SciELO - Scientific Electronic Library Online

 
vol.64 número2La eficacia de metformina y exenatida en pacientes con síndrome de ovario poliquístico (SOP)Efecto antagónico de mepiramina y tubocurarina sobre la contracción del músculo liso intestinal mediado por histamina índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Ars Pharmaceutica (Internet)

versión On-line ISSN 2340-9894

Ars Pharm vol.64 no.2 Granada abr./jun. 2023  Epub 03-Jul-2023

https://dx.doi.org/10.30827/ars.v64i2.27058 

Original Articles

Enhancing the dissolution rate of poorly soluble drug Febuxostat using spray dried amorphous solid dispersion technique

Mejora de la tasa de disolución del fármaco poco soluble Febuxostat utilizando la técnica de dispersión de sólido amorfo secado por aspersión

Vaishali Patel (orcid: 0000-0003-2812-207X)1  , Anita Patel (orcid: 0000-0001-7957-5883)2  , Ashish Shah (orcid: 0000-0003-0817-5940)3  * 

1Department of Pharmaceutics, Laxminarayandev college of Pharmacy, Bharuch, Gujarat, India

2Department of Pharmaceutics, Nootan Pharmacy College, Sankalchand Patel University, Visnagar, Gujarat, India

3Department of Pharmacy, Sumandeep Vidyapeeth, Vadodara, Gujarat, India

Abstract

Introduction:

Febuxostat belongs to Biopharmaceutical classification system (BCS) class II drugs, which have low solubility and high permeability. Amorphous solid dispersion is one of the techniques which can be useful to improve solubility and powder characteristics.

Objective:

To optimize the concentration of hydrophilic and hydrophobic polymers to improve the dissolution rate and solubility of febuxostat tablets.

Methods:

The amorphous solid dispersion of febuxostat was prepared by spray drying method using Kolliphor P237 (1:2). This amorphous solid dispersion was further used to compress the tablet. To improve solubility and dissolution rate, a full factorial design was applied to optimize the critical concentration of Kollidon SR and hydroxypropyl methyl cellulose (HPMC K4M). The prepared tablets were characterized by pre-compression and post-compression parameters.

Result:

The rate of drug release was sustained by formulating an amorphous solid dispersion technique. The optimized batch (FSRT-OB) was found to be fit for release average 93.30 % of the drug in sustain release manner up to 12hrs. The release kinetic data suggests that the drug release was controlled by combination of diffusion and chain relaxation mechanism. The optimized concentration for Kollidon SR and HPMC K4Mwas found to be 38.50 % and 7.72 % respectively.

Conclusion:

Amorphous solid dispersion technique is useful to enhance solubility, dissolution rate, and bioavailability of the Febuxostat tablet.

Keywords: Febuxostat; Amorphous Solid dispersion; Kollidon SR; HPMC K4M; Full factorial design

Resumen

Introducción:

El febuxostat pertenece a los fármacos clase II del Sistema de Clasificación Biofarmacéutica, los cuales presentan baja solubilidad y alta permeabilidad. La dispersión sólida amorfa es una de las técnicas que pueden ser útiles para mejorar la solubilidad y las características del polvo.

Objetivo:

optimizar la concentración de polímeros hidrofílicos e hidrofóbicos para mejorar la velocidad de disolución y la solubilidad de las tabletas de febuxostat.

Métodos:

La dispersión sólida amorfa de febuxostat se preparó mediante el método de secado por aspersión utilizando Kolliphor P237 (1:2). Esta dispersión sólida amorfa se utilizó además para comprimir el comprimido. Para mejorar la solubilidad y la tasa de disolución, se aplicó un diseño factorial completo para optimizar la concentración crítica de KollidonSR e hidroxi propil metil celulosa (HPMC K4M). Los comprimidos preparados se caracterizaron por parámetros de precompresión y poscompresión.

Resultados:

La velocidad de liberación del fármaco se mantuvo mediante la formulación de una técnica de dispersión sólida amorfa. Se encontró que el lote optimizado (FSRT-OB) era apto para la liberación promedio del 93,30 % del fármaco en forma de liberación sostenida hasta 12 horas. Los datos de la cinética de liberación sugieren que la liberación del fármaco estuvo controlada por una combinación de mecanismo de relajación de cadena y difusión. Se encontró que la concentración optimizada para Kollidon SR y HPMC K4M era 38,50 % y 7,72 % respectivamente.

Conclusión:

La técnica de dispersión sólida amorfa es útil para mejorar la solubilidad, la velocidad de disolución y la biodisponibilidad de la tableta de Febuxostat.

Palabras clave: Febuxostat; dispersión sólida amorfa; Kollidon SR; HPMC K4M; diseño factorial completo

Highlights

The bioavailability of febuxostat is approx. 49 % which is determined by the low dissolution rate of the drug. There is a need to explore another method that can produce amorphous solid dispersion of Febuxostat with minimum residual solvent. There are various techniques used to increase the dissolution rate of the drugs out of that amorphous solid dispersion is one of the powerful techniques to enhance the solubility of the drugs. The successful improvement in the rate of dissolution of febuxostat was obtained by preparing amorphous solid dispersion with Kollidon SR (38.50 % w/w) and HPMC K4M (7.72 %) using the spray drying technique.

