Social & Pharmacy

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

World Journal of Pharmaceutical Research

Sahu et al. SJIF Impact


World Journal of Pharmaceutical Factor 8.084
Research
Volume 9, Issue 5, 1142-1163. Research Article ISSN 2277– 7105

FORMULATION AND EVALUATION OF TRANSDERMAL PATCHES


OF DICLOFENAC SODIUM

Akarshi Shrivastava1, Harish Sharma2 and Gyanesh Kumar Sahu1*

1
Shri Shankracharya Technical Campus Shri Shankaracharya group of Institutions, Faculty of
Pharmaceutical Sciences, Junwani, Bhilai (C.G.).
2
MJ College of Pharmacy, Junwani, Bhilai (C.G.).

ABSTRACT
Article Received on
28 Feb. 2020, Objective: The main objective of this work is to fabricate Diclofenac
Revised on 19 March 2020, Sodium transdermal patch to avoid first pass metabolism and to
Accepted on 07 April 2020,
DOI: 10.20959/wjpr20205-17296 overcome the problems associated with its biological short half-life
and fluctuations in plasma concentration upon oral administration.
Method: The patch were prepared using Solvent Casting Method,
*Corresponding Author
Gyanesh Kumar Sahu using Methyl cellulose as polymer, Dibutyl phthalate as permeation
Shri Shankracharya enhancer and Ethanol: distilled water as solvent. The physical
Technical Campus Shri evaluation of the prepared patch include organoleptic observation,
Shankaracharya Group of
moisture uptake, moisture loss, content uniformity test, stability
institutions, Faculty of
studies, swelling index, folding endurance. The drug release was
Pharmaceutical Sciences,
Junwani, Bhilai (C.G.). determined using Franz diffusion cells in phosphate buffer (pH 7.4).
Result: The result of physiochemical parameters of the transdermal
patch were found satisfactory. Formula F2 produce best result among all patchs with smooth
transparent texture and maximum folding endurance with minimum moisture loss and
moisture uptake. The patch weight, thickness were found to be uniform. The drug content in
F2 was 98%. Stability study indicates that drug remain stable for six months. Conclusion:
This work is further aimed to analyse, concentration of drug reaching in the body and to
study its effect.
KEYWORDS: Diclofenac Sodium, Transdermal patch and conventional dosage Form

1. INTRODUCTION
Since last decade, TDDS acquired a lot of interest due over the conventional dosage forms
and oral controlled release delivery, specifically for the avoidance of hepatic first pass

www.wjpr.net Vol 9, Issue 5, 2020. 1142


Sahu et al. World Journal of Pharmaceutical Research

effect.[1] Optimization of drug release through the skin directly into the systemic circulation
and parallel minimize the retention and metabolism of the drug in the skin is the major goal
of transdermal products.[2]

Administration of drugs in the conventional dosage forms as compare to the controlled or


sustain release form usually results in large scale fluctuations in plasma drug concentrations
leading to unwanted toxicity or poor effectiveness along with limitations such as repetitive
dosing at certain time interval and unpredictable absorption, led to the concept of the
controlled drug delivery system or therapeutic system.[3] The successful development of
transdermal therapeutic system mainly depends on choice of drug, which should be non-
irritant, non-toxic and must cross the various skin layer to produce the desired therapeutic
effect at specific period of time. Drugs which produce these effects in small amounts with
molecular weight range of 100-800 Da are ideal candidates for TDDS.[4]

Transdermal patches were developed in the 1970’s and the first transdermal system,
Transderm Scop (ciba, now Novartis) was approved by Food and Drug Administration
(FDA) in 1979 for prevention of nausea and vomiting associated with travel, particularly by
sea.[5] A Transdermal patch is a medicament adhesive patch which when applied to the skin
utilizes passive diffusion of drug at controlled rate through the skin and into the blood
stream.[6]

