Next Article in Journal
Another Move towards Bicalutamide Dissolution and Permeability Improvement with Acetylated β-Cyclodextrin Solid Dispersion
Next Article in Special Issue
Quality by Design-Driven Zeta Potential Optimisation Study of Liposomes with Charge Imparting Membrane Additives
Previous Article in Journal
Extracellular Alterations in pH and K+ Modify the Murine Brain Endothelial Cell Total and Phospho-Proteome
Previous Article in Special Issue
Raman Mapping-Based Reverse Engineering Facilitates Development of Sustained-Release Nifedipine Tablet
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design Space Approach for the Optimization of Green Fluidized Bed Granulation Process in the Granulation of a Poorly Water-Soluble Fenofibrate Using Design of Experiment

1
Department of Pharmaceutics, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt
2
Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
3
Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(7), 1471; https://doi.org/10.3390/pharmaceutics14071471
Submission received: 1 June 2022 / Revised: 7 July 2022 / Accepted: 13 July 2022 / Published: 15 July 2022
(This article belongs to the Special Issue Aspects and Implementation of Pharmaceutical Quality by Design)

Abstract

:
In the pharmaceutical industry, the systematic optimization of process variables using a quality-by-design (QbD) approach is highly precise, economic and ensures product quality. The current research presents the implementation of a design-of-experiment (DoE) driven QbD approach for the optimization of key process variables of the green fluidized bed granulation (GFBG) process. A 32 full-factorial design was performed to explore the effect of water amount (X1; 1–6% w/w) and spray rate (X2; 2–8 g/min) as key process variables on critical quality attributes (CQAs) of granules and tablets. Regression analysis have demonstrated that changing the levels of X1 and X2 significantly affect (p ≤ 0.05) the CQAs of granules and tablets. Particularly, X1 was found to have the pronounced effect on the CQAs. The GFBG process was optimized, and a design space (DS) was built using numerical optimization. It was found that X1 and X2 at high (5.69% w/w) and low (2 g/min) levels, respectively, demonstrated the optimum operating conditions. By optimizing X1 and X2, GFBG could enhance the disintegration and dissolution of tablets containing a poorly water-soluble drug. The prediction error values of dependent responses were less than 5% that confirm validity, robustness and accuracy of the generated DS in optimization of GFBG.

1. Introduction

More than 70% of drugs are administered as solid dosage forms, such as tablets, which implies the extensive use of this tablet dosage form in healthcare sectors [1]. Prior to the tableting process, it is necessary to improve the flow properties of the materials so that the press can be evenly filled, resulting in tablets with uniform weight that can be manufactured [2]. This can be accomplished through the granulation process, which can be divided into two types: dry and wet granulation, depending on whether or not a liquid binder is utilized [3].
Wet granulation is a key unit process in the manufacture of solid dosage forms, particularly tablets and capsules [4]. The main goals of the granulation process are to enhance powder flow, increase material density, reducing segregation, improve content uniformity and enhance the tablets’ mechanical strength [5]. High-shear wet granulation (HSWG), twin-screw granulation and fluid-bed granulation (FBG) are the most common wet granulation technologies [6].
The FBG is one of the most widely used granulation technologies [7]. It converts poor flow powders to free-flowing granules, providing a single-step alternative to separate HSWG and fluid bed drying processes [8]. In addition, it produces porous granules, resulting in highly compactible granules and rapidly disintegrating tablets. The FBG is promising with respect to switching towards continuous processing. However, in the FBG, the drying process results in the granules’ attrition, an undesired size reduction phenomenon that results in the formation of fines, which is a quality-degrading side effect. The deteriorating effects of attrition on powder flow and granules homogeneity are previously reported [9,10,11].
To prevent undesirable attrition during the fluid-bed granulation/drying process, green fluidized-bed granulation (GFBG) is a possible alternative technique. The GFBG applies the concept of a moisture-activated dry granulation process, and mostly consists of two stages: granulation and moisture absorption [12,13]. During the granulation stage, drug, water-soluble diluents and binder are agglomerated by a little amount of water (1–4% w/w). In the absorption stage, the moisture available in the granules is absorbed by the addition of water-insoluble excipients, enabling the drying step to be omitted [14]. On the other hand, the GFBG technique does not involve a high amount of water, which helps to avoid issues, such as hydrolysis and degradation. Therefore, the GFBG technique may be a viable choice in terms of product quality and stability. However, the GFBG process needs more investigation, as it has the potential to avoid the issues face by conventional FBG processes.
Understanding the influence of process variables on granules’ and tablets’ attributes is critical for optimizing a process and increasing manufacturing efficiency [4]. In the pharmaceutical industry, the quality-by-design (QbD) approach has received more attention as a technique for developing products using a science and risk-based approach [15]. Implementation of QbD involves defining the critical process parameters (CPPs) that have a significant effect on the critical quality attributes (CQAs) of products. The design-of-experiment (DoE) approach has an important role in exploring the effects of large numbers of CPPs on CQAs of the product [16]. DoE aids in the development of high-quality product by decreasing the number of costly and time-consuming experiments [16]. The combinatorial effects of key process variables of GFBG on the CQAs of granules and tablets have largely been unexplored. Therefore, the main objective of the present study is to investigate the influence of CPPs of GFBG on CQAs of granules and corresponding tablets using a DoE approach in order to understand the influence of independent process variables on product quality and the implementation of effective control. Further, the GFBG process was optimized through the development of the process design space to produce desirable granules and tablets. In the present research, Fenofibrate was used as a model drug. It is categorized as a class II drug on the Biopharmaceutical Classification System, with a poor water solubility of <0.1 μg/mL [17]. It is widely used for the treatment of hyperlipidemia by reducing the blood levels of cholesterol and triglyceride [18].

2. Materials and Methods

2.1. Materials

Fenofibrate was kindly donated from JPI Co. (Riyadh, Saudi Arabia). Polyvinylpyrrolidone, Kollidon® 12 and D-α-Tocopherol polyethylene glycol 1000 succinate, and Kolliphor® TPGS were obtained as a gift sample from BASF Co. (Ludwigshafen, Germany). Microcrystalline cellulose and Avicel® PH-102 were gained as a gift sample from DuPont Nutrition USA, Inc. (Wilmington, NC, USA). Lactose monohydrate and Granulac 200® were obtained from Meggle (Wasserburg, Germany). Colloidal silicon dioxide and Aerosil 200® were purchased from Evonik (Hanau-Wolfgang, Germany). Sodium stearyl fumarate, Pruv® and sodium starch glycolate, and Explotab® were obtained as a gift sample form JRS pharma (Rosenberg, Germany). Sodium dihydrogen phosphate dehydrate, disodium hydrogen phosphate dodecahydrate and sodium hydroxide were purchased from Merck (Darmstadt, Germany).