Introduction

Febuxostat is used in the treatment of arthritis which targets the xanthine oxidase enzyme. The recommended dose of febuxostat is 40-80 mg per day, administered with or without food. It is mainly used in the treatment of hyperuricemia which is characterized by an increased amount of uric acid due to the breakdown of certain chemical products (purines) in the body1.These uric acid crystals are accumulated in joints and tissues which can cause gouty arthritis. Febuxostat inhibits the enzyme xanthine oxidase which is responsible for the formation of uric acid crystals from purine-like compounds in the human body. According to the Biopharmaceutical classification system (BCS), febuxostat is classified under class II drugs that possess high intestinal permeability and low aqueous solubility. The bioavailability of febuxostat is approx. 49 % which is determined by the low dissolution rate of the drug.

Dissolution rate can be considered as one of the important parameters, which determine the bioavailability of the drugs across the biological membrane. Approximately, 30-40 % of new chemical entities coming into the market have the problem of poor aqueous solubility. Therefore, improving the solubility and dissolution rate of the poorly soluble drug is one of the most challenging aspects of modern pharmaceutics. There are various techniques used to increase the dissolution rate of the drugs out of that amorphous solid dispersion is one of the powerful techniques to enhance the solubility of the drugs. In this technique, the drug is dispersed throughout the matrix to change drug particles from crystalline to amorphous form with an advantage of particle size reduction2.

According to the literature review, solvent evaporation and hot melt technique are useful to enhance the solubility and dissolution rate of Febuxostat but these techniques require the use of organic solvents. There is a need to explore another method by which we can produce amorphous solid dispersion of Febuxostat with minimum residual solvent3. Spray drying is also an efficient method to obtain the amorphous particles of the drug molecules. Spray drying is the transformation of liquid feed material into solid particulate form by atomization through an atomizer into a hot drying gas medium. The solid particles obtained from the spray drying method show narrow particle size distribution4.Tablets can be prepared by three techniques; wet granulation, dry granulation, and direct compression5. The solid dispersion of febuxostat obtained from the spray drying method can be used to produce a controlled-release tablet of febuxostat6. Febuxostat can be mixed with directly compressible excipients to form sustained release dosage form7.

Full factorial design is response surface design which provides individual effect, combined or interaction effect, and also curvilinear effect. It is an efficient, fast, and convenient method used for optimization study in the pharmaceutical research field8. This design is ideal because, in the smaller number of a process run, it can give accurate results. It provides information on experimental variable effects, overall experimental error, and the minimum number of runs required for the optimization study9. Therefore, the full factorial design is suitable for formulation optimization of febuxostat tablets prepared using spray-dried amorphous solid dispersion10. In this research, we intended to modulate the release kinetics by a combined mechanism: use of the porous structure generated with the insoluble polymer (Kollidon SR) and the other from the hydrophilic gelling polymer (hydroxypropyl methyl cellulose, HPMC grade K4M or hydroxypropyl cellulose, HPC) which imparts slow drug diffusion11.

Materials and methods

Febuxostat was obtained from Balaji Drugs, Surat, India. The polymers Kolliphor P 237 and Eudragit RLPO were procured from BASF/ Sigma-Aldrich, Bangalore. Silicon dioxide and magnesium stearate were obtained from Loba chemicals/ Durga Scientific, Vadodara. Avicel PH 102 was procured from Astron Chemicals, Ahmedabad. All other ingredients used were of analytical grade12.

Preparation of amorphous solid dispersion of febuxostat by spray drying method

Solid dispersion was prepared using drug (febuxostat) polymer (Kolliphor P 237) ratio 1:2. The required amount of polymer was weighed and mixed with enough acetone (200 ml) to make a clear solution. Solidification of solid dispersion was done using a spray dryer (Model: LU222 Advance, make: Labultima), equipped with a high-performance cyclone. The liquid was then atomized in a spray tower using a hollow cone pressure nozzle (bore diameter 0.6 mm) at a pressure of 100 bar. The spray tower was operated with nitrogen at an inlet temperature of 140°C and an outlet temperature of approx. 100 °C. The spray-dried powder was subsequently filtered using a tube filter. Solid dispersion was collected and stored in desiccators. This solid dispersion was further used to produce sustained release tablets of febuxostat13.

Full factorial design

To optimize the formulation in the minimum number of trials, the screening was done by applying a full factorial design. The full factorial design comprised of; two quantitative factors (X1, the concentration of Kollidon SR; and X3, the concentration of hydrophilic polymer) and one qualitative factor (X2, the type of hydrophilic polymer: HPC or HPMC). The surfaces would be graphical representations of the response as a function of X1 and X3, whereas X2 would be consecutively HPC or HPMC14. The Quadratic equations were generated for the measured responses as a function of dependent and independent variables. From the full factorial design run, response surface graphs were generated. For the optimization, 20 batches were prepared by varying the factor levels as shown in Table 1.15.

Table 1. Full factorial Design set-up for optimization. 

