DOE for Extended Release

Embed Size (px)

Citation preview

  • 8/9/2019 DOE for Extended Release

    1/9

    1

    PHARMAGENE Vol: 1 Issue: 2

    Optimization of Formulation Variables of Ranolazine Extended-Release

    Tablets by 32Full Factorial Design

    Shah Pranav*, Naik Bhargavi, Zalak Chandarana

    Maliba Pharmacy College, Bardoli, Gujarat

    ABSTRACT

    Ranolazine is antianginal drug, approved by US FDA in 2006. It is marketed as extended release tablets (Ranexa 500mg/1gm).

    Ranolazine is extensively metabolized in the liver and its absorption is highly variable. The present study was aimed to applyexperimental design in the development and optimization of drug release from extended release matrix tablets of Ranolazine(antianginal drug) using two factor three level (32) full factorial design. The extended release matrix tablets of Ranolazine were

    formulated using pH dependent polymer (Eudragit L 100-55), Sodium hydroxide, MCC, HPMC 5 cps and Magnesium stearate.

    The amount of independent variables, Eudragit L100-55 (X1) and Sodium Hydroxide (X2) were optimized on the basis of drug

    release profiles at 0.5, 4, 12, 24 hours (dependent variables ) of different tablets batches as per 32 full factorial design. Tabletswere prepared by wet granulation technique and evaluated for various physicochemical parameters and in vitro drug release.Polynomial equations and contour plots derived from the data obtained from 13 batches were used to predict the values of

    independent variables and their effect on dependent variables for the formulation of optimized tablets with desired properties.

    Optimized formulation from DOE had identical dissolution profile (f2 = 85.95 and f1 = 2.29) with innovators tablet. Stability

    studies of optimized batch were conducted at accelerated conditions for three months and tablets were found to be stable. Thus

    the study revealed that experimental design could efficiently be applied for optimization of amount of excipients affecting drug

    release. Also, it is an economical way of obtaining the maximum amount of information in a short period of time and with the

    few experiments.

    KEY WORDS: Ranolazine extended release tablet, Experimental design, Full Factorial Design,

    Received on 03-05-2013 Modified on 04-06-2013 Accepted on 10-06-2013

    INTRODUCTION

    Oral administration of drugs is strongly preferred because

    of its convenience, relatively low production cost and the

    high level of patient safety. However there are someproblems associated with the oral drug delivery such as

    poor bioavailability , high first pass metabolism, frequent

    drug administration etc. Extended-release systems allow

    the drug to be released over prolonged time periods. Byextending the release profile of a drug, the frequency of

    dosing can be reduced. Extended release can be achieved

    using sustained or controlled release dosage forms [1].

    *Address for correspondence:

    Dr. Pranav Shah, Professor,

    Maliba Pharmacy College, Bardoli, Gujarat, India

    Email:pranav.shah@ut u.ac.in

    The oral extended release system shows a typical pattern of

    drug release in which the drug concentration can be

    maintained in the therapeutic window for a prolongedperiod of time (extended release), thereby ensuring

    controlled t herapeutic action.

    Ranolazine is antianginal drug, approved by US FDA in2006. It is marketed as extended release tablets (Ranexa

    500mg/1gm). Ranolazine is extensively metabolized in the

    liver and its absorption is highly variable. The apparentterminal half-life of Ranolazine is 7 hrs. Ranolazine has

    relatively high solubility (42.08 mg/ml in 0.1 N HCl) at low

    pH in the stomach (pH 1.2 3). The high acid soluble

    property of ranolazine results in rapid drug absorpt ion and

    clearance, causing large and undesirable fluctuations in

    PHARMAGENEVol: 1 Issue: 2

    www.genesisjournals.org

    ISSN-2321-0966 (Print) ISSN-2321-0974 (Online)

