Formulation and Evaluation of Cinitapride Controlled Release Tablets

 

Mercy Mathew1, Ravikumar2*, Simila Madathil1, Anju Govind1, Narayana Swamy VB3

1M. Pharm (Pharmaceutics) Research Scholar, Karavali College of Pharmacy, Mangalore.

2Department of Pharmaceutics, Karavali College of Pharmacy, Mangalore.

3Department of Pharmacognosy, Karavali College of Pharmacy, Mangalore.

*Corresponding Author E-mail: ravikumar300@gmail.com

 

ABSTRACT:

The present study aimed at Formulation Development and Evaluation of controlled release tablets of Cinitapride for the treatment of ulcer. Cinitapride is a gastroprokinetic agent and antiulcer agent of the benzamide class. It act as an agonist of the 5- HT1 and  5- HT4  receptors and an antagonist of the 5- HT2  receptors. It is used in the treatment of gastrointestinal disorders associated with motility disturbances such as gastro esophageal reflux disease, non- ulcer dyspepsia and delayed gastric emptying. The matrix tablets of Cinitapride were prepared using wet granulation. Physical characterization of tablet and powder blends used to form the matrix tablet was under taken using a range of experimental techniques. Granules were evaluated for Bulk density, Tapped density, Compressibility index and Hausner’s ratio. Tablets were tested for weight variation, hardness, thickness and friability as per official procedure. The tablets were evaluated for in-vitro drug release profile. Dissolution studies of Cinitapride controlled release tablets in media with different dissolution media 0.1N HCl, Phosphate buffer pH (6.8) as per US Pharmacopoeia.  The dissolution data revealed that the ratio of polymers is very important to achieve a optimum formulation.  The formulation of Cinitapride CR tablets shown that formulation F23 with Methocel K100M (20%) shown good drug release profile. Formulation F23, shown similar dissolution profile when compared with the marketed product (Cintapro). Stability study of the formulation F23 indicated no significant difference in release profile after a period of 3 months.

 

KEY WORDS: Cinitapride, Gastritis, Methocel K4M, K15M and K100M, Carbopol, and Methyl cellulose.

 

 


INTRODUCTION:

Oral route is the most preferred route for  administration of controlled delivery of drugs because of convenience and ease of administration, greater flexibility in dosage form design and ease of production and low cost of such a system1.

 

Pharmaceutical products designed for oral delivery are mainly immediate release type or conventional drug delivery systems, which are designed for immediate release of drug for rapid absorption2. In order to overcome the drawbacks of conventional drug delivery systems, several technical advancements have led to the development of controlled drug delivery system that could revolutionize method of medication and provide a number of therapeutic benefits.

 

Controlled Release Dosage form:

The United States Pharmacopoeia (USP) defines the modified-release (MR) dosage form as the one for which the drug release characteristics of time course and/or location are chosen to accomplish therapeutic or convenience objectives not offered by conventional dosage forms such as solutions, ointments, or promptly dissolving dosage forms”. One class of MR dosage form is an extended-release (ER) dosage form and is defined as the one that allows at least a 2-fold reduction in dosing frequency or significant increase in patient compliance or therapeutic performance when compared with that presented as a conventional dosage form (a solution or a prompt drug-releasing dosage form). The terms “controlled release (CR)”, “prolonged release”, “sustained or slow release (SR)” and “long-acting (LA)” have been used synonymously with “extended release”. Controlled Drug Delivery (CDD) occurs when a polymer, whether natural or synthetic, is judiciously combined with a drug or other active agent in such a way that the active agent is released from the material in a predesigned manner. The release of the active agent may be constant over a long period, it may be cyclic over a long period, or it may be triggered by the environment or other external events. In any case, the purpose behind controlling the drug delivery is to achieve more effective therapies while eliminating the potential for both under and overdosing3,4.

