14
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=idrd20 Download by: [Ahmed Ali] Date: 12 March 2016, At: 12:32 Drug Delivery ISSN: 1071-7544 (Print) 1521-0464 (Online) Journal homepage: http://www.tandfonline.com/loi/idrd20 In vitro/in vivo evaluation of an optimized fast dissolving oral film containing olanzapine co- amorphous dispersion with selected carboxylic acids Eman Magdy Maher, Ahmed Mahmoud Abdelhaleem Ali, Heba Farouk Salem & Ahmed Abdelbary Abdelrahman To cite this article: Eman Magdy Maher, Ahmed Mahmoud Abdelhaleem Ali, Heba Farouk Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing olanzapine co-amorphous dispersion with selected carboxylic acids, Drug Delivery, DOI: 10.3109/10717544.2016.1153746 To link to this article: http://dx.doi.org/10.3109/10717544.2016.1153746 Published online: 10 Mar 2016. Submit your article to this journal Article views: 10 View related articles View Crossmark data

In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=idrd20

Download by: [Ahmed Ali] Date: 12 March 2016, At: 12:32

Drug Delivery

ISSN: 1071-7544 (Print) 1521-0464 (Online) Journal homepage: http://www.tandfonline.com/loi/idrd20

In vitro/in vivo evaluation of an optimized fastdissolving oral film containing olanzapine co-amorphous dispersion with selected carboxylicacids

Eman Magdy Maher, Ahmed Mahmoud Abdelhaleem Ali, Heba FaroukSalem & Ahmed Abdelbary Abdelrahman

To cite this article: Eman Magdy Maher, Ahmed Mahmoud Abdelhaleem Ali, Heba FaroukSalem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fastdissolving oral film containing olanzapine co-amorphous dispersion with selected carboxylicacids, Drug Delivery, DOI: 10.3109/10717544.2016.1153746

To link to this article: http://dx.doi.org/10.3109/10717544.2016.1153746

Published online: 10 Mar 2016.

Submit your article to this journal

Article views: 10

View related articles

View Crossmark data

Page 2: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

http://informahealthcare.com/drdISSN: 1071-7544 (print), 1521-0464 (electronic)

Drug Deliv, Early Online: 1–13! 2016 Taylor & Francis. DOI: 10.3109/10717544.2016.1153746

RESEARCH ARTICLE

In vitro/in vivo evaluation of an optimized fast dissolving oral filmcontaining olanzapine co-amorphous dispersion with selectedcarboxylic acids

Eman Magdy Maher1, Ahmed Mahmoud Abdelhaleem Ali1,2, Heba Farouk Salem1, andAhmed Abdelbary Abdelrahman3

1Department of Pharmaceutics, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt, 2Department of Pharmaceutics, Faculty of Pharmacy,

Taif University, Taif, Saudi Arabia, and 3Department of Pharmaceutics, Faculty of Pharmacy, Cairo University, Cairo, Egypt

Abstract

Improvement of water solubility, dissolution rate, oral bioavailability, and reduction of first passmetabolism of OL (OL), were the aims of this research. Co-amorphization of OL carboxylic aciddispersions at various molar ratios was carried out using rapid solvent evaporation.Characterization of the dispersions was performed using differential scanning calorimetry(DSC), Fourier transform infrared spectrometry (FTIR), X-ray diffractometry (XRD), and scanningelectron microscopy (SEM). Dispersions with highest equilibrium solubility were formulated asfast dissolving oral films. Modeling and optimization of film formation were undertaken usingartificial neural networks (ANNs). The results indicated co-amorphization of OL-ascorbicacid through H-bonding. The co-amorphous dispersions at 1:2 molar ratio showed more than600-fold increase in solubility of OL. The model optimized fast dissolving film prepared fromthe dispersion was physically and chemically stable, demonstrated short disintegration time(8.5 s), fast dissolution (97% in 10 min) and optimum tensile strength (4.9 N/cm2). The results ofin vivo data indicated high bioavailability (144 ng h/mL) and maximum plasma concentration(14.2 ng/mL) compared with the marketed references. Therefore, the optimized co-amorphousOL-ascorbic acid fast dissolving film could be a valuable solution for enhancing thephysicochemical and pharmacokinetic properties of OL.

Keywords

Amorphous, bioavailability, fast dissolvingfilm, neural networks, olanzapine,pharmacokinetics, solid dispersion

History

Received 9 December 2015Revised 4 February 2016Accepted 9 February 2016

Introduction

Typical antipsychotic drugs are usually classified by their

chemical structure and the potency of binding to the

dopamine type 2 (D2) receptors, while new antipsychotic

agents differ from selective dopamine antagonist in having a

broader receptor affinity and hence called atypical antipsych-

otics. The atypical antipsychotics are characterized by

improved clinical efficacy against schizophrenia and bipolar

disorders with fewer side effects such as hallucinations and

delusions (Worrel et al., 2000). These are also better than the

typical analogs at relieving the negative symptoms of the

illness, such as withdrawal, thinking problems, and lack of

energy (Ayala et al., 2006). Olanzapine (OL) is one of the

recent atypical antipsychotics that belongs to the thienoben-

zodiazepine-class(2-methyl-4-(4-methyl-1-piperazinyl)-10H-

thieno-[2,3-b][1,5] benzodiazepine) (Ayala et al., 2006). It is

widely used in the treatment of schizophrenia and acute

mixed or manic episodes. It is highly efficient with no or

minimal side effects such as weight gain and agranulocytosis

being similar to the first line treatment such as clozapine

(Volavka et al., 2004). However, OL exhibits very slight

solubility in water and suffers from extensive first pass

metabolism and, therefore, possesses low bioavailability

(40%) after oral administration (Sood et al., 2013).

Numerous trials were reported in the literature for improving

bioavailability of OL using solid lipid nanoparticles or

through formation of solid dispersions with various polymeric

carriers (Krishnamoorthy et al., 2009; Harde et al., 2011;

Cavallari et al., 2013; Sood et al., 2013).

Intraoral fast dissolving films (FDF) are non-bulky oral

dosage forms that have several advantages over conventional

oral dosage forms including the ease of administration with no

need for water thus improving patient compliance particularly

elderly and pediatrics. It also enables availability of larger

surface area that leads to rapid disintegration and release of

the drug into the oral cavity within seconds and hence a rapid

onset of action could be achieved (Liang & Chen, 2001; Dixit

& Puthli, 2009; Hoffmann et al., 2011). Owing to pregastric

or oro-mucosal absorption, drugs can directly enter systemic

circulation avoiding first-pass metabolism thus improving

bioavailability with possibility of reduced dosing and fewer

side effects (Saurabh et al., 2011). Many studies on

orodispersible polymer films have been conducted for various

reasons; as enhancing solubility of poorly soluble BCS class

Address for correspondence: Ahmed Mahmoud Abdelhaleem Ali,Department of Pharmaceutics, Faculty of Pharmacy, Taif University,Saudi Arabia. Email: [email protected]

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 3: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

II drugs, taste masking of antihistamines, or increasing

patient compliance for administration of antidepressant

drugs (El-Setouhy & El-Malak, 2010; Preis et al., 2012;

Sievens-Figueroa et al., 2012). Optimized formulations of

orodispersible polymer films were also studied in the

literature using simple methodologies such as solvent casting,

hot-melt extrusion, and freeze drying (ElMeshad & El

Hagrasy, 2011; Low et al., 2013; Kumria et al., 2016;

