The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

Embed Size (px)

Citation preview

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    1/13

    The effect of the oral administration of polymeric nanoparticles on the efficacy

    and toxicity of tamoxifen

    Amit K. Jain, Nitin K. Swarnakar, Chandraiah Godugu, Raman P. Singh, Sanyog Jain*

    Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar (Mohali),

    Punjab 160062, India

    a r t i c l e i n f o

    Article history:

    Received 8 July 2010

    Accepted 19 September 2010

    Available online 8 October 2010

    Keywords:

    PLGA nanoparticles

    Tamoxifen

    Oral administration

    Nuclear localization

    a b s t r a c t

    The present investigation reports on the conditions for preparation of tamoxifen loaded PLGA nano-

    particles (Tmx-NPs) for oral administration. Tmx-NPs with >85% entrapment efficiency and

    165.58 3.81 nm particle size were prepared and freeze dried. Freeze dried Tmx-NPs were found to be

    stable in various simulated GIT media (pH 1.2, pH 3.5, pH 6.8, SGF & SIF). No significant changes in

    characteristics of Tmx-NPs were observed after 3 months accelerated stability studies. The cell viability

    in C127I cells was found to be relatively lower in Tmx-NP treated cells as compared to free Tmx treated

    cells. CLSM imaging reveled that nanoparticles were efficiently localized into the nuclear region of C127I

    cells. Oral bioavailability of Tmx was increased by 3.84 and 11.19 times as compared to the free Tmx

    citrate and Tmx base respectively, when formulated in NPs. In vivo oral antitumor efficacy of Tmx-NPs

    was carried out in DMBA induced breast tumor model and tumor size was reduced up to 41.56% as

    compared to untreated groups which showed an increase in tumor size up to 158.66%. Finally, Tmx-NPs

    showed the marked reduction in hepatotoxicty when compared with free Tmx citrate as evidenced by

    histopathological examination of liver tissue as well as AST, ALT and MDA levels. Therefore Tmx-NPs

    could have the significant value for the oral chronic breast cancer therapy with reduced hepatotoxicity.

    2010 Elsevier Ltd. All rights reserved.

    1. Introduction

    Breast cancer is the second leading cause of cancer deaths today

    after lung cancer and is the most common cancer among women

    [1]. For over a quarter of a century, tamoxifen (Tmx) has been

    prescribed to treat patients with advanced breast cancer. Tmx

    belongs to a class of non-steroidal triphenylethylene derivatives

    and is the first selective estrogen receptor modulator (SERM) [2].

    The US Food and Drug Administration (FDA) approved Tmx for the

    treatment of advanced breast cancer in late 1998 [3]. Tmx shows its

    potential effects in patient who possess estrogen receptors (ER)positive cancer cells by competing with estrogen to bind with

    estrogen receptor in breast cancer cells [4].

    As Tmx therapy is chronic one (3e5 years), oral delivery is the

    most preferredroute of administration and its solubility problem in

    aqueous milieu has been overcome by forming its salt form,

    tamoxifen citrate (Tmx citrate). Commercially, Tmx is available only

    as tablet and oral solution containing Tmx citrate in a daily dose of

    10e20 mg. However Tmx citrate also showed the poor oral

    bioavailability (20e30%) due to its precipitation as free base in the

    acidic environment of stomach and also due to extensive hepatic

    and intestinalfirst pass metabolism, so as to increase its does [5]. So

    in spite of a clinical choice in advanced and metastatic stages of

    breast cancer, it suffers from large inter subject variability and

    several dose and concentration dependent side effects [6e8]. It

    mainly causes oxidative stress mediated hepatotoxicity, i.e. toxic

    hepatitis, multifocal hepatic fatty infiltration, sub massive hepatic

    necrosis and cirrhosis [9]. Tmx is also having high risk of causing

    endometrial cancer which depends mainly upon treatment dura-tion and dose accumulation [10]. Thus, existing therapy renders its

    difficult to administer in minimum effective dose, leading to liver

    toxicity. Thus an alternate delivery system is essential for optimal

    oral chronic therapy of Tmx with improved bioavailability and

    reduced side effects especially hepatotoxicity.

    Biodegradable polymeric nanoparticles (NPs) have gained

    a considerable interest in this regard [11]. Amongst them poly

    (lactic-co-glycolic acid) (PLGA) is an approved biodegradable

    polymer with good biocompatibility and widely employed for

    loading and encapsulation of variety of anticancer drugs [12e14].

    When polymeric NPs are administered by oral route, the M-cells* Corresponding author. Tel.: 172 2292055; fax: 172 2214692.

    E-mail addresses: [email protected] , [email protected] (S. Jain).

    Contents lists available at ScienceDirect

    Biomaterials

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / b i o m a t e r i a l s

    0142-9612/$ e see front matter 2010 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.biomaterials.2010.09.037

    Biomaterials 32 (2011) 503e515

    mailto:[email protected]:[email protected]://www.sciencedirect.com/science/journal/01429612http://www.elsevier.com/locate/biomaterialshttp://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://dx.doi.org/10.1016/j.biomaterials.2010.09.037http://www.elsevier.com/locate/biomaterialshttp://www.sciencedirect.com/science/journal/01429612mailto:[email protected]:[email protected]
  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    2/13

    (specialized cells staying over mucosa-associated lymphoid tissue)

    in Payers patches uptake the nano/microparticle and transport

    them from the gut lumen to intra-epithelial lymphoid cells and

    afterward through the lymphatic system into the blood stream

    [15e17]. NPs follow this special pathway and thus enhance the

    bioavailability of encapsulated drug and also avoid the enzymatic

    degradation in enterocytes, first pass metabolism in liver thus

    decrease the dose and ultimately the drug related toxicity.

    In the present work tamoxifen loadedPLGA nanoparticles (Tmx-

    NPs) have been prepared,characterized and freezedried. The freeze

    dried Tmx-NPs were evaluated for in vitro release characteristics,

    GIT stability and accelerated stability study. In vitro antitumor

    activity was evaluated on mouse breast cancer cells C127I [18].

    Pharmacokinetics, in vivo antitumor efficacy and hepatotoxicity

    were also evaluated after oral administration.

    2. Materials and methods

    2.1. Materials

    PLGA 50/50 (inherent viscosity 0.41 dl/g in chloroform at 25 C) was used from

    Boehringer Ingelheim (Ingelheim, Germany). Tamoxifen (Z)-2-[4-(1, 2-diphenyl-1-

    butenyl)phenoxy]-N,N dimethylethylamine (free base and citrate salt), Didode-cyldimethylammonium bromide (DMAB) (98%), Polyvinyl alcohol (PVA) (MW.

    30000e70000), Pluronic F-68, 7, 12-dimethylbenz[a]anthracene (DMBA), Trypsin-

    EDTA, MTT (3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide),

    coumarin-6, triton X-100 and propidiumiodide (PI)were obtainedfrom Sigma, USA.

    Dulbeccos modified Eagles medium (DMEM), fetal bovine serum (FBS), antibiotics

    (Antibioticeantimycotic solution) and Hankss balanced salt solution (HBSS) were

    purchased from PAA, Austria. Tissue culture plates and 8-well culture slides were

    procured from Tarsons and BD Falcon, respectively. Ethyl acetate (LR grade),

    Acetonitrile (HPLC grade), methanol (HPLC grade) were purchased from Ranchem

    Fine Chemicals, India. Ultra pure water (SG water purification system, Barsbuttel,

    Germany) was used for all the experiments. All other reagents used were of

    analytical grade.

    2.2. Preparation of Tmx loaded nanoparticles

    Tmx-NPs were prepared by emulsion diffusion evaporation method as reported

    earlier in literature [19] with slight modification according the laboratory condi-

    tions. Briefly,50 mgof PLGA alongwith5 mgof Tmxwere dissolved in2.5 mlof ethyl

    acetate (EA) at room temperature. The organic phase was then added to 5 ml of an

    aqueous phase containing the stabilizer. The resulting o/w emulsion was stirred at

    1000 rpm for 20 min. The droplet size reduction of resulting emulsion was carried

    out either by homogenization (high-speed homogenizer, Polytron PT 4000,

    Switzerland) or sonication (Misonix, USA). The resulting emulsion was poured into

    25 ml of water with constant stirring to diffuse and finally evaporating the organic

    solvent. This resulted in nanoprecipitation and formation of NPs. The NPs suspen-

    sion was then centrifuged and washed repeatedly to remove the excess surfactant

    and finally dispersed in 2 ml distilled water and freeze dried (FD).