    Research rticle

  • 8/9/2019 DOE for Extended Release

    2/9

    2

    PHARMAGENE Vol: 1 Issue: 2

    plasma concentration of ranolazine and short duration of

    action, thus necessitating frequent oral administration for

    adequate treatment [2]. The present study was aimed to

    develop a matrix tablet using pH-dependent polymer whichis insoluble at low pH and begins to dissolve at about pH >

    5. The extended release tablets were formulated using pH

    dependent polymer (Eudragit L 100-55), pH independent

    binder (HPMC 5 cps) and sodium hydroxide as neutralizing

    agent. Sodium hydroxide facilitates the conversion of the

    Eudragit L 100-55 into the latex like film formed around

    the individual granules which controls the drug release

    from the formulation above pH 4.5 [3].Statistical experimental design methodologies are

    powerful, efficient and systematic tools in the design of

    pharmaceutical dosage forms, allowing a rationalistic study

    of the influence of formulation and/or processing

    parameters on the selected responses with short experiment

    time and improvement in the research and development

    work [4-6]. The main objective of the experimental design

    strategies is to plan experiments in order to obtain themaximum information regarding the considered

    experimental domain with the lowest number of

    experiments [7]. Moreover, the multi-variant strategy of

    experimental design enables the simultaneous evaluation of

    the influence of the different variables involved in any

    process, being therefore part icularly useful when, as in the

    case of pre-formulation studies, multiple factors have to beevaluated simultaneously.

    In particular, optimization by means of statistical

    experimental design methodologies has been successfully

    applied in the development of different kinds of modified

    release dosage forms, allowing a quick and efficient

    quantification and prediction of the effects of formulationchanges on t he considered crucial responses [8-13].

    MATERIALS AND METHODS

    Materials

    Ranolazine was purchased from Virdev Intermediates,

    Surat, India. Eudragit L 100-55 was obtained from Evonik

    Industries, Germany. Microcrystalline cellulose was

    purchased from FMC Biopolymer, India. HPMC 5cps was

    purchased from Colorcon, Goa, India. Sodium hydroxidewas purchased from Cadila Pharmaceutical Ltd., Dholka,

    India and Magnesium stearate was purchased from Skant

    Healthcare Ltd., M umbai, India.

    Experimental Design

    Two factor three level (32) full factorial design was

    employed for development of ranolazine extended release

    matrix tablets. A translation of coded values of independentvariables and experimental design is executed as in t able 1.

    Independent variables were as follow:

    X1 : Amount of Eudragit L 100-55 (mg/tab)

    X2 : Amount of Sodium hydroxide (mg/tab)

    Dependent variables evaluated were as follows:

    Y1 = % drug released in 0.5 hours

    Y2 = % drug released in 4 hours

    Y3 = % drug released in 12 hours

    Y4 = % drug released in 24 hours

    Preformulation studiesPreformulation studies were designed to identify

    physicochemical properties of Ranolazine and excipients

    that may influence formulation design and method of

    manufacture.

    Table 1: Execution of experimental design and coding

    of actual values of independent variables for factorial

    design

    Batch Level of Factor

    X1

    Amount of

    Eudragit L 100-55 (mg/tablet)

    Level of Factor

    X2

    Amount of

    Sodiumhydroxide

    (mg/tablet)

    F1 -1(67) -1(2.6)

    F2 0(83.50) -1(2.6)

    F3 +1(100) -1(2.6)

    F4 -1(67) 0(3.9)

    F5 0(83.50) 0(3.9)

    F6 +1(100) 0(3.9)

    F7 -1(67) +1(5.2)

    F8 0(83.50) +1(5.2)

    F9 +1(100) +1(5.2)

    F10* 0(83.50) 0(3.9)

    F11* 0(83.50) 0(3.9)

    F12* 0(83.50) 0(3.9)

    F13* 0(83.50) 0(3.9)

    *Centre point batches

    Analytical method development

    HPLC method was developed to perform assay and

    analysis of dissolution samples of Ranolazine. HPLCanalysis of samples were done using Phenomenex Luna

    ODS 250 mm 4.6 mm, 5 microns column with mobile

    phase flow rate 1.0 ml/min. Mobile phase consist of buffer

    pH 7.5 and acetonitrile in the ratio of 40:60. Sample andstandards were dissolved in mobile phase and detected

    using UV detector at wavelength 225nm. Fig. 1 shows the

    HPLC chromatogram of standard ranolazine (100g/ml)

    with retention time of 8.487 minutes.