Shamma & Elkasabgy, 2016). The aim of the current study

was to improve the dissolution properties of the poorly water-

soluble OL via formation of stable OL co-amorphous

dispersions (COADs) from successful solid dispersions with

some polycarboxylic acids (CAPs) (Ali et al., 2015). Solid

dispersion method adjustment could result in co-amorphiza-

tion of the active pharmaceutical ingredients using numerous

coformers and many successful examples were employed in

the pharmaceutical research for improving the physicochem-

ical properties especially for poorly soluble drugs (Jensen

et al., 2015; Qian et al., 2015). Co-milling and freeze drying

with hydrophilic polymers were used to form amorphous

glass solutions and hot-melt extrusion of drugs using mixtures

of low-melting polymers and surfactants were also employed

for co-amorphization (Zhang et al., 2014). Some di and tri-

carboxylic acids have the ability to adjust the glass transition

temperature of the composite solid dispersion by the

antiplasticizing effects thus protecting the amorphous state

of the resulting dispersion (Curtin et al., 2012). In this study,

solvent evaporation under vacuum was used for the prepar-

ation of solid dispersions of OL and poly carboxylic acids

followed by incorporation of successful co-amorphous dis-

persions into different film formulations, followed by incorp-

oration in different film formulations. Modeling and

optimization of the effects of film formulation parameters

on its mechanical strength, disintegration, and dissolution

properties were extensively studied using response surface

models (Hosny et al., 2016). However, in this work, modeling

was undertaken using artificial neural networks and genetic

algorithms (Abdelrahman et al., 2015). Oral bioavailability

and pharmacokinetics of OL from the optimized fast

dissolving films (OFDF) were evaluated in human volunteers

in comparison with marketed products of OL.

Materials and method

Materials

Olanzapine was obtained as a free sample from the Egyptian

Pharmaceutical Industries Company (EPICO, Cairo, Egypt).

Hydroxy propyl methyl cellulose (HPMC-E5) and sodium

carboxy methyl cellulose (NaCMC) were purchased from

Sigma-Aldrich, Darmstadt, Germany. Glycerol, menthol,

propylene glycol, anhydrous carboxylic acids (CAPs); ascor-

bic acid, tartaric acid, and citric acid were purchased from

Natco Pharma, Hyderabad, India. Ethanol (95%) and acetone

were purchased from El-Gomhoria Company (Cairo, Egypt).

Methanol HPLC grade was purchased from Sigma Aldrich,

Gillingham, UK. Other chemicals were of analytical grade

and were used as obtained.

Preparation of OL/carboxylic acid solid dispersions

Solid dispersion preparation was carried out using fast solvent

evaporation under reduced pressure according to a reported

method (Ali et al., 2015). The calculated equivalent amounts

of anhydrous CAPs, ascorbic, citric and D-tartaric acid

according to molar ratios (Table 1), were dissolved in ethanol

95% (10 mL). The weighted amounts of OL (200 mg) were

dissolved in acetone (10 mL) and then mixed with the

ethanolic solution of CAPs in a rotary evaporation flask.

The flask contents were sonicated for 10 min in a water bath

sonicator (Ultrasonic Cleaner Model 57 H, Ney Instruments

Co. Ltd, Melville, NY) until all contents were completely

dissolved. The resulting solution was evaporated under

vacuum (0.25 MPa) using arotary evaporator (Barloworld

Scientific Ltd., Stone, UK). The water bath temperature was

kept at 50 �C. After complete dryness, the collected mass was

pulverized and passed through sieve number 60 (250 mm

apertures), then kept in a desiccator until further examination.

Characterization of OL-carboxylic acid soliddispersions

Saturated solubility

Saturated solubility of the dispersions was measured by

adding known excess amount of each formula to the

dissolution medium (10 mL). The dispersion was kept in a

shaking water bath (37 �C) for 24 h. The samples were taken

out of the shaker and left aside for an extra 12 h to equilibrate

then filtrated using membrane filter (0.45mm, Millipore�,

Billerica, MA). The filtrate was analyzed spectrophotomet-

rically at lmax 273 nm (Seju et al., 2011).

Scanning electron microscopy (SEM)

The morphology of the prepared dispersions was examined

using SEM (Analytical Scanning Microscope, JEOL-JSM-

6510LA, JEOL, Tokyo, Japan). Few specks from each

formulation were placed on the carbon stubs and then

coated using a gold sputter (SPI-Module Sputter Coater, SPI

Supplies Inc., West Chester, PA) followed by microscopical

scanning.

Table 1. Composition of OL-carboxylic acid solid dispersions.

Formula Olanzapine (mg) Ascorbic acid (mg) Citric acid (mg) Tartaric acid (mg) Stoichiometric ratio

Mr 312.44 176.12 192.12 150.09 –F1 200 112.74 – – 1.00:1.00F2 200 225.48 – – 1.00:2.00F3 200 – 122.98 1.00:1.00F4 200 – 245.96 1.00:2.00F5 200 – – 96.08 1.00:1.00F6 200 – – 192.16 1.00:2.00

2 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 4: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Differential scanning calorimetry (DSC)

DSC analysis of pure OL, CAPs, physical mixtures, and the

prepared dispersion formulations (5 mg) were carried out

using DSC (TA-60WSI, Shimadzu, Tokyo, Japan). The

instrument was calibrated using purified Indium (99.99%).

Samples were sealed in a flat bottom aluminum pan

(Shimadzu DSC-60, Tokyo, Japan). The pan was placed in

the DSC instrument and scanned between 30 and 300 �C at a

rate of 10 �C/min. Dry nitrogen was used as a carrier gas

to eliminate the oxidative and pyrrolytic effects with a

flow rate of 10 mL/min. The melting and transition point

measurements were performed using the software provided

with the device.

Fourier transform infrared analysis (FTIR)

Sample (5 mg) of OL, CAPs, and prepared dispersions were

individually mixed with 100 mg dry potassium bromide. The

powder mixtures were compressed into discs under a pressure

of 10 000–15 000 PSI. The infrared spectrum was determined

at a scanning range of 400–4000 cm�1 using a Fourier

Transform Infrared instrument (IR435-U-04, Shimadzu,

Kyoto, Japan).

X-ray diffraction (XRD)

Samples of OL powder, CAPs as well as the solid dispersion

formulations were subjected to X-ray diffraction analysis

using Shimadzu XRD-6000 X-ray powder diffractometer

(XRD-610, Shimadzu, Tokyo, Japan). The equipment was

coupled with a standard Cu sealed X-ray tube with 40 kV

voltage and 30 mA current (Ali et al., 2015). Data collection

was performed at 2� 5–80� in steps of 0.05� and a scanning

speed of 0.5 s per step.

Preparation of OL fast dissolving films (FDF)

Solvent casting method was adopted for the preparation of

OL FDF using mixtures of the successful co-amorphous

dispersions (COADs) with film-forming polymers, hydroxyl

propyl methyl cellulose (HPMC) and sodium carboxy

methyl cellulose (Na–CMC) as shown in Table 2.

Glycerin, propylene glycol (PG), or PEG 400 were used as

plasticizers, citric acid as saliva stimulant, Na-saccharine as

sweetening agent, and menthol as flavoring agent. The film-

forming polymer was dissolved in cold water then OL

co-amorphous dispersion (COAD) was added in the required

quantity. Menthol was dissolved in 1 mL ethanol, and then

added to the aqueous dispersion. The mixture was kept

under magnetic stirring at room temperature until complete

dissolution. Before film casting, the aqueous solution was

sonicated for 1 h to ensure complete removal of the

entrapped air bubbles. To cast the film, the aqueous solution

was poured into a dry clean Teflon plate and then kept

in hot air oven at 60 �C for the first 30 min and then

the temperature was decreased to 40 �C for the next 24 h

(El-Setouhy & El-Malak, 2010). After complete drying, the

films were peeled off carefully from the Teflon plates,

wrapped in aluminum foil, and stored in airtight containers

at room temperature until further investigations.