Sr. No. Name of Factor Unit Level Remark
Low High
Independent Factors
1 Con. of Kollidon SR (X1) % 25 40 Insoluble polymer SR polymer
2 Con. of Hydrophilic polymer (X2) % 0 10 Hydrophilic, gelling SR polymer
3 Type of Hydrophilic polymer (X3) - HPC HPMC-K4M Hydrophilic, gelling SR polymer
4 Drug solid dispersion mg Equivalent to 40 mg of Febuxostat Drug
5 Silicon dioxide % 0.5 Flow enhancer
6 Magnesiumstearate % 0.25 Antiadharent
7 Avicel PH 102 mg QS (up to 250 mg) Directly compressible Diluent
Dependent Factor
1 CDR 2hr (Y1) % - 10-20 % (15%)
2 CDR 4hr (Y2) % - 21-30 % (25%)
3 CDR 6hr (Y3) % - 31-50 % (40%)
4 CDR 8hr (Y4) % - 51-75 % (62.5%)
5 CDR 12hr (Y5) % - NLT 85 % (>85 %)

CDR: Controlled Drug Release; HPC: hydroxy propyl cellulose; HPMC K4M: hydroxy propyl methyl cellulose, grade K4M; NLT: Not Less Than; QS: Quantity Sufficient.

Total weight of each Tablet is 250 mg. Tablets were compressed using direct compression method, 9 mm standard concave round shaped punch.

Manufacturing of the sustained release solid dispersion tablets

The 120 mg of solid dispersions were mixed thoroughly with the required quantity of Avicel PH 102 using polybag for 10 min. Then 0.5 % of each Aerosil and Magnesium stearate was mixed with the previous blend using polybag for 10 min. The tablets of desired weight (250 mg) were compressed on rotary tablet press by direct compression method using 9 mm standard concave round-shaped punch. Twenty different batches (FSRT1-FSRT20), having a different concentration of sustained-release polymer (Kollidon SR) and different hydrophilic polymer (HPC or HPMC-K4M) were prepared to evaluate the effect of polymer on drug release2. The actual composition of all optimization batches is shown in table 2.

Table 2. Actual composition of optimization batches. 

Batch Code IndependentFactors ConstantFactors Total weight per Tablet (mg)
Con. of Kollidon SR (mg) (X1) Con. of Hydrophilic polymer (mg) (X2) Type of hydrophilic polymer (X3) SD Equivalent to 40 mg of Febuxostat (mg) Silicon dioxide (mg) Magnesiumstearate (mg) Avicel PH 102 (mg)
FSRT1 62.5 25 HPMC-K4M 120 1.25 1.25 40 250
FSRT2 62.5 25 HPC 120 1.25 1.25 40 250
FSRT3 100 0 HPC 120 1.25 1.25 27.5 250
FSRT4 81.25 25 HPMC-K4M 120 1.25 1.25 21.25 250
FSRT5 100 12.5 HPMC-K4M 120 1.25 1.25 15 250
FSRT6 81.25 25 HPC 120 1.25 1.25 21.25 250
FSRT7 81.25 0 HPC 120 1.25 1.25 46.25 250
FSRT8 100 25 HPMC-K4M 120 1.25 1.25 2.5 250
FSRT9 81.25 12.5 HPC 120 1.25 1.25 33.75 250
FSRT10 62.5 12.5 HPC 120 1.25 1.25 52.5 250
FSRT11 100 12.5 HPC 120 1.25 1.25 15 250
FSRT12 100 25 HPC 120 1.25 1.25 2.5 250
FSRT13 81.25 12.5 HPC 120 1.25 1.25 33.75 250
FSRT14 81.25 0 HPMC-K4M 120 1.25 1.25 46.25 250
FSRT15 62.5 0 HPMC-K4M 120 1.25 1.25 65 250
FSRT16 81.25 12.5 HPMC-K4M 120 1.25 1.25 33.75 250
FSRT17 100 0 HPMC-K4M 120 1.25 1.25 27.5 250
FSRT18 62.5 12.5 HPMC-K4M 120 1.25 1.25 52.5 250
FSRT19 62.5 0 HPC 120 1.25 1.25 65 250
FSRT20 81.25 12.5 HPMC-K4M 120 1.25 1.25 33.75 250

FSD24: Febuxostat Solid Dispersion containing batch no.24; HPC: Hydroxypropylcellulose; HPMCK4M; Hydroxypropyl methylcellulose K4M.

Total weight of each Tablet is 250 mg; 120 mg of Solid dispersion (FSD24, Drug to polymer ratio 1:2) equivalent to 40 mg of Febuxostat added in each batch. Tablets were compressed using direct compression method, 9 mm standard concave round shaped punch.

Characterization of Tablet containing Febuxostat Solid dispersion

Pre compression tests16:

  1. Bulk density: The bulk density of a powder mixture is determined by measuring the volume of a known mass of powder sample (Quantity = 20 gm).

  2. Tapped density: The Tapped density of a powder mixture is determined by measuring the volume of a known mass of powder sample (20gm) after 100 tapings.

  3. Hausner's ratio: It is the ratio of the ease with which powder can flow. Hausner's ratio less than 1.25 indicates good flow property and greater than 1.5 indicates poor flow. To improve the flow property of powder, glidants can be added.

  4. Car's index: It is also known as the Compressibility index. By comparing the tapped density and bulk density of the powder, the compressibility of the powder can be determined.