2.2. Design of Experiments

In the present study, two key process variables, namely water amount (X1, 1–6% w/w) and spray rate (X2, 2–8 g/min), were investigated to understand their effects on CQAs of prepared granules and tablets using a 32 full-factorial design. According to the results of the preliminary experiments, the low, moderate and high levels of each independent variable were chosen. The independent process variables and their levels are listed in Table 1. For a 32 full-factorial design, nine runs were generated as shown in Table 2. The experimental design and data analysis were carried out using Design-Expert software (version 11, State-ease, Inc., Minneapolis, MN, USA). The following polynomial equation was utilized to fit the experimental data of all granule and tablet properties Equation (1):
Y = b 0 + b 1 X 1 + b 2 X 2 + b 12 X 1 X 2 + b 11 X 1 2 + b 22 X 2 2
where Y is a measured granule or tablet response associated with each variable level combination; b0 is an intercept; b1 and b2 are regression coefficients; and X1 and X2 are the coded levels of investigated variables. The terms of X1X2 and X i 2 (i = 1 and 2) symbolize the interaction and quadratic terms, respectively. The generated design was validated by calculating the prediction error (PE) using Equation (2). A PE of less than 5% proves the validity of the experimental design [19].
PE = (   Predicted   value Experimental   value   Predicted   value ) × 100

2.3. Green Fluidized Bed Granulation

Table 3 shows the formulation used in the present study. The granulation process was conducted in a fluidized bed granulator (BOSCH Packaging Technology, Schopfheim, Germany) at a 700 g scale for all trials. During the entire process, the velocity of fluidizing air was 0.3–0.4 m3/min without the use of heater. The drug, lactose monohydrate, polyvinylpyrolidone and TPGS were initially mixed (2 min) and granulated by spraying water (1–6% w/w) using a centered top spray nozzle (spray rate 2–8 g/min). For the 5 min absorption stage, colloidal silicon dioxide and microcrystalline cellulose were added as moisture absorbents. Finally, the disintegrant “sodium starch glycolate” and lubricant “sodium stearyl fumarate” were added directly into the granulator and mixed for 1.5 min and 2 min, respectively.

2.4. Tablet Compaction

The lubricated granules were used for tablet compaction on an automated single-punch tablet press (Erweka EP-1, Apparatebau, Langen, Germany) using 12 mm round flat-faced tooling at 12 kN. The target tablet weight was 500 mg. The machine was adjusted to produce approximately 50–60 tablets per minute. The produced tablets were collected, stored and evaluated.

2.5. Granules Evaluation

2.5.1. Particle Size Distribution (PSD)

The granules (n = 3) were analyzed for PSD by the laser diffraction method as described in our previous work [20]. Fines were considered as granules with a diameter smaller than 50 µm.

2.5.2. Bulk Density

Granule samples of 15 g (n = 3) were carefully fed into a 50 mL graduated cylinder. The granules weight (m) and volume (v) were determined, and bulk density (ρ) was calculated using Equation (3).
Bulk   Density =   m / v

2.5.3. Granules Flow

A static angle of repose (AoR) method was used to evaluate the flowability of prepared granules as well as the physical mixture. The powder (n = 3) was carefully poured using a dry conical funnel fixed at a constant height (H). The base of the cone’s diameter (D) was determined, and the AoR was calculated using Equation (4).
tan ( α ) = 2 H / D

2.6. Tablets Evaluation

2.6.1. Tablet Weight and Tensile Strength

The tablet weight, thickness (t), diameter (d) and breaking force (f) were measured (n = 10) by Erweka Multi-Check 5.1 (ERWEKA, Heusenstamm, Germany). The tensile strength (σ) of the prepared tablet was calculated using Equation (5).
σ   = 2 F / π td

2.6.2. Tablet Friability

Tablet friability was detected by the feeding of 10 tablets in the drum of friability tester (ERWEKA, Heusenstamm, Germany); rotating at 25 rpm for 4 min. Tablet friability was measured using Equation (6).
Friability   ( % ) = [ ( W 1 W 2 ) / W 1 ] × 100
where W1 is tablets weight before the test and W2 is the tablets’ weight after the test.

2.6.3. Tablet Disintegration Time (DT)

The DT was estimated (n = 6) utilizing the automatic disintegration apparatus (ERWEKA, Heusenstamm, Germany). The distilled water at 37 ± 0.5 °C was used as the disintegration medium. The DT was determined using a digital stopwatch. The DT was achieved when the tablets were fully disintegrated, and no lumps remained in the disintegration medium.

2.6.4. Tablet Dissolution

Tablet dissolution (n = 6) was conducted using an USP apparatus type II (ERWEKA, Heusenstamm, Germany) maintained at 37 ± 0.5 °C and 100 RPM. The test was performed in 900 mL of phosphate buffer (1.73 g L−1 of sodium dihydrogen phosphate dihydrate and 4.92 g L−1 disodium hydrogen phosphate dodecahydrate in highly purified water, pH 6.8), to simulate intestinal fluid, containing 0.75% sodium lauryl sulphate to achieve sink condition. Aliquots of 3 mL were collected at predetermined time intervals (5, 10, 15, 30, 45 and 60 min) and an equivalent volume of fresh medium was replenished at each time point. The withdrawn samples were filtered through a 0.45 µm membrane filter. The filtrate was centrifuged at 11,000 rpm for 5 min and the UV absorbance at 289 nm was used to estimate the fenofibrate content using a UV–vis spectrophotometer (Shimadzu, UV-1800, Kyoto, Japan) [21]. Dissolution profiles were compared using similarity facto (f2), which is defined by Equation (7) [22].
f 2 = 50 log { [ 1 + 1 / n   n t = 1 ( R t T t ) 2 ] 0.5 × 100 }
where n is the number of dissolution sampling times, and Rt and Tt are the percentage of drug released at each time point for the reference and test, respectively. An f2 value of more than 50 indicates that the two dissolution profiles are similar.

3. Results and Discussion

3.1. Data Fitting

The model fitting results including the coefficient of determination (R2), adjusted R2, predicted R2, associated probability (p-value) and adequate precision are summarized in Table 4. All the suggested models have R2 > 0.9, which indicates a high significance and high fitness data to the models. Additionally, the p-value for all suggested models was less than 0.05 at a 5.0 percent significance level, indicating that the models were well-fit to the data. Furthermore, the predicted R2 agrees reasonably well with the adjusted R2 (i.e., the difference is less than 0.2), which demonstrates good data fitting. As shown in Figure 1, a linear correlation between experimental results and results predicted by the model, suggests a good model fit. For all suggested models, adequate precision was more than 4.0. This indicated that the suggested models could be utilized to navigate the design space.