Physicochemical evaluation of OL fast dissolving films

Physical appearance and weight variation

The appearances of the prepared films were evaluated by

visual observation of transparency or opaqueness. Weight

variation test was carried out by individually weighing 10

films of each formula then calculating the average compared

with other formulations. The thickness of the film was

measured using Vernier caliper micrometer (Shanghai

Measuring and Cutting Tools Limited Company, Shanghai,

China). The thickness was measured at five different locations

(four corners and one at center) and the average was recorded.

Surface pH determination

A combined pH electrode was used for testing the surface pH

of the film. The prepared film (4 cm2) was immersed in 2 mL

distilled water at room temperature. The pH was measured by

bringing the electrode in contact with the surface of the oral

film (Kunte & Tandale, 2010). The experiments were

performed in triplicate and the average was recorded.

Tensile strength

Tensile strength was determined using an apparatus fabricated

in laboratory. Increased stress was applied to the point at which

the strip specimen breaks (Felton et al., 2008). The prepared

film strips with dimensions of 1 � 3 cm2 were fixed from both

ends between two clamps. During measurement (n¼ 5), the

strips were pulled at the top clamp by adding weights in pan till

the film broke. The tensile strength represented by the weight

in grams required to break the film was determined using the

following equation (Yoon et al., 2012):

TS ¼ F=A ð1Þ

Table 2. Different formulation variables included into OL thin film.

RecordPolymer

type

Polymerconcentration

(mg)Plasticizer

type

Plasticizerconcentration

(mg)

1 HPMC (E5) 100.00 PG 10.002 HPMC (E5) 100.00 PEG400 10.003 HPMC (E5) 100.00 Glycerin 10.004 HPMC (E5) 100.00 PG 20.005 HPMC (E5) 100.00 PEG400 20.006 HPMC (E5) 100.00 Glycerin 20.007 HPMC (E5) 200.00 PG 20.008 HPMC (E5) 200.00 PEG400 20.009 HPMC (E5) 200.00 Glycerin 20.00

10 HPMC (E5) 200.00 PG 40.0011 HPMC (E5) 200.00 PEG400 40.0012 HPMC (E5) 200.00 Glycerin 40.0013 NaCMC 100.00 PG 10.0014 NaCMC 100.00 PEG400 10.0015 NaCMC 100.00 Glycerin 10.0016 NaCMC 100.00 PG 20.0017 NaCMC 100.00 PEG400 20.0018 NaCMC 100.00 Glycerin 20.0019 NaCMC 200.00 PG 20.0020 NaCMC 200.00 PEG400 20.0021 NaCMC 200.00 Glycerin 20.0022 NaCMC 200.00 PG 40.0023 NaCMC 200.00 PEG400 40.0024 NaCMC 200.00 Glycerin 40.00

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 3

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 5: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

where F is the maximum load applied on the film in Newtons

(N) and A is the original cross-sectional area measured in

squared centimeter (cm2).

Folding endurance

This test was measured manually (n¼ 3) for the prepared FDF

formulae, where the prepared film (4 cm2) was folded

repeatedly at the same place for several times till visible

cracks were developed. The folding endurance is expressed as

the number of folds (number of times the film is folded at the

same place) required to break the film or to develop visible

cracks. This test was applied to check the ability of the sample

to withstand folding and also it gives an estimation of film

brittleness (Mishra & Gilhotra, 2008).

Drug content determination

Ten film pieces were used in this test, where 4 cm2 was cut

and dissolved in stoppered flask containing 100 mL of 20 mM

phosphate buffer pH 6.8. The contents were sonicated until

complete dissolution of the film. One mL of the solution was

filtered through Millipore filter (0.45 mm pore size) then

introduced into 25 mL volumetric flask and completed to

volume by the buffer solution. The absorbance of the solution

was measured using UV–visible spectrophotometer at lmax of

273 nm.

In vitro disintegration and dissolution studies

The disintegration test was carried out using disintegration

apparatus (Erweka, Milford, CT), where each one of the

six films was placed in the tubes of the container and

the cover disks were placed over it (Dahiya et al., 2009).

The disintegration test of the film was carried out according

to specifications of fast dissolving tablets reported in the

United State Pharmacopeia (USP). To mimic the mouth

saliva, 10 mM phosphate buffer pH 5.8 (900 mL) at

37 �C ± 0.5 �C was used as a disintegration medium and the

time taken for complete disappearance of the film was

measured in seconds.

The in vitro dissolution of OL from the investigated films

(cut into 4 cm2 pieces equivalent to 5 mg OL) was tested by

using USP dissolution tester, apparatus II (Hanson Research,

Chatsworth, CA). The paddles were rotated at 50 rpm in

900 mL of 10 mM phosphate buffer pH 5.8 with a temperature

of water bath kept at 37 ± 0.5 �C. Aliquots of 5 mL were

withdrawn from the dissolution medium at pre-determined

time intervals (1, 3, 5, 7, 10, 15, 20, 25, and 30 min) and then

replaced with fresh medium. The samples were filtered

through Millipore� filter (0.45mm) and OL content was

determined using UV spectrophotometer at lmax 273 nm using

simulated salivary fluid (SSF) as blank. The dissolution

profiles were plotted using average % OL released (n¼ 3)

from FDF and crystalline OL.

Modeling and optimization of OFDF preparation

The data set of OL FDF composed of 24 records (Table 3)

based on different formulation variables was subjected to the

modeling and optimization process. The inputs included

polymer type, polymer concentration, plasticizer type, and

concentration. The polymer types were coded with numerical

values as follows: HPMC E5 (1) and NaCMC (2) while the

plasticizer types were encoded as 3 for PG, 4 for PEG400, and

5 for Glycerol. The measured dependent variables included

tensile strength (N/cm2), disintegration time (s), folding

endurance, and percentage drug released after 10 min.

Modeling and optimization of the data were carried out

Table 3. Input and output film formulation variables used in the modeling and optimization process.

Input variables Output properties

RecordPolymer

typepolymer conc

(mg)Plasticizer

typePlasticizerconc (mg)

Tensile str.(N/cm2)

Folding end.(times)

Disintegrationtime (s)

% released(10 min)

1 1 100.00 3 10.00 4.73 109.00 15.00 97.542 1 100.00 4 10.00 4.05 138.00 9.00 98.133 1 100.00 5 10.00 4.35 157.00 23.00 96.134 1 100.00 3 20.00 4.65 95.00 11.00 99.795 1 100.00 4 20.00 3.89 132.00 4.00 100.236 1 100.00 5 20.00 4.22 146.00 17.00 99.317 1 200.00 3 20.00 7.08 148.00 21.00 90.488 1 200.00 4 20.00 6.12 189.00 14.00 95.449 1 200.00 5 20.00 6.95 207.00 27.00 90.05

10 1 200.00 3 40.00 6.43 135.00 18.00 92.7811 1 200.00 4 40.00 5.52 177.00 10.00 97.4312 1 200.00 5 40.00 6.03 195.00 22.00 91.2513 2 100.00 3 10.00 3.37 124.00 34.00 82.4514 2 100.00 4 10.00 2.76 181.00 28.00 85.2615 2 100.00 5 10.00 2.98 199.00 39.00 82.1916 2 100.00 3 20.00 3.24 119.00 26.00 87.9217 2 100.00 4 20.00 2.41 174.00 21.00 90.1618 2 100.00 5 20.00 2.91 188.00 33.00 86.4419 2 200.00 3 20.00 3.57 226.00 51.00 78.4320 2 200.00 4 20.00 3.11 265.00 44.00 80.9821 2 200.00 5 20.00 3.34 278.00 49.00 77.9522 2 200.00 3 40.00 3.36 213.00 46.00 83.9223 2 200.00 4 40.00 2.65 261.00 38.00 88.5224 2 200.00 5 40.00 3.04 270.00 43.00 81.49