  5. Angle of Repose: Angle of repose is defined as the maximum angle viable between the surface of a pile of the powder and the horizontal base.

Post compression tests16:

  1. Hardness testing

    The hardness of tablets was determined using a Pfizer hardness tester.

  2. Weight variation test

    Twenty tablets were selected at random, weighed, and the average weight was calculated. Not more than two of the individual weights should deviate from the average weight by more than 7.5%.

  3. Friability test

    For each formulation, a pre-weighed tablet sample (10 tablets) was placed in a friability (Electrolab, Mumbai, India), which is then operated for 100 revolutions. The tablets were de-dusted and reweighed. Compressed tablets that lose not more than 1% of their weight are considered acceptable.

  4. Assay

    Twenty tablets were weighed and powdered using glass mortar pestle. The quantity of powder equivalent to 40 mg of febuxostat was accurately weighed and transferred into a 100 ml volumetric flask. Methanol was added up to 100 ml and shaken well. The solution was filtered through a 0.45μ membrane filter. 1 ml of the above solution was transferred into a 100 ml volumetric flask to make up the final volume up to 100 ml using methanol. The absorbance of the resulting solution was measured at a λmax of 315 nm using a UV-Visible spectrophotometer (Shimadzu 1800, Kyoto, Japan). The amount of the febuxostat was calculated by using the equation obtained from the calibration curve.

  5. In-Vitro Dissolution study

    In vitro drug release study of prepared batches (n=3) was performed using USP (United States Pharmacopoeia) apparatus II (TDT-08T; Electrolab, India) fitted with a paddle (75 rpm) at 37 ± 0.5°C. In acid stage 750 ml of 0.1M HCL was used as dissolution media. The percentage drug release was calculated up to 2 hrs (Sampling time 0 hr, 1 hr, and 2 hr).

In Buffer stage 0.2 M solution of trisodium phosphate dodecahydrate (Previously warmed up to 36.5ºC to 37.5ºC, 250 ml) was added to the dissolution basket. To adjust the pH of 6.8 ± 0.05, add 2M hydrochloric acid or 2M sodium hydroxide (if necessary). The sampling was done at the time interval of 4 hr, 6 hr, 8 hr, and 12 hr. At predetermined time intervals, 5 ml samples were withdrawn, filtered through a 0.45μ membrane filter, and analyzed at the respective wavelength (0.1M HCL= 284 nm and buffer stage = 315 nm) using a UV-Visible double beam spectrophotometer (Shimadzu 1800, Kyoto, Japan). Cumulative percentage drug release was calculated using an equation obtained from a calibration curve.

Results and discussion

The powder blends were prepared by mixing all ingredients in a polythene bag. The prepared powder blend of different batches was evaluated for their angle of repose, bulk density, tapped density, compressibility index, and Hausner's ratio17.All results were within acceptance criteria. The post-compression tests were Hardness, friability, and weight variation,they were also found to be satisfactory. The in vitro drug release data for all batches were calculated as shown in table 3.

Table 3. Results of optimization batches (FSRT 1- FSRT 20). 

Batch-Code Independent Factors Dependent Factors
Con. of Kollidon SR (%) Con. of Hydrophilicpolymer (%) Type of hydrophilic polymer % CDR
2 hr 4 hr 6 hr 8 hr 12 hr
X1 X2 X3 Y1 Y2 Y3 Y4 Y5
FSRT1 25 10 HPMC-K4M 36.08 57.30 89.13 99.49 99.69
FSRT2 25 10 HPC 21.90 40.16 67.54 99.98 100.21
FSRT3 40 0 HPC 2.98 8.37 18.22 27.41 37.91
FSRT4 32.5 10 HPMC-K4M 26.72 42.44 66.02 98.81 101.76
FSRT5 40 5 HPMC-K4M 9.51 17.21 29.72 47.81 65.2
FSRT6 32.5 10 HPC 17.11 31.37 52.76 84.85 99.23
FSRT7 32.5 0 HPC 2.2 8.8 19.6 29.7 41.91
FSRT8 40 10 HPMC-K4M 20.39 32.4 50.51 77.4 101.92
FSRT9 32.5 5 HPC 7.13 17.32 32.65 55.59 78.54
FSRT10 25 5 HPC 8.57 20.81 39.17 66.71 94.25
FSRT11 40 5 HPC 5.96 14.42 27.2 46.33 65.46
FSRT12 40 10 HPC 13.81 25.30 42.55 68.43 94.30
FSRT13 32.5 5 HPC 6.71 16.29 30.68 52.25 73.83
FSRT14 32.5 0 HPMC-K4M 6.11 13.25 26.46 46.8 54.02
FSRT15 25 0 HPMC-K4M 7.5 16.54 33.02 58.6 67.5
FSRT16 32.5 5 HPMC-K4M 12.29 22.53 37.90 60.95 83.99
FSRT17 40 0 HPMC-K4M 5.18 11.32 22.03 38.81 44.87
FSRT18 25 5 HPMC-K4M 16.47 30.20 50.79 81.68 100.02
FSRT19 25 0 HPC 2.60 10.38 23.36 35.05 49.32
FSRT20 32.5 5 HPMC-K4M 12.67 23.23 39.07 62.83 86.59

FSD24: Febuxostat Solid Dispersion containing batch no.24; HPC: Hydroxypropylcellulose; HPMCK4M; Hydroxypropyl methylcellulose K4M

Total weight of each Tablet is 250 mg.120 mg of Solid dispersion (FSD24, Drug to polymer ratio 1:2) equivalent to 40 mg of Febuxostat added in each batch.

