Pharmacophore an International Research Journal
Pharmacophore
Submit Manuscript
Open Access | Published: 2021 - Issue 1

ESTIMATION OF QUALITY CHARACTERISTICS FOR SUSTAINED RELEASING AND ACTING FORMULATION OF DOMPERIDONE

Zeb-un-Nisa1, Syed Imran Ali1*, Saira Shahnaz2, Tayyaba Mumtaz3, Muhammad Mustafa Swaleh1

 

  1. Department of Pharmaceutics, Faculty of Pharmacy, Ziauddin University, Karachi, Sindh, Pakistan.
  2. Department of Pharmacy Practice, Faculty of Pharmacy, Nazeer Hussain University, Karachi, Sindh, Pakistan.
  3. Department of Pharmacognosy, Faculty of Pharmacy, Ziauddin University, Karachi, Sindh, Pakistan.

ABSTRACT

The present study aimed to design a sustained releasing and acting formulation of Domperidone with the aid of hydrophilic Methocel® K4M. Domperidone is an antiemetic drug used to treat nausea and vomiting. Generally, the dosing frequency is twice or thrice daily. Sustained- release formulation was prepared with different ratios of K4M. Furthermore, its quality and stability were determined. Pre-compression and post-compression characteristics were examined and the optimized formulation was selected for dissolution studies and also for similarity profiles. Drug dissolution studies revealed formulated tablet containing CF2 depicted the ideal release profile for once-daily consumption. Long-acting domperidone matrices were not just prepared but also displayed desirable properties required for an ideal once-daily formulation. The mechanism might be gel layer formation which afterward proceeded by gel erosion, hence giving a long slow-releasing pattern and desirable therapeutic effect. The concept of controlled release formulations helps the patients and care givers by reducing dosing frequency and chances of missed doses or overnight gap in administration of dose.

Keywords: Domperidone, Methocel®, Ethocel®, Long-acting, Erosion


Introduction

Domperidone blocks the dopamine receptors. It increases the peristaltic movement of gastrointestinal, resulting in the release of prolactin, it is an antiemetic agent and it paves a way for the study of different dopaminergic mechanisms [1].

Domperidone helps in GI emptying and also a stimulant of peristalsis. These characteristics are related to its peripheral dopamine receptor blocking properties. The drug enhances GI motility. On the contrary, it slows transit time and elevates the peristalsis of the esophagus and GIT. It also decreases the pressure of the esophageal sphincter [2-4]. The emesis controlling properties of domperidone is because it blocks the dopamine receptor. Both at the chemoreceptor trigger zone and also at the GI level. It bears bonding properties for the dopamine receptors (D2 &D3), lie within the chemoreceptor trigger zone, found near the blood-brain barrier, responsible forcontroling nausea [5, 6]. 

Domperidone is (5-chloro-1-{1-[3-(2, 3-dihydro-2-oxo-1H-benzimidazol-1-yl) propyl] -4- piperidinyl} benzimidazolin-2-one) [7]. It is low soluble and highly permeable drug with a half-life of 7 h[1].                      

The drugs which are to be taken several times in a day, such medicines could be better delivered if they modified in the once-daily formulation [8, 9]. These formulations have great benefits over conventional ones, such as lesser doses, coverage of overnight no dose period, and steady-state plasma concentration [8, 10, 11]. Controlled release dosage forms could be formulated in various ways and they follow different mechanisms such as diffusion, dissolution, erosion, ion exchange, and osmotically controlled systems. These systems mechanically control drug release in a précise and predetermined manner to maintain drug plasma levels for a prolonged period [11]. The matrix-based systems are most commonly used due to their efficacy and convenience [12]. 

The methocel® is white or off white powder [13]. It is inert and gives good control through gel formation and erosion [14].

 

Hydroxypropyl Methylcellulose - an overview | ScienceDirect Topics

Figure 1. Hydroxypropyl methylcellulose, R = H, −CH3 or - (OCH2CHCH3)xOH. [15]

 

Sol-Gel Behavior of Hydroxypropyl Methylcellulose (HPMC) in Ionic Media  Including Drug Release. - Abstract - Europe PMC

a)

b)

c)

d)

Figure 2. Physical structures of HPMC hydrogels (a), (b) at lower temperatures and (c), (d) at higher temperatures) [16]

These polymers can control the release of the active pharmaceutical ingredient [17, 18]. Methocel® and Ethocel® (EC) are effective polymers for formulating matrices. Besides this, the amount and polymer grade control the release profile. HPMC delays release due to gel formation upon contact with the surrounding medium followed by erosion [19]. The chemical and physical structures of the HPMC polymer are shown in Figures 1 and 2 respectively. Hydrophobic polymers slow the release due to their hydrophobicity which distracts the fluids and wettability. The rate of drug dissolution is calculated by the amount of dissolved drug during the time. Dissolution profiles can be examined with model-independent and model-dependent approaches [20-23]. The data shown by model parameters replicated the dissolution pattern of the drug [24-29]. When the dissolution profile at different time intervals is obtained the model-independent approach could be used. The method predicts one differentiation factor (f1), the second one is a similarity factor (f2) [30].

Materials and Methods

Chemicals

Domperidone was gifted from Medisure Pharmaceuticals, Pakistan, K4M (Methocel®), (Colorcon LTD Kent, England), Magnesium stearate, Avicel PH-10, Methanol, Potassium dihydrogen phosphate, Disodium hydrogen phosphate were purchased from Life Science, Germany.

Instruments

Electronic balance (Shimadzu, Japan), Single punch compression machine (Shanghai, China),  Vernier caliper (China), hardness tester (Fujiwara, Japan), friabilator (Curio, Pakistan), FT-IR spectrometer (Germany), disintegration tester (Germany), dissolution tester (Erweka, Germany), UV spectrophotometer (UV-1800, Shimadzu, Japan), HPLC system pump (SPD-10AVP CBM 102, Shimadzu), column (Bondapak C-18 4.6 × 250 mm 10 μm Germany), ultrasonic bath (Germany), filter assembly (Millipore, England) and microliter syringe (Switzerland). pH meter, membrane filter (USA), and vacuum pump (China) were utilized.

Softwares

Excel plugin software DDSolver, was applied for the analysis of dissolution models; MS 

Excel® was applied to estimate before-and-after-compression data [31].

Methodology

Micromeritic Evaluation of Blends

Micromeritic characteristics of powder blends were estimated through official methods. The following equations (1)–(5) were applied to estimate Bulk density, Tapped density, angle of repose, Hausner’s ratio, and Carr’s index  respectively [32]:

Bulk density = M‑ /Vbulk

(1)

Tapped density = M/‑Vtapped

(2)

tan (𝜃) = height∕0.5base

(3)

Hausner ratio =(Vo∕Vf)=(𝜌tapped∕𝜌bulk)

(4)

Carr’s index = 100 ×[(𝜌tapped − 𝜌bulk)∕𝜌tapped]

(5)

Where, Weight in grams (g) is denoted by M, the Powder volumes before tapping and after tapping are denoted by Vbulk and Vtapped in mL, and bulk and tapped densities are Pbulk and Ptapped, respectively [32].

 

Preparation of tablets

Tablet formulation blends were prepared by blending HPMC K4M in various proportions with Domperidone and Avicel PH 101 in a polybag. Magnesium stearate was added in last, mixed and compressed directly on a single punch compression machine. The final weight of the tablet was 150 mg. Table 1 shows the composition of tablets.

Table 1. Domperidone matrices

Formulation code

Ingredients percentage (%)

Active ingredient

Matrix former

Diluent

Lubricant

Domperidone

Methocel

K4M

Avicel

PH 101

Magnesium

Stearate

CF-1

23.33

20

55.33

1.3

CF-2

23.33

30

45.33

1.3

CF-3

23,33

40

35.33

1.3

CF-4

23.33

50

25.33

1.3

 

FT-IR Analysis

FT-IR analysis of domperidone drug and controlled release formulation was performed on FT-IR Spectrometer by ATR technique.

 

Scanning Electron Microscopy (SEM)

Morphological properties of optimized formulation (CF2) was evaluated through a scanning electron microscope, SEM (Japan). 

Assay of Domperidone

The domperidone content in matrices was determined by the following method [32].

Chromatographic Conditions

The mobile phase comprised of phosphate buffer and methanol (30:70v/v) and was filtered and degassed before use through a 0.2µ membrane filter. The flow rate was set at a rate of 1.0 mL/min and the wavelength was adjusted at 280 nm by a UV detector at a sensitivity of 0.0001.

Preparation of Standard Solution

The standard solution was prepared by dissolving 10mg of domperidone in 50 ml mobile phase then transfer 10ml of this solution to 100ml volumetric flask and this solution was diluted with mobile phase to obtain a solution of 20 mcg/mL.

Preparation of Sample Solution

Accurately weighed twenty tablets, which were randomly selected to obtained their average weight, then crushed them to powder. Equivalent to 10 mg of domperidone, powder of tablets was accurately weighed and taken into a volumetric flask of 50 ml containing mobile phase and shacked for about 15minutes then filtered through Whatman filter paper. The filtered solution was further diluted with mobile phase to obtain a final concentration of 20 mcg/ml.

Quality Characteristics of Formulated Tablets

Domperidone formulated tablets were assessed for prerequisite quality parameters to establish and assess weight uniformity, crushing strength, friability, disintegration time, assay, and dissolution [33]. 

Weight Uniformity

 Each tablet of the randomly selected sample was weighed individually using an analytical balance. Mean and the standard deviation was calculated on MS Excel®. 

 

Crushing Strength: The crushing strength of a randomly selected sample from each formulated experimental batch was examined and noted carefully. Mean and standard deviation was calculated on MS Excel®. 

Friability: Randomly selected tablets were weighed and subjected to friability test after test reweighed and friability was calculated according to the following equation:

 

(6)

(Where W1& W2 are the initial and final weight of tablets, respectively)

Disintegration Test: The test was conducted according to USP specifications at 37 ±2 °C in 900 mL distilled water until tablets disintegrated [33].

 

Swelling Studies

A beaker containing 250 mL distilled water was taken and a single tablet from each lot was taken and immersed in it for 10 hours at ambient conditions. The swollen tablet was reweighed after every hour. The swelling ratio was calculated by using the equation:

 

(7)

 (Weight of tablet before and after swelling denoted by W1 and W2) [34].

 

In-vitro Dissolution Studies

In vitro drug release was performed by using USP dissolution apparatus type-II (Paddle apparatus) at the rotation of 50rpm and the temperature of dissolution medium was maintained at 37±0.5°C. 900ml of Phosphate buffer pH 6.8 was used as dissolution medium and the samples were analyzed for 24 hours. The drug release was evaluated by taking a sample of 5 ml (which was replaced with fresh dissolution medium) at a predetermined time interval of 30min, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 18 and 24 hour. The sample solution was filtrated through a 0.45-μm Whatman filter and absorbance was analyzed at 384nm using UV Spectrophotometer. Cumulative drug release percentage was computed and the mean of six tablets was included in the result [35].

Dissolution Profiles Comparison

The Model-independent method was used for the similarity factor (f2). The method had been applied for comparison of the release kinetics for produced formulations (Eq. 8).

f2 is the similarity factor is the logarithmic reciprocal square root transformation of the sum of squared error. It is a determination of similarity in % age of dissolution amid two curves. If the reading is between 50 and 100, the release is determined as similar. A decreasing value of f2 is indicative of dissimilar dissolution kinetics [35].

 

(8)

 

Where, Ti denotes the % of drug under test, reference drug % is denoted by Ri, number of total samples is represented by N.

Results and Discussion

The results of pre-compression characteristics of formulation blends are given in Table 2.

Table 2. Pre compression characteristics of domperidone blends

Formulation code

Mass

Bulk volume

Tapped volume

Bulk density

Tapped density

Hausner ratio

Carr’s index

Angle of repose

Flow properties

(g)

(mL)

(mL)

(g/mL)

(g/mL)

 

(%)

(θ)

(USP35-NF30)

CF-1

10

19

16

0.53

0.63

1.19

15.7

36.3

Fair

CF-2

10

19

17

0.52

0.58

1,11

10.3

33.2

Good

CF-3

10

17

14

0.58

0.71

1.22

18.30

44.1

Passable

CF-4

10

20

18

0.50

0.55

1.1

9.09

33.12

Good

FT-IR spectrum of domperidone was analyzed and the spectrum shows all characteristic peaks. The post-compression physical parameters and assay results of formulations are given in Table 3. Results of weight uniformity were in the range of 149.78±1.82 to 153.21±1.99 mg and were according to USP's acceptable range of variation of ±5 mg. All formulations displayed friability within limits (less than 1%). Disintegration time was in the range of 4.67 to 6.89 hours and the crushing strength of tablets was in the range of 8.91±1.33 to 12.82±1.68Kg. Assay results showed that the drug concentration of formulations lied between 98.21 and 101.74%. 

Table 3. Physical parameters and assay of domperidone matrices

Formulation code

Weight

Hardness

Friability

Disintegration Time

Assay

(mg)

(kg)

(%)

(h)

(%)

CF1

151.67±1.36

8.91±1.33

0.89

4.67

99.34

CF2

150.64±.1.53

12.82±1.68

0.57

6.21

101.76

CF3

149.78±1.82

11.45±1.45

0.78

6.89

98.66

CF4

153.21±1.99

11.23±1.76

0.23

5.66

98.21

The results of the assay complied with USP specifications i.e. 95-105% [USP].

Swelling Studies

Swelling studies were conducted to estimate the tendency of swelling in HPMC matrix formulations. The Swelling behavior of formulated tablets is indicative of the release behavior of tablets. The hydration of domperidone formulations showed that               swelling percentage increase with ascending concentrations of methocel® in formulations. The polymer concentration decides the fate of release because the more the concentration of polymer the lesser the drug release.

 

Scanning Electron Microscopy (SEM)

The SEM of the optimized formulation showed uneven surface and grooves as showed in Figure 3.

Figure 3. SEM of optimized formulation CF2

Dissolution Studies

The drug release pattern from CR formulations was studied at multiple time point intervals. Release profiles were studied in pH 1.2, 4.5, and 6.8 as shown in Figure 4. Release profile of CF2(30% polymer) was 19% at 4 h, 73 % at 16 h, 80% at 18 h and 91% at 24 h). An increased concentration of polymer resulted in adequate release control. A greater concentration of the polymer is responsible for gel layer formation and then erosion. Such behavior of polymer was also seen by other researchers for CR tablets. The release profiles are shown in Figure 4. The release pattern followed the zero-order kinetics. The current study shows the release profiles of two formulations CF2 and CF3were similar while the remaining were dissimilar as shown in Figure 4.

 

Figure 4. Release profile of  domperidone controlled release matrix tablets

Conclusion

A sustained releasing and acting formulation of domperidone had formulated and estimated for various quality control tests, assay, and dissolution. The findings revealed that the formulation containing methocel® K4M 30% gives the required drug release pattern and pace due to higher viscosity grade from excess entanglement of a polymer. The optimized formulation can be used for nausea and vomiting as a once-daily formulation. It is also cost-effective and would have achieved patient compliance. 

Acknowledgments: We would like to thanks Medisure Pharmaceuticals, Pakistan for providing us the Domperidone for Product development and analysis.

Conflict of interest: None

Financial support: None

Ethics statement: In-vitro testing of the formulated tablets was performed and no animal or human testing was conducted. Ethics statement is not required for such analysis. 

