc-erol.For eachexperiment,appropriate volumes of nanodispersions,

    corresponding to an amount of LMWH in the range 2070g/mL,were put in Eppendorf tubes containing 0.5mL of SLF. The samples

    were incubated under mechanical agitation (100rpm) at 37 C. At

    scheduledtime intervals(0, 4, 6, 15 and 24h) each samplewas cen-

    trifuged at 16,000gfor 45min. The supernatant was analyzed forthecontent of LMWH usingAzure A spectrophotometric assay. The

    experiment was performed in triplicate.

    2.13. In vivo studies

    Buffer (TrisHCl, pH 7.0), LMWH or Lipoid S100-LMWH GCS

    NPs formulated in neutral conditions were administered to Swiss

    mice (25g, Charles River, Calco, Italy) using a MicroSprayerTM

    aerosoliser (IA-1C; Penn Century, Philadelphia, PA, USA) suit-

    able for mice, attached to a high-pressure syringe (FMJ-250;

    Penn Century). The mice were anaesthetized by an intraperitoneal

    injection of 0.5 mg/g body weight Avertin (2,2,2-tribromoethanol;

    SigmaAldrich, Germany)and weresuspendedat a 45 anglebythe

    upper teeth. The light sources (lamp type FLQ85E; Helmut Hund,Germany)flexible fiber-optics arm was adjusted to provide optimal

    illumination of the trachea. A small spatula was used to open the

    lower jaw of the mouse and blunted forceps were used to help dis-

    place the tongue for maximal oropharyngeal exposure. Aftera clear

    view of the trachea was obtained by an otoscope, the MicroSprayer

    tip was endotracheally inserted until it reached the carina (the first

    bronchial bifurcation) and 100L of solution (buffer or LMWH) orNP suspension was sprayed. The tip was immediately withdrawn

    and the mouse was taken off the support.

    2.13.1. Anticoagulation study

    Anticoagulation was studied with activated partial thrombo-

    plastin time (APTT). Citrate-anticoagulated blood was obtained 4 h

    after administration of buffer, LMWH or NPs. Plasma was com-bined with activated partial thromboplastin time (APTT) reagent

    (Beckman-Coulter/Instrumentation Laboratory)for three min prior

    to activation with calcium chloride and mechanical determination

    of coagulation time. At least 3 mice per group were analysed.

    2.13.2. Histopathological study

    Mice received the same treatment described above. Positive

    control mice were administered with aerosolised Lipopolysaccha-

    ride (LPS) from Pseudomonasaeruginosa (20g/mouse). Mice weresacrified 4 h after each treatment, lungs collected and inflated with

    1 mL of 10% neutral buffered formalin and fixed for a minimum

    period of 24h. Following fixation, lungs were placed in tissue cas-

    sette. These tissues were processed into paraffin, embedded in a

    paraffin block, sectioned on a microtome to a thickness of 5m,

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    4/9

    118 A. Trapani et al./ International Journal of Pharmaceutics 447 (2013) 115123

    Table 1

    Physicochemical properties of LMWH loaded CS- and GCS-based NPs. PI: polydispersity index. MeanS.D. are reported, n= 6. Control nanoparticles were unloaded CS and

    unloaded GCS, respectively).

    NPs formulation Size (nm) PI Zeta potential (mV) Yield (%) Association efficiency (%)

    CS pH 5.6 166 (21) 0.210.26 +15.2 (3.4) 64.8 (7.0)

    Lipoid S100 CS pH 3.8 209* (18) 0.310.38 +17.2 (2.5) 59.9 (13.1)

    Lipoid S100-LMWH CS pH 3.8 280** (35) 0.330.38 +16.6 (1.9) 70.9 (11.4) 100 (0.0)

    GCS pH 3.8 156 (25) 0.020.04 +22.2 (1.8) 49.4 (5.3)

    Lipoid S100-GCS pH 4.3 187 (24) 0.320.37 +21.0 (3.1) 88.0 (12.7)