 

The rationale for the Controlled Delivery of drugs is to promote therapeutic benefits while at the same time minimizing toxic effects. Controlled, sustained drug delivery can reduce the undesirable fluctuation of drug levels, enhancing therapeutic action and eliminating dangerous side effects5. The goal of any drug delivery system is to provide a therapeutic amount of drug to the proper site in the body in order to promptly achieve and there by to maintain the desired concentration. In recent years, various modified drug products have been developed to release the active drug from the product at a controlled rate. Controlled drug delivery systems aim to maintain plasma concentration of drugs within the therapeutic window for a longer period of time, thereby to ensure sustained therapeutic action and for that reason an increasing interest in their development exist. Moreover, many of new therapeutics under development are large molecules such as peptides, proteins, oligonucleotides, and vaccines. Their physical, chemical, and biopharmaceutical attributes distinct from small molecule drugs demand novel controlled release technologies to diminish barriers for oral delivery, such as instability in GI tract and poor absorption6. In the present study Cinitapride was chosen as model drug. It is a gastroprokinetic agent and antiulcer agent of the benzamide class. It is used in the treatment gastrointestinal disorders associated with motility disturbances such as gastro esophageal reflux disease, non – ulcer dyspepsia and delayed gastric emptying. The present study is an attempt that has been made to formulate controlled release tablet of Cinitapride by wet granulation method using polymers like carbopol, Methyl cellulose, HPMC K4M, HPMC K15M and HPMC K100M. Considering that an antiulcer drug it is important to improve the patient compliance by making it convenient to take and reduce adverse gastrointestinal reaction.

 

MATERIAL USED:

Cinitapride was purchased from Alkem Labs, Mumbai. All other reagents and chemicals used were Pharmacopoeial grade.

 

METHODS:

Drug - Excipient Compatibility Studies:

A successful formulation of a stable and effective solid dosage form depends on careful selection of excipients that are added to facilitate administration, promote the consistent release and bioavailability of the drug and protect it from degradation. If the excipients are new and not been used in formulation containing the active substance, the compatibility studies are of paramount importance. Compatibility of cinitpride with the respective polymers and physical mixture of main formulation was established by Infrared Absorption Spectral Analysis (FTIR). Any changes in the chemical composition after combining with the excipients were investigated with IR spectral analysis

 

Preparation of Cinitapride CR Tablets:

Cinitapride CR tablets each containing 3 mg of cinitapride were prepared by Wet granulation method. The different               polymers used diluents used was Lactose and Dicalcium phosphate were Carbopol, Methyl cellulose, HPMCK 4M, HPMC K 15M and HPMC K 100M in different concentrations. Accurate quantities of cinitapride, polymers and excipients were weighed passed through  24 # and 30 #.

 

Dry Mixing:

Mix the material into the blender at slow speed for 10 minutes

 

Granulation:

The prepared binder solution was added to the above dried mixed ingredients and makes them as wet mass. This wet mass was passed through Sieve No: # 8 and allowed to dry for 1 hour. Then it was rasped using sieve No: # 20

 

Mixing of extra granular material:

The prepared granules were mixed with extra granular material for 5 min.

 

 

 


Table 1: Composition of cintapride CR Tablets with different polymers

Ingredients (mg/tab)

FORMULATION CODE

F1

F2

F3

F4

F5

F6

F7

F8

F9

F10

F11

F12

F13

Cinitapride

3

3

3

3

3

3

3

3

3

3

3

3

3

Carbopol

10

15

20

25

30

-

-

-

-

-

-

-

-

Methyl cellulose

-

-

-

-

-

10

15

20

25

30

-

-

-

HPMCK 4 M

-

-

-

-

-

-

-

-

-

-

10

15

20

HPMC K 15M

-

-

-

-

-

-

-

-

-

-

-

-

-

HPMC K 100M

-

-

-

-

-

-

-

-

-

-

-

-

-

Lactose monohydrate

51

46

41

36

31

51

46

41

36

31

51

46

41

Dibasic calcium phosphate

30

30

30

30

30

30

30

30

30

30

30

30

30

PVP K30

2

2

2

2

2

2

2

2

2

2

2

2

2

Sodium starch glycolate

1

1

1

1

1

1

1

1

1

1

1

1

1

Magnesium stearate

1

1

1

1

1

1

1

1

1

1

1

1

1

Talc

2

2

2

2

2

2

2

2

2

2

2

2

2

Total weight  (mg)