    2.3. Optimization of process variables

    2.3.1. Effect of droplet size reduction process

    Screening of the droplet size reduction processes (i.e. either homogenization or

    sonication) was carried out to get the optimum size (below 200 nm). For this, NPswere prepared following the above described process keeping other experimental

    parameters like aqueous to organic phase ratio 1:2, final volume of dilution 25 ml

    and stabilizer concentration (2% w/v PVA) constant. Different homogenization

    speeds and sonication (60% amplitude for 1 min) were employed to prepare NPs

    dispersion. Finally particle size and PDI of NPs dispersion was measured using zeta

    sizer (Nano ZS, Malvern, UK).

    2.3.2. Screening of suitable stabilizer

    Tmx-NPs were prepared by using different type and concentration of stabilizers

    like DMAB, PVA and Pluronic F-68. The best suitable stabilizer was identified based

    on the optimum particle size, zeta potential and entrapment efficiency.

    2.3.3. Screening of optimum concentration of stabilizer

    The best suitable stabilizer identified as above was then screened for the

    optimum concentration of the stabilizer required for the preparation of Tmx-NPs.

    The optimum concentration of stabilizer was determined on the basis of particle

    size, size distribution and encapsulation effi

    ciency.

    2.3.4. Optimization of drug loading

    Finally, Tmx-NPs were prepared using different Tmx loading i.e. 5%, 10% and 15%

    w/w ofpolymerand itseffecton particlesizeand entrapmentefficiency wasstudied.

    The other experimental parameters likesonication time (1 cycle at 60% of amplitude

    for 60 s), stabilizer concentration (2% PVA) and aqueous to organic phase ratio 1:2

    were kept constant.

    2.4. Characterization of nanoparticles

    2.4.1. Particles size and zeta potential measurementTmx-NPs were evaluated for their mean particle size and polydispersity index

    (PDI) by using Zeta Sizer (Nano ZS, Malvern Instruments, UK). All the values were

    taken by the average of 6 measurements. Zeta potential was estimated on the basis

    of electrophoretic mobility under an electric field, as an average of 30 measure-

    ments. Zeta potential was also determined by using Zeta Sizer (Nano ZS, Malvern

    Instruments, Malvern, UK).

    2.4.2. Entrapment efficiency

    The percentage of drug encapsulated in PLGA NPs was determined by using

    a validated HPLC method reported in literature with slight modifications [20].

    Briefly, Tmx-NPs suspension was centrifuged and the obtained pellet was dissolved

    in acetonitrile furthermore analyzed by Waters high-performance liquid chroma-

    tography (HPLC) system consisting of 996 Photodiode Array Detector and dVR

    Agilent Technologies Lichrospher 100 RP-18e end capped 5 mm column (Lot No. L

    54921633) (Germany). Acetonitrile and methanol (containing 0.02% triethylamine)

    (70:30) were used as the mobile phase with a flow rate of 0.7 ml/min. The injection

    volume was 10 ml and retention time of Tmx was found to be 5.1 min. The detection

    wavelength (lmax) for Tmx was 281 nm.

    2.4.3. Morphology of nanoparticles

    The surface morphology of nanoparticles was analyzed by atomic force micro-

    scope (Veeco Bioscope II, USA). The nanoparticles suspension were placed on the

    silicon wafer with the help of a pipette and allowed to dry in air. The microscope is

    vibration damped and measurements were madeusing commercial pyramidal Si3N4tips (Veecos CA, USA). The cantilever used for scanning was having length 325 mm

    and width 26 mm with a nominal force constant 0.1 N/m. Images were obtained by

    displaying the amplitude signal of the cantilever in the trace direction, and the

    height signal in the retrace direction, both signals being simultaneously recorded.

    2.5. Freeze drying of NPs

    Tmx-NPs were freeze dried (Vir Tis, Wizard 2.0, New York, USA freeze dryer)

    following an optimized freeze dried cycle (Table 1) [21]. The condenser temperature

    was 60 C and pressure applied in each step was 200 Torr. 2 ml of washed NPs

    suspension was filled in 5 ml glass vials and subjected to freeze drying using 5% w/v

    of trehalose. After freeze dying the Tmx-NPs were characterized for the appearance

    of the cake, reconstitution time, size after freeze drying, entrapment efficiency,

    nature of drug in nanoparticles using DSC and XRD analysis.

    2.6. DSC analysis

    Differential scanning calorimetry (DSC) thermogram of the freeze dried Tmx-

    NPs, physical mixture, pure tamoxifen and trehalose was carried out using a Mettler

    Toledo differential scanning calorimeter calibrated with indium standards.

    Measurements were performed at heating rate of 10 C/min from 0 to 200 C.

    2.7. XRD analysis

    The X-ray diffraction patterns of pure tamoxifen, PLGA, blank nanoparticles,

    drugloaded freeze dried nanoparticleswere obtainedusing theX-raydiffractometer

    Table 1

    Optimized freeze drying cycle.

    Thermal treatment Primary drying

    Step Temperature (C) Time

    (Min)

    Step Temperature (C) Time

    (Min)

    1 20 30 1 45 60

    2 15 60 2 30 360

    3 10 60 3 20 360

    4 5 120 4 10 420

    5 15 60 5 5 360

    6 25 60 6 0 180

    7 45 30 7 5 120

    Secondary drying 8 10 60

    1 25 120 9 15 60

    10 20 30

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515504

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    3/13

    (Bruker D8 advance, Bruker, Germany). Measurements were performed at a voltage

    of 40 kV and 25 mA. The scanned angle was set from 3 2q! 40 , and the scanned

    rate was 2 min1.

    2.8. In vitro release of Tmx

    In vitro release of Tmx form PLGA NPs was performed in phosphate buffer saline

    (PBS) pH 7.4, under sinkcondition. The studywas performed usingfreezedried drug

    loadedNPs (correspondingto 1 mgof entrapped drug).The freezedried drug loaded

    nanoparticles were suspended in dialysis tube bag (MWCS 12000 Da). The bag was

    suspended in 10 ml of PBS pH 7.4, containing 1% w/v Tween 80 at 37 C in shaking

    water bath at 100 rpm. Aliquots of 200 ml of sample were withdrawn and estimated

    by HPLC method for amount of drug released.

    2.9. pH dependent stability of freeze dried Tmx-NPs

    Freezedried Tmx-NPs were evaluated for their stability in various simulated GIT

    fluids (pH 1.2, pH 3.5, pH 7.4, simulated gastric fluids (SGF), and simulated intestinal

    fluids (SIF)) to assess the stability of NPs under various GIT pH and enzymatic

    conditions that can influence their particle size and drug release characteristics.

    Briefly, 10 ml of simulated fluids were added to 2 ml of reconstituted freeze dried

    Tmx-NPs. An incubation time of 2 h was employed for pH 1.2, pH 3.5 and SGF while

    6 h for pH 7.4 [13,22]. Particle size, PDI and entrapment efficiency were determined

    after the incubation of freeze dried Tmx-NPs with different simulated fluids.

    2.10. Accelerated stability studies

    Freeze dried Tmx-NPs were assessed for accelerated stability studies over

    a period of 3 months, according to the some protocols reported in the literature

    [13,21,23]. Briefly, freeze dried Tmx-NPs were transferred to 5 ml glass vials sealed

    with plastic caps and were kept in stability chamber with temperature of 25 2 C

    and RH 60 5%. The different formulations were monitored for changes in particle

    size, PDI and entrapment efficiency in addition to for physical appearances and ease

    of reconstitution.

    2.11. Cell culture experiments

    2.11.1. Cells

    C127I mouse breast cancer cell line was obtained from National Centre for Cell

    Sciences, Pune, India. The cells were maintained in complete medium containing

    Dulbeccos modified Eagles medium (DMEM; PAA, Austria), 10% fetal bovine serum

    (FBS; PAA, Austria), and antibiotics (Antibiotice

    antimycotic solution; PAA, Austria).