3.2. Influence of Process Variables on Granule Properties

3.2.1. Mean Granule Size (y1) and Percent Fines (y2)

As depicted in Table 5, the mean granule size (D50) ranged from 54.11 ± 0.25 to 142.13 ± 0.35. Compared with the mean size of physical mixture, it can be seen that granule size was significantly increased with the increasing water amount from 1–6% w/w and spray rate from 2–8 g/min. The influence of X1 and X2 on D50 was demonstrated by the following polynomial Equation (8).
D 50 ( μ m ) = 109.38 + 36.37   X 1 + 7.33 X 2 + 2.28   X 1   X 2 15.53   X 1 2 + 1.7   X 2 2
Regression analysis (Table 6) demonstrated that all process variables had significant effects on D50 (p < 0.0001 for amount of water and p = 0.0005 for spray rate). However, the sum of square values suggested that the amount of water was the most significant process variable (7935.93 for water amount and 322.52 for spray rate). All the significant parameters had a positive influence as indicated by the positive sign of the regression coefficient (+36.37 for water amount and +7.33 for spray rate), indicating that the increasing level of these variables results in a larger granule size. Similarly, the interaction terms X1X2 (p = 0.026) and the quadratic term X12 (p = 0.0003) increase the mean granule size. Combined with the contour plot (Figure 2), it is clear that D50 attained its maximum value with the concurrent increase in the investigated process variables (X1 and X2). This could be due to the fact that water induced the powder to stick and coalesce more easily [20,23]. In addition, increasing the spray rate results in more availability of water in the spraying zone and, hence, more wetting and sticking of the particles [23]. This result is in agreement with previously reported work [23,24].
As shown in Table 5, the percent fines ranged from 7.84 ± 0.028 to 18.32 ± 0.052 %. The impact of the process parameters on percent fines was described by the following first-order polynomial Equation (9).
Percent   fines   ( % ) = 13.11 4.94   X 1 + 0.19   X 2
Regression analysis (Table 6) demonstrates that the amount of fines was mainly influenced by water amount (p < 0.0001), while spray rate had no significant effect (p = 0.5248). According to the negative sign of the regression coefficient (−4.94 for water amount), the significant term had a negative effect, indicating that as the amount of water increased, the amount of fines decreased. This could be attributed to an inadequate amount of water, which results in weak and fragile granules with a high percentage of fines [25].

3.2.2. Bulk Density (y3)

The bulk density of produced granules ranged from 0.443 ± 0.009 to 0.523 ± 0.016 g/mL, as indicated in Table 5. The following polynomial equation was used to demonstrate the impact of process parameters on the granule density Equation (10).
Bulk   density   ( g / Ml ) = 0.4634   0.0323   X 1 0.0062   X 2 + 0.0047   X 1   X 2 + 0.0143   X 1 2 + 0.0008   X 2 2
Regression analysis (Table 6) indicated that all process variables had a significant negative effect (p = 0.0002 for amount of water and p = 0.0214 for spray rate) on the bulk density of granules with respect to the negative sign of the regression coefficient (−0.0323 for the amount of water and −0.0062 for the spray rate). However, the amount of water had the predominant effect based on the sum of square values (0.0063 for water amount and 0.0002 for spray rate). Combined with the contour plot shown in Figure 2, the granule bulk density decreased with increasing water amount. The fluid bed produces small granules that have a low free-fall velocity and a high surface-to-mass ratio; this results in a lower bulk density of prepared granules (25). The two-way interaction between the investigated factors had no significant effect (p = 0.0686) on granules bulk density.

3.2.3. Flowability (y4)

The flow of powder during die filling, one of the crucial steps in the manufacturing of tablets, can affect not only the overall efficiency of the tableting process, but also the final product quality, such as weight and content uniformity [26,27]. The flowability of powders used in the manufacturing of tablets has a significant impact on the uniformity of tablet weight and drug content [28]. The angle of repose is an important test for determining the flowability of powders and granules [19]. Its value below 35 indicates good powder flowability, while a value above 35 indicates poor flowability [29]. Comparing the flowability of the physical mixture (the angle of repose of the physical mixture = 35.76 ± 0.311) with the flowability of granules prepared by GFBG, it can be seen that the flowability of granules prepared under various process variables ranged from 26.12 ± 0.295 to 33.11 ± 0.523° (Table 5). This indicates that the flowability of the granules obtained by the GFBG process were significantly improved compared to flowability of the physical mixture. The first-order polynomial equation generated by regression analysis of granules angle of repose is described by Equation (11).
Angle   of   repose   ( ° ) = 29.77     3.38   X 1 + 0.3117   X 2
The regression analysis (Table 6) indicates that the amount of water had a significant effect on granules’ flowability (p < 0.0001 for amount of water and p = 0.4050 for spray rate). According to the polynomial equation, the negative sign of regression coefficients (−3.38) showed that the water amount has an inverse relationship with the granule angle of repose. Combined with the contour plot (Figure 2), it is clear that angle of repose reaches its minimum value with the concurrent increase in water amount, as displayed on the right side of the contour plot. This indicates that, the granules produced using green fluid bed granulation with spraying of a water amount of more than 3.5% has better flowability because of uniform and high granule size. It is reported that increasing water amount improves granules flowability [14].

3.3. Influence of Process Variables on Tablet Properties

3.3.1. Weight Variation

The tablet weight ranged from 497.56 ± 1.35 to 501.71 ± 1.33 mg (Table 7). Based on the USP criteria for tablet weight variation, all prepared tablets achieved an acceptable weight variation with an SD of less than 2. This indicates that the prepared granules demonstrated an acceptable flow and bulk density [30]. The empirical model of first-order polynomial Equation (12) described the impact of the process parameters on weight variation of prepared tablets.
SD   of   weight   variation   = 1.10     0.2767   X 1 + 0.01   X 2
As shown in Table 8, only the amount of water had a significant effect on variation of tablet weight (p = 0.0002 for amount of water and p = 0.7725 for spray rate). The regression coefficients in Equation (12) indicated that the amount of water had negative impact on tablet weight variation according to the negative sign of coefficient estimate (−0.2767). Combined with the contour plot (Figure 2) it is clear that tablet weight variation attained its minimum value with the simultaneous increase in the water amount. By contrast, weight variation was highest when water amount was at its minimum value. This result was consistent with the results of powder flow (Section 3.2.3).