4 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 6: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

using artificial neural networks (ANNs)-Genetic algorithm

software package (INForm V3.71, Intelligensys Ltd., London,

UK) (Ali & Ali, 2013). The experimentally collected data set

was divided into training records (80%), testing records

(10%), and validation records (10%) for model training,

testing, and model validation, respectively. Predictability of

trained models was evaluated using the correlation coefficient

(R2) values computed automatically during training, testing,

and validation steps (Equation (2)). High R2 values closer to

unity indicate appropriate predictability of the trained model

(Plumb et al., 2002):

R2 ¼ 1�Pn

i�1 ðyi � y�i Þ2

Pni�1 ðyi � y�i Þ

2� 100 ð2Þ

where yi is the individual value of the dependent variable, yi is

the predicted value from the model, and yi- is the mean of the

dependent variable. In this formula, the numerator represents

the sum of squares for the error term (SSE) and the

denominator represents the total sum of variable is accounted

for in the model. The artificial neural network structure I(4)–

HL(3)–O(1) was used for model training (linking inputs and

the out-put properties), with four nodes representing the input

layer, three nodes in the hidden layer, and one node in the

output layer. Trusted models should result in validation

correlation R2 as high as those obtained during model training

and testing. The root mean-squared errors (RMSE) were also

calculated and compared with those of training and testing

(Ali & Abdelrahim, 2014).

After developing of the predictive models for each

property of the film, optimization was carried out by setting

the desired range for each of the out-put properties into the

model optimization screen and the desirability function was

selected as ‘‘tent’’ in the model optimization window (Plumb

et al., 2005). The desired minimum and maximum values for

the out-put properties were assigned as follows: tensile

strength (4–5 N/cm2), disintegration time (20–30 s), and

percentage dissolved in 10 min (95–100%). The model

generated solution represented a suggested optimized formula

for the FDF which was then prepared, characterized, and the

experimentally obtained properties were compared to those

previously predicted by the model.

Accelerated stability testing

The optimized FDF formula obtained by the model was chosen

for testing the physicochemical stability. First, the film was

wrapped in butter paper as one layer and above which a second

layer of aluminum foil was tightly applied. Samples of the

optimized films were stored at 40 �C/75% RH for a period of

12 weeks (Akil et al., 2011; Farid et al., 2015). Periodically,

samples were withdrawn at different predetermined time

intervals (0, 1, 2, 4, 6, 9, and 12 weeks) and examined

physically for any changes in color, appearance thickness, and

surface pH as well as chemically for their drug content.

In vivo evaluation of OL film in human volunteers

This study was carried out to compare the pharmacokinetics

of OL from the optimized fast dissolving film formulation

(FDF) to the reference products: A (Olazine oral tablets,

EIPICO, Cairo, Egypt) and product B (fast dissolving tablet

Zyprexa� Velotab, Lilly, Indianapolis, IN). A single oral

dose equivalent to 10 mg OL was given to the volunteers

using randomized crossover design in three phases with 1

week washout period between phases. Nine healthy man

volunteers aged between 23 and 34 years (median weight:

75 kg and median height: 183 cm) were chosen and divided

into three groups each containing three volunteers. Health

status of the volunteers was confirmed by complete medical

history, physical examination, and laboratory analysis for

complete hematological and biochemical examination. The

subjects were instructed to take no other drugs for 1 week

prior to and during the course of study. No consumption of

nicotine was permitted 12 h before and 24 h after drug

intake. Moreover, on each test day, coffee, tea, and cola

beverages were withheld from subjects 12 h before the

administration and till the blood sampling was completed.

The protocol of the study was conducted according to

Helsinky agreement protocol for human subjects and

according to the requirements of the ethical committee of

faculty of medicine, Assuit University, Assiut, Egypt. The

OFDF and the references were administered orally to the

volunteers after fasting overnight. Venous blood samples

(5 mL) were collected into heparinized tubes at certain time

intervals (0, 0.5, 1, 2, 3, 4, 6, 8, 12, and 24 h). Plasma was

obtained by centrifugation at 3000 rpm (centrifuge R32,

Bombay, India) for 10 min and then samples were stored at

�20 �C until the time of analysis.

Chromatographic conditions

Plasma samples were analyzed using a sensitive, reprodu-

cible, and accurate LC-MS/MS method, developed, and

validated before the study. The isocratic mobile phase

consisted of methanol and 0.1% formic acid pH 4.3 (90:10

v/v), which was delivered at a flow rate of 0.2 mL/min into

the mass spectrometer’s electrospray ionization chamber.

Quantitation was achieved by LC-MS/MS detection in

positive ion mode for both OL and atorvastatin using a QT

mass detector (Matuszewski et al., 1998). The ion spray

voltage was set at 3500 V. The common parameters: nebulizer

N2 gas temperature: 350 �C, drying N2 gas flow: 200 ml/min,

sheath gas pressure: 30 Arb, and auxiliary gas pressure: 5 Arb.

The peak area of transition from the m/z 313.17 precursor ion

to m/z 256.13, with an collision energy of 30 eV for OL and

the m/z 559.39 precursor ion to m/z 440.27, with an collision

energy of 20 eV for atorvastatin.

A calibration curve of OL in plasma was conducted in

concentrations ranged from 0.1 to 30 ng/ml. A solvent

extraction procedure was used. Human plasma samples

(500 mL) and 50 mL of internal standard solution were placed

in 10 mL glass tubes then vortexed for 15 s. Five mL methyl-

ter-butyl ether was added and samples were then vortexed for

1 min. The tubes were then centrifuged for 5 min at 4000 rpm.

The upper organic phases were then transferred to clean glass

tubes and evaporated to dryness at 45 �C. Dry residues were

dissolved in 150mL of mobile phase and vortexed for 1 min to

reconstitute residues, and 10 mL were injected using the

autosampler.

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 5

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 7: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Pharmacokinetic analysis

Pharmacokinetic parameters were calculated from plasma

data using WinNonlin� (version 1.5, Scientific consulting,

Inc., Cary, NC). Non-compartmental analysis was adopted for

the calculation of the observed maximal drug concentration

Cmax (ng/mL) and the time needed to reach this concentration

tmax (h). The relative bioavailability was calculated from the

24 h area-under-the-plasma concentration versus time curve as

shown in the following equation:

Rel:Bioavailability ¼ AUCtest=AUCstandardð Þ�100 ð3Þ

Statistical analysis of the data was undertaken using

analysis of variance (ANOVA) test performed for the

untransformed data for the pharmacokinetic parameters

Cmax, tmax, AUC0–24, and t1/2 using the software SPSS 11.0

(SPSS Inc., Chicago, IL). The significance level was set at

p value¼ 0.05.

Result and discussion

Characterization of the co-amorphous dispersions

Saturated solubility

All the tested COAD samples showed increased OL water

solubility over the crystalline form (Table 4). The maximum

solubility (602-folds) was observed with ascorbic acid at a

molar ratio 1:2 (COAD2), so it was selected for performing

further solid state analysis and formulation of the oral fast

dissolving film.

Scanning electron microscopy

The results of SEM (Figure 1) indicated that large difference

in size and shape existed between the crystalline OL and the

ascorbic acid with smaller OL and larger cubic crystals of

ascorbic acid (Figure 1A and B). The co-amorphous disper-

sions COAD1 and COAD2 demonstrated the characteristic

amorphous aggregates and absence of the defined shape of

crystals (Figure 1C and D) The OFDF prepared from both

dispersions showed almost clear, transparent glassy, and

homogenous layer with completely dissolved OL into the

polymer matrix (Figure 1E and F).