    Lipoid S100-LMWH GCS pH 4.3 214*

    (22) 0.220.28 +29.9**

    (1.3) 85.2 (12.3) 99(0.2)Lipoid S100-LMWH GCS pH 7.0 385** (27) 0.250.35 +11.6** (0.8) 95.0 (5.0) 43 (2.9)

    * Significantly different from the control (i.e., CS- or GCS-NPs,p

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    5/9

    A. Trapani et al. / International Journal of Pharmaceutics447 (2013) 115123 119

    Fig. 1. (a) Image of Lipoid S100-LMWHCS (pH3.8) nanoparticles, 1.5m1.5m topography obtained in resonant mode AFM; (b) image in phase contrast demonstrating

    mechanical properties of the surface; (c) 3D representation of thesame nanoparticles and (d) corresponding cross section.

    time points (Fig. 3a). The incubation of the particles at 25C and

    37 C displayed retention of particle sizes with the exception of

    Lipoid S100 LMWH GCS NPs formulated in neutral conditionswhere after an initial increase the size resulted constant (Fig. 3b

    and c).

    3.4. Mucoadhesion studies of LMWHloadedNPs

    The degree of interaction between NPs and mucin was studied

    by recording the variations in zeta potential of the nanoparticles

    upon interaction with a mucin solution. As canbe seenin Fig.4, the

    zeta potential values changed after addition of increasing amounts

    of NPs to the mucin dispersion. This effect was more intense for

    Lipoid S100LMWHGCSNPspreparedin acidicmedium, thanfor the

    same formulation prepared in neutral medium and also for Lipoid

    S100 LMWH CS NPs.

    3.5. In vitro release studies

    In vitro release studies from Lipoid S100-LMWH GCS NPs pre-

    pared under neutral conditions were carried outin SLFand showed

    that these nanocarriers exhibited a progressive LMWH release up

    to 6h, followedby a plateaufor upto 24h (Fig. 5).

    3.6. In vivo studies

    In vivo preliminary experiments were performed with LipoidS100 LMWH GCS NPs prepared either in acidic or neutral condi-

    tions, and the administration to the lung was done in the form of

    liquid instillation and aerosolisation. The results showed that NPs

    administered as a bolus andalso those prepared in acidic conditions

    andadministeredby aerosolizationdid notlead to a change in coag-

    ulation times (data not shown). However, as shown in Fig. 6, the

    aerosol-type administration of free LMWH and Lipoid S100-LMWH

    GCS NPs (LMWH dose of 8 mg/kg) prepared in neutral conditions

    led to a significant elongation of the coagulation time as compared

    to the control (buffer) (Fig. 6).

    Additional experiments were intended to study whether the

    passage of LMWH to the blood circulation could be due to the leak-

    age through a damaged lung. Fig. 7a shows a tissue section taken

    from a lung of an animal treated with aerosolized buffer (negativecontrol) and examined by microscopy. The section did not exhibit

    any abnormalities. The alveolar sacs are well defined with no signs

    of hemorrhage, flooding, or collapse of the alveolar spaces which

    all can be defined as a manifestation of pulmonary toxicity. On the

    contrary, the animals treated with LPS, which is able to trigger a

    strong inflammatory response, showed a severe cell infiltration

    predominated by neutrophils accompanied by edema which was

    Table 2

    Surface elemental composition of Lipoid S100-LMWHCS and GCS NPs. Theerror associated to each percentage is 0.5%.

    NPs formulation % C % O % N % P % S % Na % Cl

    Lipoid S100-LMWH CS pH 3.8 72.6 20.2 3.6 1.8 0.5 0.4 0.9

    Lipoid S100-LMWH GCS pH 4.3 65.3 24.7 2.1 4.4 2.7 0.9

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    6/9

    120 A. Trapani et al./ International Journal of Pharmaceutics 447 (2013) 115123

    Fig. 2. C1sXP spectra of thecomponents andof theresulting NPsformulations: (a)

    Lipoid S100, (b) GCS, (c) Lipoid S100-LMWH GCS, and (d) Lipoid S100-LMWH CS.