    2.11.2. In vitro anticancer activity

    C127I cells were harvested from confluent cultures by trypsinization and

    adjusted to 50,000 cells/ml in complete medium. The cell suspension was added in

    96 well tissue culture plates (0.2 ml/well) and incubated overnight for cell attach-

    ment. Following attachment, the medium was replaced with complete medium

    (0.2 ml) containing the free Tmx or Tmx-NPs at the desired concentration. The cells

    were incubated with free Tmx-NPs for 24 or 72 h and cell viability was determined

    by MTT assay. In another set of experiments, the recovery of cells after free Tmx and

    Tmx-NPs treatment was determined. The cells were incubated with free Tmx-NPs

    for 24 h, washed with Hankss balanced salt solution (HBSS; PAA, Austria) and

    furtherincubated in complete medium (withoutdrug/NPs) for 48 h. Thecell viability

    was assessed by MTT assay.

    2.11.3. MTT assay

    Following treatment, the cells were washed with HBSS and incubated with

    0.2 ml fresh DMEM containing 0.5 mg/ml MTT (Sigma, USA). The MTT-containingmedium was removed after 3 h incubation. The MTT formazon was dissolved in

    0.2 ml dimethylsulfoxide (CDH, India) and opticaldensitywas determined at 550 nm

    using a Bio-Tek ELISA plate reader.

    2.11.4. Cell uptake studies

    Fluorescent NPs were prepared by co-encapsulation of coumarin-6 with Tmx in

    PLGA (coumarin-6-Tmx-NPs). The dye was added in the organic phase (100 mg/

    50 mg polymer) and coumarin-6-Tmx-NPs were prepared following the optimized

    protocol as described earlier. Cumulative dye release from coumarin-6-Tmx-NPs

    was determined in phosphate-buffered saline (pH 7.4) by fluorimetry (excitation/

    emission458/505nm) after 24h C127Icells wereseededin 8-wellcultureslides (BD

    Falcon) and allowed to attach overnight. The cells were incubated with coumarin-6-

    Tmx-NPs for 3 h and extracellular particles were removed by washing with HBSS

    (5). The cells were fixed with 3% paraformaldehyde (Merck, India) and per-

    meabilized with0.2% Triton X-100 (Sigma, USA). The nuclei were stained with10 mg/

    ml propidium iodide (Sigma, USA). The cells were observed under the confocal laser

    microscope (CLSM) (Olympus FV1000).

    2.12. In vivo pharmacokinetic after oral administration

    2.12.1. Animals and dosing

    Female Sprague Dawley (SD) rats of 220e230 g and 4e5 weeks old were

    supplied by the central animal facility (CAF), NIPER, India. All the animal studies

    protocols were duly approved by the Institutional Animal Ethics Committee (IAEC),

    National Institute of Pharmaceutical Education & Research (NIPER), India. The

    animals were acclimatized at temperature of 25 2 C and relative humidity of

    50e60% under natural light/dark conditions for one week before experiments. The

    animals were randomly distributed into three groups each containing 6 animals.First group of animals received oral free Tmx base (suspension) while another

    second group of animals received free Tmx citrate (suspension) and third group

    received Tmx-NPs. All the formulations were administered orally at a dose of 10mg/

    Kg bodyweight. Theblood samples (approximately 0.25ml) were collected fromthe

    retro orbital plexus under the mild anesthesia into the micro centrifuge tubes

    containing heparin (40 IU/ml blood). Plasma was separated by centrifuging the

    blood samples at 5000 rpm for 5 min at 4 C. To 100 ml of plasma, 200 ml of aceto-

    nitrile was added to precipitate proteins and 25 ml of 10 mg/ml of internal standard

    (estradiol) was added. The samples were vortexed and centrifuged at 10,000 rpm for

    15min. The supernatantswere separatedand analyzedfor drugcontentby validated

    RP-HPLC [24].

    2.12.2. HPLC quantification of Tmx in plasma samples

    Calibration curves were used for the conversion of the Tmx/estradiol chro-

    matographic area to the concentration of Tmx. Calibrator and quality control

    samples were prepared by adding of appropriate volumes of standard Tmx solution

    in acetonitrile to drug free plasma. Calibration curves were designed over the range

    of25e1000ng/ml (r2 0.998). Briefly,an aliquot (100 ml) of plasmasample was mixed

    with 25 ml of internal standard solution (estradiol 1 mg/ml) and 25 ml of drug solu-

    tion. After vortexing for 30 s a protein precipitating agent acetonitrile (100 ml) was

    added vortexed for 5 min. The mixture was centrifuged for 10 min at 10,000 rpm.

    After centrifugation supernatant was transferred to autosampler vials, capped and

    placed in the HPLC autosampler. An 80 ml aliquot of each sample was injected onto

    the HPLC column. Mobile phase employed for analysis was the mixture of acetoni-

    trile and methanol, containing 0.02% triethylamine (85:15).

    2.12.3. Pharmacokinetic data analysis

    The pharmacokinetic analysis of plasma concentrationetime data was analyzed

    by one compartmental model, using Kinetica software (Thermo scientific). Required

    pharmacokinetics parameters like total area under the curve (AUC)0eN, terminal

    phase half-life (t1/2), peak plasma concentration (Cmax) and time to reach the

    maximum plasma concentration (Tmax) were determined.

    2.13. In vivo antitumor efficacy

    Female Sprague Dawley (SD) rats of 45e50 day age were used for the induction

    of chemical induced breast cancer. 7,12-dimethylbenz[a]anthracene (DMBA) in soya

    bean oilwasadministeredorally toratsat 45 mg/kgdoseat weekly intervalfor three

    consecutive weeks. Measurable tumor size was observed in animals and tumor

    bearing animals were separated and divided randomly into different treatment

    groups. Thetumor width( W) and length(L) were recordedwith an electronic digital

    caliper and tumor size was calculated using the formula (L W2/2). Drug treatment

    was given after 10 weeks of the last dose of DMBA. Animals were treated with

    a repeated (once in 3 days) dose of Tmx citrate suspension (group A) and Tmx-NPs

    (group B) both in a dose equivalent to 3 mg/Kg body weight, of Tmx. The control

    group C received a samerepeated oraladministration of PBS (pH 7.4).The tumor size

    was calculated as described above. The tumor size was measured up to 30 days

    (during the treatment period). Further, survival rate was observed in another group

    of animals up to 60 days.

    2.14. Toxicity evaluation

    Next day after administration of the last dose the animals were sacrificed and

    blood was collected by cardiac puncture. Liver toxicity markers ALT and AST were

    estimated in plasma samples by commercially available diagnostic kits (Accurex Pvt.

    Ltd., India). Fromthe same groupof animals liverwas isolatedand homogenizedin 5

    volumes of PBS (pH 7.4). The total homogenate was used for the oxidative stress

    (MDA levels) estimation. Enzyme activities in plasma were evaluated by a UV kinetic

    method. Representative liver tissues from each group were excised and fixed in 10%

    (v/v) formalin saline and processed for routine histopathological procedures.

    Paraffin embedded specimen were cut into 5 mm sections and stained with hema-

    toxylin and eosin (H&E) for histopathological evaluations.

    2.15. Statistical analysis

    All the results were expressed as mean standard deviation (SD). Statistical

    analysis was performed with Sigma Stat (Version 2.03) using one-way ANOVA fol-

    lowed by TukeyeKramer multiple comparison test. P < 0.05 was considered as

    statistically signifi

    cant difference.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 505

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    4/13

    3. Results

    3.1. Preparation and optimization of Tmx-NPs

    3.1.1. Effect of droplet size reduction process

    Tmx-NPs were prepared by using both homogenization and

    sonication as a tool for droplet size reduction. Two different

    homogenization speed10,000 rpmand 15,000 rpmweretested. Fig.1

    shows significant reduction in particle size upon increasing the

    homogenization speed. Howeverthe desiredparticle size(0.4) and unacceptable. But no significant

    difference (p > 0.05) was observed in particle size and PDI with

    respect to the 2% and 3% PVA. So 2% w/v PVA was optimized for

    Tmx-NPs preparation.

    3.1.4. Effect of drug loading

    Tmx-NPs were prepared using different theoretical drug loading

    i.e. 5%, 10% and 15% w/w of polymer to determine the optimum

    percentage of Tmx in PLGA matrix. As shown in the Table 4, theo-

    retical drug loading didnt affect the particle size significantly

    (p > 0.05) when it was increased form 5e15% but there was

    significant change (p < 0.05) in PDI. The entrapment of efficiency

    was also increased when drug loading was increased from 5% to

    10% but it decreased on further increasing it to 15%.