3.3.2. Tensile Strength and Friability

The tensile strength of the tablet is an important indicator to evaluate the mechanical strength of the obtained tablet [19]. As shown in Table 7, the tablet tensile strength ranged from 2.51 ± 0.13 to 3.23 ± 0.24 MPa. This indicates that granules obtained by the green fluidized bed granulation method could be compressed into tablets with acceptable mechanical strength. On the other hand, regression analysis (Table 8) showed that the amount of water had a significant impact (p < 0.0001) on tablet tensile strength. The empirical model of the first-order polynomial Equation (13) describes the effect of the process parameters on tablet tensile strength.
Tensile   strength   ( MPa ) = 2.88 + 0.2867   X 1 + 0.0517   X 2
As shown in Equation (12), the positive coefficients of water amount (+0.2867) revealed that a higher value for this variable results in a higher tensile strength. Combined with the contour plot in Figure 2, it is concluded that increasing the water amount sprayed (up to 6%) could improve the tablet mechanical strength. It was reported that green fluidized bed granulation has the potential to increase the granules compaction properties by enhancing plastic deformability [13]. Additionally, the present granulation method produces looser and large surface area granules that contribute to the strong inter-particle bonding [14].
Friability of tablet is also an important index to evaluate the tablet mechanical strength [31]. As shown in Table 7, the friability of tablet ranged from 0.71 ± 0.01 to 1.24 ± 0.02 %. Runs 1, 2 and 3 (granulated with 2% water amount) produces friable tablets since the prepared tablets lose more than 1.0 % of weight after undergoing the friability test. This is due to inadequate wetting of powder particles and the generation of large amounts of fines (17–18 %) [20,32]. However, runs 4–9 (3.5–6 % water amount) produced acceptable tablets since the tablet weight loss was less than 1.0%. An ANOVA analysis (Table 8) revealed that the amount of water had a significant effect on tablet friability (p < 0.0001). The empirical model of first-order polynomial Equation (14) describes the impact of the process parameters on tablet tensile strength.
Friability   ( % ) = 0.9689 0.2417   X 1 0.0150   X 2
As shown in Equation (13), the negative coefficient of water amount (−0.2417) demonstrates that a higher value for this variable results in a lower weight loss of tablet. Combined with the contour plot in Figure 2, it is concluded that increasing the water amount (more than 2%) could decrease the friability of the prepared tablets and provide acceptable tablet in term of the USP criteria.

3.3.3. Disintegration Time

As shown in Table 7, disintegration time (DT) of the obtained tablet ranged from 31.69 ± 1.31 to 143.12 ± 0.86 s. It was observed that as the water amount increased, the DT was significantly increased. Regression analysis shown in Table 8 indicates that water amount had a significant effect (p < 0.0001) on the DT of the tablets. The empirical model of polynomial Equation (15) describes the influence of the process parameters on DT.
DT   ( s ) = 52.0 + 50.42   X 1 + 4.51   X 2 + 0.1975   X 1   X 2 + 0.3373   X 1 2 + 2.45   X 2 2
Based on the polynomial Equation (15), the positive coefficient of water amount (+50.42) demonstrates that a higher value for this variable results in longer tablet disintegration. On the other hand, the two-way interaction between the investigated variables had no significant (p = 0.9235) effect on tablet disintegration. The contour plot shown in Figure 2 shows the influence of water amount and spray rate on the DT. It is observed that tablet DT attained its minimum value with the simultaneous decrease in the water amount. The rapid disintegration may be due to the low tensile strength of tablets that prepared a low amount of water.

3.3.4. In Vitro Drug Release

Figure 3 shows the release profiles of fenofibrate at different process variables. As shown in Table 7 and Figure 3, only runs 5–9 demonstrate an acceptable drug release profile (i.e., 85 % release after 30 min), while runs 1, 2, 3 and 4 released 73.23 ± 3.15, 75.15 ± 1.14, 76.66 ± 2.13 and 81.25 ± 2.35 %, respectively, after 30 min. Regression analysis (Table 8) indicates that the water amount and spray rate had a significant effect (p = 0.0003 for the amount of water and p = 0.0376 for spray rate) on fenofibrate dissolution. However, water amount was the most significant factor with respect to the sum of square values (281.67 for water amount and 37.20 for spray rate). The following first-order polynomial equation was applied to describe the relationship between the independent process parameters and drug release Equation (16).
Percent   drug   release   after   30   min   ( % ) = 83.00 + 6.85   X 1 + 2.49   X 2
According to the polynomial Equation (16), the positive sign of regression coefficients (+6.85 for water amount and +2.49 for spray rate) shows that water amount and spray rate had a positive effect on fenofibrate release. Combined with the contour plot (Figure 2), it is clear that drug release attained its maximum value with the concurrent increase in the investigated process variables as exhibited in the upper right corner of the contour plot. The result suggests that dissolution of fenofibrate from prepared tablets can potentially be improved by increasing the water amount and spray rate. This could be attributed to high levels of water amount and spray rate produced by low-density granules, as previously described (Section 3.2.2), which rapidly eroded and rapidly released the drug. This resulted in the rapid release and maximum concentration of fenofibrate in dissolution medium. It was reported that granulation using fluid-bed results in the formation of low density and high porous granules that rapidly erode [31,33]. On the other hand, incorporation of TPGS into the granulation process could improves the dissolution profile of fenofibrate. TPGS was found to improve the dissolution and apparent solubility of poorly water-soluble drugs in the granulation process [34]. As shown in Table 9, the comparison of the obtained dissolution profiles clearly revealed a significant increase in the dissolution rates upon increasing the water amount from 1 to 6% w/w (as seen in run 1 # 7, run 2 # 8 and run 3 # 9, and the dissolution profile was dissimilar since f2 was less than 50). The results of f2 similarity confirm the results of regression analysis.

3.4. Process Optimization, Design Space and Validation

The objective of the optimization step is to define the optimum level of each independent variable, in order to produce a product of desired quality attributes [35]. The process parameters should be operated within a defined design space (DS) to obtain the desired quality attributes [15]. Optimization of investigated key process variables was carried out using a numerical optimization method. As shown in Table 10, values of SD of weight variation, tensile strength, friability, DT and percent release within 30 min were set to the target of 0.8, 3 MPa, 0.75% 120 sec and 85%, respectively, to generate the optimized process conditions using desirability function. According to the set criteria, water amount and spray rate at high (5.69% w/w) and low levels (2 g/min), respectively, demonstrate the optimized conditions with a desirability value close to one (Figure 4). As shown in Table 11, granulation at optimized conditions results in tablets with an SD of weight variation of 0.82, tensile strength of 3.20 ± 0.46, friability of 0.74 ± 0.05, DT of 125.12 ± 2.43 and percent release of 88.21 ± 3.31 after 30 min. In addition, the prediction error between the experimental value and the corresponding predicted value was less than 5.0%, which was acceptable. This revealed that the model was valid with reliable and effective predictive power [36].

4. Conclusions

In the present study, the DoE approach as a part of the QbD paradigm was used to optimize the GFBG process. A 32 full-factorial design was performed to explore the effect of water amount (1–6% w/w) and spray rate (2–8 g/min) as key process variables of GFBG on CQAs of granules (D50, percent fines, bulk density and flowability) and tablets (weight variation, mechanical strength, DT and drug release). Regression analysis demonstrated that the investigated process variables have significant effect (p ≤ 0.05) on CQAs of granules and corresponding tablets. Particularly, water amount was found to have the more pronounced effect on granules’ and tablets’ properties. The results reveal that the desired quality attributes of granules and tablets could be optimized by adjusting the process variables of water amount and spray rate during the GFBG process. A design space was developed based on tensile strength, disintegration and dissolution of prepared tablets using desirability function. The results of the prediction error test have shown the validity, reliability and effectiveness of the generated design space in optimizing the GFBG process. By optimizing the water amount and spray rate, GFBG could improve the disintegration and dissolution of tablets containing a poorly water-soluble drug. Compared with the conventional fluid-bed granulation process, GFBG was found to be simple, cost-effective and a less energy consuming process, yielding granules and tablets with desirable quality attributes as it requires no heating step. Thus, this granulation process is a promising method for the granulation of moisture and heat sensitive materials. In summary, the current study increases knowledge of the impact of the GFBG process parameters on properties of granules and tablets, and it might be used to construct a mechanistic model for the granulation process.