 

[1] DrugBank.Com 2020

References

1.        Cavallotti C, Nuti F, Bruzzone P, Mancone M. Age-related changes in dopamine D2 receptors in rat heart and coronary vessels. Clin Exp Pharmacol Physiol. 2002;29(5-6):412-8.

2.        Osinski MA, Uchic ME, Seifert T, Shaughnessy TK, Miller LN, Nakane M, et al. Dopamine D2, but not D4, receptor agonists are emetogenic in ferrets. Pharmacol Biochem Behav. 2005;81(1):211-9.

3.        de Mey C, Enterling D, Meineke I, Yeulet S. Interactions between domperidone and ropinirole, a novel dopamine D2-receptor agonist. Br Clin Pharmacol. 1991;32(4):483-8.

4.        Chen X, Ji ZL, Chen YZ. TTD: Therapeutic Target Database. Nucleic Acids Res. 2002;30(1):412-5.

5.        Ali I, Gupta VK, Singh P, Pant HV. Screening of domperidone in wastewater by high performance liquid chromatography and solid phase extraction methods. Talanta. 2006;68(3):928-31. doi:10.1016/j.talanta.2005.06.027

6.        Martindale WH. The Extra Pharmacopoeia, 36th ed. The Royal Pharmaceutical Society, London; 2009. p.1726.

7.        National Center for Biotechnology Information. PubChem Compound Summary for CID 46878575. Retrieved June 23, 2021. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/46878575.

8.        Vyas SP, Khar RK. Controlled drug delivery concepts and advances. Vallabh Prakashan. 2002;1:411-47.

9.        Subash Chandran MP, Prasobh GR, Jaghatha T, Aswathy BS, Remya SB. An Overview on Liposomal Drug Delivery System. Int J Pharm Phytopharmacol Res. 2019;9(2):61-8.

10.     Al Zahrani S, Eid Alosaimi M, Alamrim AA, Alotaibi M, Almatar EA, Almanea BA. Association between knowledge and drug adherence in patients with hypertension in Saudi Arabia. Arch Pharm Pract. 2019;10(3):71-6.

11.     Shah SU, Shah KU, Rehman A, Khan GM. Investigating the in vitro drug release kinetics from controlled release diclofenac potassium-ethocel matrix tablets and the influence of co-excipients on drug release patterns. Pak J Pharm Sci. 2011;24(2):183-92.

12.     Kanakagiri D, Omprakash H, Kumar KM, Vishwanadham Y. Formulation and evaluation of pantoprazole delayed release tablets using Eudragit L30D55. Res J Pharm Technol. 2017;10(2):421-5.

13.     Siepmann J, Peppas N. Modeling of drug release from delivery systems based on hydroxypropyl methylcellulose (HPMC). Adv Drug Deliv Rev. 2012;64:163-74.

14.     Chaerunisaa AY, Ali R, Dashevskiy A. Release adjustment of two drugs with different solubility combined in a matrix tablet. AAPS Pharm Sci Tech. 2019;20(4):142.

15.     Deshmukh K, Ahamed MB, Deshmukh RR, Pasha SK, Bhagat PR, Chidambaram K. Biopolymer composites with high dielectric performance: interface engineering. In Biopolymer composites in electronics. Elsevier. 2017;1:27-128.

16.     Joshi SC. Sol-gel behavior of hydroxypropyl methylcellulose (HPMC) in ionic media including drug release. Mater. 2011;4(10):1861-905.

17.     Aulton ME, Taylor KM, editors. Aulton's Pharmaceutics E-Book: The Design and Manufacture of Medicines. Elsevier Health Sciences; 2017.

18.     Klančar U, Baumgartner S, Legen I, Smrdel P, Kampuš NJ, Krajcar D, et al. Determining the polymer threshold amount for achieving robust drug release from HPMC and HPC matrix tablets containing a high-dose BCS class I model drug: in vitro and in vivo studies. AAPS Pharm Sci Tech. 2015;16(2):398-406.

19.     Bae Y. Drug delivery systems using polymer nanoassemblies for cancer treatment. Ther Deliv. 2010;1(3):361-3.

20.     Ritger PL, Peppas NA. A simple equation for description of solute release I. Fickian and non-fickian release from non-swellable devices in the form of slabs, spheres, cylinders or discs. J Control Release. 1987;5(1):23-36.

21.     Fotaki N, Aivaliotis A, Butler J, Dressman J, Fischbach M, Hempenstall J, et al. A comparative study of different release apparatus in generating in vitro–in vivo correlations for extended release formulations. Eur J Pharm Biopharm. 2009;73(1):115-20.

22.     Kostewicz ES, Brauns U, Becker R, Dressman JB. Forecasting the oral absorption behavior of poorly soluble weak bases using solubility and dissolution studies in biorelevant media. Pharm Res. 2002;19(3):345-9.

23.     Kostewicz ES, Abrahamsson B, Brewster M, Brouwers J, Butler J, Carlert S, et al. In vitro models for the prediction of in vivo performance of oral dosage forms. Eur J Pharm Sci. 2014;57:342-66.

24.     Murari K, Mishra GA. U. Development of Sustained Release Floating Tablet for Cefpodoxime Proxetil (CP). Int J Pharm Phytopharmacol Res. 2019;9(2):96-105.

25.     Arti M, Ashwini A. Formulation and evaluation of Nitazoxanide sustained-release matrix tablets. Int J Pharm Phytopharmacol Res. 2019;9(3):153-61.

26.     Mastropietro D, Park K, Omidian H. Polymers in oral drug delivery. Comprehensive Biomaterials II, Elsevier. 2017:430-44.

27.     Ford JL. Design and evaluation of hydroxypropyl methylcellulose matrix tablets for oral controlled release: a historical perspective. InHydrophilic matrix tablets for oral controlled release. Springer, New York, NY. 2014:17-51.

28.     Krese A, Kovačič NN, Kapele T, Mrhar A, Bogataj M. Influence of ionic strength and HPMC viscosity grade on drug release and swelling behavior of HPMC matrix tablets. J Appl Polym Sci. 2016;133(26).

29.     Saeidipour F, Mansourpour Z, Mortazavian E, Rafiee-Tehrani N, Rafiee-Tehrani M. New comprehensive mathematical model for HPMC-MCC based matrices to design oral controlled release systems. Eur J Pharm Biopharm. 2017;121:61-72.

30.     Reddy BB, Nagoji KE, Sahoo S. Preparation and in vitro & in vivo evaluation of cephalexin matrix tablets. Braz J Pharm Sci. 2018;54(3):e17277.

31.     The United States Pharmacopeia USP 35/The National Formulary, NF 30. US Pharmacopeial Convention, Rockville, MD 2012.

32.     Saurí J, Millán D, Suñé-Negre JM, Colom H, Ticó JR, Miñarro M, et al. Quality by design approach to understand the physicochemical phenomena involved in controlled release of captopril SR matrix tablets. Int J Pharm. 2014;477(1-2):431-41.

33.     Almutairi FM. Biopolymer nanoparticles: a review of prospects for application as carrier for therapeutics and diagnostics. Int J Pharm Res Allied Sci. 2019;8(1):25-35.

34.     EL Korso FN, Sebba FZ, Rechache M. Study inhibition of Armco iron corrosion by some polymers based on poly (4-Vinyl Pyridine) (PVP) in 0.5M Sulfuric Acid medium. World J Environ Biosc. 2019;8(1):37-45.

35.     Khan A, Iqbal Z, Khan A, Mugha MA, Khan A, Ullah Z, et al. Modulation of pH-independent release of a class ΙΙ drug (domperidone) from a polymeric matrix using acidic excipients. Dissolut Technol. 2016;23(1):32-40.

QR code:

Short Link:
Quick Access

Pharmacophore
ISSN: 2229-5402

Pharmacophore
© 2024 All rights reserved
Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.