  • 8/9/2019 DOE for Extended Release

    3/9

    3

    PHARMAGENE Vol: 1 Issue: 2

    Preparation of tablets

    Tablets were prepared by wet granulation technique. Each

    batch of tablets (F1F13) (Table 2) has varied amount of

    Eudragit L100-55 and sodium hydroxide. All ingredientswere weighed accurately in required quantity. Ranolazine,

    Eudragit L 100-55, Avicel PH 101 and HPMC 5 cps were

    sifted through 20# sieve. The materials were mixed in r apid

    mixer granulator (RMG) at 75 rpm impeller speed. Binder

    solution was prepared by dissolving sodium hydroxide in

    sufficient amount of water. Granulation was done in RMG.

    The wet mass was dried in FBD at 60C for 20 minutes and

    the semi dried mass was passed through 16# and further

    dried at the same temperature till LOD value below 2% wasobtained on moisture balance. The dried granules were

    passed through 16# sieve. Sized granules were then mixed

    with previously sifted magnesium stearate (60#) for 3

    minutes. The tablets were compressed with 16.4 8 mm

    capsule shaped, standard concave punches with break line

    on one side and plain on other side (D tooling).

    Table 2: Formulation of tablets batches

    Ingredients

    (mg)

    Batches

    F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 F12 F13

    Ranolazine* 506 506 506 506 506 506 506 506 506 506 506 506 506

    EudragitL 100-55

    67 83.5 100 67 83.5 100 67 83.5 100 83.5 83.5 83.5 83.5

    HPMC 5 cps 13 13 13 13 13 13 13 13 13 13 13 13 13

    SodiumHydroxide

    2.6 2.6 2.6 3.9 3.9 3.9 5.2 5.2 5.2 3.9 3.9 3.9 3.9

    Magnesium

    Stearate

    13 13 13 13 13 13 13 13 13 13 13 13 13

    MCC PH 101 58.4 41.9 25.4 57.1 40.6 24.1 55.8 39.3 22.8 40.6 40.6 40.6 40.6

    Total tablet

    weight

    660 660 660 660 660 660 660 660 660 660 660 660 660

    * The potency of Ranolazine was found to be 506 mg for actual dose of 500 mg/tablet; LOD: 0.29%; Assay: 99.1%

    Figure 1: Chromatogram of standard Ranolazine (100g/mL) (Retention time: 8.487 min)

    Assay

    Assay of ranolazine was done using HPLC method.Standard solution was prepared by dissolving 50 mg of

    Ranolazine standard in 50 ml mobile phase in volumetric

    flask. Tablet powder equivalent to 50 mg of ranolazine was

    accurately weighed and transferred into 50 ml volumetric

    flask containing 25ml mobile phase (Buffer:Acetonitrile;

    40:60), sonicated for 30 minutes, allowed to cool to room

    temperature and diluted upto 50ml volume with mobilephase and mixed. Resulting solution was filtered through

    0.45 m PVDF Millipore filter discarding first few ml of

    the filtrate. 5.0 ml of clear filtrate was diluted to 50.0 ml

    with mobile phase and mixed. 20L of sample and standard

    preparation were injected into the column. Chromatogram

    was recorded and the response was measured at 225nm.

    (Figure: 1) Content of ranolazine per tablet was calculated.

    I n vitro drug release study (I n vi tro dissolution study) [14]

    In vitro drug release study was performed as per the

    following specifications of OGD, 900 ml of 0.1 N HCl

    medium at 50 rpm for 24 hours at 370.5 C. A 10 ml of

    sample from dissolution medium was withdrawn at

    predetermined time intervals (0.5, 2, 4, 8, 12, 20, 24 hours)

    and replaced by an equal volume of dissolution medium.