Author Contributions

Conceptualization, M.H.F., A.A. and D.A.H.; methodology, M.H.F., A.A. and D.A.H.; software, M.H.F., A.A. and Z.S.A.; validation, M.H.F., A.A. and Z.S.A.; formal analysis, M.H.F., A.A. and D.A.H.; investigation, A.A. and Z.S.A.; resources, M.H.F., A.A. and Z.S.A.; data curation, M.H.F., A.A., Z.S.A. and D.A.H.; writing—original draft preparation, M.H.F., A.A., Z.S.A. and D.A.H.; writing—review and editing, M.H.F., A.A., Z.S.A. and D.A.H.; visualization, M.H.F., A.A., Z.S.A.; supervision, A.A., Z.S.A.; project administration, A.A.; funding acquisition, A.A., Z.S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research funded by the Deputyship for Research & Innovation, Ministry of Education in Saudi Arabia (project number 2021/03/18868).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Acknowledgments

The authors extend their appreciation to the Deputyship for Research & Innovation, Ministry of Education in Saudi Arabia for funding this research work through the project number (2021/03/18868).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Paul, S.; Baranwal, Y.; Tseng, Y.-C. An insight into predictive parameters of tablet capping by machine learning and multivariate tools. Int. J. Pharm. 2021, 599, 120439. [Google Scholar] [CrossRef] [PubMed]
  2. Ficzere, M.; Mészáros, L.A.; Madarász, L.; Novák, M.; Nagy, Z.K.; Galata, D.L. Indirect monitoring of ultralow dose API content in continuous wet granulation and tableting by machine vision. Int. J. Pharm. 2021, 607, 121008. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, B.; Wang, J.; Zeng, J.; Zhao, L.; Wang, Y.; Feng, Y.; Du, R. A review of high shear wet granulation for better process understanding, control and product development. Powder Technol. 2021, 381, 204–223. [Google Scholar] [CrossRef]
  4. Liu, H.; Galbraith, S.C.; Ricart, B.; Stanton, C.; Smith-Goettler, B.; Verdi, L.; O’Connor, T.; Lee, S.; Yoon, S. Optimization of critical quality attributes in continuous twin-screw wet granulation via design space validated with pilot scale experimental data. Int. J. Pharm. 2017, 525, 249–263. [Google Scholar] [CrossRef]
  5. Meng, W.; Rao, K.S.; Snee, R.D.; Ramachandran, R.; Muzzio, F.J. A comprehensive analysis and optimization of continuous twin-screw granulation processes via sequential experimentation strategy. Int. J. Pharm. 2019, 556, 349–362. [Google Scholar] [CrossRef]
  6. Singh, M.; Shirazian, S.; Ranade, V.; Walker, G.M.; Kumar, A. Challenges and opportunities in modelling wet granulation in pharmaceutical industry—A critical review. Powder Technol. 2022, 403, 117380. [Google Scholar] [CrossRef]
  7. Burggraeve, A.; Monteyne, T.; Vervaet, C.; Remon, J.P.; De Beer, T. Process analytical tools for monitoring, understanding, and control of pharmaceutical fluidized bed granulation: A review. Eur. J. Pharm. Biopharm. 2013, 83, 2–15. [Google Scholar] [CrossRef]
  8. Kemp, I.C.; van Millingen, A.; Khaled, H. Development and verification of a novel design space and improved scale-up procedure for fluid bed granulation using a mechanistic model. Powder Technol. 2020, 361, 1021–1037. [Google Scholar] [CrossRef]
  9. Nieuwmeyer, F.; van der Voort Maarschalk, K.; Vromans, H. The consequences of granulate heterogeneity towards breakage and attrition upon fluid-bed drying. Eur. J. Pharm. Biopharm. 2008, 70, 402–408. [Google Scholar] [CrossRef]
  10. De Leersnyder, F.; Vanhoorne, V.; Bekaert, H.; Vercruysse, J.; Ghijs, M.; Bostijn, N.; Verstraeten, M.; Cappuyns, P.; Van Assche, I.; Vander Heyden, Y.; et al. Breakage and drying behaviour of granules in a continuous fluid bed dryer: Influence of process parameters and wet granule transfer. Eur. J. Pharm. Sci. 2018, 115, 223–232. [Google Scholar] [CrossRef]
  11. Vandevivere, L.; Denduyver, P.; Portier, C.; Häusler, O.; De Beer, T.; Vervaet, C.; Vanhoorne, V. The effect of binder types on the breakage and drying behavior of granules in a semi-continuous fluid bed dryer after twin screw wet granulation. Int. J. Pharm. 2022, 614, 121449. [Google Scholar] [CrossRef]
  12. Takasaki, H.; Sakurai, A.; Katayama, T.; Matsuura, Y.; Ohyagi, N.; Wada, K.; Ishikawa, A.; Yonemochi, E. Novel, lean and environment-friendly granulation method: Green fluidized bed granulation (GFBG). Int. J. Pharm. 2019, 557, 18–25. [Google Scholar] [CrossRef] [PubMed]
  13. Ishikawa, A.; Takasaki, H.; Sakurai, A.; Katayama, T.; Wada, K.; Furuishi, T.; Fukuzawa, K.; Obata, Y.; Yonemochi, E. Manufacturability and Properties of Granules and Tablets Using the Eco-Friendly Granulation Method Green Fluidized Bed Granulation Compared to Direct Compression. Chem. Pharm. Bull. 2021, 69, 447–455. [Google Scholar] [CrossRef] [PubMed]
  14. Takasaki, H.; Sakurai, A.; Katayama, T.; Matsuura, Y.; Ohyagi, N.; Mizoguchi, M.; Takano, J.; Wada, K.; Matsui, K.; Nagato, T.; et al. Importance of free water in controlling granule and tablet properties in a novel granulation method, green fluidized bed granulation (GFBG). Int. J. Pharm. 2019, 570, 118647. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, H.; Ricart, B.; Stanton, C.; Smith-Goettler, B.; Verdi, L.; O’Connor, T.; Lee, S.; Yoon, S. Design space determination and process optimization in at-scale continuous twin screw wet granulation. Comput. Chem. Eng. 2019, 125, 271–286. [Google Scholar] [CrossRef]