100

100

100

100

100

100

100

100

100

100

100

100

100

 

Table 2: Composition of cintapride CR Tablets with different polymers

Ingredients (mg/tab)

FORMULATION CODE

F14

F15

F16

F17

F18

F19

F20

F21

F22

F23

F24

F25

Cinitapride

3

3

3

3

3

3

3

3

3

3

3

3

Carbopol

-

-

-

-

-

-

-

-

-

-

-

-

Microcrystalline cellulose

-

-

-

-

-

-

-

-

-

-

-

-

HPMC K 4 M

25

30

-

-

-

-

-

-

-

-

-

-

HPMC K 15M

-

-

10

15

20

25

30

-

-

-

-

-

HPMC K 100M

-

-

-

-

-

-

-

10

15

20

25

30

Lactose monohydrate

36

31

51

46

41

36

31

51

46

41

36

31

Dibasic calcium phosphate

30

30

30

30

30

30

30

30

30

30

30

30

PVP K30

2

2

2

2

2

2

2

2

2

2

2

2

Sodium starch glycolate

1

1

1

1

1

1

1

1

1

1

1

1

Magnesium stearate

1

1

1

1

1

1

1

1

1

1

1

1

Talc

2

2

2

2

2

2

2

2

2

2

2

2

Total weight  (mg)

100

100

100

100

100

100

100

100

100

100

100

100

 

 


Lubrication:

After mixing of extra granular material the blend was lubricated by using magnesium stearate, talc and sodium starch glycolate for 5 min. Before  compression, all precompressional parameters were adjusted.3 mg of cinitapride Cr tablets were compressed on single rotary punching machine, each weighing 100mg. the composition of cinitapride CR tablets was given in Table 1 and 2.

 

EVALUATION OF PRECOMPRESSIONAL PARAMETERS:7-18

PRECOMPRESSIONAL STUDIES:

1.    Angle of repose:

The angle of repose of  granules was determined by the funnel method. Accurately weighed granules were placed in a plugged glass funnel which had a distance of 2 cm from the flat surface. The granules was  allowed  to flow through the 8 mm funnel orifice by removing the cotton plug from the funnel orifice. The height of the heap (h) formed as well as the radius of the heap (r) were noted. The data obtained was used for calculating angle of repose.

 

θ = tan-1(h/r)

 

2.    Bulk Density and Tapped  Density:

Accurately weighed granules were previously passed through 20 # sieve, was transferred in 100 ml graduated cylinder. The powder in the cylinder was leveled without compacting, and the unsettled apparent volume was noted.  The cylinder was dropped on a wooden platform from a height of 2.5 cm, three times at 2s interval. The volume occupied by the granules was recorded as the bulk volume. The cylinder was then tapped on the wooden platform until the volume occupied by the granules was remained constant. This was repeated three times for granules. The data generated were used in calculating the Carr’s compressibility index and Hausner’s ratio. The LBD and TBD were calculated in g/ml  by the following formula

 

 

Bulk density = Weight of powder / Bulk volume

Tapped Density = Weight of powder / Tapped volume

 

 

3.    Carr’s Index:

The Compressibility Index of the powder blend was determined by Carr’s compressibility index. It was a simple test to evaluate the BD and TD of a powder and the rate at which it was packed down. The formula for Carr’s Index is as below

 

Carr’s Index (%) =  [(TD-BD) x100]/TD

 

4.    Hausner’s Ratio:

The Hausner’s ratio is a number that is correlated to the flowability of a powder or granular material.

 

Hausner’s Ratio = Tapped Density / Bulk Density

 

POST-COMPRESSIONAL STUDIES:7-18

A.   Tablet thickness:

The thickness of 10 tablets of each selected at random from the formulated tablets was determined using a vernier caliper and the mean of these readings was taken as the mean tablets thickness.

 

B.   Tablet weight uniformity:

Twenty tablets were weighed individually using a digital balance with the precision of 0.05 mg and readability of 0.1 mg, from which the mean was calculated and percentage deviations determined.