The graphical representation of comparative % drug release of all batches is shown in figure 1.

Figure 1. Comparative % Controlled drug release at different time Interval of all 20 batches (CDR: Controlled Drug Release). 

The 32 full factorial design (X1: concentration of sustained-release polymer; Kollidon SR, X2: Concentration of hydrophilic polymer) with additional one categorical factor (X3: Type of hydrophilic polymer; HPMC K4M or HPC) was constructed using Design expert® demo version 11 software (Stat-ease, MN, US). The 20 batches (18 design point batches with additional 2 replication of centre point for lack of fit test) (FSRT 1- FSRT 20) containing different compositions was suggested by the software. The design output with the level of actual factors and results for all responses are shown in table 3. Additionally, some measures of the influence on the response of single/individual components and in combination with other components were measured. The standard form of the quadratic equation was:

Quadratic:y=i=1qbixi+i<jq1jqbijxixj (1)

Where Y represents the response variable of the process. Βirepresents the coefficients of the factor's response to the pure blend Xi=1 and Xj=0 whenji. The portion ∑i=1βiXiis called a linear blending portion. When there is curvature arising from non linear blending between component pairs, the parameters βij represent either synergistic or antagonistic blending. Therapeutic values of the regression coefficients were determined to evaluate the significance of the factors on the responses. ANOVA was also applied to determine the significance of the model.

Model fitting and regression analysis

The experiments were performed in random order and it was observed that in all cases there exists a reasonable impact of independent variables. The results were fitted to different models and the residual errors were estimated to examine the goodness of fit for each model. The software suggests that the best-fitted model was quadratic for Y1 to Y5. The model summary statistics are given in table 4. The regression coefficients for each of the responses were shown in table 5.

Table 4. Model summary statistics and Model selection. 

Responses Models SD R2 Adjusted R2 Predicted R2 PRESS Observation
Y1: % CDR 2hr Linear 3.37 0.88 0.855 0.791 310.72
2FI 2.37 0.95 0.928 0.858 211.80
Quadratic 0.84 0.99 0.991 0.974 38.20 Suggested
Cubic 0.32 1.00 0.999 0.992 12.38 Aliased
Y2: % CDR 4hr Linear 4.510 0.895 0.875 0.819 560.54
2FI 3.050 0.961 0.943 0.884 360.02
Quadratic 1.213 0.995 0.991 0.975 79.09 Suggested
Cubic 0.454 1.000 0.999 0.996 11.61 Aliased
Y3: % CDR 6hr Linear 6.429 0.896 0.877 0.822 1133.54
2FI 4.551 0.958 0.938 0.876 789.56
Quadratic 1.831 0.994 0.990 0.975 159.84 Suggested
Cubic 0.755 0.999 0.998 0.996 26.74 Aliased
Y4: % CDR 8hr Linear 5.663 0.947 0.937 0.913 843.68
2FI 5.057 0.966 0.950 0.906 911.56
Quadratic 4.620 0.976 0.958 0.896 1013.16 Suggested
Cubic 3.657 0.992 0.974 0.814 1809.29 Aliased
Y5: % CDR 12hr Linear 7.186 0.913 0.897 0.858 1353.47
2FI 7.020 0.933 0.902 0.805 1851.68
Quadratic 6.260 0.955 0.922 0.809 1818.97 Suggested
Cubic 2.321 0.997 0.989 0.960 380.68 Aliased

Model was selected based on Low PRESS value, Low SD, Highest R2. Software have also suggested Aliased models too, which was omitted from selection criteria. CDR: Controlled Drug Release

Table 5. Regression analysis for Factorial design. 

Modelterm ▶ Model Intercept Main effect terms Twofactors Interactionterms Square effect Terms
Response ▼ X1 X2 X3 X1X2 X1X3 X2X3 X12 X22
Y1: % CDR 2hr Quadratic FM 9.60 -2.94 9.12 3.20 -2.73 -1.22 1.61 0.62 3.53
p-value < 0.0001 < 0.0001 < 0.0001 < 0.0001 < 0.0001 0.0004 < 0.0001 0.1357 < 0.0001
RM 9.91 -2.94 9.12 3.20 -2.73 -1.22 1.61 - 3.63
Y2: % CDR 4hr FM 19.72 -5.53 13.36 3.66 -4.07 -1.65 1.81 1.07 4.37
p-value < 0.0001 < 0.0001 < 0.0001 < 0.0001 < 0.0001 0.0006 0.0003 0.0435 < 0.0001
Y3: % CDR 6hr FM 34.95 -9.40 18.82 4.55 -5.94 -2.38 1.87 1.90 6.39
p-value < 0.0001 < 0.0001 < 0.0001 < 0.0001 < 0.0001 0.0009 0.0046 0.0469 < 0.0001
Y4: % CDR 8hr FM 59.23 -11.28 24.38 5.34 -3.28 -1.35 -2.47 0.09 4.49
p-value < 0.0001 < 0.0001 < 0.0001 < 0.0001 0.070 0.334 0.091 0.968 0.059
RM 59.27 -11.28 24.38 5.34 -3.28 - -2.47 - 4.51
Y5: % CDR 12hr FM 80.81 -8.44 25.13 3.53 3.80 -0.76 -2.30 0.35 -6.66
p-value < 0.0001 0.001 < 0.0001 0.028 0.114 0.683 0.229 0.907 0.042
RM 80.99 -8.44 25.13 3.53 3.795 - - - -6.598