Differential scanning calorimetry

DSC thermograms of the drug, ascorbic acid, physical

mixture, and solid dispersion systems are shown in Figure

2. The thermogram of OL showed a highly crystalline

component with a sharp endothermic peak at 194.01 �C(DH¼ 126.83 J/g) corresponding to its melting point. The

thermogram of ascorbic acid also showed a melting endo-

thermic peak at 191.24 �C. Physical mixtures showed broad

endotherm without sharp melting indicating interactions and/

or miscibility of the two components. Thermograms of the

dispersions containing OL–ascorbic acid in molar ratios; 1:1

(COAD1)and 1:2 (COAD2), showed broad peaks with

complete disappearance of the characteristic melting endo-

therms of parent components.

Fourier transform infra-red analysis

The FTIR spectra of OL, ascorbic acid, physical mixture

(1:1), and solid dispersions are presented in Figure 3. Pure OL

showed characteristic absorption bands at 3217 cm�1 (NH

stretching), 2929, 2836, and 2791 cm�1 (C–H stretching),

1586 cm�1 (C¼C stretching), 1461 cm�1 (C¼N stretching),

and 1283 cm�1 C–N stretching.(Ayala et al., 2006; Hiriyanna

et al., 2008) The FTIR spectra of ascorbic acid indicated a

characteristic peak at 1754 cm�1 which is attributed to the

C¼O stretching vibration commonly observed for carboxyl

groups (Zhang et al., 2015). The characteristic peaks of pure

OL and ascorbic acid were found to be present in the spectra

of the physical mixture; however, they were shorter than

original components. The prepared solid dispersion formula-

tions (COAD1 and COAD2) showed shortening, broadening,

and shifting of certain peaks of OL.

X-ray diffraction

The diffraction spectrum of pure OL (Figure 4) showed highly

crystalline component as demonstrated by the characteristic

intense peaks at 2� 8.67, 17.09, 19.87, 21.05, 21.54, and

23.95� which were identical to those reported in the literature

(Tiwari et al., 2007). The XRD pattern for ascorbic acid also

showed multiple sharp characteristic diffraction lines at 2�10.63, 15.89, 17.59, 25.33, 27.25, 28.19, 30.18, and 34.85�

indicating a purely crystalline compound. The XRD spectra of

the drug–ascorbic acid 1:1 physical mixture showed less

intense spectrum compared with the parent components. The

diffraction lines of the expected COADs demonstrated the

disappearance of the characteristic peaks of both OL and

ascorbic acid.

Physicochemical evaluation of the prepared films

The prepared films were elegant, transparent, and flexible

with smooth surface and showed no blooming. The measured

average thickness of the films was 0.18 ± 0.02 mm. The

surface pH of all prepared films was 5.90–6.90 which is in the

range of salivary pH, hence no expected sensation of irritation

to the oral cavity was found. The drug content values were in

the range of 96.79–99.37% for the different formulations with

standard deviation less than 2% indicating that the drug was

uniformly dispersed throughout the film. The TS of the films

was found to be in the ranged 2.41–7.08 N/cm2, which is

considered adequate TS for handling and certain flexibility to

guarantee patient compliance (Lim & Hoag, 2013). The

results of folding endurance were found to be between 95 and

278 times, which could be considered relatively variable due

Table 4. Saturated solubility of co-amorphous dispersions compared topure OL.

Type offormulation

Conformeracid

Molarratio

Saturatedsolubility(mg/mL)

Numberof folds

increase insolubility

Percentageincrease insolubility

Olanzapine – – 0.0334 ± 0.015 – –F1 Ascorbic 1:1 6.303 ± 0.38 191 530F2 Ascorbic 1:2 19.855 ± 0.52 602 1886F3 Citric 1:1 4.293 ± 0.23 130 329F4 Citric 1:2 15.742 ± 0.42 477 1474F5 Tartaric 1:1 2.837 ± 0.11 86 184F6 Tartaric 1:2 12.489 ± 0.44 378 1149

6 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 8: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Figure 1. Scanning electron micrographs of pure olanzapine (A), its physical mixture with ascorbic acid (B), co-amorphous dispersion 1:1 (C),co-amorphous dispersion 1:2 (D), 1:1 film (E), and 1:2 film (F).

Figure 3. IR spectra for olanzapine (OL), ascorbic acid (AS), their 1:1physical mixture (PM), and co-amorphous dispersions COAD1 (1:1) andCOAD2 (1:2).

Figure 2. DSC thermograms for olanzapine (OL), ascorbic acid (As),(1:1) physical mixture (PM), and co-amorphous dispersions COAD 1(1:1) and COAD 2 (1:2).

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 7

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 9: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

to subject hand errors. The folding endurance was directly

proportional to concentration of polymer and inversely to

plasticizer concentration.

Assessment of in vitro disintegration and dissolutionof the film

The disintegration time of the films was in the ranged of

4.00–51.00 s which is less than 1 min and considered accept-

able time as reported in previous studies (Liew et al., 2012).

It was clear that the time was increased as the polymer

concentration increased. The in vitro dissolution profiles of

OL from the optimized film in SSF (pH¼ 5.8) compared with

OL powder are shown in Figure 5. The optimized fast

dissolving film achieved almost complete dissolution within

10 min compared with only 55.34% dissolved for those

containing crystalline drug (30 min) which were also com-

patible with fast dissolving films reported in the literature

(Vila et al., 2014). These results indicated that the COAD

used to prepare the films greatly enhanced the extent and rate

of dissolution of OL from the prepared OFDF. This

enhancement was a result of the synchronized dissolution of

OL and ascorbic acid brought about by the co-amorphous

dispersion (Lobmann et al., 2011). The cumulative percentage

of drug released after 10 min (Q10min) was evaluated among

different films. The drug dissolution from all formulae

decreased as the concentration of the polymer increased,

this expected result was in agreement with the data reported in

the literature for fast dissolving oral films (Scott et al., 2013).

Modeling of film formulation

The relationships between film formulation variables and

each of the output properties could be explained from the

response surface plots shown in Figure 6. The results of the

modeling experiment indicated a highly trusted predictive

model as confirmed by high training correlation R2 (92–

99%), testing R2 (78–94%), and validation R2 (87–89.90%).

The model trustability is also confirmed by the low values

of the RMSE of training, testing, and validation data sets

(Tables 5 and 6). The effects of polymer type and

concentration on the film disintegration time demonstrated

that polymer 1 (HPMC E15) resulted in lower disintegration

time than polymer 2 (NaCMC). The time was also increased

at higher concentration of polymers (200 mg) as shown in

Figure 6(A). The plasticizer type 4 (PEG 400) showed

lowest values of disintegration time of the film compared

with PG (type 3) and glycerin (type 5) and the time linearly

decreased with increased plasticizer concentration (Figure

6B). For tensile strength, both polymer type and concentra-

tion were found to directly increase film tensile strength

(Figure 6C). As previously noted with disintegration time,

the tensile strength of the film had its lowest values with

plasticizer 4 (PEG 400) as demonstrated by the large

curvature of Figure 6(D). It was also noticed that PG showed

somewhat higher tensile strength than glycerin and the

increased concentration of any of the plasticizers led to a

direct decrease in film strength. On one hand, for, percent-

age OL released from the film, the relationship observed

with polymer type indicated that HPMC E5 (type 1) resulted

in higher % drug release than type 2 (NaCMC) as illustrated

in Figure 6(E). This could be due to the formation of

stronger matrix layer with higher tortuosity and poor water

porosity for diffusion of drug caused by more intimate

contact between particles of HPMC at high concentration

(Sapkal et al., 2011). Moreover, higher polymer concentra-

tion resulted in viscous environment of the system retarding

water movement into the matrix and diffusion of the drug

into the surroundings (Dunn & English, 2002). On the other

hand, NaCMC has higher molecular weight than HPMC E5

which supports the formation of stronger matrix and lower

release rates (Table 7).