    In each panel the peak attributions to the corresponding chemical functional groupare indicated: peak (1) C C; peak (2) C OH, C N; peak (3) C OP; peak (4) C O,

    O C O; peak (5) COOH; peak (6) N C OOH.

    more prominent in the alveolar region (Fig. 7b). The histological

    analysis of the mouse lungs treated with either LMWH alone or

    LMWH loaded NPs (at the same LMWH dose range used in the

    APTT experiments) suggests that no gross damage and inflamma-

    tory infiltration in the lung is induced by these treatments. For a

    comparison with negative and positive controls, Fig. 7c and d show

    tissue sections taken from animal treated with either LMWH alone

    or LMWH loaded NPs at the highest dose (8mg/kg) respectively.

    These tissues appear very similar to what was seen in the negative

    control group.

    Fig. 3. Particle size of LMWH loaded NPs upon incubation in NaCl 0.9% at different

    temperatures: 4 C (a); 25C ( b) and 37C (c). Values represent meansSD (n=3).

    Whitebars:acidic LipoidS100-LMWH CS NPs;black bars:acidic LipoidS100-LMWH

    GCS NPs; grey bars: neutral Lipoid S100-LMWHGCS NPs.

    Fig.4. Change of zetapotential of coarse mucinparticles whenmixedwith nanopar-

    ticles. () Lipoid S100-LMWH CS (pH 3.8); () acidic Lipoid S100-LMWH GCS; ()

    neutral Lipoid S100-LMWHGCS NPs.

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    7/9

    A. Trapani et al. / International Journal of Pharmaceutics447 (2013) 115123 121

    Fig. 5. In vitro release profile of LMWH from Lipoid S100 loaded GCS NPs in SLF

    bufferat pH7.2in 24h.

    4. Discussion

    In the context of anticoagulant therapy, the development of

    a non-invasive drug delivery system for LMWH could represent

    a useful approach to enhance patient compliance and minimize

    adverse effects. The purpose of this work was to evaluate the

    potential of CS and GCS NPs containing the pulmonary surfactant

    Lipoid S100 for the systemic delivery of heparin upon pulmonaryadministration. To this end, LMWH loaded CS- and GCS-based NPs

    were formulated using an adaptation of the ionic gelation tech-

    nique (Mao et al., 2010; Trapani et al., 2009; Gan et al., 2005).

    This technique, which has been previously used for the associa-

    tion of heparin to CS (Paliwal et al., 2012) and CS-hyaluronic acid

    nanoparticles (Oyarzun-Ampuero et al., 2009), was also found to

    be appropriate for the formation of GCS NPs.

    Asindicated,the size ofthe NPs preparedin acidicis smaller than

    thatof NPspreparedin neutral conditions. In acidic conditions poly-

    cations (CS and GCS) have a high charge density, a fact that favors

    Fig. 6. Changes in plasma activated partial thromboplastin time (APTT) values

    after pulmonary administration of free LMWH and LMWH-loaded Lipoid-GCS

    NPs. Mice were aerosolised either with buffer (n=6), 18mg/kg LMWH (n=34

    per group), or GCS loaded with 18mg/kg (n=36 per group). APTT values are

    expressedin seconds.The control groupwith buffer is representedby a graycolumn.

    KruskallWallis:p= 0.0003. **p< 0.01 forbuffer vs.GCSwith LMWH 8 mg/kg andvs.

    free LMWH 8 mg/kg (MannWhitney).

    their interaction with TPP and also with LMWH. As a result, the

    nanoparticleshave a smallsize anda highly compacted structure.Incontrast, the GCS-based NPs formulated in neutral medium might

    have a less dense structure because of the limited protonation of

    the primary amino groups of GCS, thus leading to an enlargement

    in their size.