 

C.   Hardness (Crushing strength):

The crushing strengths of tablets were determined individually with the Monsanto hardness tester. 6 tablets were used and the mean crushing strength was calculated.

 

D.   Friability:

For each tablet formulation the friability of 10 tablets were determined. Friability can be determined by the following equation

 

                    W(I) – W(F)

        F =  ------------------------ X 100

                         W(I)

 

E.   Drug content uniformity:

The drug content of the matrices determined  by equilibrating an accurately weighed quantity of the tablet in appropriate dissolution medium. The samples were filtered, suitably diluted and assayed spectrophotometrically.

 

F.    Tablet dosage forms assay:

Tablet containing 3 mg of drug was dissolved in 100 ml of 0.1N HCl .The drug was allowed to dissolve in the solvent, the solution was filtered, and 1ml of filtrate was suitably diluted with phosphate buffer pH 6.8 and analyzed spectrophotometrically at 264 nm. The amount of cinitapride was estimated by using standard calibration curve of the drug. Drug content studies were carried out for each batch of formulation

G.   In-vitro drug release studies of tablets:

Drug release studies of  tablets were carried out using a USP II dissolution rate test apparatus (50 rpm, 37 °C) for 2 hr in 0.1 N HCl (900 ml) as the average gastric emptying time is about 2 hr. Then the dissolution medium was replaced phosphate buffer pH-6.8 (900 ml) for 12 hrs tested for drug release up to complete drug release. At the end of the time period 10 ml of the samples were taken and analyzed for cinitapride content. A 10 ml Volume of fresh and filtered dissolution medium was added to make the volume after each sample withdrawal. Sample was analyzed using UV spectrophotometer at 264 nm.

 

H.   Comparison of in-vitro dissolution of formulation with the marketed product:

Comparison of dissolution profile of optimized formulation (F23) with marketed product (Cintapro OD) was done.

 

I.     Stability study of optimized formulation:

The purpose of stability testing was to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a variety of environmental factors such as temperature, humidity, light, to establish re-set period for drug substances or a shelf life for the drug product and recommended storage conditions. The storage condition used for stability studies were 40°C, 75±5% RH, for 3 months. The tablets were withdrawn for analysis of following parameters:

1. Physical characteristics

2. Assay

3. Dissolution

 

Tablets were evaluated for physical characteristics; Assay, and in-vitro drug release after 3 months.

 

RESULTS AND DISCUSSION:

The present study was to formulate controlled release tablets of cinitapride using different polymers in different concentrations and diluent Lactose and DCP along with other excipients by wet granulation method. The prepared tablets were evaluated for physic chemical properties, in-vitro dissolution studies and stability studies.

 

DRUG- EXCIPIENT COMPATIBILITY STUDIES:

The optimized formulation F23 was subjected for IR Spectroscopic analysis to ascertain whether there was any interaction between drug and excipients used. The IR Spectra showed similar characteristic peaks at their respective wavelength with minor difference. The similarity in the peaks indicated the compatibility of drug with formulation excipients.

 


 

Figure 1: FTIR Spectra of Cinitapride

 

 

Figure 2:  FTIR Spectra of drug and F23

 