    3.2. Shape and morphology of Tmx-NPs

    AFM image of nanoparticles showed distinct spherical particles

    with smooth surface (Fig. 2). A good correlation was obtained in the

    particle size as observed by both zeta sizer and AFM.

    3.3. Freeze drying of Tmx-NPs

    The Tmx-NPs were freeze dried using optimized stepwise freezedrying cycle developed previously by our group [21]. A 5% w/v

    trehalose was added to 2 ml of NPs suspension. After freeze drying

    the obtained cake was redispersed in 2 ml distilled water and

    particle size along with PDI after freeze drying was analyzed using

    zeta sizer. Different properties of freeze dried NPs like physical

    appearance, reconstitution nature and size ratio (before and after

    freeze drying) are given in Table 5. It is clear form Table 5 that

    lyophilization using 5% trehalose as a lyoprotectant produced intact

    fluffy cake which was easily redispersed to form NPs by mere

    manual shaking (upside down for 20 s). No significant changes in

    particle size, PDI and entrapment efficiency were observed after

    freeze drying. Moreover, Sf/Si (ratio of particle size after and before

    freeze drying) remained almost unity (1.005) when Tmx-NPs were

    lyophilized in presence of 5% trehalose. In contrast, particle size ofNPs was increased significantly (p < 0.05) with unacceptable PDI

    for freeze dried Tmx-NPs without trehalose. No significant differ-

    ence in percentage entrapment efficiency (p > 0.05) was observed

    before and after freeze drying in all cases.

    Fig. 1. Effect of droplet size reduction process on particle size and PDI.

    Table 2

    Effect of stabilizer type on particle size, PDI, entrapment efficiency and zeta

    potential.

    Surfactants Size (nm) PDI Entrapment

    efficiency (%)

    Zeta potential

    2% PVA 165.58 3.81 0.085 0.07 86.20 1.450 3.26 0.95

    1% DMAB 120.00 4.10 0.015 0.04 15.30 0.312 45.57 4.68

    PF-68 130 5.60 0.136 0.08 38.56 0.245 3.45 0.67

    Values are in mean

    SD (n

    6)

    Table 3

    Effect of PVA concentration on particle size, PDI and entrapment efficiency.

    Surfactants

    concentration

    Size (nm) PDI Entrapment

    efficiency (%)

    1% PVA 195.50 9.20 0.425 0.90 86.20 1.550

    2% PVA 165.58 3.81 0.079 0.07 85.78 2.540

    3% PVA 168.00 3.10 0.055 0.05 84.94 1.550

    Values are in mean SD (n 6)

    Table 4

    Effect of initial drug loading on particle size and entrapment efficiency.

    Drug loading

    (% w/w)

    Particle

    size (nm)

    PDI Zeta

    potential (mV)

    Entrapment

    efficiency (%)

    5% 153.23 4.35 0.034 0.056 3.26 0.95 65.32 2.23

    10% 165.58 3.81 0.085 0.070 3.50 0.50 86.20 1.45

    15% 190 1.58 0.198 0.011 3.34 0.89 60.45 1.38

    Values are in mean

    SD (n

    6)

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515506

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    5/13

    3.4. DSC analysis

    Fig. 3 shows DSC thermograms of pure Tmx, Tmx-NPs, PLGA,

    trehalose and their physical mixture. Tmx-NPs showed no melting

    peak indicating absence of crystallinity. Whereas, Tmx in pure form

    exhibited a melting peak around at 98 C indicating crystalline

    nature of the drug.

    3.5. XRD analysis

    To study the characteristic of drug inside the NPs, X-ray diffrac-

    tion (XRD) pattern of pure Tmx, PLGA, Tmx-NPs, physical mixture

    andtrehalose were studied. ThecharacteristicXRD pattern of Tmxis

    shown in Fig. 4, while in the case of freeze dried nanoparticles, no

    characteristic peaks of Tmx were observed.

    3.6. In vitro release of Tmx

    The release profile of optimized Tmx-NPs is shown in Fig. 5. The

    formulation exhibited sustained release profile over a period of

    time. The Tmx released from PLGA nanoparticles showed the

    biphasic release pattern with 23.65% of drug released in 24 h fol-

    lowed by sustained release up to 23 days (94.25%). No initial burst

    release was obtained from the formulation. The cumulative drug

    release was fitted into different release models namely zero order,

    first order, Higuchis square root plot and Hixson Crowell cube root

    plot and the model giving a correlation coefficient close to unity

    was taken as order of release. An initial rapid release was found in

    formulation, followed by Higuchis square root pattern with r2

    0.989 values and zero order patterns with r2 0.951 values.

    3.7. pH dependent stability of freeze dried Tmx-NPs

    Table 6 shows the change in particle size and PDI after theincubation of Tmx-NPs in different simulated GIT fluids. It is clear

    form the Table 6, no significant change (p > 0.05) in the particle

    size, PDI and entrapment efficiency of Tmx-NPs were observed

    upon incubation with the various GI fluids.

    3.8. Accelerated stability studies

    Freeze dried nanoparticles containing trehalose as a lyopro-

    tectant were used for accelerated stability studies. After 3 months

    of storage in accelerated conditions, freeze dried Tmx-NPs with 5%

    trehalosewere found to be stable without any collapse or shrinkage

    of dried cake. No changes in the physical appearance as well as

    encapsulation efficiency, particle size and PDI before and after

    storage were observed with freeze dried Tmx-NPs (Table 7).

    3.9. Cell culture experiments

    3.9.1. Cell cytotoxicity assay (MTT assay)

    The results demonstrate that free Tmx and Tmx-NPs showed

    similar effect on cell viability after 72 h of incubation (Fig. 6A).

    Regression analysis showed similar trends in concentration

    dependent cytotoxicity and IC50 values were also similar (w0.1 mg/

    ml) in both cases. However, in recovery experiments, the cell

    viability was relatively lower in Tmx-NP treated cells as compared

    to free Tmx treated cells (Fig. 6B). The regression analysis showed

    higher activity of Tmx-NPs as compared to free drug. Further, the

    IC50 value of Tmx-NPs was lower as compared to free Tmx by

    w0.2 mg/ml.

    3.9.2. Cell uptake studies by confocal laser microscopy

    The cellular uptake of Tmx-NPs was evident within 3 h of

    incubation with cells (Fig. 7AeD). Further, the Tmx-NPs showed

    good nuclear co-localization after 3 h of incubation (Fig. 8AeD).

    Nearly 80% of the green fluorescence (coumarin-6) co-localized

    with the red fluorescence (propidium iodide) (Fig. 8E) indicating

    rapid internalization and nuclear transport of Tmx-NPs. The

    nuclear co-localization was further confirmed by line series anal-

    ysis (Fig. 8FeI).

    3.10. In vivo pharmacokinetics after oral administration

    The plasma concentration profiles of Tmx after a single oral

    administrationof the Tmx-NPs,free Tmxcitratesuspension,and free

    Tmx base at 10 mg/kg are shown in Fig. 9. Table 8 summarizes the

    pharmacokinetic parameters estimated with one compartmental

    Fig. 2. AFM image of Tmx-NPs.

    Table 5

    Particle size and PDI of Tmx-NPs before and after freeze drying.

    Formulation Before freeze drying After freeze drying Ratio

    (Sf/Si)

    Physical

    Appearance

    Reconstitution

    ScoreParticle size

    (nm)

    PDI Entrapment

    efficiency

    Particle

    size (nm)

    PDI Entrapment

    efficiency

    NPs with trehalose 168.45 1.54 0.045 0.002 86.20 1.45 172.65 2.78 0.135 0.052 84.30 1.65 1.005 Intact floppy cake a

    NPs without trehalose 165.60 2.56 0.069 0.014 86.20 1.45 228.34 3.56 0.45 0.127 85.56 2.65 1.564 Collapsed cake b

    Values are in mean SD (n 6); Sf/Sieratio of particle size after freeze drying to particle size before freeze drying;a Reconstitution in 1 ml water and cake is easily redispersed within 20 sec by mere shaking.b

    Not reconstitute.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 507

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    6/13

    analysis of the experimental data obtained after the oral adminis-

    tration of Tmx-NPs, Tmxcitrate and Tmx base to rats. The data were

    adjusted by one compartmental model. The peak plasma concen-

    tration (Cmax) ofTmx citrate was about48.34ng/ml. Themeanvalues

    obtained for AUC0eN andhalf-life(t1/2) were 891.87ng ml1 h1 and

    11.29 h, respectively. Where as Tmx base showed the Cmax about

    23.56 ng/ml with AUC0eN andhalf-life (t1/2) 306.55ng ml1 h1 and

    7.58 h respectively. On the contrary, when Tmx was loaded in PLGA

    NPs formulation, sustained plasma Tmx levels for at least 72 h were

    observed (Fig. 9). The plasma level curve obtained by the adminis-

    tration Tmx-NPs could be described as divided into two parts; the

    first part involves the absorption phase until the achievement of

    Cmax (18 h) followed by maintenance of plasma concentration up to

    72 h. Upon comparing the AUC0eN it was observed that the nano-particles formulation increased the bioavailability of Tmx by 3.84

    and 11.19 times for Tmx citrate and Tmx base respectively.