  16. Fayed, M.H.; Abdel-Rahman, S.I.; Alanazi, F.K.; Ahmed, M.O.; Tawfeek, H.M.; Al-Shdefat, R.I. New gentle-wing high-shear granulator: Impact of processing variables on granules and tablets characteristics of high-drug loading formulation using design of experiment approach. Drug Dev. Ind. Pharm. 2017, 43, 1584–1600. [Google Scholar] [CrossRef]
  17. Mamidi, H.K.; Palekar, S.; Nukala, P.K.; Mishra, S.M.; Patki, M.; Fu, Y.; Supner, P.; Chauhan, G.; Patel, K. Process optimization of twin-screw melt granulation of fenofibrate using design of experiment (DoE). Int. J. Pharm. 2021, 593, 120101. [Google Scholar] [CrossRef]
  18. Bin Ahn, J.; Kim, D.-H.; Lee, S.-E.; Pyo, Y.-C.; Park, J.-S. Improvement of the dissolution rate and bioavailability of fenofibrate by the supercritical anti-solvent process. Int. J. Pharm. 2019, 564, 263–272. [Google Scholar] [CrossRef]
  19. Fayed, M.H.; Aldawsari, M.F.; AlAli, A.S.; Alsaqr, A.; Almutairy, B.K.; Aodah, A.H.; Tawfeek, H.M.; Khafagy, E.-S.; Helal, D.A. Design-of-experiment approach to quantify the effect of nano-sized silica on tableting properties of microcrystalline cellulose to facilitate direct compression tableting of binary blend containing a low-dose drug. J. Drug Deliv. Sci. Technol. 2022, 68, 103127. [Google Scholar] [CrossRef]
  20. Almutairy, B.K.K.; Khafagy, E.-S.; Alalaiwe, A.; Aldawsari, M.F.; Alshahrani, S.M.; Alsulays, B.B.B.; Alshetaili, A.S.; AlShehri, S.M.; Fayed, M.H.H. Enhancing the Poor Flow and Tableting Problems of High Drug-Loading Formulation of Canagliflozin Using Continuous Green Granulation Process and Design-of-Experiment Approach. Pharmaceuticals 2020, 13, 473. [Google Scholar] [CrossRef]
  21. Tran, B.N.; Van Pham, Q.; Tran, B.T.; Le, G.T.; Dao, A.H.; Tran, T.H.; Nguyen, C.N. Supercritical CO2 impregnation approach for enhancing dissolution of fenofibrate by adsorption onto high-surface area carriers. J. Supercrit. Fluids 2022, 184, 105584. [Google Scholar] [CrossRef]
  22. Srinarong, P.; Faber, J.; Visser, M.; Hinrichs, W.; Frijlink, H.W. Strongly enhanced dissolution rate of fenofibrate solid dispersion tablets by incorporation of superdisintegrants. Eur. J. Pharm. Biopharm. 2009, 73, 154–161. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, J.; Li, W.; Qu, H.; Tian, G.; Wei, Y. Application of definitive screening design to quantify the effects of process parameters on key granule characteristics and optimize operating parameters in pulsed-spray fluid-bed granulation. Particuology 2019, 43, 56–65. [Google Scholar] [CrossRef]
  24. Vengateson, U.; Mohan, R. Experimental and modeling study of fluidized bed granulation: Effect of binder flow rate and fluidizing air velocity. Resour. Technol. 2016, 2, S124–S135. [Google Scholar] [CrossRef] [Green Version]
  25. Gao, J.Z.; Jain, A.; Motheram, R.; Gray, D.; Hussain, M. Fluid bed granulation of a poorly water soluble, low density, micronized drug: Comparison with high shear granulation. Int. J. Pharm. 2002, 237, 1–14. [Google Scholar] [CrossRef]
  26. Schomberg, A.K.; Kwade, A.; Finke, J.H. The Challenge of Die Filling in Rotary Presses—A Systematic Study of Material Properties and Process Parameters. Pharmaceutics 2020, 12, 248. [Google Scholar] [CrossRef] [Green Version]
  27. Morin, G.; Briens, L. The Effect of Lubricants on Powder Flowability for Pharmaceutical Application. AAPS PharmSciTech 2013, 14, 1158–1168. [Google Scholar] [CrossRef] [Green Version]
  28. Kudo, Y.; Yasuda, M.; Matsusaka, S. Effect of particle size distribution on flowability of granulated lactose. Adv. Powder Technol. 2020, 31, 121–127. [Google Scholar] [CrossRef]
  29. Moravkar, K.K.; Shah, D.S.; Magar, A.G.; Bhairav, B.A.; Korde, S.D.; Ranch, K.M.; Chalikwar, S.S. Assessment of pharmaceutical powders flowability and comparative evaluation of lubricants on development of gastro retentive tablets: An application of powder flow tester. J. Drug Deliv. Sci. Technol. 2022, 71, 103265. [Google Scholar] [CrossRef]
  30. Yu, Y.; Zhao, L.; Lin, X.; Wang, Y.; Du, R.; Feng, Y. Research on the powder classification and the key parameters affecting tablet qualities for direct compaction based on powder functional properties. Adv. Powder Technol. 2021, 32, 565–581. [Google Scholar] [CrossRef]
  31. AlAli, A.S.; Aldawsari, M.F.; Alalaiwe, A.; Almutairy, B.K.; Al-Shdefat, R.; Walbi, I.A.; Fayed, M.H. Exploitation of Design-of-Experiment Approach for Design and Optimization of Fast-Disintegrating Tablets for Sublingual Delivery of Sildenafil Citrate with Enhanced Bioavailability Using Fluid-Bed Granulation Technique. Pharmaceutics 2021, 13, 870. [Google Scholar] [CrossRef] [PubMed]
  32. Basim, P.; Haware, R.V.; Dave, R.H. Tablet capping predictions of model materials using multivariate approach. Int. J. Pharm. 2019, 569, 118548. [Google Scholar] [CrossRef] [PubMed]
  33. Morin, G.; Briens, L. A Comparison of Granules Produced by High-Shear and Fluidized-Bed Granulation Methods. AAPS PharmSciTech 2014, 15, 1039–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sotthivirat, S.; Ramesh, R.; Wasylaschuk, W.; Bottone, C.; Xia, B.; Stellabott, J.; McNevin, M.; Skomski, D.; Brown, C. Effect of TPGS surfactant on dissolution sensitivity of a poorly water-soluble drug using high-shear wet granulation. Powder Technol. 2020, 375, 302–309. [Google Scholar] [CrossRef]
  35. Alalaiwe, A.; Fayed, M.H.; Alshahrani, S.M.; Alsulays, B.B.; Alshetaili, A.S.; Tawfeek, H.M.; Khafagy, E.-S. Application of design of experiment approach for investigating the effect of partially pre-gelatinized starch on critical quality attributes of rapid orally disintegrating tablets. J. Drug Deliv. Sci. Technol. 2019, 49, 227–234. [Google Scholar] [CrossRef]