The type of plasticizer showed a slow increase in % drug

released moving from plasticizer 3-4-5 (PG-PEG400-Gly) and

the increased concentration of the three polymers led to a

direct increase in % OL released up to a plasticizer

concentration of 20–22% followed by a slow decrease

possibly due to increased viscosity of the film matrix

(Figure 6F). Folding endurance unfortunately did not result

in a good model during preliminary model training most

probably due to high variability and scatter of its values and

hence was excluded from the final model.

Figure 4. X-ray diffraction lines of olanzapine (OL), ascorbic acid (AS),1:1 physical mixture (PM), 1:1 co-amorphous dispersion (COAD1), and1:2 co-amorphous dispersion (COAD2).

Figure 5. Dissolution profiles of pure olanzapine (OL) compared withthe optimized fast dissolving film (OFDF).

8 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 10: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Model optimization

The model-generated optimized formulation for OL film was

composed of HPMC E5 or polymer 1 (178.01 mg) and PEG

400 or plasticizer type 4 (40 mg) as demonstrated in Table 7.

The desirability of the obtained model exceeded 99% which

represents high closeness of the obtained model predictions

from the desired values entered during optimization. The

experimental testing of the optimized formulation indicated

similar properties to those predicted by the model. The actual

tensile strength was found to be 4.93 N/cm2, the disintegration

time was 8.52 (s), and the percentage OL released was

97.47(%), respectively. The difference between the actual and

the model predicted properties was found to be statistically

insignificant (p50.05) which ensured model trustability.

Accelerated stability studies

Fast dissolving films were found to be physically and

chemically stable at the selected temperature and humidity

(40 �C/75%RH) with no significant change in terms of

physical characteristics and drug content (98.33%). The

percentage OL remaining up to the end of the storage

period (3 months) at the above-mentioned storage conditions

was found to be within the USP-permitted values (90–110%)

of the original content in the film (5 mg/4 cm2).

Figure 6. 3D response surface plots showing effects of different input variables on film disintegration time (A and B), tensile strength (C and D), and %OL released (E and F).

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 9

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 11: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

In vivo evaluation of the film

The mean plasma concentration–time curves of OL following

oral administration of the optimized formula (OFDF), the oral

generic product A, and the fast dissolving Product B tablets are

shown in Figure 7. Higher mean Cmax were obtained from the

OFDF followed by product B and product A tablets. However,

the differences between the three treatments for Cmax were

found to be statistically insignificant (p50.05). The mean

AUC0–24 h which reflects the total amount of drug absorbed

over the 24 h time period was significantly higher for the fast

dissolving film under test compared with the two reference

formulations (Table 8). Shorter tmax was observed with the

film followed by the references B and A tablets.

Table 5. Model training and testing ANOVA statistics for OL fast dissolving film output properties.

Output property Source of variation Sum of squares Degrees of freedom Mean squares RMSE Computed f ratio

Tensile strength (N/cm2) Model 43.4954 19 2.28923 1.513 28.8634Error 0.0793126 1 0.07931 0.282Total 43.6201 20

Covariance term Sum of errors0.0453962 0.0014496

Train Set R2 99.82%Test Set R2 94.69%

Disintegration time (s) Model 3955.37 19 208.177 14.428 10.0595Error 20.6946 1 20.6946 4.549Total 3977.24 20

Covariance term Sum of errors1.17717 0.0488199

Train set R2 99.48%Test set R2 91.40%

% Released (10 min) Model 979.519 19 51.5537 7.180 0.672918Error 76.6121 1 76.6121 8.753Total 1054.28 20

Covariance term Sum of errors1.85219 0.308051

Train Set R2 92.73%Test Set R2 78.65%

Table 6. Model validation ANOVA statistics for each output property.

Output property Source of variation Sum of squares Degrees of freedom Mean squares RMSE Computed f ratio

Tensile strength (N/cm2) Model 5.7283 19 0.30149 0.549 11.81Error 0.43385 17 0.02552 0.160Total 3.8653 2

Covariance term Sum of errors2.29694 0.51135

Validation R2 88.78%Disintegration time (s) Model 353.2 19 18.5895 4.312 6.79

Error 46.517 17 2.7363 1.654Total 368.452 2

Covariance term Sum of errors31.2657 7.8533

Validation R2 87.37%% Released (10 min) Model 83.5956 19 4.3997 2.098 4.81

Error 15.5611 17 0.9154 0.957Total 140.236 2

Covariance term Sum of errors41.0797 0.4412

Validation R2 88.90%

Table 7. Model optimized solution for desirable film properties.

X1 X2 X3 X4 Y1 Y2 Y3

Optimizedsolution Desirability

Polymertype

polymerconcentration (mg)

Plasticizertype

Plasticizerconcentration (mg)

Tensilestrength (N/cm2)

Disintegrationtime (s)

% released(10 min)

Model generated 0.99 1.00 178.01 4.00 40.00 5.00 8.06 95.04Experimental – 1.00 178.01 4.00 40.00 4.93 8.52 97.47

10 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 12: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Discussion

The use of ascorbic acid in the preparation solid dispersions

and successful OL COAD systems was found to be useful in

enhancing water solubility of OL by more than 600-folds

which led to high dissolution and expected maximized

absorption. This approach is considered a highly effective

method usually used for the formation of cocrystals to

improve solubility of poorly soluble drugs (McNamara et al.,

2006). However, in this study, a co-amorphous dispersion was

obtained at molar ratios of OL:ascorbic acid; 1:2 and 1:3

using rapid solvent evaporation under vacuum. The inter-

action between ascorbic acid and OL was effected most

probably by H-bonding of the NH group of OL and the

carbonyl group of ascorbic acid (Forster et al., 2001). In

addition to its solubility enhancing effect in the prepared

dispersions, ascorbic acid also is expected to provide

antioxidant and neuroprotective effects to the final film

formulation (Allahtavakoli et al., 2015; Xu et al., 2015).

The DSC results above confirmed the formation of

homogenous single phases between OL and ascorbic acid

through complete amorphization and absence of true melting

endotherms. This complete miscibility between the two

components at 1:1 and 1:2 molar ratios as indicated by the

broad DSC thermograms of the dispersions (Figure 2) may

suggest the formation of homogenous co-amorphous disper-

sions (Ali et al., 2015; Jensen et al., 2015).

From the IR data, it becomes obvious also that interactions

between functional groups of OL and As acid have taken

place. For example, the C–H stretching and C¼N stretching

were shortened and broadened and the shallow peak of N–H

stretching also demonstrated shifting to 3394 cm�1 instead of

3217 cm�1 (Figure 3). These results strongly suggest that the

interaction between OL and ascorbic acid may have taken

place through H-bonding (Mistry et al., 2015).

The XRD results (Figure 4) also confirm the interactions

and formation of single homogenous phase by the appearance

of the amorphous halo characteristic of single co-amorphous

dispersions (Lobmann et al., 2013; Dengale et al., 2014).

The incorporation of OL–ascorbic acid co-amorphous

dispersions in fast dissolving oral film was intended to

provide a simple easily administered dosage form for the

application inside the mouth. The formulated film was found

to have acceptable transparent appearance and mechanical

properties, disintegration, and dissolution. Almost complete

dissolution from the film was attained within 10 min. Also,

the drug was found to be stable in the film for up to 12 weeks

at 40 �C and 75% RH which could be attributed to compati-

bility between polymers used in the film (PEG 400 and

HPMC E5) and the COAD system. This increased phase

stability possibly occurred through extending the supersatur-

ation state (Kawakami, 2012).