    3.11. In vivo antitumor efficacy

    Fig. 10 shows the antitumor activity of Tmx-NPs and Tmx citrate

    suspension after oral administration. Orally administered Tmx-NPs

    suppressed tumor growth significantly (p < 0.05) as compared to

    untreated control group and free Tmx citrate solution. After 30

    days, tumor size was reduced up to 41.56% with Tmx-NPs whereas

    untreated groups showed an increase in tumor size up to 158.66%

    as compared to tumor volume before the start of treatment which

    was considered to be 100%. Fig. 11 represents the tumor burden on

    rats after 30 days after the start of treatment. Significant reduction

    (p< 0.05) in tumor burdenwas observed after 30 days for Tmx-NPs

    group. The survival of animals was monitored for 60 days after the

    start of the treatment in another group of animals. Kaplane

    Meiersurvival curve (Fig.12) was plotted for survival analysis of Tmx-NPs.

    The Tmx-NPs enhanced the survival of 83.33% of animals up to 58th

    Fig. 3. Overlay of DSC thermograms of Tmx, PLGA, trehalose, physical mixture and freeze dried Tmx-NPs.

    Fig. 4. Overlay of X-ray diffraction pattern of Tmx, PLGA, trehalose, physical mixture and freeze dried Tmx-NPs.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515508

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    7/13

    day as compared to the free drug where almost complete loss of

    animal took place at end of the study.

    3.12. Toxicity evaluations

    Tmx-NPs showed a marked reduction in hepatoxicity as

    compared to free Tmx citrate. AST, ALT levels in plasma while MDA

    level in liver are normally increased in case of hepatotoxicity. As

    observed in Fig. 13AeC, the levels of these markers increased

    significantly in the case of Tmx citrate given orally while no

    significant (p< 0.05) increase wasobserved in the case of Tmx-NPs.

    Conventional histopathological examinations were carried out

    to determine the possibility of Tmx induced hepatotoxicity in rats,

    treated chronically with Tmx-NPs and Tmx Citrate solutions. As

    evident from the control group liver sections, normal parenchymal

    cells, portal system, and blood sinusoids were observed (Fig. 14A).

    Fig. 14B shows the liver section of rats treated with Tmx citrate

    orally. The liver sections clearly indicate the appearance of edema

    and swelling of hepatocytes, necrosis, kupffer cells showed hyper-

    plasia and apoptosis. Liver sections of rats treated with oral Tmx-

    NPs showed normal histopathological appearance (Fig. 14C).

    4. Discussion

    Different process variables like droplet size reduction process,

    stabilizer type/concentration and % of theoretical drug loading for

    the preparation of Tmx-NPs were optimized [19]. The NPs formu-

    lation involves the formation of initial o/w emulsion. Further, the

    droplet size of emulsion has to be reduced toget a proper size range

    of the NPs. Two methods were employed for reducing the droplet

    size i.e. homogenization and sonication. Firstly homogenization

    was carried out at twodifferent speeds (10,000 and 15,000 rpm) for

    5 min. Reduction in the particles size was observed upon increasing

    the homogenization speed from 10,000 rpm to 15,000 rpm. This

    could be due to increased shear provided at higher speed of

    homogenization. The effect of homogenization and sonication on

    particles size and PDI is previously discussed. Upon comparison of

    both the methods, it was observed that sonication for 1 min, half

    cycle at 60% amplitude, produced the NPs of lesser sized and better

    PDI as compared to homogenization (165.6 4.67 nm with PDI

    0.029 0.001). Moreover, sonication provides high energy as

    compared to homogenization in shorter period of time. Therefore

    the sonication was selected for the droplet size reduction of o/w

    emulsion. While screening of different stabilizers i.e. PVA,

    DMAB and PF-68, it was observed that DMAB produced the

    smallest size particles as compared to PVA and PF-68 but at the

    same time entrapment efficiency was considerably reduced

    (15.28 0.425%). The lower entrapment of Tmx in PLGA NPs couldbe due the increased aqueous solubility in presence of DMAB

    (0.45 0.056 mg/ml in 1% w/v DMAB solution at 25 C). To prove

    this hypothesis we have also prepared Tmx-NPs with different

    concentration of DMAB ranging from 0.2% w/v to 1% w/v. It was

    observed that, as the concentration of DMAB was increased form

    0.2%e1% w/v, entrapment efficiency decreased very significantly

    (p < 0.01) (complete data are not shown). This could be attributed

    tothe higher affinity of Tmx for DMAB as compared toPLGA matrix.

    DMAB is cationic surfactant having a positively charged amino

    group and a long hydrophobic alkyl chain. If we consider the

    orientation of DMAB on the PLGA nanoparticles, hydrophobic alkyl

    chain is more oriented or penetrated inside the PLGA matrix and

    positive charged amino group is present on the surface of nano-

    particles and imparts the cationic charge to nanoparticles. Due tothe excessive penetration or high affinity of hydrophobic chain of

    DMAB inside the PLGA matrix, Tmx was unable to retain inside the

    PLGA matrix and thrown to outer environment where DMAB was

    present and due to its higher solubility in DMAB, it showed a lesser

    entrapment in the PLGA nanoparticles. On the other hand PVA has

    short hydrocarbon chain as compared to DAMB and Tmx was also

    having significantly lesser solubility (25.45 4.56 mg/ml in 1% w/v

    PVA solution at 25 C) in PVA as compared to DMAB. So due to the

    higher solubility of Tmx in DMAB, lesser entrapment was observed

    in PLGA.

    Effect of theoretical drug loading on the particles size, PDI and

    entrapment efficiency was also studied. At 15% drug loading due to

    the saturation of the polymer matrix with drug the entrapment

    efficiency was not increased upon increase in drug loading. More-

    over, the total amount of the drug inside the NPs, at 10% and 15%

    drug loading was found to be almost constant. Taking this into

    consideration 10% w/w theoretical drug loading was optimized.

    Fig. 5. In vitro release of Tmx-NPs in PBS (pH 7.4).

    Table 6

    Initial and final particle size/PDI of Tmx-NPs after exposure to simulated GIT media.

    Medium Particle size PDI Entrapment ef ficiency

    Initial Final Initial Final Initial Final

    pH 1.2 171.65 3.78 175.34 4.65 0.135 0.052 0.171 0.067 84.30 1.65 85.90 1.30

    pH 3.5 171.65 3.78 176.78 3.23 0.135 0.052 0.178 0.098 84.30 1.65 83.56 2.01

    pH 7.4 171.65 3.78 180.12 4.83 0.135 0.052 0.167 0.034 84.30 1.65 86.23 3.10

    SGF 171.65 3.78 179.89 2.45 0.135 0.052 0.198 0.023 84.30 1.65 83.76 3.78

    SIF 171.65 3.78 181.73 2.45 0.135 0.052 0.213 0.056 84.30 1.65 86.12 2.40

    Values are in mean

    SD (n

    6)

    Table 7

    Characterization of formulation after 3 months of accelerated stability studies.