    On the other hand, the size increase observed upon introducing

    the surfactant Lipoid S100 in the formulation might be due to the

    interference of the surfactant in the interaction of the counterions.

    The additionof Lipoid S100did not change significantlythe positive

    zeta potential values with respect to the controls except for Lipoid

    S100-LMWH GCS NPs prepared in acidic conditions, which showed

    Fig. 7. Lung microscopy of experimental mice. Hematoxylin-stained lung sections (5-m thickness) from the following conditions: (a) animals treated with buffer; (b)

    animals treated with LPSalone; (c)animalstreated with 8 mg/kg ofLMWH alone;and (d)animals treated with LMWH NPs. Panelsare representative ofthreemiceper group.

    Original magnification: 20.

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    8/9

  • 8/10/2019 Systemic Heparin Delivery by the Pulmonary Route Using Chitosan and Glycol

    9/9

    A. Trapani et al. / International Journal of Pharmaceutics447 (2013) 115123 123

    Oyarzun-Ampuero, F.A., Brea, J., Loza, M.I., Torres, D., Alonso, M.J., 2009. Chitosan-hyaluronic acid nanoparticles loaded with heparin for the treatment of asthma.Int. J. Pharm. 381, 122129.

    Paliwal, R., Paliwal, S.R., Agrawal, G.P., Vyas, S.P., 2012. Chitosan nanoconstructs forimproved oral delivery of low molecular weight heparin: in vitro and in vivoevaluation. Int. J. Pharm. 422, 179184.

    Qi, Y., Zhao, G., Liu, D., Shriver, Z., Sundaram, M., Sengupta, S., Venkataraman, G.,Langer, R., Sasisekharan, R., 2004. Delivery of therapeutic levels of heparin andlow-molecular-weight heparin through pulmonary route. Proc. Natl. Acad. Sci.USA (PNAS) 101, 98679872.

    Ramadan, A., Lagarce, F., Tessier-Marteau, A., Thomas, O., Legras, P., Macchi, L.,

    Saulnier, P., Benoit, J.P., 2011. Oral fondaparinux: use of lipid nanocapsulesas nanocarriers and in vivo pharmacokinetic study. Int. J. Nanomedicine 6,29412951.

    Schulman,R.I., 2000.Assessment of low-molecular-weight heparin trialsin cardiol-ogy. Pharm.Ther. 87, 19.

    Siew, A., L e, H., Thiovolet, M., Gelle rt , P. , S chatzlein, A., Uche gbu, I. , 2012.E nhanced or al abso rption of hy dro pho bic and hy dro philic drugs using

    quaternary ammonium palmitoyl glycol chitosan nanoparticles. Mol. Pharm. 9,1428.

    Smart, J.D., 2005. The basics and underlying mechanisms of mucoadhesion. Adv.Drug Del. Rev. 57, 15561568.

    Sogias, I.A., Williams, A.C., Khutoryanskiy, V.V., 2008. Why is chitosan mucoadhe-sive? Biomacromolecules 9, 18371842.

    Takeuchi, H., Thongborisute, J., Matsui, Y., Sugihara, H., Yamamoto, H., Kawashima,Y., 2005. Novel mucoadhesion tests for polymers and polymer coated particlesto design optimal mucoadhesive drug delivery systems. Adv.Drug Del. Rev. 57,15831594.

    Trapani, A., Sitterberg, J., Bakowsky, U., Kissel, T., 2009. The potential of glycol chi-

    tosan nanoparticles as carrier for low water soluble drugs. Int. J. Pharm. 375,97106.

    Wagner, C.D., Naumkin, A.V., Kraut-Vass, A., Allison, J.W., Powell, C.J., Rumble, J.R.,2003. NIST X-ray Photoelectron Spectroscopy Database. Standard ReferenceDatabase,20 Version 3.5.

    Yang, T., Mustafa, F., Bai, S., Ahsan, F., 2004. Pulmonary delivery of low molecularweight heparins.Pharm. Res. 21, 20092016.


Recommended