  36. Aodah, A.; Fayed, M.; Alalaiwe, A.; Alsulays, B.; Aldawsari, M.; Khafagy, E.-S. Design, Optimization, and Correlation of In Vitro/In Vivo Disintegration of Novel Fast Orally Disintegrating Tablet of High Dose Metformin Hydrochloride Using Moisture Activated Dry Granulation Process and Quality by Design Approach. Pharmaceutics 2020, 12, 598. [Google Scholar] [CrossRef]
Figure 1. Linear correlation plot relating; mean granule size, percent fines, bulk density, angle of repose, SD of weight variation, tensile strength, friability, disintegration time and drug release after 30 min, between the predicted and the actual values.
Figure 1. Linear correlation plot relating; mean granule size, percent fines, bulk density, angle of repose, SD of weight variation, tensile strength, friability, disintegration time and drug release after 30 min, between the predicted and the actual values.
Pharmaceutics 14 01471 g001
Figure 2. Contour plots showing the effect of water amount (X1) and spray rate (X2) on mean granule size (y1), percent fines (y2), bulk density (y3), angle of repose (y4), SD of weight variation (y5), tensile strength (y6), friability (y7), disintegration time (y8) and percent release after 30 min (y9).
Figure 2. Contour plots showing the effect of water amount (X1) and spray rate (X2) on mean granule size (y1), percent fines (y2), bulk density (y3), angle of repose (y4), SD of weight variation (y5), tensile strength (y6), friability (y7), disintegration time (y8) and percent release after 30 min (y9).
Pharmaceutics 14 01471 g002
Figure 3. In vitro release profiles of fenofibrate tablets based on 32 full-factorial Design.
Figure 3. In vitro release profiles of fenofibrate tablets based on 32 full-factorial Design.
Pharmaceutics 14 01471 g003
Figure 4. Optimization plot showing the influence of water amount (X1) and spray rate (X2) on overall desirability.
Figure 4. Optimization plot showing the influence of water amount (X1) and spray rate (X2) on overall desirability.
Pharmaceutics 14 01471 g004
Table 1. Key process variables used in the design of experiments.
Table 1. Key process variables used in the design of experiments.
Coded LevelsWater Amount (% w/w)Spray Rate (g/min)
−112
03.55
168
−1: factor at low level; 0: factor at medium level; 1: factor at high level.
Table 2. A 32 full-factorial experimental design.
Table 2. A 32 full-factorial experimental design.
RunWater Amount (% w/w)Spray Rate (g/min)
11.02
21.05
31.08
43.52
53.55
63.58
762
865
968
Table 3. Formulation used in the granulation experiments.
Table 3. Formulation used in the granulation experiments.
Ingredients % w/wFunction
Fenofibrate32Poorly water-soluble drug
Lactose monohydrate 46.5Water soluble filler
Microcrystalline cellulose PH-102 10Moisture absorbents
Polyvinylpyrrolidone K125Binder
D-α-Tocopherol polyethylene glycol 1000 succinate2Non-ionic surfactant
Sodium starch glycolate 2Super-disintegrant
Colloidal silicone dioxide 1.5Moisture absorbents
Sodium stearyl fumarate1Hydrophilic lubricant
Total 100-
Table 4. Summary of model fitting and statistical analysis.
Table 4. Summary of model fitting and statistical analysis.
ResponsesSuggested Modelp-ValueR2Adjusted R2Predicted R2Adequate Precision
Y1:D50Quadratic<0.00010.99960.99890.996596.642
Y2: Percent finesLinear<0.00010.98090.97460.959625.775
Y3: Bulk densityQuadratic0.00120.99500.98680.942827.664
Y4: Angle of reposeLinear0.00020.94050.92070.877614.986
Y5: SD of weight variationLinear0.00050.92120.89490.830812.261
Y6: Tensile strengthLinear<0.00010.95940.94590.902419.571
Y7: FriabilityLinear<0.00010.99340.99130.987545.194
Y8: Disintegration timeQuadratic0.00040.99760.99350.971335.529
Y9: Percent release after 30 minLinear0.00140.90990.87980.799214.105
Table 5. Measured response results of physical mixture and prepared granules (mean ± SD).
Table 5. Measured response results of physical mixture and prepared granules (mean ± SD).
FormulaD10
(µm ± SD )
D50
(µm ± SD )
D90
(µm ± SD )
Percent Fines
(% ± SD )
Bulk Density
(g/mL ± SD)
AoR
(Degree ± SD)
Physical Mixture12.22 ± 0.3151.15 ± 0.22107.36 ± 0.3321.13 ± 0.0220.411 ± 0.03135.76 ± 0.311
114.31 ± 0.2954.11 ± 0.25114.25 ± 0.2418.11 ± 0.0340.523 ± 0.01632.87 ± 0.331
217.12 ± 0.3157.32 ± 0.34116.43 ± 0.3618.32 ± 0.0520.511 ± 0.00433.11 ± 0.523
319.02 ± 0.3364.42 ± 0.31123.11 ± 0.2917.92 ± 0.0230.498 ± 0.02232.73 ± 0.027
423.11 ± 0.28103.37 ± 0.25205.33 ± 0.2711.67 ± 0.0910.468 ± 0.03328.71 ± 0.515
523.76 ± 0.26110.54 ± 0.27225.42 ± 0.3113.77 ± 0.0370.462 ± 0.00530.88 ± 0.347
627.32 ± 0.31117.63 ± 0.25221.15 ± 0.3413.45 ± 0.0150.462 ± 0.03831.14 ± 0.342
736.15 ± 0.19122.71 ± 0.29227.71 ± 0.388.66 ± 0.0510.449 ± 0.00426.54 ± 0.263
842.21 ± 0.26129.22 ± 0.38235.86 ± 0.397.84 ± 0.0280.446 ± 0.04525.79 ± 0.532
9112.23 ± 0.31142.13 ± 0.35256.15 ± 0.348.21 ± 0.0240.443 ± 0.00926.12 ± 0.295
Table 6. Regression analysis of granules measured responses.
Table 6. Regression analysis of granules measured responses.
VariablesCoefficient EstimateSum of SquaresStandard ErrorF-Valuep-Value95 % CI Low95 % CI High
Y1: D50 (Quadratic model)
Intercept109.38-0.8256--106.75112.01
X136.377935.930.45226468.77<0.000134.9337.81
X27.33322.520.4522262.890.00055.898.77
X1 X22.2820.750.553816.910.02600.5154.04
X12−15.53482.260.7832393.100.0003−18.02 −13.04