    Parameters Initial Final

    Particle size 175.65 3.78 178.45 4.34

    PDI 0.235 0.042 0.305 0.056

    Entrapment efficiency 85.30 2.55 84.23 3.45

    Physical appearance Intact cake Intact cake

    Ease of reconstitution By mere shaking By mere shaking

    Values are in mean SD (n 6)

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 509

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    8/13

    Tmx-NPs formulation was freeze dried by an optimized step-

    wise freeze drying cycle (Table 1) using 5% trehalose as a cryopro-

    tectant. When NPs are subjected to rapid freezing, a large

    iceewater interface is formed thus producing small ice crystals

    which determines the size of the formed pores after the sublima-

    tion of ice crystals [25]. On the other hand a small interface is pro-

    duced by slow freezing. Cryoprotectants form a glassy amorphous

    matrix around the NPs and also form the hydrogen bonds with the

    polar groups of nanoparticles at the end of the drying process

    [26,27]. It isclearfrom Table 5, that size of Tmx-NPs after the freeze

    drying without the trehalose changed very significantly (p > 0.05)

    as compared to the NPs freeze dried with the trehalose, as the later

    provided a barrier for the formation of aggregates of NPs after the

    process. Entrapment efficiency of NPs after freeze drying were also

    Fig. 6. Concentration dependent effect of Tmx and Tmx-NPs on cell viability. (A) Cell viability of Tmx and Tmx-NPs after 72 h of incubation; (B) Cell viability in recovery exper-

    iments; the dashed and solid lines show regression lines for Tmx and Tmx-NPs, respectively.

    Fig. 7. Cellular uptake of Tmx-NPs. (A) A single cell with Tmx-NPs; (B) Nuclear staining with propidium iodide; (C) Differential interference contrast image of the same cell; (D) 3D

    image of (A) obtained by Z-stacking.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515510

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    9/13

    not changed significantly (p < 0.05) and NPs was easily dispersed

    after reconstitution due to the formation offlaccid cake.

    DSCstudieswereperformed to study thephysical state of drug in

    nanoparticles whether it was present in amorphous or crystalline

    state [28] and also to confirm the possible interaction between the

    drug and the polymer within the matrix [29]. The disappearance of

    endothermic peak of the free drug at 98 C confirmed the entrap-

    ment of Tmx in the amorphous form inside PLGA nanoparticles.

    PLGA and trehalose showed endothermic peak at around 56 C and

    102 C. All the three endothermic peaks i.e. of free drug, trehalose

    and PLGA were clearly observed in DSC thermogram of physical

    mixture as above mentioned. The DSC thermogram of physical

    mixture clearly indicated the absence of the physical interaction

    between the drug and polymer when it was entrapped in PLGA

    nanoparticles.

    In the XRD pattern (Fig. 4), no characteristic peaks of Tmx wereobserved in case of freeze dried nanoparticles. That could be due to

    entrapment of Tmx in molecular form in the PLGA nanoparticles

    during the preparation of nanoparticles. Our finding was also

    supported by the DSC analysis of freeze dried Tmx-NPs which

    clearly indicated the presence of amorphous nature of drug while

    encapsulated in nanoparticles. The present finding was further

    supported by earlier reports in literature [21,29,30].

    In vitro drug release from the freeze dried Tmx-NPs (10% drug

    loading) was estimated in phosphate buffer saline (pH 7.4) under

    sink condition. The Tmx-NPs exhibited Higuchi release pattern and

    showed sustained drug release for 20 days. Although there is no

    clear defined drug release mechanism is proposed for PLGA nano-

    particles. Several release mechanisms could be involved including

    surface and bulk erosion, disintegration, diffusion, and desorption.

    Fig. 8. Nuclear co-localization of Tmx-NPs. (A) Tmx-NPs uptake by cells; (B) shows the nucleus; (C) Differential interference contrast of the same field; (D) Overlay offigure A, B and

    C. (E) Scatter plot of overlap between green (A) and red (B) channel fluorescence. (FeI) Line series analysis offluorescence. (FeH) show the same fields as AeC along with the line

    (red line) along which the analysis was performed. (I) Results of line series analysis showing co-localization of green and red fluorescence.

    Fig. 9. Plasma concentration time profiles of Tmx after oral administration to SD rats at

    10 mg/kg dose formulated in the PLGA NPs, compared with the oral administration of

    Tmx free base and Tmx salt.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 511

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    10/13

    The best described release mechanism of Tmx from the PLGA NPs is

    by diffusion and in the initial phases followed by the diffusion and

    degradation in later phase of release [19,21].

    Data presented in Table 6 confirms the stability of formulated

    Tmx-NPs in simulated GIT fluids. The present work mainly focuses

    on the design of Tmx loaded PLGA for oral administration so it is

    necessary that NPs would resist to their aggregation in presence of

    various GIT environment so as to facilitate their intestinal absorp-

    tion [31].

    As a final exercise of characterization we have evaluated accel-

    erated storage stability of freeze dried Tmx-NPs for 3 months at

    temperature of 25 2 C and RH 60 5%. The study demonstrated

    the stable nature of nanoparticles in presence of trehalose afteraccelerated stability studies (Table 7).

    Free Tmx and Tmx-NPs were further evaluated for their in vitro

    cellular viability assay on C127I cell line by MTT assay [32]. After

    72 h of incubation, both Tmx and Tmx-NPs showed concentration

    dependent reduction in cell viability to similarextent. These results

    suggest that Tmx retained its antitumor efficacy even after its

    encapsulation in polymeric NPs. Further, in recovery experiment,

    the Tmx-NPs resulted in significantly lower cell viability at 1 mg/ml

    as compared to Tmx. In general, the recovery of cells after an initial

    exposure to Tmx-NPs was lower as compared to free Tmx. The

    lower recovery in NPs-treated cells may be attributed to internal-

    ization followed by retention of NPs attributed to retention of NPs

    inside the cell. Thus, the drug is slowly released inside the cells

    even when extracellular NPs have been washed away.Further, the NPs accumulated in cell nucleus which is also the

    site for pharmacological action of the drug as the estrogen receptor

    is a nuclear receptor. When applied to in vivo systems, this indi-

    cated that the cellular uptake and nuclear delivery of NPs in cancer

    cells leads to retention of NPs while the free drug is cleared by

    metabolism or excretion. This could ultimately lead to a reduction

    in both dose as well as dosing frequency of drug loaded NPs as

    compared to free drug.

    In vitro cellular uptake of NPs was carried out by co-encapsu-

    lation of hydrophobic fluorescent marker, coumarin-6 with Tmx in

    PLGA (coumarin-6-Tmx-NPs) with the same procedure employed

    as for the Tmx-NPs preparation [19,33] with slight modification. It

    is clear from the Fig. 7AeD that the Tmx-NPs incubated with C127I

    cells showed the significant internalization after 3 h. Fluorescence

    after 3 h, inside the cells is an indication of rapid internalization of

    Tmx-NPs which were co-encapsulated with the coumarin-6. In

    vitro release of coumarin-6 from the PLGA NPs showed the negli-

    gible release (

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    11/13

    inside the cells for longer period of time as evident by recovery cell

    viability experiments.The Tmx-NPs and free Tmx base were evaluated for their phar-

    macokinetics after oral administration. The market availability of

    Tmx is in a tamoxifen citrate tablets (10 mg, 20 mg) which are

    administered for 2e3 years. Thus theoral pharmacokinetics of Tmx-

    NPs was compared with both the freebase and salt form of the drug

    in the same dose (10 mg/Kg). The plasma level of Tmx after the

    administration of free Tmx base was detected only up to 12 h of the

    oraladministrationwith the Cmax 23.56ng/ml. The Cmax and AUC0eNof thedrug were further increasedto 48.34 ng/ml,when salt form of

    the drug was administered with the same dose. The Cmax and

    AUC0eN of the drug were obtained in line with the data reported in

    theliterature forTmx citrate salt [24]. On the other hand, whenTmx

    was loaded in PLGA NPs and orally administered to rats, the drug

    plasma concentrations dramatically increased (Fig. 10). Thus, the

    AUC0eN for Tmx-NPs was drastically increased to 3431.11 ng/ml h

    from 891.87 ng/ml h that was obtained when Tmx citrate salt was

    administered orally (Table 8). So administration of Tmx by loadinginto the PLGA nanoparticles leads to enhancement of oral bioavail-

    ability of Tmx by 3.84 and 11.19 times as compared to the free Tmx

    citrate and free Tmx base respectively. The increased bioavailability

    of Tmx was observed with O/W microemulsion by Araya et al. when

    they employed medium chain fatty acid triglyceride (MCT), digly-

    ceryl monooleate (DGMO-C), polyoxyethylene hydrogenated castor

    oil 40 (HCO-40) for the formulation Tmx O/W microemulsion [37].