From the above response surface plots, it could be

concluded that HPMC E5 as the film forming polymer and

PEG 400 as the plasticizer had the major effects on optimum

film properties (tensile strength, disintegration time, and %

OL released). Therefore, the model optimization resulted in a

suggested formula with maximum dissolution and fast

disintegration containing the above-mentioned polymers.

This formula when tested for the actual properties was

found to have the required disintegration, dissolution, and

mechanical strength as those predicted by the model. The

comparative pharmacokinetic evaluation of the film proved

that formulation of OL in the form of OFDF using the co-

amorphous phase was a successful strategy for enhancing the

pharmacokinetic parameters. The higher Cmax, short tmax, and

the improved bioavailability observed for OFDF relative to

the commercially available formulations could be attributed to

the rapid disintegration and dissolution of OL in saliva

brought about by the COAD system incorporated into the

optimized film leading to fast absorption.

Figure 7. Plasma concentration versus timecurves of olanzapine following oral adminis-tration of Olazine tablets (product A),Zyprexa� velotab (product B), and fastdissolving film (OFDF) to human volunteers.

Table 8. Comparative pharmacokinetics following oral administration ofOL film and reference formulations (products A and B) to humanvolunteers.

Formula

Parameter Product A Product B OF FDF

Cmax (ng/ml) 10.85 ± 1.29 12.96 ± 2.64 14.22 ± 2.95tmax (h) 5.50 ± 1.00 3.75 ± 0.50 2.75 ± 0.50t½ el (h) 8.29 ± 0.84 7.67 ± 0.96 7.74 ± 1.44AUC(0–24) (ng h/ml) 113.61 ± 24.82 125.34 ± 34.18 144.44 ± 43.01R. bioavailability 90.64% 100% 115.83%

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 11

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 13: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Conclusion

The physicochemical properties of OL were highly improved

using the principle of solid dispersion amorphization with a

highly soluble coformer such as ascorbic acid. The results of

solid state characterization of OL COADs indicated the

formation of single homogenous phase through H-bonding

interactions. This homogenous dispersion was further stabi-

lized through incorporation with selected polymers to form a

fast dissolving oral film. The optimized fast dissolving films

were successful formulations which provided highly dissol-

ving OL in a simple and easily administered dosage form for

psychotic patients. The pharmacokinetics data indicated that

the OFDF ensured improved bioavailability of OL compared

with the marketed reference products. This can be attributed

to faster dissolution leading to rapid absorption of OL from

the buccal mucosa which undoubtedly resulted in a decreased

pre-systemic biotransformation and maximized the

bioavailability.

Acknowledgements

The authors of this manuscript acknowledge the help given by

the technicians working at the central laboratory of Beni Suef

University for help and support during use of the LC-MS and

SEM equipment. Also, a special acknowledgment is directed

to all members of the human research ethical committee at

Faculty of medicine, Assiut University, Egypt. The human in

vivo study performed in this research paper was undertaken

according to the guidelines outlined in the Helsinki agreement

for human research.

Declaration of interest

The authors report that they have no conflicts of interest

References

Abdelrahman AA, Salem HF, Khallaf RA, Ali AMA. (2015). Modeling,optimization, and in vitro corneal permeation of chitosan–lomefloxa-cin HCl nanosuspension intended for ophthalmic delivery. J PharmInnov 10:254–68.

Akil A, Parniak MA, Dezzutti CS, et al. (2011). Development andcharacterization of a vaginal film containing dapivirine, a non-nucleoside reverse transcriptase inhibitor (NNRTI), for prevention ofHIV-1 sexual transmission. Drug Deliv Transl Res 1:209–22.

Ali AA, Ali AM. (2013). Optimization of propranolol HCl releasekinetics from press coated sustained release tablets. Pharm DevTechnol 18:1238–46.

Ali AMA, Abdelrahim MEA. (2014). Modeling and optimization ofterbutaline emitted from a dry powder inhaler and influence onsystemic bioavailability using data mining technology. J Pharm Innov9:38–47.

Ali AMA, Ali AA, Maghrabi IA. (2015). Clozapine–carboxylic acidplasticized co-amorphous dispersions: preparation, characterizationand solution stability evaluation. Acta Pharm 65:133–46.

Allahtavakoli M, Amin F, Esmaeeli-Nadimi A, et al. (2015). Ascorbicacid reduces the adverse effects of delayed administration of tissueplasminogen activator in a rat stroke model. Basic Clin PharmacolToxicol 117:335–9.

Ayala AP, Siesler H, Boese R, et al. (2006). Solid state characterizationof olanzapine polymorphs using vibrational spectroscopy. Int J Pharm326:69–79.

Cavallari C, Fini A, Ceschel G. (2013). Design of olanzapine/lutrol soliddispersions of improved stability and performances. Pharmaceutics 5:570–90.

Curtin V, Amharar Y, Hu Y, et al. (2012). Investigation of thecapacity of low glass transition temperature excipients to minimize

amorphization of sulfadimidine on comilling. Mol Pharm 10:386–96.

Dahiya M, Saha S, Shahiwala AF. (2009). A review on mouth dissolvingfilms. Curr Drug Deliv 6:469–76.

Dengale SJ, Ranjan OP, Hussen SS, et al. (2014). Preparation andcharacterization of co-amorphous Ritonavir–Indomethacin systems bysolvent evaporation technique: improved dissolution behavior andphysical stability without evidence of intermolecular interactions. EurJ Pharm Sci 62:57–64.

Dixit R, Puthli S. (2009). Oral strip technology: overview and futurepotential. J Control Release 139:94–107.

Dunn RL, English JP. (2002). Biodegradable polymer composition, USPatent 20,020,090,398.

El-Setouhy DA, El-Malak NSA. (2010). Formulation of a noveltianeptine sodium orodispersible film. AAPS PharmSciTech 11:1018–25.

El Meshad AN, El Hagrasy AS. (2011). Characterization and optimiza-tion of orodispersible mosapride film formulations. AAPSPharmSciTech 12:1384–92.

Farid M, El-Setouhy DA, El-Nabarawi MA, El-Bayomi T. (2015).Particle engineering/different film approaches for earlier absorption ofmeloxicam. Drug Deliv. [Epub ahead of print]. doi: 10.3109/10717544.2014.982262.

Felton LA, Donnell PBO, McGinity JW. (2008). Mechanical propertiesof polymeric films prepared from aqueous dispersions. Drugs PharmSci 176:105.

Forster A, Hempenstall J, Rades T. (2001). Characterization of glasssolutions of poorly water-soluble drugs produced by melt extrusionwith hydrophilic amorphous polymers. J Pharm Pharmacol 53:303–15.

Harde H, Das M, Jain S. (2011). Solid lipid nanoparticles: anoral bioavailability enhancer vehicle. Expert Opin Drug Deliv 8:1407–24.

Hiriyanna S, Basavaiah K, Goud P, et al. (2008). Identification andcharacterization of olanzapine degradation products under oxidativestress conditions. Acta Chromatogr 20:81–93.

Hoffmann EM, Breitenbach A, Breitkreutz J. (2011). Advances inorodispersible films for drug delivery. Expert Opin Drug Deliv 8:299–316.

Hosny KM, El-Say KM, Ahmed OA. (2016). Optimized sildenafil citratefast orodissolvable film: a promising formula for overcoming thebarriers hindering erectile dysfunction treatment. Drug Deliv 23:355–61.

Jensen KT, Blaabjerg LI, Lenz E, et al. (2015). Preparation andcharacterization of spray-dried co-amorphous drug–amino acidsalts. J Pharm Pharmacol. [Epub ahead of print]. doi: 10.1111/jphp.12458.