Table 3: Pre-compressional studies of Cinitapride granules

Formulation

Angle of Repose

Bulk Density

Tapped Density

Compressibility Index

Hausner's Ratio

LOD

F1

28.02

0.5581

0.6775

17.62

1.214

2.0234

F2

28.80

0.5665

0.6813

16.85

1.203

2.0202

F3

28.65

0.5562

0.6714

16.36

1.207

2.0240

F4

28.70

0.5602

0.6698

17.11

1.196

2.0002

F5

28.71

0.5620

0.6787

17.19

1.207

2.0198

F6

28.40

0.5702

0.6782

15.92

1.189

2.02

F7

28.46

0.5725

0.6909

17.14

1.206

1.9952

F8

28.48

0.5698

0.6701

14.96

1.176

1.998

F9

28.46

0.5710

0.6897

17.21

1.121

2.012

F10

28.48

0.5711

0.6780

15.76

1.187

2.032

F11

27.855

0.5612

0.6616

15.107

1.178

1.88

F12

27.85

0.5642

0.6592

14.41

1.168

2.002

F13

27.96

0.5602

0.6680

16.13

1.1922

1.98

F14

27.90

0.5600

0.6596

15.10

1.178

1.970

F15

27.77

0.5611

0.6593

14.89

1.175

2.011

F16

26.70

0.5590

0.6618

15.53

1.183

1.985

F17

25.98

0.5602

0.6603

15.15

1.178

1.9086

F18

26.12

0.5597

0.6660

15.96

1.189

1.889

F19

26.04

0.5600

0.6596

15.10

1.178

1.980

F20

26.34

0.5610

0.6627

15.34

1.181

1.995

F21

26.03

0.5604

0.6603

15.13

1.178

2.002

F22

25.95

0.5661

0.6599

14.21

1.165

1.877

F23

26.91

0.5624

0.6591

14.67

1.171

1.903

F24

26.73

0.5654

0.6603

14.37

1.167

1.930

F25

26.11

0.5655

0.6589

14.17

1.165

1.919

 


 

 

EVALUATION OF CORE TABLETS:

A)  PRECOMPRESSIONAL PARAMETERS:

Granules of all the formulations were subjected  for  various  precompressional evaluations such as  angle  of repose,  bulk  and  tapped density, compressibility index and Hausners Ratio. Results of all the pre-compression parameters performed on the granules for batches are shown in Table 3. The result of angle of repose was found to be 25.95 to 28.80 for batches F1 to F25 respectively. Compressibility index was found to be 14.17 to 17.62 for batches F1 to F25 respectively. The results of Hausners ratios were found to be 1.121  to 1.214 respectively for batch F1 to F25 respectively. This was further supported by lower compressibility index values. Generally, compressibility index values up to 15% results in good flow properties.

 

B)   POST-COMPRESSIONAL PARAMETERS:

All the tablet formulations were subjected for  evaluation  according  to various official specifications  and  other parameters. Shape, thickness, hardness, friability, weight variation, tablet dosage form assay.

 

a)    Shape and appearance:

Formulations prepared were randomly picked from  each batch were examined in order to find their shape, in presence of light for colour. Tablets shown round concave shape. Tablets were white in colour.

b)   Uniformity of thickness:

Thickness of the tablets was measured using calibrated vernier caliper by picking three tablets randomly from all the batches. The results of thickness for tablets are shown in  Table 4.  The mean thickness of tablets lied between 3.48 ±0.14 mm to 3.62 ±0.16 mm. The standard deviation values indicated that all the formulations were within the range.

 

c)    Weight variation test:

The weight variation of uncoated tablets all the formulations is shown in Table 15. The results lied between 98.90±2.03 mg to 99.87±1.43 mg. All the tablets passed the weight variation test, i.e., average percentage weight variation was found within the pharmacopoeial limits of ±7.5%.

 

d)   Hardness test:

Hardness or crushing strength of the tablets of both the formulations were found to be lied between 5.60 ±0.6 kg/cm2 to 6.37±0.65 kg/cm2. The mean hardness test results are tabulated in Table 15. The  low standard  deviation values indicated that the hardness of all the formulations were almost uniform and the tablets  possessed good  mechanical strength  with sufficient hardness.

 


 

Table 4: Post-compression evaluation of the prepared Tablets F1 – F25

Formulation

Thickness (mm)

Weight of Tablet (mg)

Friability (%)

Hardness (kg/cm2)

Assay (%)

F1

3.53±0.18

99.84 ± 1.43

0.215

5.75 ± 0.6

    98.39

F2

3.48± 0.17

99.06 ± 1.95

0.120

5.80 ±0.72

99.53

F3

3.48± 0.17

99.46 ± 1.33

0.094

  5.83 ±0.91

99.80

F4

3.52±0.18

  99.18 ± 2.00

0.117

5.84±0.17

99.68

F5

3.53±0.18

98.78 ± 2.05

0.276

5.92 ±0.80

98.50

F6

3.48± 0.14

99.87 ± 1.43

0.295

5.60 ± 0.6

  100.36

F7

3.62±0.17

99.17 ± 1.80

0.164

5.72 ±0.53

99.82

F8

3.57 ±0.14

99.26 ± 1.33

0.305

  5.70± 0.65

   98.88

F9

3.60±0.18

  99.18 ±   2.10

0.229

5.75±0.70

   99.18

F10

3.54 ±0.18

99.86 ± 1.62

0.177

  5.78 ±0.80

98.70

F11

3.62±0.15

99.27 ± 1.43

0.175

  5.90 ± 0.6

   99.26

F12

3.60 ±0.17

99.26 ± 1.85

0.098

5.86 ±0.72

99.72

F13

3.62±0.14

99.46 ± 1.30

0.087

5.97 ±0.65

  100.80

F14

3.58± 0.17

  99.28 ±   1.68

0.131

6.12±0.70

   99.68

F15

3.58± 0.17

99.26 ± 1.32

0.108

6.19 ±0.90

99.12

F16

3.62±0.16

99.07 ± 1.63

0.085

5.90 ± 0.6

   99.16

F17

3.60±0.18

99.26 ± 1.93

0.093

  5.98 ±0.47

99.12

F18

3.53±0.19

98.90 ± 2.03

0.139

  5.99 ±0.45

  101.0

F19

3.58± 0.14

  98.98 ±   2.22

0.106

6.02±0.72

99.68

F20

3.60± 0.15

99.16 ± 1.32

0.089

6.17 ±0.04

  100.32

F21

3.63±0.18

99.27 ± 1.43

0.085

6.0 ± 0.6

    99.36

F22

3.58± 0.16

99.26 ± 1.85

0.086

6.10 ±0.77

99.42

F23

3.60± 0.17

99.56 ± 1.33

0.086

6.37 ± 0.65

   100.30

F24

3.62±0.15

    98.98 ± 2.10

0.088

6.30±0.70

99.28

F25

3.58±0.18

99.66 ± 1.32

0.097

6.31±0.80

 98.98

 


e)    Friability test:

Friability values for each batch were found to be lies between 0.085% to 0.305% respectively. The obtained results were found to be well within the approved range (<1%) in all the designed formulations. That indicated tablets possess good mechanical strength.

 

f)    Tablet dosage form assay:

Tablet dosage form assay for both the formulations was carried out. In assay of formulations lies in between 98.39 % to 101 % drug content respectively. The cumulative percentage drug released from each tablet  in the  in-vitro release studies was based on the average drug content present in the tablet.

 

The In-vitro release studies:

The in-vitro release studies were carried out using USP II dissolution assembly. The results obtained in the   in-vitro drug release study are shown in Figure 3 to Figure 7.

 

 

Figure 3: In-vitro drug release profile of Cinitapride trial batches F1 to F5

 

 

Figure 4: In-vitro drug release profile of Cinitapride trial batches F6 to F10

 

Figure 5: In-vitro drug release profile of Cinitapride trial batches F11 to F15

 

 

 

Figure No. 6: In-vitro drug release profile of Cinitapride trial batches F16 to F20

 

 

 

Figure 7: In-vitro drug release profile of Cinitapride trial batches F21 to F25

 

Stability study of Cinitapride controlled release tablets:

The stability study was carried out at 40°C/75% RH for formulation F23 up to 90 days. At every 30 days time interval, the devices were analyzed for Physical Properties and in-vitro drug release. Release profile of F23 after stability study is plotted as shown in the Figure 13.The results of accelerated  stability study showed that there was no change in the formulation after 3 months. In-vitro drug release study throughout 2  hours,  after 30, 6 0 and 9 0 days, showed the values 21.88, 21.33 and 22.09 respectively. The drug release throughout 2  hours  obtained the values within range of  targeted  release profile. In-vitro drug release study throughout  12  hours,  after 30, 6 0 and 90 days, showed the values 98.07%, 97.91% and 98.76% respectively. The drug release throughout 12 hours obtained the values within range of targeted release  profile. Assay result after 30, 60 and 90 days, showed the values 99.78%, 99.15% and 99.07% respectively. After 90 days accelerated stability study the assay result was stable.

 

 

Figure 8: Comparison of In-vitro drug release of Cinitapride CR formulation (F23) before and after  stability studies

 

CONCLUSION:

The study was undertaken with an aim of Formulation, Development and Evaluation of Cinitapride controlled release tablets, using different polymers as release retarding agent. Preformulation study was done initially and results directed for the further course of formulation. Based on preformulation studies different  batches  of  Cinitapride were  prepared  using  selected  excipients. Granules were evaluated for Bulk density, Tapped density, Compressibility index, and Hausners ratio. Tablets were tested for weight variation, thickness, hardness and friability as per official procedure. Dissolution was carried out in phosphate buffer pH 6.8. Based on dissolution tests, it was concluded that F23 satisfactorily release drug through 12 hrs. From the above results and discussion, it was concluded that formulation of controlled release tablet Cinitapride of formulation F23 can be taken as an ideal formulation of controlled release tablets for 12 hour release, as it fulfills all the requirements for controlled release tablet and our study encourages for in-vivo studies. Stability study was carried out after 3 months of time.  Stability study report confirmed  that  formulation  was  not  shown  any  colour  change  and  no significant release profile, assay from initial period. Thus it confirms that prepared formulation was stable.

 

ACKNOWLEDGEMENT:

The authors are thankful to the Management and Principal of Karavali college of Pharmacy, Mangalore for providing all the facilities to conduct the research work and the authors are also thankful to Alkem Labs, Mumbai, for generous gift sample of Cinitapride.

 

REFERENCES:

1.     Lachman L. Liberman HA, Kanig JL. The Theory and Practice of Industrial Pharmacy, 3rd Ed, Varghese Publishing House 1990; 430-453.

2.     Brahmankar DM, Jaiswal SB. Biopharmaceutics and pharmacokinetics a treaties, 2nd Ed, Vallabh prakashan 2010; 397–515.

3.     Debjit B, Harish G, Kumar BP, Duraivel S, Kumar KPS. Controlled Release Drug Delivery Systems. Pharm Innovation J 2012; 1(10):24- 32.

4.     Notari R. Biopharmaceutics and Clinical Pharmacokinetics an Introduction, 3rd ed, Marcel Dekker Inc1971;152-154.

5.     Martin Alford N, Sinko Patrick J. Martins Physical Pharmacy and Pharmaceutical Sciences. 5th Ed, Lippincott Williams and Wilkins 2006; 667-672.

6.     Kumar KPS, D Bhowmik, A Dutta, S Paswan, L Deb. Recent trends in scope and opportunities of Control Release Oral Drug Delivery Systems 2012; 1:20-33.

7.     Remington, The Science and Practice of Pharmacy. 20th Ed, Lippincott Willams and Wilkins2007 vol.1:903-914

8.     Jain NK. Controlled And Novel Drug Delivery, CBS Publication 2010;1(2):27-4

9.     www.nlm.nih.gov/medlineplus/ency/article/007193.htm.

10.   British Pharmacopoeia 2012 ;1665-1667

11.   European Pharmacopoeia 5.0: 2190-2192.

12.   Prescribing Information of Paxil

13.   Prescribing Information of Paxil CR

14.   www.rxlist.com

15.   Leonard, Gramham S, David P. Paroxetine Controlled Release Compositions EP0839039 A1. 1998.

16.   Subramaniam K, M Rangasamy, G Kugalur, KN Parthiban, S Natesan. Formulation and evaluation of sustained release tablets of Aceclofenac using hydrophilic matrix system. Int J Pharm Tech Res 2010; 2(3):1775-1780.

17.   Indian Pharmacopoeia. 5th Ed, Ghaziabad.. Indian Pharmacopoeia  Commission 2007;.

18.   ICH guidelines for Stability testing of new drug substances and products.

 

 

 

 

 

Received on 16.03.2016       Accepted on 08.04.2016     

© Asian Pharma Press All Right Reserved

Asian J. Pharm. Res. 2016; 6(2): 87-94

DOI: 10.5958/2231-5691.2016.00015.0