    Here we have reported the oral bioavailability enhancement of

    poorly water soluble drug Tmx by its encapsulation in the PLGA

    nanoparticulate matrix. When nanoparticles are administered by

    oral route, they are absorbed through specialized M-cells of the

    peyers patches in the small intestine [15]. Absorption through the

    M-cells directly drains into the lymphatics thus prevents the drug

    fromthefirst passmetabolism in the hepatic tissue and thereforethe

    chances of drug induced hepatotoxicity are also reduced.

    Fig. 13. (A) ALT levels in plasma after one month of the treatment using Tmx formulations. (*p < 0.05 a Vs control, b Vs Oral Tmx citrate group). (B) AST levels in plasma after one

    month of the treatment using Tmx formulations. (**p < 0.01,*p < 0.05 a Vs control, b Vs Oral Tmx citrate group). (C) Lipid Peroxidation products (MDA) in liver homogenates after

    one month of the treatment. (**p < 0.01.*p < 0.05 a Vs control and b Vs oral Tmx citrate group).

    Fig. 14. Liver microscopic sections after treatment of different formulations.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 513

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    12/13

    Since pharmacokinetics studies of Tmx-NPs revealed significant

    enhancement of bioavailability of Tmx as compared to free Tmx

    base and Tmx citrate. So they were further evaluated for their in

    vivo antitumor efficacy in breast cancer induced animal model. The

    in vivo antitumor tumor efficacy of Tmx-NP offered a proof of

    concept of their effectiveness as appropriate delivery carriers for

    breast cancer. Since, the bioavailability of Tmx base was found

    significantly lower (p < 0.05) as compared to Tmx citrate and only

    salt form of the drug is commercially available the later only was

    employed for evaluation of antitumor efficacy of Tmx and for

    comparison purpose. A chemical mutagen (7,12-dimethylbenz-

    anthracene) (DMBA) was employed as a suitable breast cancer

    animal model because of its structural resemblance to the human

    breasts cancer [38e40]. Oral administration of Tmx-NPs at an

    interval of 2 days for 28 days significantly reduced the tumor

    burden as compared to the Tmx citrate. On the contrary, continue

    increase in the tumor volumewas observed in DMBA control group.

    The enhanced efficacy of Tmx-NPs could be attributed to increased

    bioavailability of Tmx by Tmx-NPs. The increased absorption of

    particulate carriers through a specific reign of GIT (Payers patches)

    lead to their increased availability in the central compartment [40].

    Since the Tmx-NPs showed the sustained pharmacokinetic pattern,

    thus had the longer circulation time in the blood compartment soas to have greater tumor accumulation. The greater tumor accu-

    mulation of Tmx-NPs could be attributed to their enhanced

    permeation and retention (EPR) [41]. The extensive metabolism of

    free Tmx citrate in the liver lead to reduced antitumor efficacy as

    compared to Tmx-NPs and subsequent enhanced hepatotoxicity.

    KaplaneMeier survival curve (Fig. 12) showed enhanced survival

    time of tumor bearing rats following oral administration of Tmx-

    NPs as compared to Tmx citrate administration. These observations

    are also reliable dueto the higher buildup of the drug concentration

    in a discriminating manner in tumor tissue when animal were

    treated with Tmx-NPs, compared to free Tmx-NPs.

    Tmx increases some hepatotoxicity markers levels like ALT, AST

    and MDA in the plasma as well as in liver homogenate [8,42,43].

    AST, ALT and MDA level were not significantly increased (p > 0.05)when Tmx-NPs were administered orally repetitively for 30 days

    whereas these levels increased for free Tmx citrate administered in

    similar manner. The increased hepatotoxicity marker levels for Tmx

    citrate could be attributed to oxidative reactions that takes place

    during its metabolism in the liver [44]. It is acting as an uncoupling

    agent and powerful inhibitor of mitochondrial electron transport

    chain. This ultimately results in oxidative stress mediated damage

    of mitochondria [9]. Decreased hepatotoxicty of Tmx after the

    encapsulation in PLGA NPs could be attributed to its escape from

    first pass metabolism in the liver as NPs are absorbed viaa different

    route. Certainly a tissue biodistribution study would be required to

    determine the actual concentration of Tmx in hepatic tissues after

    administration of Tmx-NPs and Tmx citrate formulations.

    The reduced hepatotoxicity associated after the chronic oraladministration of Tmx-NPs was further established by histopa-

    thology of the liver tissues after one month of oral administration.

    Tmx citrate showed marked changes in cellular integrity leading to

    the hepatotoxicty (Fig. 14) whereas the Tmx-NPs ameliorated the

    changes in hepatic ultra structure by preserving cellular integrity

    and preventing oxidative stress and ultimately inhibited the

    hepatic inflammation.

    5. Conclusions

    In summary, Tmx-NPs can be prepared by solvent-diffusion

    evaporation method with high encapsulation efficiency with 2%

    PVA as a stabilizer with probe sonication method. After freeze

    drying of Tmx-NPs with 5% trehalose, NPs were found to be stable

    in simulated GIT medium hence are suitable for oral administra-

    tion. In vitro release study showed Higuchis release pattern for

    more than 20 days. Freeze dried Tmx-NPs were also stable in

    accelerated stability condition after 3 months. Tmx-NPs can be

    efficiently delivered, retained and localized in the nuclear region

    when tested in mouse breast cancer cells. In vivo pharmacokinetics

    of freezedried Tmx-NPs showed 3.84 and 11.19 times enhancement

    in oral bioavailability as compared toTmx citrate and free Tmx base.

    Further enhanced accumulation of Tmx-NPs in tumor tissue might

    have taken place due to combined effect of enhanced oral

    bioavailability and enhanced permeation and retention (EPR)

    effect. Increased Tmx-NPs accumulation in tumor cells led to

    enhanced antitumor activity with reduced Tmx related heapto-

    toxicity as compared to the marketed Tmx citrate. Therefore Tmx-

    NPs can be of significant value in chronic Tmx therapy for the

    treatment of breast cancer with reduced side effects.

    Acknowledgement

    Authors are thankful to Director, NIPER for providing necessary

    infrastructure facilities and Department of Science & Technology

    (DST), Government of India, New Delhi, India, for financial support.Authors are also thankful forhelp and co-operation renderedby Mr.

    Dinesh Singh and Rahul Mahajan. The histopathological examina-

    tion carried out at Dr. Vijay Malhotras Lab, Chandigarh is also duly

    acknowledged.

    Appendix

    Figures with essential colour discrimination. Figs. 1,2,7e14 in

    this article have parts that are difficult to interpret in black and

    white. The full colour images can be found in the online version, at

    doi:10.1016/j.biomaterials.2010.09.037.

    References

    [1] Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, et al. Cancer statistics. CACancer J Clin 2008;58:71e96.

    [2] Osborne CK, Fuqua SAW. Selective estrogen receptor modulators: structure,function, and clinical use. J Clin Oncol 2000;18:3172e86.

    [3] Jordan VC. Tamoxifen: a most unlikely pioneering medicine. Nat Rev DrugDiscov 2003;2:205e13.

    [4] Desai PB, Nallani SC, Sane RS, Moore LB, Goodwin BJ, Buckley DJ, et al.Induction of cytochrome P450 3A4 in primary human hepatocytes and acti-vation of the human pregnane X receptor by tamoxifen and 4-hydrox-ytamoxifen. Drug Metab Dispos 2002;30:608e12.

    [5] Shin SC, Choi JS. Effects of epigallocatechin gallate on the oral bioavailabilityand pharmacokinetics of tamoxifen and its main metabolite, 4-hydrox-ytamoxifen, in rats. Anticancer Drugs 2009;20:584e8.

    [6] McVie JG, Simonetti GP, Stevenson D, Briggs RJ, Guelen PJ, de Vos D. Thebioavailability of tamoplex (tamoxifen) part 1 a pilot study. Methods Find ExpClin Pharmacol 1986;8:505e12.

    [7] Tukker JJ, Blankenstein MA, Nortier JW. Comparison of bioavailability in man

    of tamoxifen after oral and rectal administration. J Pharm Pharmacol1986;38:888e92.

    [8] Hard GC, Iatropoulos MJ, Jordan K, Radi L, Kaltenberg OP, Imondi AR, et al.Major difference in the hepatocarcinogenicity and DNA adduct forming abilitybetween toremifene and tamoxifen in female Crl: CD (BR) rats. Cancer Res1993;53:4534e41.