Jensen KT, Larsen FH, Cornett C, et al. (2015). Formation mechanism ofcoamorphous drug-amino acid mixtures. Mol Pharm 12:2484–92.

Kawakami K. (2012). Modification of physicochemical characteristics ofactive pharmaceutical ingredients and application of supersaturatabledosage forms for improving bioavailability of poorly absorbed drugs.Adv Drug Deliv Rev 64:480–95.

Krishnamoorthy V, Nagalingam A, Priya Ranjan Prasad V, et al. (2009).Characterization of olanzapine-solid dispersions. Iran J PharmaceutResh: IJPR 10:13–24.

Kumria R, Nair AB, Goomber G, Gupta S. (2016). Buccal films ofprednisolone with enhanced bioavailability. Drug Deliv 23:471–8.

Kunte S, Tandale P. (2010). Fast dissolving strips: a novel approach forthe delivery of verapamil. J Pharm Bioallied Sci 2:325–8.

Liang AC, Chen LlH. (2001). Fast-dissolving intraoral drug deliverysystems. Expert Opin Ther Pat 11:981–6.

Liew KB, Tan YTF, Peh KK. (2012). Characterization of oraldisintegrating film containing donepezil for Alzheimer disease.AAPS PharmSciTech 13:134–42.

Lim H, Hoag SW. (2013). Plasticizer effects on physical-mechanicalproperties of solvent cast Soluplus� films. AAPS PharmSciTech 14:903–10.

Lobmann K, Grohganz H, Laitinen R, et al. (2013). Amino acids as co-amorphous stabilizers for poorly water soluble drugs – Part 1:preparation, stability and dissolution enhancement. Eur J PharmBiopharm 85:873–81.

Lobmann K, Laitinen R, Grohganz H, et al. (2011). Coamorphous drugsystems: enhanced physical stability and dissolution rate of indo-methacin and naproxen. Mol Pharm 8:1919–28.

12 E. M. Maher et al. Drug Deliv, Early Online: 1–13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6

Page 14: In vitro/in vivo evaluation of an optimized fast ...€¦ · Salem & Ahmed Abdelbary Abdelrahman (2016): In vitro/in vivo evaluation of an optimized fast dissolving oral film containing

Low AQJ, Parmentier J, Khong YM, et al. (2013). Effect of type andratio of solubilising polymer on characteristics of hot-melt extrudedorodispersible films. Int J Pharm 455:138–47.

Matuszewski B, Constanzer M, Chavez-Eng C. (1998).Matrix effect in quantitative LC/MS/MS analyses of biologicalfluids: a method for determination of finasteride in humanplasma at picogram per milliliter concentrations. Anal Chem 70:882–9.

McNamara DP, Childs SL, Giordano J, et al. (2006). Use of a glutaricacid cocrystal to improve oral bioavailability of a low solubility API.Pharm Res 23:1888–97.

Mishra D, Gilhotra R. (2008). Design and characterization ofbioadhesive in-situ gelling ocular inserts of gatifloxacin sesquihy-drate. DARU J Pharm Sci 16:1–8.

Mistry P, Mohapatra S, Gopinath T, et al. (2015). Role of the strength ofdrug–polymer interactions on the molecular mobility and crystalliza-tion inhibition in ketoconazole solid dispersions. Mol Pharm 12:3339–50.

Plumb AP, Rowe RC, York P, Brown M. (2005). Optimisation of thepredictive ability of artificial neural network (ANN) models: acomparison of three ANN programs and four classes of trainingalgorithm. Eur J Pharm Sci 25:395–405.

Plumb AP, Rowe RC, York P, Doherty C. (2002). The effect ofexperimental design on the modeling of a tablet coating formulationusing artificial neural networks. Eur J Pharm Sci 16:281–8.

Preis M, Pein M, Breitkreutz J. (2012). Development of a taste-maskedorodispersible film containing dimenhydrinate. Pharmaceutics 4:551–62.

Qian S, Heng W, Wei Y, et al. (2015). Coamorphous lurasidonehydrochloride� saccharin with charge-assisted hydrogen bondinginteraction shows improved physical stability and enhanced dissol-ution with pH-independent solubility behavior. Cryst Growth Des 15:2920–28.

Sapkal NP, Kilor VA, Daud AS, Bonde MN. (2011). Development of fastdissolving oral thin films of ambroxol hydrochloride: effect offormulation variables. J Adv Pharm Res 2:102–9.

Saurabh R, Malviya R, Sharma PK. (2011). Trends in buccal film:formulation characteristics, recent studies and patents. Eur J Appl Sci3:93–101.

Scott RA, Park K, Panitch A. (2013). Water soluble polymer films forintravascular drug delivery of antithrombotic biomolecules. Eur JPharm Biopharm 84:125–31.

Seju U, Kumar A, Sawant KK. (2011). Development and evaluation ofolanzapine-loaded PLGA nanoparticles for nose-to-brain delivery: invitro and in vivo studies. Acta Biomater 7:4169–76.

Shamma R, Elkasabgy N. (2016). Design of freeze-dried Soluplus/polyvinyl alcohol-based film for the oral delivery of an insoluble drugfor the pediatric use. Drug Deliv 23:489–99.

Sievens-Figueroa L, Bhakay A, Jerez-Rozo JI, et al. (2012). Preparationand characterization of hydroxypropyl methyl cellulose films con-taining stable BCS Class II drug nanoparticles for pharmaceuticalapplications. Int J Pharm 423:496–508.

Sood S, Jawahar N, Jain K, et al. (2013). Olanzapine loaded cationicsolid lipid nanoparticles for improved oral bioavailability. CurrNanosci 9:26–34.

Tiwari M, Chawla G, Bansal AK. (2007). Quantification of olanzapinepolymorphs using powder X-ray diffraction technique. J PharmBiomed Anal 43:865–72.

Vila MM, Tardelli ER, Chaud MV, et al. (2014). Development of abuccal mucoadhesive film for fast dissolution: mathematical rationale,production and physicochemical characterization. Drug Deliv 21:530–9.

Volavka J, Czobor P, Sheitman B, et al. (2004). Clozapine, olanzapine,risperidone, and haloperidol in the treatment of patients with chronicschizophrenia and schizoaffective disorder. Focus 2:59–67.

Worrel JA, Marken PA, Beckman SE, Ruehter VL. (2000). Atypicalantipsychotic agents: a critical review. Am J Health Syst Pharm 57:238–55.

Xu KX, Tao J, Zhang N, Wang JZ. (2015). Neuroprotective properties ofvitamin C on equipotent anesthetic concentrations of desflurane,isoflurane, or sevoflurane in high fat diet fed neonatal mice. Int J ClinExp Med 8:10444–58.

Yoon SD, Park MH, Byun HS. (2012). Mechanical and water barrierproperties of starch/PVA composite films by adding nano-sized poly(methyl methacrylate-co-acrylamide) particles. Carbohydr Polym 87:676–86.

Zhang C, Dabbs DM, Liu LM, et al. (2015). Combined effects offunctional groups, lattice defects, and edges in the infrared spectra ofgraphene oxide. J Phys Chem C 119:18167–76.

Zhang Y, Liu Y, Luo Y, et al. (2014). Extruded Soluplus/SIM as an oraldelivery system: characterization, interactions, in vitro and in vivoevaluations. Drug Deliv. [Epub ahead of print]. doi: 10.3109/10717544.2014.960982.

DOI: 10.3109/10717544.2016.1153746 Evaluation of an optimized fast dissolving oral film 13

Dow

nloa

ded

by [

Ahm

ed A

li] a

t 12:

32 1

2 M

arch

201

6