X221.75.790.78324.720.1182−0.79084.19
Y2: Percent fines (Linear model)
Intercept13.11-0.2298--12.5413.67
X1−4.94146.420.2815308.04<0.0001−5.63−4.25
X20.190.210.28150.450.5248−0.490.87
Y3: Bulk density (Quadratic model)
Intercept0.463-0.0012--0.4550.4715
X1−0.0320.00630.0014539.800.0002−0.0368−0.0279
X2−0.0060.00020.001419.640.0214−0.0106−0.0017
X1 X20.0040.00010.00177.770.0686−0.00070.0102
X120.01430.00040.002435.360.00950.00670.0220
X220.00081.389 × 10−61.389 × 10−60.11950.7524−0.00680.0085
Y4: Angle of repose (Linear model)
Intercept29.77-0.2842--29.0730.46
X1−3.3868.410.348194.12<0.0001−4.23−2.52
X20.310.58280.34810.800.4050−0.541.16
X1 and X2 are water amount and spray rate, respectively; X1X2 is the interaction effect.
Table 7. Measured response results of prepared tablets (mean ± SD).
Table 7. Measured response results of prepared tablets (mean ± SD).
FormulaWeight
(mg ± SD)
Thickness
(mm ± SD)
Tensile Strength
(MPa ± SD)
Friability
(% ± SD)
Disintegration Time
(s ± SD)
%release at 30 min
(% ± SD)
1497.56 ± 1.354.12 ± 0.0112.53 ± 0.641.22 ± 0.0234.25 ± 2.1373.23 ± 3.15
2498.93 ± 1.374.31 ± 0.0042.51 ± 0.131.24 ± 0.0231.69 ± 1.3175.15 ± 1.14
3501.71 ± 1.334.33 ± 0.0062.73 ± 0.651.19 ± 0.0244.91 ± 1.2676.66 ± 2.13
4499.78 ± 1.034.23 ± 0.022.89 ± 0.380.97 ± 0.0251.36 ± 1.6781.25 ± 2.35
5498.84 ± 1.184.12 ± 0.0032.86 ± 0.580.94 ± 0.0453.21 ± 1.4185.11 ± 1.84
6497.75 ± 1.214.31 ± 0.0072.88 ± 0.910.96 ± 0.0556.33 ± 1.3489.45 ± 2.48
7498.45 ± 0.884.32 ± 0.0043.11 ± 0.610.76 ± 0.01131.67 ± 2.1986.76 ± 2.81
8498.63 ± 0.734.41 ± 0.0013.15 ± 0.290.73 ± 0.02138.57 ± 1.5789.32 ± 2.21
9501.23 ± 0.784.33 ± 0.033.23 ± 0.240.71 ± 0.01143.12 ± 0.8690.07 ± 3.17
Table 8. Regression analysis of tablets measured responses.
Table 8. Regression analysis of tablets measured responses.
VariablesCoefficient EstimateSum of SquaresStandard ErrorF-Valuep-Value95 % CI Low95 % CI Low
Y5:SD of weight variation (Linear model)
Intercept1.10-0.027--1.031.16
X1−0.270.45930.03370.020.0002−0.3576−0.1958
X20.010.01600.0330.090.7725−0.07090.0909
Y6:Tensile strength (Linear model )
Intercept2.88-0.020--2.832.93
X10.280.49310.024137.49<0.00010.22680.3465
X20.051.050.0244.470.0790−0.00820.1115
Y7:Friability (Linear model)
Intercept0.96-0.006--0.95280.9849
X1−0.240.35040.008905.38<0.0001−0.2613−0.2220
X2−0.010.00140.0083.490.111−0.03470.0047
Y8:Disintegration time (Quadratic model)
Intercept52.00-2.82--43.0260.98
X150.4215252.051.551063.40<0.000145.5055.34
X24.51122.221.558.520.0615−0.4079.43
X1 X20.190.151.890.010.923−5.836.22
X1233.732276.102.68158.690.001125.2142.26
X222.4512.002.680.83700.4277−6.0710.97
Y9:Percent release at 30 min (linear model)
Intercept83.00-0.7647--81.1384.87
X16.85281.670.936653.520.00034.569.14
X22.4937.200.93667.070.03760.19824.78
X1 and X2 represent the amount of water and the spray rate, X1X2 is the effect of interaction and X12& X22 are the sum of effects.
Table 9. Similarity factor (f2) of dissolution profiles of different runs.
Table 9. Similarity factor (f2) of dissolution profiles of different runs.
ComparisonF2 ValueDissolution Profile
Run 1 # 459Similar
Run 1 # 749Disimilar
Run 2 # 554Similar
Run 2 # 846Disimilar
Run 3 # 654Similar
Run 3 # 946Disimilar
Run 4 # 770Similar
Run 5 # 870Similar
Run 6 # 962Similar
Table 10. The constraints adopted for developed design space.
Table 10. The constraints adopted for developed design space.
VariablesTargetRangeWeightImportance Co-Efficient
In-put
Water amountIn range1–6% w/w1Not applicable
Spray rateIn range2–8 g/min1Not applicable
Out-put
SD of weight variation0.80.73–1.371+++
Tensile strengthMaximize2.51–3.23 MPa1+++
Friability0.750.71–1.24%1+++
Disintegration time12031.69–143.12 s1+++
Percent release after 30 min8573.23–90.07%1+++
Table 11. Predicted and experimental values of optimized run with their prediction errors.
Table 11. Predicted and experimental values of optimized run with their prediction errors.
VariablesValue
Water amount (% w/w)5.69
Spray rate (g/min)2.0
Overall desirability = 0.876
ResponsesPredicted ValuesExperimental Values *Prediction Error (%)
SD of weight variation0.840.822.38
Tensile strength (MPa)3.073.20 ± 0.46−4.23
Friability (%)0.770.74 ± 0.053.89
Disintegration time (sec)120125.12 ± 2.43−4.26
Percent release after 30 min (%)86.5288.21 ± 3.31−1.95
* Experimental (actual) values are presented as mean ± SD.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fayed, M.H.; Alalaiwe, A.; Almalki, Z.S.; Helal, D.A. Design Space Approach for the Optimization of Green Fluidized Bed Granulation Process in the Granulation of a Poorly Water-Soluble Fenofibrate Using Design of Experiment. Pharmaceutics 2022, 14, 1471. https://doi.org/10.3390/pharmaceutics14071471

AMA Style

Fayed MH, Alalaiwe A, Almalki ZS, Helal DA. Design Space Approach for the Optimization of Green Fluidized Bed Granulation Process in the Granulation of a Poorly Water-Soluble Fenofibrate Using Design of Experiment. Pharmaceutics. 2022; 14(7):1471. https://doi.org/10.3390/pharmaceutics14071471

Chicago/Turabian Style

Fayed, Mohamed H., Ahmed Alalaiwe, Ziyad S. Almalki, and Doaa A. Helal. 2022. "Design Space Approach for the Optimization of Green Fluidized Bed Granulation Process in the Granulation of a Poorly Water-Soluble Fenofibrate Using Design of Experiment" Pharmaceutics 14, no. 7: 1471. https://doi.org/10.3390/pharmaceutics14071471

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop