Int J App Pharm, Vol 15, Issue 3, 2023, 35-42Original Article

IMPLEMENTING CENTRAL COMPOSITE DESIGN FOR THE DEVELOPMENT OF TACROLIMUS FILM FOR SUBLINGUAL ADMINISTRATION

JISHA MOHANAN1, GOPINATH SUBRAMANIYAN2*, ARUL KUTTALINGAM3, DAMODHARAN NARAYANASAMY4

1,3College of Pharmaceutical Sciences, Government Medical College, Thiruvananthapuram 695011, Kerala, India. 2Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, Tamil Nadu, India. 4SRM College of Pharmacy, SRM University, Kattankulathur 603203, Tamil Nadu, India
*Email: gopinath.s@sriramachandra.edu.in

Received: 04 Jan 2023, Revised and Accepted: 09 Mar 2023


ABSTRACT

Objective: The study aimed to develop fast-dissolving films (FDFs) of the immunosuppressant drug tacrolimus monohydrate for sublingual administration, employing central composite design (CCD), to improve its bioavailability.

Methods: Tacrolimus: β-cyclodextrin inclusion complexes prepared earlier were formulated into FDFs. CCD was used for developing optimal film formulation with the desired characteristics. The solvent casting method was used for the preparation of films. For optimization, the independent variables selected were the concentration of hydroxy propyl methyl cellulose E5 (HPMC E5) (X1) and concentration of croscarmellose sodium (CCS) (X2) and the responses were disintegration time (Y1) and percentage drug release at 5 min (Y2). The suggested optimal films were subjected to further characterization.

Results: All the formulations showed good mechanical properties. The composition of optimized FDF constituted 3.016% w/v of HPMC and 11.731%w/w of CCS and its average disintegration time was 27.28s and showed 83.13% mean drug release at 5 min. Differential Scanning calorimetry (DSC) analysis showed complete dispersion and partial conversion into the amorphous form of the drug, which was also confirmed by X-ray diffraction (XRD) studies. Scanning Electron Microscopy (SEM) revealed the smooth and porous nature of the film.

Conclusion: The developed FDF may be used sublingually for delivering tacrolimus efficiently, avoiding its oral bioavailability problems.

Keywords: Tacrolimus, Croscarmellose sodium, Sublingual, Fast dissolving films, Central composite design


INTRODUCTION

The sublingual route of administration has the advantage of bypassing first-pass metabolism and thereby increasing the bioavailability of drugs. Also, the sublingual mucosa is having relatively increased permeability due to the thin membrane and supply of vascular and lymphatic drainage. Additionally, reduced drug dosage to achieve similar target levels compared with oral administration may result in lower medical costs and patient compliance will be more especially in patients facing difficulty in swallowing and chewing [1, 2].

The commonly used sublingual dosage forms are orodispersible films and tablets. As the fast-dissolving film (FDF) is administered through the sublingual route, rapid absorption of the drug is possible, which finally leads to the quick onset of drug action [3]. They are taken without water and due to their ability to disintegrate within a few minutes, they quickly release medication in the mouth [4, 5]. Various superdisintegrants are also employed in the formulation of fast-dissolving films [6].

Tacrolimus is an immunosuppressant having poor and erratic bioavailability mainly due to low solubility, the influence of food intake and concomitant medication, and heapto first-pass metabolism. Tacrolimus has played a key role in today’s high success rate of solid organ transplantation. A variety of clinical situations requiring nonoral medication delivery arise, presenting the need for reliable alternative routes of tacrolimus administration. Many clinical studies suggest the use of sublingual administration of tacrolimus producing comparable blood-drug levels with similar or even lower doses than the oral route in the lung as well as liver transplant recipients [7–9].

Response surface methodology (RSM) is an efficient technique for formulation optimization by efficiently exploring the relationships between the investigated factors and measured responses using the minimum number of experimental trials. Central composite design (CCD) is one of the most pertinent designs of RSM, which consists of factorial and axial points as well as one point at least at the center of the experimental region that provides properties like orthogonality and rotatability for fitting quadratic polynomials. This diversity of points is useful for providing complete knowledge of responses using the least number of experiments [10].

In the present work, an attempt was made to formulate tacrolimus into FDF formulation for sublingual application. CCD was employed to analyze the effect of the independent variables on the selected responses and to suggest the optimized production parameters for attaining a film formulation with desirable responses.

MATERIALS AND METHODS

Materials

Tacrolimus monohydrate was purchased from Yarrowchem, Mumbai, India. Hydroxy propyl methyl cellulose E5 (HPMC E5) and saccharine sodium were procured from Loba Chemie, Mumbai, India, croscarmellose sodium (CCS) from Astron Chemicals, Ahmedabad, India, and Menthol from the Sisco Research Laboratories, India. Polyethylene glycol 400 (PEG 400) and methanol were purchased from Kanton Laboratories, India.

Methods

Formulation of sublingual ODFs

To increase the solubility of tacrolimus, it was converted to an inclusion complex with β-cyclodextrin as the carrier by the kneading method. The drug and the carrier were triturated with a mixed solution of 1:1 (v/v) methanol: water in a glass mortar to obtain a thin slurry. The slurry was kneaded thoroughly for around 40 min. After the evaporation of the solvents, the complexes were dried at room temperature for a period of 1 d [11].

FDF formulations loaded with β-cyclodextrin inclusion complex of tacrolimus monohydrate were formulated with HPMC E5 as the film-forming polymer, PEG 400 as the plasticizer, and CCS as the superdisintegrant [12]. Menthol was used as the flavouring agent to give a mouth refreshment feeling and saccharine sodium was used as the sweetening agent.

The films were prepared by solvent casting method. An accurately weighed quantity of HPMC E5 was soaked in distilled water for 30 min for proper dispersion of the polymer. Added PEG 400, inclusion complex of the drug and other excipients, and the final volume was adjusted with methanol. The dispersion was sonicated to remove the air-entrapped bubbles.

The casting solution (10 ml) was poured into a glass mold (area =32 cm2, every 2 cm2 contains 1 mg of drug) and the solvent was allowed to evaporate by placing the glass mold in an oven for 24 h. The dried films were stored in a desiccator at room temperature.

Experimental design using CCD

CCD was used for the formulation and optimization of FDF using design expert software (version 13, Statease Inc, Minneapolis, USA) to investigate the influence of two independent variables i.e., the concentration of HPMC E5 (X1) and concentration of CCS (X2) on two responses, disintegration time (Y1) and in vitro percentage drug release at 5 min (Y2). A total of thirteen experimental runs were generated by the software, including 5 replicates of the center points, 4 axial, and 4 factorial points, and were carried out in randomized order. The different levels of variables are presented in table 1 and the actual composition of each formulation batch (F1-F13) is given in table 2.

Table 1: Variables used in the CCD

Levels
-1 0 +1
Independent variables
X1= Conc. of HPMC E5(%w/v) 2.58579 3 4 5 5.41421
X2= Conc. of CCS (%w/w) 6.75736 8 11 14 15.2426
Dependent variables Desirability constraints
Y1= Disintegration time (s) Minimise
Y2= In vitro drug release at 5 min (%) Maximise

Table 2: Composition of different batches of formulations*

Run Batch

HPMC E5

(%w/v)

CCS

(%w/w)

PEG 400

(%v/v)

Menthol (mg) Saccharine sodium (mg) Distilled water (ml) Methanol q. s. (ml)
1 F1 3 8 2 100 80 3 10
2 F2 4 11 2 100 80 3 10
3 F3 3 14 2 100 80 3 10
4 F4 5 8 2 100 80 3 10
5 F5 4 6.75736 2 100 80 3 10
6 F6 4 11 2 100 80 3 10
7 F7 2.58579 11 2 100 80 3 10
8 F8 4 15.2426 2 100 80 3 10
9 F9 4 11 2 100 80 3 10
10 F10 4 11 2 100 80 3 10
11 F11 5 14 2 100 80 3 10
12 F12 4 11 2 100 80 3 10
13 F13 5.41421 11 2 100 80 3 10

*note: the dose of the drug-β-cyclodextrin inclusion complex was calculated to contain 1 mg of the drug in a single film of 2 cm2 area

Evaluation of sublingual films

Physical and mechanical properties

The prepared films were evaluated for physical characteristics, weight uniformity, thickness, and folding endurance.

The prepared films were tested visually for their physical appearance and surface texture as well as for any drug precipitation and air bubble entrapment. Weight variation was calculated by weighing the film formulations in electronic analytical balance. The films of 2 × 1 cm2 size were cut from three different positions of each batch prepared and analyzed. The film thickness was measured using a micrometer screw gauge [13]. Thickness was measured at different locations of a film and the average value was determined. Folding endurance was measured manually by repeated folding of the films at the same place till they broke. The folding endurance value is the number of times the film is folded without breaking [14, 15]. All the evaluations were done in triplicate and the results were expressed as average values and SD.

Surface pH

The surface pH of the film and the sublingual pH (6.8) should be closer so that the film dissolves quickly and be compatible. The surface pH is determined to evaluate the possible irritative effects of the formulations on the mucosae.

The film was placed in a Petri dish and was moistened with 1 ml of distilled water and kept for 30 seconds. The pH was noted after bringing the electrode of the pH meter in contact with the surface of the formulation and allowing equilibration for 1 min [16, 17].

Drug content uniformity

The film was placed in a 100 ml volumetric flask and then dissolved in 50 ml methanol using a magnetic stirrer. The volume was completed with methanol and the solution was filtered and drug content was estimated by HPLC (instrument model: HPLC Agilent 1260 infinity).

The estimation of tacrolimus in the samples was done by the HPLC method according to USP. The mobile phase was constituted from different compositions of 6 mmol phosphoric acid and acetonitrile and tertiary butyl methyl ether (81:19). The diluent used was acetonitrile and water (7:3). The column employed was of dimension 15 cm X 4.6 mm and the column temperature, 60 °C. 20 µl of injection volumes were used and the eluents were examined at 220 nm by a stream rate of 1.5 ml/minute [16, 18].

In vitro disintegration time

No official guidelines are available for the determination of in vitro disintegration time of sublingual films. So, a non-official method was adopted. The disintegration test was performed by placing a film of size 2 × 1 cm2 in a glass Petri dish containing 10 ml of phosphate buffer pH 6.8. The time required for the film to disintegrate was recorded [14, 19].

In vitro drug release studies

The drug release study from the prepared FDFs was performed in beakers, each containing 100 ml of phosphate buffer at pH 6.8 [4, 14]. The beakers were placed in a shaking water bath set at 37±0.5 °C and 100 rpm. Samples were removed at predetermined time intervals and filtered through a Whatman filter paper and the drug content of sample solutions was determined after suitable dilutions by HPLC. The aliquots withdrawn were replaced with fresh medium to maintain sink condition.

The determinations were conducted in triplicates. The amount of drug released at each time interval as well as the cumulative amount of drug released, was calculated as a function of time and the drug release profile graphs were constructed.

Statistical analysis

The experimental data obtained from the evaluations of all the film formulations (13 batches) were analyzed using Design Expert software. The optimum concentration of the independent variables, concentrations of HPMC E5 and CCS, was chosen based on the condition of the responses in obtaining maximum in vitro drug release at 5 min and minimum disintegration time. The response surface behavior was investigated for the response function (Y) employing the polynomial equation (1) and the generalized response surface model;

Y = β01X12X211X1222X2212X1X2 …. (1)

Where Y is the predicted response; β0 is constant; β1 and β2 are the linear and quadratic coefficients and β11, β22, and β12 are the interaction coefficients.

The analysis of variance (ANOVA) was used to identify significant effects of factors on response regression coefficients. All significant independent variable effects (p<0.05) were included in the reduced model. To visualize the interaction effect of the variables on the responses, three-dimensional (3D) response surface plots were composed.

With the application of desirability constraints on in vitro drug release at 5 min and disintegration time, a numerical optimization analysis was then employed by Design-Expert® software to develop the optimal ODF formulation with the desired responses. The disintegration time and drug release studies were carried out on the prepared optimized formulation to verify the theoretical prediction. The relative errors (%) between the predicted and observed values for each response were calculated [20].

Differential scanning calorimetry (DSC)

The prepared optimized formulation was subjected to DSC analysis (instrument model Q20 TA instruments) and compared with the thermogram of pure tacrolimus. Film strips weighing 10–12 mg were hermetically sealed in a standard aluminium pan and were heated at a scanning rate of 10 °C/min from room temperature to 225 °C under nitrogen gas flow.

X-ray diffraction (XRD) analysis

The roentgen graphic analyses were conducted with bruker model D8 Advance X-ray diffract meter (The samples were analyzed utilizing monochromatic Cu K-radiation at 40 mA and 40 kV within the region of 2.5° ≤ 2θ ≤ 40° at 25 °C in continuous scan mode with a step size of 0.02°. The diffractogram of the film was compared to the pure tacrolimus.

Surface morphology by scanning electron microscopy (SEM)

SEM studies of optimized FDF and that of pure tacrolimus were done with the Scanning Electron Microscope (JOEL JSM-6390; JAPAN), samples were mounted with double-sided carbon tape on aluminium stubs, and all the samples were sputtered with a thin layer of gold in auto fine coater JEOL JFC 1600 and the images were examined under vacuum at an accelerating voltage of 10 kV.

RESULTS AND DISCUSSION

Physical and mechanical properties

FDFs should possess suitable mechanical strength to resist handling during processing, packing, and transit without being damaged.

All the films were smooth, transparent, homogeneous, and flexible. The results of evaluations of weight variation, film thickness, folding endurance, surface pH, and drug content uniformity are presented in table 3. The weight variation was found to be acceptable as indicated by the small standard deviations. This shows the uniform distribution of the ingredients in the prepared films. All the films were of uniform thickness, in the range of 0.090 to 0.113 mm. Folding endurance is used to estimate the mechanical properties of a film and all the batches showed satisfactory flexibility. The surface pH of all the formulations was in the sublingual pH range; therefore, no possibility of irritation may be expected when administered sublingually and thus, patient acceptance will not be affected. The drug content of the ODFs was in the range of 95.71–99.12%, implying uniform distribution of the drug in the films.

Table 3: Evaluation of weight variation, film thickness, folding endurance, surface pH, and drug content uniformity of films

Batch Weight variation(mg) Film thickness (mm) Folding endurance Surface pH Drug content uniformity (%)
F1 19.77±1.2494 0.097±0.0058 30±3.5498 6.89±0.0582 98.16±0.9301
F2 23.10±2.7118 0.107±0.0058 33±5.4654 6.64±0.0747 97.89±1.5396
F3 20.56±0.7409 0.010±0.0100 28±3.8173 6.71±0.0151 97.66±0.7723
F4 27.67±1.3255 0.113±0.0058 38±4.8875 6.65±0.0473 96.82±1.8630
F5 22.58±0.9754 0.103±0.0058 32±7.5878 6.85±0.0354 98.47±1.0792
F6 23.14±1.3230 0.107±0.0058 28±2.5473 6.72±0.0620 95.91±0.8334
F7 18.98±2.1204 0.090±0.0100 25±10.554 6.69±0.0645 99.12±1.0414
F8 24.43±1.1412 0.107±0.0058 28±6.4979 6.73±0.0745 98.44±1.4462
F9 23.87±1.4390 0.107±0.0058 30±5.3556 6.67±0.0954 98.48±0.7456
F10 24.91±1.2871 0.107±0.0058 29±7.9854 6.76±0.0453 97.65±2.0165
F11 26.24±2.4746 0.113±0.0058 39±9.2591 6.81±0.0980 98.44±1.4166
F12 23.31±1.2146 0.107±0.0058 34±10.655 6.64±0.0655 95.67±0.8747
F13 28.82±1.6756 0.113±0.0058 38±4.9645 6.84±0.0873 96.96±1.6874

Data are expressed as mean±SD, n=3

In vitro disintegration time

The basic requisite of FDF is its short disintegration time in the saliva when placed in the mouth. But none of the official guidelines states the acceptable range of disintegration time. In the present study, almost all the batches have shown a disintegration time of fewer than 60 seconds, and the results obtained are presented in table 5. This response was selected as one of the independent variables, X1, for the optimization of the formulation.

In vitro drug release studies

No official compendial methods have been reported for release studies of FDF dosage forms. The literature reports various approaches, including the use of different volumes from 5 to 900 ml, different apparatus (pharmacopoeial or custom-made), and different media [19].

A fast release of the drug from the sublingual film is necessary so that the dose is not swallowed into the stomach. Moreover, since tacrolimus is a BCS class II drug, once the drug is released into the sublingual mucosa it will be rapidly absorbed and the higher permeability of the sublingual mucosa also aids the absorption process. So, the greater the drug release, the better we can expect bioavailability.

The cumulative percentage of drug release from all the prepared batches is shown in fig. 1a, 1b, and 1c. For all the batches, more than 95% of drug release was obtained in 30 min and almost 80% of drug content was released at around 5 min and this % drug release at 5 min was selected as one of the independent variables (X2) for the optimization of the formulation.

The fast drug release from the film can be attributed to the hydrophilic nature of the used polymer, HPMCE5, which absorbs water, dissolves rapidly and, thus introducing porosity into the film. The external solvent then diffuses into the film, thereby accelerating the drug dissolution [21].

Sublingual administration of tacrolimus FDF is expected to improve the drug’s bioavailability by bypassing first-pass hepatic metabolism. In addition, the inclusion of the drug into β-cyclodextrin complexes before its formulation as a film enhances the drug's solubility too.

Fig. 1: In vitro drug release profiles of sublingual films a) from batches F1-F5 b) from batches F6-F9 and 1c) from batches F10-F13 (mean±SD, n=3)

Table 4: ANOVA analysis of responses

Variables Disintegration time (s) In vitro drug release at 5 min (%)
Regression coefficients P-value
Intercept 37.82 <0.0001
X1-HPMC E5 11.08 <0.0001
X2-CCS -5.56 <0.0001
X1 X2 -5.94 <0.0001
X1² -1.39 0.0089
X2² 2.57 0.0003
0.9949
Adjusted R² 0.9913
Predicted R² 0.9828
Std. Dev. 1.02
CV% 2.65

Statistical analysis

Based on CCD analysis, the effect of concentration of HPMC E5 (X1) and concentration of CCS (X2) on two responses disintegration time (Y1) and percentage drug release at 5 min (Y2) was studied and evaluated the main effects, interaction effects, and quadratic effects of the process variables on selected responses.

Fitting model to the data

The Design expert software suggested a quadratic model fitting best for both responses (disintegration time and percentage drug release at 5 min) as this model maximized adjusted R2 and the predicted R2 values.

ANOVA analysis

Analysis of variance (ANOVA) was performed to assess the significance of the quadratic polynomial models developed. The regression coefficients and p-values obtained are given in table 4. The small p-value (p<0.05) of the terms in the models indicated a significant influence on the response variables.

Sufficiency of models

Diagnostic plots (such as predicted vs. actual) are useful graphic tools to observe the adequacy of developed models. It shows the relationship between predicted and experimental values and describes model suitability. Table 5 shows the predicted and experimental values of disintegration time and in vitro drug release of the films and fig. 2a and 2b show the predicted vs. actual plots for all responses. The closer the points are to the diagonal line, the better the model fits the experimental data [22].

Table 5: Predicted and experimental values of disintegration time and in vitro drug release of the films

Batch Independent variables Response variables
X1 X2 Y1 Y2
Actual Predicted Actual
F1 3 8 27.53 27.02 82.60
F2 4 11 39.20 37.82 80.80
F3 3 14 28.64 28.30 83.90
F4 5 8 62.14 63.84 68.20
F5 4 6.75736 50.60 50.81 74.40
F6 4 11 37.38 37.82 78.80
F7 2.58579 11 19.31 19.37 84.40
F8 4 15.2426 34.42 35.10 79.00
F9 4 11 36.34 37.82 80.20
F10 4 11 37.61 37.82 80.50
F11 5 14 39.47 38.58 69.90
F12 4 11 38.56 37.82 77.80
F13 5.41421 11 49.88 50.72 66.70

Fig. 2: Predicted and experimental values of a) disintegration time and b) drug release

Effect of factors on responses

The 3D response surface plots were used to study the effects of independent variables on responses. These plots are known to study the interaction effects of the factors on the responses [23]. The 3D graphs for both responses are given in fig. 3a and 3b

Effect on disintegration time (Y1)

The following polynomial equation was proposed for the disintegration time of the FDFs:

37.818+11.0841X1-5.55525 X2-5.945X1 X2-1.38838X12+2.56912 X22

The Model F-value of 275.76 implies the model is significant. P-values less than 0.05 indicate model terms are significant. In this case X1, X2, X1X2, X1², X22 are significant model terms. The Lack of Fit F-value of 0.66 implies the Lack of Fit is not significant.

Fig. 3a shows the response surface plot of the effect of different independent variables on the disintegration time of the FDFs. From the figure, it can be seen that the disintegration time (Y1) was prolonged with the increase in the concentration of film-forming polymer, HPMC E5 (X1)because, with an increase in the amount used in the formulation, the viscous gel produced after the film’s swelling in water and subsequent retardation of penetration of water resulted in the delay of disintegration. With the increase of the super disintegrant (CCS) concentration, there were changing tendencies in the disintegration time which first descended and then ascended. The reasons might be that CCS was swelled in water due to its good water uptake ability, which is beneficial to disintegration, but an excess quantity would lead to high viscosity with swelling, which would suppress the water penetration.

Effect on in vitro drug release at 5 min (Y2)

The polynomial equation proposed for the % in vitro drug release at 5 min was:

79.62-6.67895X1+1.18817X2+0.1X1X2-2.02875X12-1.45375X22

The Model F-value was 60.31 implies that the model is significant. In this case, the significant model terms are X1, X2, X1², and X22and their P-values were less than 0.05. The Lack of Fit F-value of 0.61 implies the Lack of Fit is not significant.

Fig. 3b is the 3-D surface plot of the effect of the two independent variables on the cumulative % release of the drug from the films within 5 min. Response surface 3D plot reveals that in vitro drug release at 5 min (Y2) significantly decreases with the increase in HPMC (X1) concentration. On contact with the release medium, the viscous nature of the film-forming polymer (HPMC) creates thick matrix gel fluid, which governs drug release by resisting drug diffusion or matrix erosion and may lead to a delay in the drug release from the films. Another reason contributing to decreased drug release at increased polymer concentration may be the increase in the time required for wetting and dissolving the drug molecules present in the polymer matrices [15].

Fig. 3: 3D response surface plots of FDFS for the effect of independent variables on a) disintegration time and b) drug release at 5 min

Selection of optimum formulation

The optimized formulation was selected with the following criteria: minimum disintegration time and the maximum cumulative drug release of the FDFs and from among the independent variables’ concentration solutions suggested by the software, the following composition was selected for the preparation of the optimal formulation: 3.016% w/v of HPMC E5 (X1) and 11.731%w/w CCS (X2) with the desirability of 1. With these values, 25.79 sec for disintegration time (Y1) and 84.41% for in vitro drug release at 5 min (Y2) were the responses predicted.

Validation of optimum formulation

To validate these predicted values generated according to the results of the CCD design, the optimum formulation batches were prepared according to the optimized concentrations of the factors HPMC E5 and CCS and subjected to the disintegration and drug release tests. The data means of 27.7 sec for disintegration time and 83.13% for in vitro drug release at 5 min were obtained. These differences in the film’s responses were not significantly deviated from the predicted responses of the optimized formula (table 6). From the confirmational analysis, it was shown that the above data means were within the range of 95% low and high prediction intervals. The results indicate the effective use of DOE for investigating variables’ effects and optimization.

DSC analysis of optimized FDF

DSC thermograms of pure tacrolimus and optimized film are shown in fig. 4. DSC of pure tacrolimus showed a broad endothermic event attributed to dehydration happening at temperatures between 75 °C and 122 °C followed by melting of the anhydrate form with a sharp endothermic peak at 129.46 °C which corresponds to its melting point and indicating its crystalline nature. The optimized film formula showed the disappearance of the characteristic peak of tacrolimus which might be due to the homogenous dispersion of the drug in the formed film.

Table 6: Comparison of predicted and observed response values of FDFs prepared at optimal conditions

Response Predicted values Observed values Prediction error (%)
Disintegration time (sec) 25.79 27.7 -6.9
In vitro drug release at 5 min (%) 84.41 83.13 1.54

Fig. 4: DSC thermogram of a) tacrolimus monohydrate b) optimized film

XRD

To obtain further information on the solid-state changes, XRD spectra were carried out on the optimized FDF formulation. The diffractograms of the drug and the optimized films are shown in fig. 5. The presence of numerous distinct peaks in the spectrum of tacrolimus indicates that the drug is present as a crystalline material. The FDF formulation revealed a decrease in the intensity of diffraction peaks, indicating that the drug completely dispersed in the film matrix. This corresponds with the results obtained from DSC testing. Apart from the reduction of peak intensities, they were significantly broadened, which indicated that due to the inclusion complex formation, the drug was converted to a partial amorphous form.

Fig. 5: XRD pattern of a) tacrolimus monohydrate b) optimized film

SEM

The SEM image of the pure drug as well as the morphology of the optimized FDF formula, is given in fig. 6a and 6b. From fig. 6a, it was found that tacrolimus occurred as cuboidal-shaped crystals, and the SEM image of the film revealed a smooth, slightly porous surface, indicating complete and uniform imgment of the drug: β-cyclodextrin complexes in the polymeric film matrix. The porous nature indicates a uniform distribution of the drug complexes that help the medium to get penetrate inside the film. Very few drug crystals were found in the film, probably formed during the drying of the film.

Fig. 6: Scanning electron micrograph of a) tacrolimus monohydrate, b) optimized film (magnification 2700×)

CONCLUSION

In the present work, FDF formulations containing tacrolimus-β-cyclodextrin inclusion complexes for sublingual application were developed and optimized employing the central composite statistical design. The multiple regression analysis of the results provided equations describing the influence of the selected variables on the responses under study. The desirability function led to the optimum values of the factors at which the prepared film showed minimum disintegration time and maximum drug release. DSC evaluation suggested the homogenous dispersion of the drug in the FDFs. The XRD analysis suggested that the drug in the FDFS was in a partial amorphous state. SEM analysis revealed the smooth and porous nature of the film. Accordingly, it could be concluded that FDFs, optimized using CCD, may provide a novel sublingual delivery system of tacrolimus for avoiding its oral bioavailability problems.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICT OF INTERESTS

The authors declare no conflict of interest.

REFERENCES

  1. Dharmasthala S, Shabaraya AR, Andrade GS, Shriram RG, Hebbar S, Dubey A. Fast dissolving oral film of piroxicam: solubility enhancement by forming an inclusion complex with β-cyclodextrin, formulation and evaluation. J Young Pharm. 2018;11(1):1-6. doi: 10.5530/jyp.2019.11.1.

  2. Londhe V, Shirsat R. Formulation and characterization of fast-dissolving sublingual film of iloperidone using box–behnken design for enhancement of oral bioavailability. AAPS PharmSciTech. 2018;19(3):1392-400. doi: 10.1208/s12249-018-0954-y, PMID 29396734.

  3. Suresh BS, Hemraj MT. Design and statistical optimization of mouth dissolving sublingual film of fixed dose combination of doxylamine succinate and pyridoxine hydrochloride using design of experiment in the treatment of nausea and vomiting in pregnancy. Asian J Pharm Clin Res. 2018;11(12). doi: 10.22159/ajpcr.2018.v11i12.27978.

  4. Panraksa P, Tipduangta P, Jantanasakulwong K, Jantrawut P. Formulation of orally disintegrating films as an amorphous solid solution of a poorly water-soluble drug. Membranes. 2020;10(12):1-17. doi: 10.3390/membranes10120376, PMID 33261025.

  5. Mashru RC, Sutariya VB, Sankalia MG, Parikh PP. Development and evaluation of fast-dissolving film of salbutamol sulphate. Drug Dev Ind Pharm. 2005;31(1):25-34. doi: 10.1081/ddc-43947, PMID 15704855.

  6. PH, Fatima S. Formulation and evaluation of oral fast dissolving films of naproxen sodium. Int J Curr Pharm Sci. 2022;14:48-53. doi: 10.22159/ijcpr.2022v14i2.1953.

  7. Watkins KD, Boettger RF, Hanger KM, Leard LE, Golden JA, Hoopes CW. Use of sublingual tacrolimus in lung transplant recipients. J Heart Lung Transplant. 2012;31(2):127-32. doi: 10.1016/j.healun.2011.10.015. PMID 22177691.

  8. Nasiri Toosi Z, Dashti Khavidaki S, Nasiri Toosi M, Khalili H, Jafarian A, Irajian H. Clinical pharmacokinetics of oral versus sublingual administration of tacrolimus in adult liver transplant recipients. Exp Clin Transplant. 2012;10(6):586-91. doi: 10.6002/ect.2012.0032, PMID 22770208.

  9. Solari S, Cancino A, Wolff R, Norero B, Vargas JI, Barrera F. Sublingual tacrolimus administration provides similar drug exposure to per-oral route employing lower doses in liver transplantation: a pilot study. Aliment Pharmacol Ther. 2017;45(9):1225-31. doi: 10.1111/apt.14022. PMID 28261844.

  10. Aziz DE, Abdelbary AA, Elassasy AI. Implementing central composite design for developing transdermal diacerein-loaded niosomes: ex vivo permeation and in vivo deposition. Curr Drug Deliv. 2018;15(9):1330-42. doi: 10.2174/1567201815666180619105419, PMID 29921206.

  11. Mohanan J, Palanichamy S, Kuttalingam A, Narayanasamy D. Development and characterization of tacrolimus tablet formulations for sublingual administration. Int J App Pharm. 2021;13(6):89-97. doi: 10.22159/ijap.2021v13i6.42429.

  12. Jadhav YG, Galgatte UC, Chaudhari PD. Overcoming poor solubility of dimenhydrinate: development, optimization and evaluation of fast dissolving oral film. Adv Pharm Bull. 2018;8(4):721-5. doi: 10.15171/apb.2018.081, PMID 30607345.

  13. Haju SS, Yadav S. Formulation and evaluation of cilnidipine mucoadhesive buccal film by solvent casting technique for the treatment of hypertension. Int J Pharm Pharm Sci. 2021;13(9):34-43. doi: 10.22159/ijpps.2021v13i9.42641.

  14. Allam A, Fetih G. Sublingual fast dissolving niosomal films for enhanced bioavailability and prolonged effect of metoprolol tartrate. Drug Des Devel Ther. 2016;10:2421-33. doi: 10.2147/DDDT.S113775. PMID 27536063.

  15. Al-Mogherah AI, Ibrahim MA, Hassan MA. Optimization and evaluation of venlafaxine hydrochloride fast dissolving oral films. Saudi Pharm J. 2020;28(11):1374-82. doi: 10.1016/j.jsps.2020.09.001. PMID 33250644.

  16. Senthilkumar K, Vijaya C. Formulation development of mouth dissolving film of etoricoxib for pain management. Advances in Pharmaceutics. 2015;2015:1-11. doi: 10.1155/2015/702963.

  17. Abouhussein DMN, El-Bary AA, Shalaby SH, El Nabarawi MAE. Chitosan mucoadhesive buccal films: effect of different casting solvents on their physicochemical properties. Int J Pharm Pharm Sci. 2016;8(9):206-13. doi: 10.22159/ijpps.2016.v8i9.12999.

  18. The United States pharmacopoeia. The United States of America Pharmacopoeial Convention; 2013. p. 3-5.

  19. Brniak W, Maslak E, Jachowicz R. Orodispersible films and tablets with prednisolone microparticles. Eur J Pharm Sci. 2015;75:81-90. doi: 10.1016/j.ejps.2015.04.006. PMID 25889975.

  20. Bhargav E, Prakash Reddy CS, Sowmya C, Haranath C, Ahmed Khan KA, Rajesh K. Formulation and optimization of piroxicam orodispersible tablets by central composite design. J Young Pharm. 2017;9(2):187-91. doi: 10.5530/jyp.2017.9.37.

  21. Chaudhary H, Gauri S, Rathee P, Kumar V. Development and optimization of fast dissolving oro-dispersible films of granisetron HCl using box-behnken statistical design. Bull Fac Pharm Cairo Univ. 2013;51(2):193-201. doi: 10.1016/j.bfopcu.2013.05.002.

  22. Rezaee F, Ganji F. Formulation, characterization, and optimization of captopril fast-dissolving oral films. AAPS PharmSciTech. 2018;19(5):2203-12. doi: 10.1208/s12249-018-1027-y, PMID 29728997.

  23. Shen C, Shen B, Xu H, Bai J, Dai L, Lv Q. Formulation and optimization of a novel oral fast dissolving film containing drug nanoparticles by box–behnken design–response surface methodology. Drug Dev Ind Pharm. 2014;40(5):649-56. doi: 10.3109/03639045.2014.884116, PMID 24506458.