    The samples were filtered through 0.45m whatman filterpaper and 5.0 ml of filtrate was diluted to 20.0ml with

    dissolution medium. Samples were analyzed using HPLC.

    Mechanism of drug release [15-17]

    To evaluate the mechanism of drug release from the dosage

    form, data for the first 60% of drug release were plotted in

    Korsmeyers equation as log cumulative percentage of drug

  • 8/9/2019 DOE for Extended Release

    4/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    4

    PHARMAGENE Vol: 1 Issue: 2

    released vs log time, and the exponent n was calculated

    from the slope of the straight line.

    / = Where, Mt/M is the fractional solute release, t is the

    release time, K is a kinetic constant characteristic of the

    drug/ polymer system, and n is an exponent that

    characterizes the mechanism of release.

    Stability Study

    The optimized formulation was subjected to short term

    accelerated stability study (40C/75% RH) for the period of

    three months as per ICH guidelines. Physical stability was

    analyzed by recording the change in appearance, hardness,friability and chemical stability was analyzed by the change

    in the assay and in vitro drug dissolution at the end of three

    months.

    RESULTS AND DISCUSSION

    Preformulation studies

    Ranolazine was found to have very poor compressibility

    and flow properties (Carrs index: 36.67, Hausners ratio:

    1.5789), hence wet granulation method was opted for better

    compression and good flow property for the preparation of

    the Ranolazine matrix tablet.

    Precompression evaluation of granules exhibited good flow

    property (Hausners ratio < 1.25) and good compress ibility

    (Carrs index < 20%). Post compression evaluation of

    tablets such as appearance, dimensions, weight variation,

    hardness, friability, and assay were within thespecifications (Table 3).

    Table 3: In process quality control of Tablets

    Batch

    Average

    tablet weight

    (mg)

    n = 20

    Hardness

    (kg/cm2)

    n = 10

    Dimensions

    n = 6 % Friabil ity Assay

    n = 5Length

    (mm)

    Width

    (mm)

    Thickness

    (mm)

    F1 662.62.88 17.20.5 16.400.01 8.010.02 5.710.02 0.064 99.800.10

    F2 659.22.38 17.50.2 16.420.01 8.020.01 5.720.01 0.073 98.320.20

    F3 663.13.27 17.80.4 16.390.03 8.010.03 5.750.03 0.068 100.440.23

    F4 661.42.25 17.40.3 16.410.01 7.990.03 5.840.01 0.066 99.600.33

    F5 658.52.60 17.30.1 16.380.03 7.980.02 5.770.02 0.055 99.210.19

    F6 660.32.96 17.30.1 16.410.03 8.010.03 5.850.01 0.062 98.680.22

    F7 662.63.20 17.40.4 16.420.04 7.990.01 5.740.01 0.059 100.340.30

    F8 659.82.49 17.30.2 16.380.02 8.030.01 5.720.03 0.053 100.810.36F9 658.22.31 17.10.4 16.410.03 8.020.02 5.810.01 0.056 99.480.20

    F10 662.62.84 17.80.4 16.430.01 8.010.02 5.710.03 0.073 99.610.33

    F11 659.22.58 17.40.3 16.420.02 8.010.01 5.730.01 0.068 99.210.19

    F12 663.12.99 17.30.1 16.390.01 8.020.03 5.760.02 0.066 98.680.22

    F13 661.42.34 17.10.1 16.410.03 7.980.01 5.790.01 0.055 100.340.30

    I n vitro drug release study (I n vitro dissolution)

    In v itrodrug release data (Figure 2) showed that the drug

    release from all the formulated batches (F1-F13) (n = 3)

    was extended upto 24 hours.

    Figure 2: Comparative % cumulative drug release

    profile of formulations (F1F13)

  • 8/9/2019 DOE for Extended Release

    5/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    5

    PHARMAGENE Vol: 1 Issue: 2

    (a)

    (b)

    (c)

    (d)Figure 3: Linear correlation plots of dependent variables Y1-Y4 (a-d) between actual and predicted value

    Drug Release kinetics

    Dissolution data were fitted to zero order, first order,Higuchi and Korsmeyer kinetic treatment for all the

    formulations and different kinetic equat ions were app lied to

    interpret the release rate. The formulation with higher

    correlation coefficient R2 was found with Higuchis Model

    as shown in Table 4 indicating that release from gel

    forming system is based on diffusion mechanism for allformulations. The value of release exponent was more than

    0.45 and less than 0.798 indicating non-fickian anomalous

    release from all the formulations except F1.

    Statistical Analysis

    The two factor three level full factorial design allowed the

    development of mathematical equations, where predictedresults (Y) were assessed as a function of amount ofEudragit (X1) and amount of Sodium hydroxide (X2) and

    calculated as the sum of a constant, two first-order effects

    (terms in X1 and X2), one interaction effect (X12) and two

    second-order effects (X12and X22).

    The relationship between the two independent variables

    (amount of Eudragit L 100-55 and sodium hydroxide) and

    the four dependent variables (% drug release at 0.5, 4, 12

    and 24 hour) were analyzed using response surfacemethodology

    Data given in Table 5 depicts that all the models were

    significant at the 5% confidence level since P values were

    less than 0.05. The large P values for lack of fit (>0.05)

    presented in T able 5 (PLOF) show that theF-statistic was

    insignificant, implying significant model correlationbetween the variables and process responses. Adequate

    Precision (AP) values higher than four (Table 5) for all the

    responses confirmed that all predicted models can be used

    to navigate the design space defined by the full factorial

    design. For all the models % CV were not greater than 10%

    (Table 5) which indicate that models are reproducible.

    Model equation for the all the variables showed thenegative co-efficient terms for the first order effect,interaction term and second order effects which indicated

    the negative effect on the response with respect to the

    independent variable.

    Contour Plots and Response Surface Analysis

    Figure 4, 5, 6 and 7 (a and b) represents the response

    surface plot and contour plot of dependent variables Y1, Y2,

  • 8/9/2019 DOE for Extended Release

    6/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    6

    PHARMAGENE Vol: 1 Issue: 2

    Y3 and Y4 respectively. For responses Y1, Y2, Y3 and Y4

    drug release decreases rapidly with increase in amount of

    one variable while other at low level. This showed that both

    the variables (Eudragit L 100-55 and sodium hydroxide)had prominent negative effect on Y1, Y2, Y3 and Y4.These

    results are in confirmation with mechanism.

    As the concentration of pH dependent binder increases in

    the formulation, there is decrease in the release rate of

    ranolazine at pH below 4.5 as enteric coating formed by the

    binder was less soluble in acidic pH. Partial neutralizingagent, sodium hydroxide facilitated the conversion of the

    binder into the latex like film formed around the individual

    granules which controled the drug release from the

    formulation above pH 4.5.

    Table 4: Release kinetic data for F1-F13 formulations

    Batch no. Zero order

    kinetic R2

    First order

    kinetic R2

    Higuchi

    kinetic R2

    Korsmeyer Peppas

    R n

    F1 0.868 0.888 0.984 0.976 0.426

    F2 0.928 0.954 0.993 0.997 0.452

    F3 0.951 0.958 0.993 0.998 0.604

    F4 0.882 0.885 0.991 0.997 0.453

    F5 0.895 0.97 0.994 0.997 0.465

    F6 0.972 0.936 0.981 0.996 0.707F7 0.92 0.959 0.997 0.998 0.485

    F8 0.913 0.973 0.996 0.997 0.499

    F9 0.99 0.939 0.95 0.998 0.798

    F10 0.895 0.972 0.994 0.997 0.458

    F11 0.891 0.932 0.993 0.997 0.458

    F12 0.898 0.969 0.994 0.997 0.463

    F13 0.895 0.964 0.994 0.997 0.455

    Table 5: ANOVA analysis of data

    ANOVA results for dependent variablesY Mathematical

    model

    P value PLOF R Adjusted

    R2

    Predicted

    R2

    AP S.D CV

    %

    PRES

    S

    Y1 Y1= +16.01-5.75X1-2.56X2 + 4.49X12-4.01X1

    2-0.74X22

  • 8/9/2019 DOE for Extended Release

    7/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    7

    PHARMAGENE Vol: 1 Issue: 2

    Figure 4: Response surface plot (a) and Contour plot (b)

    for response Y1

    Figure 5: Response surface plot (a) and Contour plot (b)

    for response Y2

    Figure 6: Response surface plot (a) and Contour plot (b)

    for response Y4

    Figure 7: Response surface plot (a) and Contour plot (b)

    for response Y3

  • 8/9/2019 DOE for Extended Release

    8/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    8

    PHARMAGENE Vol: 1 Issue: 2

    Optimization

    After generating the model polynomial equations to relate

    the dependant and independent variables, the process wasoptimized for all four responses. The final optimal

    experimental parameters were calculated using Design-

    Expert V8 (8.071).

    The optimized batch F14 contains:

    Amount of Eudragit L 100-55 (X2) : 80 mg/tab

    Amount of NaOH : 3.25 mg/tab

    Dissolution profile of Ranexa (innovators formulation) and

    optimized formulation F14 were compared using the FDA

    recommended similarity factor (f2) (figure 8). The value of

    f2 was found to be 85.95 which was above the critical value

    (50) indicating an equivalence to the release profile of theoptimum formulation and the innovator p rofile.

    Figure 8: Dissolution profile comparison of optimized

    batch and innovators product

    Figure 9: Overlay plot of optimized batch

    Stability study

    Stability study of the optimized formulation proved the

    physical and chemical integrity of the developed ranolazine

    extended release matrix tablet with no significant change in

    the assay and dissolution profile.

    Validation of Response Surface Methodology

    Five check point batches were formulated for the validation

    of response surface methodology. Actual experimentalresponses and predicted responses were then compared to

    validate design (Table 6). For all the 5 checkpoint

    formulations, the results of the dependent variables were

    found to be within limits. For validation of RSM results,

    the experimental values of the responses were compared

    with the anticipated values and the prediction error was

    found to vary between -5.62 and +5.17. These results

    demonstrate the reliability of the optimization procedure inpredicting the effect of process variables on the dissolution

    behavior of the ranolazine extended release tablet p rofile.

    Table 6: Composition of check point batches and

    comparison of experimental and predicted values of

    response variables

    Check pointFormulations

    Response

    Variabl

    es

    Experi

    mental

    values

    Predict

    ed

    values

    %predicti

    on

    ErrorX1 X2

    67.83 2.96

    Y1 19.14 19.65 -2.70

    Y2 49.63 48.66 1.94

    Y3 76.8 77.62 -1.07

    Y4 98.48 97.09 1.40

    87.62 3.12

    Y1 16.37 15.52 5.17

    Y2 44.03 42.57 -3.29

    Y3 70.65 71.72 -1.51

    Y4 90.57 95.66 -5.62

    71.95 4.81

    Y1 14.93 15.67 -4.99

    Y2 43.61 42.31 2.96

    Y3 70.99 70.14 1.19

    Y4 95.55 93.29 2.35

    76.90 4.55

    Y1 15.88 16.10 -1.41

    Y2 42.42 42.86 -1.03

    Y3 72.35 71.04 1.79

    Y4 96.06 94.08 2.05

    83.50 2.60

    Y1 17.56 17.83 -1.53

    Y2 45.18 45.90 -1.57

    Y3 76.36 75.37 1.31

    Y4 94.30 96.80 -2.59

    CONCLUSION

    Ranolazine extended release tablets were manufactured by

    wet granulation technique. The tablets exhibited drugrelease for a period of 24 hours and followed Higuchi

    kinetics. The amount of Eudragit L100-55 and Sodium

    hydroxide was optimized by 32 full factorial design based

    on the drug release. The contour plots represented the

    influence of the amount of the independent variable on the

  • 8/9/2019 DOE for Extended Release

    9/9

    Optimization of Formulation Variables of Ranolazine Extended-Release Tablets by 32Full Factorial Design

    9

    PHARMAGENE Vol: 1 Issue: 2

    drug release. The design was also validated by the check

    point batches. The optimized formulation exhibited drug

    release similar to the innovator (f2= 85.95). The accelerated

    stability studies suggested no significant change in the drugcontent, physical properties and drug release.

    REFERENCES1. Perrie Y, Rades T. FASTtrack-Pharmaceutics-:

    Drug Delivery and Targeting: Pharmaceutical Press;

    2012.

    2. Goodman LS. Goodman and Gilman's the

    pharmacological basis of therapeutics: PergamonPress New York; 1990.

    3. Andrew A. Wolf, Sustained Release Ranolazine

    Formulation, United States Patent 6864258 B2,

    2005 M arch 8.

    4. Lewis GA, Mathieu D, Phan RTL. Pharmaceutical

    experimental design: CRC Press; 1999.

    5. Schwartz JB, O'Connor RE. Optimization

    techniques in pharmaceutical formulation andprocessing. DRUGS AND THE

    PHARMACEUTICAL SCIENCES. 1996;72:727-

    52.

    6. Gabrielsson J, Lindberg NO, Lundstedt T.

    Multivariate methods in pharmaceutical

    applications. Journal of chemometrics.

    2002;6(3):141-60.7. Lundstedt T, Seifert E, Abramo L, Thelin B,

    Nystrm , Pettersen J, et al. Experimental design

    and optimization. Chemometrics and Intelligent

    Laboratory Systems. 1998;42(1):3-40.

    8.

    Renoux R, Demazieres J, Cardot J, Aiache J.Experimentally designed optimization of direct

    compression tablets. Drug development andindustrial pharmacy. 1996;22(2):103-9.

    9. Sastry SV, Reddy IK, Khan MA. Atenolol

    gastrointestinal therapeutic system: optimization of

    formulation variables using response surface

    methodology. Journal of controlled release.1997;45(2):121-30.

    10. Takahara J, Takayama K, Nagai T. Multi-objective

    simultaneous optimization technique based on an

    artificial neural network in sustained release

    formulations. Journal of controlled release.

    1997;49(1):11-20.

    11. Geoffroy J-M, Fredrickson JK, Shelton JT. A

    mixture experiment approach for controlling thedissolution rate of a sustained-release tablet. Drug

    development and industrial pharmacy.

    1998;24(9):799-806.

    12. Hamed E, Sakr A. Application of multiple response

    optimization technique to extended release

    formulations design. Journal of controlled release.

    2001;73(2):329-38.

    13.

    Kramar A, Turk S, Vreer F. Statist ical optimisationof diclofenac sustained release pellets coated with

    polymethacrylic films. International journal of

    pharmaceutics. 2003;256(1):43-52.

    14. https://www.accessdata.fda.gov/scripts/cder/dissolut

    ion/index.cmf

    15. Costa P, Sousa Lobo JM. Modeling and comparison

    of dissolution profiles. European journal ofpharmaceutical sciences. 2001;13(2):123-33.

    16. Korsmeyer RW, Gurny R, Doelker E, Buri P,

    Peppas NA. Mechanisms of solute release from

    porous hydrophilic polymers. International Journal

    of Pharmaceutics. 1983;15(1):25-35.

    17.

    Korsmeyer R, Peppas N. Solute and penetrantdiffusion in swellable polymers. III. Drug release

    from glassy poly (HEMA-co-NVP) copolymers. JControlled Release. 1984;1(2):89-98.