FM; Full model; RM: Reduced model

Term with p-value greater than 0.05 was omitted from full model

As per software suggestion, some insignificant terms were kept as it is required to maintain the hierarchy of model.

A positive value denotes an effect that favours the optimization, while a negative value indicates an inverse relationship between the factor and the response. The polynomial equation of the full model generated for each response is given below.

Y1=9.60C-2.94X1+9.12X2+3.20X3-2.73X1X2-1.22X1X3+1.61X2X3+0.62X12+3.53X22 (2)

Y2=19.72C-5.53X1+13.36X2+3.66X3-4.07X1X2-1.65X1X3+1.81X2X3+1.07X12+4.37X22 (3)

Y3=34.95C-9.40X1+18.82X2+4.55X3-5.94X1X2-2.38X1X3+1.87X2X3+1.90X12+6.39X22 (4)

Y4=59.23C-11.28X1+24.38X2+5.34X3-3.28X1X2-1.35X1X3+2.47X2X3+0.09X12+4.49X22 (5)

Y5=80.81C-8.44X1+25.13X2+3.53X3+3.80X1X2-0.76X1X3-2.30X2X3+0.35X12-6.66X22 (6)

The polynomial equation generated from the experimental design was validated by ANOVA and F statistics. ANOVA result and lack of fit tests of the models for all responses are shown in table 6.

Table 6. Model Testing Summary. 

Source SS df MS F Value p-value R2 Adj R2 Pred R2 F- Statistics
Y1:% CDR 2hr
Regression FM 1480.32 8.00 185.04 264.55 < 0.0001 0.995 0.991 0.974 FCal= 2.303 FTab= 4.844 DF = (1, 11) α =0.05
RM 1478.51 7.00 211.22 266.62 < 0.0001 0.994 0.990 0.974
Residual FM 7.69 11 0.70
RM 9.51 12 0.79
LackofFit 9.35 10 0.93 11.65 0.082
Pure Error 0.16 2 0.08
Y2: % CDR 4hr
Regression FM 3085.885 8 385.74 262.26 < 0.0001 0.995 0.991 0.975 No need modelreduction
Residual FM 16.179 11 1.471
LackofFit 15.404 9 1.712 4.4142 0.19830
Pure Error 0.775 2 0.388
Y3: % CDR 6hr
Regression FM 6347.21 8 793.40 236.73 < 0.0001 0.994 0.990 0.975 No need model reduction
Residual FM 36.87 11 3.352
LackofFit 34.242 9 3.80 2.90 0.283
Pure Error 2.625 2 1.31
Y4: % CDR 8hr
Regression FM 9509.56 8 1188.70 55.69 < 0.0001 0.976 0.958 0.896 FCal= 0.512 FTab= 3.982 DF = (2,11) α =0.05
RM 9487.71 6 1581.29 80.10 < 0.0001 0.974 0.962 0.930
Residual FM 234.777 11 21.343
RM 256.628 13 19.741
LackofFit 249.283 11 22.662 6.17 0.148
Pure Error 7.345 2 3.673
Y5: % CDR 12hr
Regression FM 9079.32 8 1134.92 28.96 < 0.0001 0.955 0.922 0.809 FCal= 0.604 FTab= 3.587 DF = (3,11) α =0.05
RM 9008.27 5 1801.65 50.23 < 0.0001 0.947 0.928 0.879
Residual FM 431.06 11 39.19
RM 502.11 14 35.865
LackofFit 487.639 12 40.64 5.62 0.161
Pure Error 14.472 2 7.24

SS: sum of squares; df: Degree soffreedom; MS: mean of squares; F: Fischer's ratio; R2: Regression coefficient; FM: Full model; RM: Reduced model; FTab: Table value of F; FCal: calculated value of F. Details of calculations are shown by MendenhallWandSincich. If FTab is greater than the FCal that indicating the reduced term does not contribute significantly to the prediction of responses and therefore can be omitted from the full model and reduced model can be used for optimization prediction.

It has indicated significant effects of the independent factors (P > F) on response Y1 to Y5. The larger F-value recommends that the data fit to the model were significant and leads to a good correlation with a high R2 value. For all responses, adjusted and predicted R2 values were in reasonable agreement, demonstrating the mathematical model describes the data adequately. However, certain model terms for Y1, Y4, and Y5 having P>0.05 require a model reduction to improve the model. Removal of this insignificant term improved the model for Y1, Y4, and Y5 responses. The polynomial equation of the reduced model was generated for each response as given below.

Y1=9.91C-2.94X1+9.12X2+3.20X3-2.73X1X2-1.22X1X3+1.61X2X3+3.53X22 (7)

Y2=19.72C-5.53X1+13.36X2+3.66X3-4.07X1X2-1.65X1X3+1.81X2X3+1.07X12+4.37X22 (8)

Y3=34.95C-9.40X1+18.82X2+4.55X3-5.94X1X2-2.38X1X3+1.87X2X3+1.90X12+6.39X22 (9)

Y4=59.27C-11.28X1+24.38X2+5.34X3-3.28X1X2-2.47X2X3+4.51X22 (10)

Y5=80.99C-8.44X1+25.13X2+3.53X3+3.80X1X2-6.66X22 (11)

The F statistics was used to test the generated reduced model, shows that the FTab was greater than the FCal for all the responses indicating for the reduced term which does not contribute significantly to the prediction of responses and therefore can be omitted from the full model (Table 6). An insignificant lack of fit for all responses also implies that the models were adequate for the prediction with the range of experimental variables.

Direct interpretation of reduced polynomial equations may lead to errors since interaction and polynomial terms are also significant. Therefore, contour and response surface plots were drawn. Nonlinear relationship is visible in all contour and 3D surface plots (Figure 2).

Figure 2. Contour Plots and 3D surface plots for Responses [For Y1 (A, B), Y2 (C, D), Y3 (E, F), Y4 (G, H) and Y5 (I,J)]. 

Design space can be identified based on the highest and the lowest range of variables set by the user. These plots help to constitute desired responses and formulation compositions. In the two-dimensional view of the contour plots, constant responses are connected to construct the contour line. On the other hand, a 3D view of the surface plot may serve a clearer picture of the response surface.After generating the reduced model polynomial equations to relate the dependent and independent variables, the formulation was optimized using all five responses. The formulation was optimized based on the constraints set on the independent variable as shown in table 7.

Table 7. Target responses for selection of optimum formulation. 

Response Variables Range Target TI Low TI High
Y1: % CDR 2hr 10-20 15 13.24 17.79
Y2: % CDR 4hr 21-30 25 22.95 29.25
Y3: % CDR 6hr 31-50 40 37.11 46.61
Y4: % CDR 8hr 51-75 62.5 63.54 71.34
Y5: % CDR 12hr NLT 85 >85 86.18 96.1

CDR: Controlled Drug Release ;TI: Tolerance interval

The optimized composition of febuxostat sustained-release tablet [FSRT-OB; Concentration of Kollidon SR (X1) = 38.50 %w/w, Concentration of hydrophilic polymer (X2) = 7.72 %w/w, Type of hydrophilic polymer (X3) = HPMC K4M] was used for formulation development and it was evaluated for physical and chemical characteristics18.The composition of optimized batch (FSRT-OB) is shown in table 8. Checkpoint validation results suggest that there was reasonable agreement between predicted and experimental (percentage bias < 10%) in all responses. So, the model can be said to be valid for the given factorial design19.The optimized powder blend was prepared by mixing all ingredients in a polythene bag. The prepared powder blend of optimized batch (FSRT-OB) was compressed using direct compression method and evaluated for physical and chemical characteristics (Angle of repose, Compressibility index, Hausner's ratio, Hardness Friability, Weight variation, and Assay)20.All results were within acceptance criteria.

Table 8. Composition for optimized batch (FSRT-OB). 

Ingredients Qty Qty/Tab
Febuxostat solid dispersion (FSD24) equivalent to 40mg of febuxostat 120 mg 120
Avicel PH 102 QS 11.95
Kollidon SR (X1) 38.50 %w/w 96.25
HPMC K4M (X2, X3) 7.72%w/w 19.3
Silicon dioxide 0.5 %w/w 1.25
Magnesium stearate 0.5%w/w 1.25
Total Weight each Tablet 250 mg
Desirability 1

HPMC K4M; Hydroxypropyl methylcellulose K4M

Conclusion

The successful improvement in the rate of dissolution of febuxostat was obtained by preparing amorphous solid dispersion with Kollidon SR (38.50%w/w) and HPMC K4M (7.72%) using the spray drying technique. In all the prepared batches, it was clear that the solubility of the drug was sustained in the case of sustained-release tablets prepared with HPMC K4M as compared to HPC. Product properties including solubility, dissolution rate, and amorphous characteristics were improved by applying full factorial design and results showed good agreement with the prediction of the models. From the evaluation parameters of all batches, it can be concluded that the Kollidon SR provided a sustained release to the tablets. It can be concluded that the optimization provides help in selecting the appropriate number of dependent variables to achieve the required goal.

Acknowledgement

We would like to thank Laxminarayandev College of Pharmacy for providing necessary facilities

References

1. Bisht M, Bist SS. Febuxostat: a novel agent for management of hyperuricemia in gout. Indian JPharm Sci. 2011;73(6):597-600. doi: 10.4103/0250-474X.100231. [ Links ]

2. Paradkar A, Ambike AA, Jadhav BK, Mahadik KR. Characterization of curcumin-PVP solid dispersion obtained by spray drying. Int J Pharm. 2004;271(1-2):281-6. doi: 10.1016/j.ijpharm.2003.11.014. [ Links ]

3. Chiou WL, Riegelman S. Pharmaceutical applications of solid dispersion systems. J Pharm Sci. 1971;60(9):1281-302. doi: 10.1002/jps.2600600902. [ Links ]

4. Shah JC, Chen JR, Chow D. Preformulation study of etoposide: II. Increased solubility and dissolution rate by solid-solid dispersions. Int J Pharm. 1995;113(1):103-11. [ Links ]

5. Sastry SV, Nyshadham JR, Fix JA. Recent technological advances in oral drug delivery - a review. Pharm Sci Technol Today. 2000;3(4):138-45. doi: 10.1016/s1461-5347(00)00247-9. [ Links ]

6. Habib W, Khankari R, Hontz J. Fast-dissolve drug delivery systems. Crit Rev Ther Drug Carrier Syst. 2000;17(1):61-72. doi: 10.1615/CritRevTherDrugCarrierSyst.v17.i1.20. [ Links ]

7. Dass R, Jaiswal S, Gupta G. Formulation and evaluation of febuxostat fast disintegrating tablet. Indo american journal of pharmaceutical research. 2014;4:2928-36. [ Links ]

8. Eriksson L. Design of experiments: principles and applications. MKS Umetrics AB. 2008. [ Links ]

9. Ahuja BK, Jena SK, Paidi SK, Bagri S, Suresh S. Formulation, optimization and in vitro-in vivo evaluation of febuxostat nanosuspension. Int J Pharm. 2015;478(2):540-52. doi: 10.1016/j.ijpharm.2014.12.003. [ Links ]

10. Saag KG, Becker MA, Whelton A, Hunt B, Castillo M, Kisfalvi K, et al. Efficacy and Safety of Febuxostat Extended and Immediate Release in Patients With Gout and Renal Impairment: A Phase III Placebo Controlled Study. Arthritis Rheum. 2018;71(1):143-53. doi: 10.1002/art.40685. [ Links ]

11. Song SH, Chae BR, Sohn SI, Yeom DW, Son HY, Kim JH, et al. Formulation of controlled-release pelubiprofen tablet using Kollidon((R)) SR. Int J Pharm. 2016;511(2):864-75. doi: 10.1016/j.ijpharm.2016.07.074. [ Links ]

12. Jung JY, Yoo SD, Lee SH, Kim KH, Yoon DS, Lee KH. Enhanced solubility and dissolution rate of itraconazole by a solid dispersion technique. Int J Pharm. 1999;187(2):209-18. doi: 10.1016/s0378-5173(99)00191-x. [ Links ]

13. Sahoo J, Murthy PN, Biswal S, et al. Formulation of sustained-release dosage form of verapamil hydrochloride by solid dispersion technique using Eudragit RLPO or Kollidon SR. AAPS PharmSciTech. 2009;10(1):27-33. doi: 10.1208/s12249-008-9175-0. [ Links ]

14. Shamma RN, Basha M. Soluplus®: a novel polymeric solubilizer for optimization of carvedilol solid dispersions: formulation design and effect of method of preparation. Powder Tech. 2013;237:406-14. [ Links ]

15. Siepmann J, Peppas NA. Modeling of drug release from delivery systems based on hydroxypropyl methylcellulose (HPMC). Adv DrugDeliv Rev. 2001;48(2-3):139-57. doi: 10.1016/s0169-409x(01)00112-0. [ Links ]

16. Sahoo J, Murthy PN, Biswal S, Sahoo SK, Mahapatra AK. Comparative study of propranolol hydrochloride release from matrix tablets with KollidonSR or hydroxy propyl methyl cellulose. AAPS PharmSciTech. 2008;9(2):577-82. doi: 10.1208/s12249-008-9092-2. [ Links ]

17. Shah N, Sandhu H, Phuapradit W, Pinal R, Iyer R, Albano A, et al. Development of novel microprecipitated bulk powder (MBP) technology for manufacturing stable amorphous formulations of poorly soluble drugs. Int J Pharm. 2012;438(1-2):53-60. doi: 10.1016/j.ijpharm.2012.08.031. [ Links ]

18. Song Y, Wang L, Yang P, Wenslow RM, Tan B, Zhang H, et al. Physicochemical characterization of felodipine-kollidon VA64 amorphous solid dispersions prepared by hot-melt extrusion. J Pharm Sci. 2013;102(6):1915-23. doi: 10.1002/jps.23538. [ Links ]

19. Six K, Verreck G, Peeters J, Brewster M, Van Den Mooter G. Increased physical stability and improved dissolution properties of itraconazole, a class II drug, by solid dispersions that combine fast- and slow-dissolving polymers. J Pharm Sci. 2004;93(1):124-31. doi: 10.1002/jps.10522. [ Links ]

20. Patel J, Dhingani A, Garala K, Raval M, Sheth N. Design and development of solid nanoparticulate dosage forms of telmisartan for bioavailability enhancement by integration of experimental design and principal component analysis. Powder Technol. 2014;258:331-43. doi: 10.1016/j.powtec.2014.03.001. [ Links ]

FundingNot applicable

Received: January 07, 2023; Accepted: March 02, 2023; pub: March 20, 2023

Corresponding Ashish Shah ashishshah.dop@sumandeepvidyapeethdu.edu.in

Conflict of interest

The authors declare no conflict of interest.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License