    [9] Parvez S, Tabassum H, Rehman H, Banerjee BD, Athar M, Raisuddin S. Catechinprevents tamoxifen-induced oxidative stress and biochemical perturbationsin mice. Toxicol 2006;225:109e18.

    [10] Shin SC, Choi JS, Li X. Enhanced bioavailability of tamoxifen after oraladministration of tamoxifen with quercetin in rats. Int J Pharm 2006;313:144e9.

    [11] Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers asan emerging platform for cancer therapy. Nat Nanotech 2007;2:751e60.

    [12] Astete CE, Sabliov CM. Synthesis and characterization of PLGA nanoparticles.J Biomater Sci Polym Ed 2006;17:247e89.

    [13] Kalaria DR, Sharma G, Beniwal V, Ravi Kumar MNV. Design of biodegradablenanoparticles for oral delivery of doxorubicin: in vivo pharmacokinetics and

    toxicity studies in rats. Pharm Res 2009;26:492e

    501.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515514

  • 7/31/2019 The Effect of the Oral Administration of Polymeric Nanoparticles Efficacy and Toxicity

    13/13

    [14] Govender T, Stolnik S, Garnett MC, Illum L, Davis SS. PLGA nanoparticlesprepared by nanoprecipitation: drug loading and release studies of a watersoluble drug. J Control Release 1999;57:171e85.

    [15] Florence AT. The oral absorption of micro-and nanoparticulates: neitherexceptional nor unusual. Pharm Res 1997;14:259e66.

    [16] Clark M, Jepson MA, Hirst BH. Exploiting M cells for drug and vaccine delivery.Adv Drug Deliv Rev 2001;50:81e106.

    [17] Hussain N, Jaitley V, Florence AT. Recent advances in the understanding ofuptake of microparticulates across the gastrointestinal lymphatics. Adv DrugDeliv Rev 2001;50:107e42.

    [18] Kogure K, Hama S, Manabe S, Tokumura A, Fukuzawa K. High cytotoxicity of[alpha]-tocopheryl hemisuccinate to cancer cells is due to failure of theirantioxidative defense systems. Cancer Lett 2002;186:151e6.

    [19] Hariharan S, Bhardwaj V, Bala I, Sitterberg J, Bakowsky U, RaviKumar MNV. Design of estradiol loaded PLGA nanoparticulate formula-tions: a potential oral delivery system for hormone therapy. Pharm Res2006;23:184e95.

    [20] Fried KM, Wainer IW. Direct determination of tamoxifen and its four majormetabolites in plasma using coupled column high-performance liquid chro-matography. J Chromatogr B Biomed Sci Appl 1994;655:261e8.

    [21] Jain S, Mittal A, Jain AK, Mahajan RR, Singh D. Cyclosporin A loaded PLGAnanoparticle: preparation, optimization, in vitro characterization and stabilitystudies. Curr Nanoscience 2010;6:422e31.

    [22] Zimmermann E, Mller RH. Electrolyte-and pH-stabilities of aqueous solidlipid nanoparticle (SLN (TM)) dispersions in artificial gastrointestinal media.Eur J Pharm Biopharm 2001;52:203e10.

    [23] Abdelwahed W, Degobert G, Fessi H. Investigation of nanocapsules stabiliza-tion by amorphous excipients during freeze-drying and storage. Eur J PharmBiopharm 2006;63:87e94.

    [24] Buchanan CM, Buchanan NL, Edgar KJ, Little JL, Malcolm MO, Ruble KM, et al.Pharmacokinetics of tamoxifen after intravenous and oral dosing of tamox-ifenehydroxybutenylecyclodextrin formulations. J Pharm Sci 2006;96:644e60.

    [25] Willemer H. Measurements of temperatures, ice evaporation rates andresidual moisture contents in freeze-drying. Dev Biol Stand 1992;74:123e36.Discussion.

    [26] Ford AW, Dawson PJ. The effect of carbohydrate additives in the freeze-dryingof alkaline phosphatase. J Pharm Pharmacol 1993;45:86e93.

    [27] Crowe JH, Crowe LM, Carpenter JF. Preserving dry biomaterials: the waterreplacement hypothesis, part 1. Biopharm-Eugene 1993;6:28.

    [28] Layre AM, Gref R, Richard J, Requier D, Chacun H, Appel M, et al. Nano-encapsulation of a crystalline drug. Int J Pharm 2005;298:323e7.

    [29] Chawla JS, Amiji MM. Biodegradable poly (-caprolactone) nanoparticles fortumor-targeted delivery of tamoxifen. Int J Pharm 2002;249:127e38.

    [30] Kojima T, Katoh F, Matsuda Y, Teraoka R, Kitagawa S. Physicochemical prop-erties of tamoxifen hemicitrate sesquihydrate. Int J Pharm 2008;352:146e51.

    [31] Jain AK, Goyal AK, Mishra N, Vaidya B, Mangal S, Vyas SP. PEGePLAePEG blockcopolymeric nanoparticles for oral immunization against hepatitis B. Int JPharm 2010;387:253e62.

    [32] Westedt U, Kalinowski M, Wittmar M, Merdan T, Unger F, Fuchs J, et al. Poly(vinyl alcohol)-graft-poly (lactide-co-glycolide)nanoparticles for local deliveryof paclitaxel for restenosis treatment. J Control Release 2007;119:41e51.

    [33] Panyam J, Sahoo SK, Prabha S, Bargar T, Labhasetwar V. Fluorescence andelectron microscopy probes for cellular and tissue uptake of poly (D, L-lactide-

    co-glycolide) nanoparticles. Int J Pharm 2003;262:1e

    11.[34] Htun H, Holth LT, Walker D, Davie JR, Hager GL. Direct visualization of the

    human estrogen receptor alpha reveals a role for ligand in the nucleardistribution of the receptor. Mol Biol Cell 1999;10:471e86.

    [35] Gupta R, Mishra P, Mittal A. Enhancing nucleic acid detection sensitivity ofpropidium iodide by a three nanometer interaction inside cells and in solu-tions. J Nanosci Nanotechnol 2009;9:2607e15.

    [36] Razandi M,PedramA, GreeneGL, Levin ER.Cell membraneand nuclearestrogenreceptors (ERs) originate from a single transcript: studies of ERa and ERbexpressed in Chinese hamster ovary cells. J Mol Endocrinol 1999;13:307e19.

    [37] Araya H, Tomita M, Hayashi M. The novel formulation design of O/W micro-emulsion for improving the gastrointestinal absorption of poorly watersoluble compounds. Int J Pharm 2005;305:61e74.

    [38] Kaufmann Y, Kornbluth J, Feng Z, Fahr M, Schaefer RF, Klimberg VS. Effect ofglutamine on the initiation and promotion phases of DMBA-inducedmammary tumor development. JPEN J Parenter Enteral Nutr 2003;27:411e8.

    [39] Rehm S. Chemically induced mammary gland adenomyoepitheliomas andmyoepithelial carcinomas of mice. Immunohistochemical and ultrastructuralfeatures. Am J Pathol 1990;136:575e84.

    [40] Bhardwaj V, Ankola DD, Gupta SC, Schneider M, Lehr CM, Ravi Kumar MNV.PLGA nanoparticles stabilized with cationic surfactant: safety studies andapplication in oral delivery of paclitaxel to treat chemical-induced breastcancer in rat. Pharma Res 2009;26:2495e503.

    [41] Maeda H. The tumor blood vessel as an ideal target for macromolecularanticancer agents. J Control Release 1992;19:315e24.

    [42] Recknagel RO, Glende Jr EA, Dolak JA, Waller RL. Mechanisms of carbontetrachloride toxicity. Pharmacol Ther 1989;43:139e54.

    [43] Albukhari AA, Gashlan HM, El-Beshbishy HA, Nagy AA, Abdel-Naim AB. Caffeicacid phenethyl ester protects against tamoxifen-induced hepatotoxicity inrats. Food Chem Toxicol 2009;47:1689e95.

    [44] Elefsiniotis IS, Pantazis KD, Ilias A, Pallis L, Mariolis A, Glynou I, et al.Tamoxifen induced hepatotoxicity in breast cancer patients with pre-existingliver steatosis: the role of glucose intolerance. Eur J Gastroenterol Hepatol2004;16:593e8.

    A.K. Jain et al. / Biomaterials 32 (2011) 503e515 515