NSAIDs (Non-steroidal anti-inflammatory drugs) are mostly used for the preparation of
transdermal patches for the treatment of inflammation or pain. The NSAIDs patches are safer
and convenient to use than its oral dosage form. NSAIDs tablets for rheumatism leads to
various side effects like internal stomach bleeding, increased acidity, ulcers can be avoided
by using transdermal patches of NSAIDs.[7] On the other hand, the analgesic patch
of NSAIDs can be used on the site of sprain or strain. These patch when applied on the skin
in form of transdermal patch, without reaching higher plasma drug concentrations the drug
penetrate the various layer of skin in sufficient amount to exert local therapeutic effect.
Hence NSAIDs transdermal patches offers advantages of painless drug delivery, is easy to
apply, provides faster and longer relief, and have no or few gastrointestinal side effects from
the drug itself. That’s the reason now a days patients are advised to take the NSAIDs patch
over the other route because of its minimal side effects related to systemic toxicity and GIT
irritation.[8]

www.wjpr.net Vol 9, Issue 5, 2020. 1143


Sahu et al. World Journal of Pharmaceutical Research

Mainly, nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most frequently used
drugs, which are prescribed in both acute and chronic condition of rheumatoid arthritis,
osteoarthritis, dysmenorrhea treatment because of their analgesic, antipyretic and anti-
inflammatory effect. Diclofenac (2-[2-(2,6 dichlorophenyl amino) phenyl]acetic acid) is one
of the most likely and commercially successful drug in the family of NSAIDs. The main
mechanism of action of diclofenac is to inhibit the activity of cyclooxygenase (COX) by
interdicting the prostaglandin (PG) synthesis thereby controlling the synthesis of
thromboxanes and prostaglandins. The cyclooxygenase is an enzyme released during pain
and inflammation whereas, thromboxanes and prostaglandins are mediators of
[9-11]
vasoconstriction and inflammation. The drug undergoes substantial hepatic first-pass
metabolism and only about 50% of administered dose reaches systemic circulation. Its
biological half-life is also very short, it is considered as a suitable candidate to formulate it
into a sustained release matrix type transdermal patch system.[12]

The Diclofenac sodium also possesses the ideal characteristics such as poor bioavailability,
short biological half-life and smaller dose etc., to be formulated in to a transdermal patch.
Main added advantage of transdermal patches as it maintain constant and prolonged drug
level, and dose frequency is reduced, self-administration, quick onset of action and easy
termination of medication leads to provide higher level of patient compliance.[13,14]

Diclofenac was chosen as a model drug for present study as it possess near ideal
characteristic, that the drug must have in formulation to develop different transdermal matrix
films with different ratios of hydrophilic and hydrophilic –lipophilic combination containing
the drug and to perform the physicochemical and in-vitro, in-vivo evaluation along with study
of the prepared films.[15]

Diclofenac is efficiently and rapidly absorbed after conventional oral, rectal or intramuscular
administration.[16,17] After intramuscular administration peak plasma concentrations are
attained after 10 to 15 minutes. With the enteric-coated formulation peak concentration are
reached after 1.5 to 2.5 hours, and this is delayed by food to 2.5 to 12 hours. After a single
50mg dose of this formulation, mean peak plasma concentration of unchanged diclofenac is
0.7 to 1.5 mg/L.[18-21] Like other NSAIDs, diclofenac is highly (>9.5%) protein bound. The
mean total volume of distribution is 0.12 to 0.17 L/kg and that of the central compartment is
0.04 L/kg. The drug efficiently penetrates inflamed synovial fluid where high concentrations

www.wjpr.net Vol 9, Issue 5, 2020. 1144


Sahu et al. World Journal of Pharmaceutical Research

are maintained compared with plasma concentrations. Diclofenac and its metabolites cross
the placenta in animals, and small amount may be found in the breast milk of women.[22-26]

In healthy volunteers, mean plasma clearance of diclofenac is 16 L/h, and the mean
elimination half-life of the terminal phase is 1.1 to 1.8 hours. The mean elimination half-life
after a radiolabelled dose is about 30 hours for the tracer.[27-32] The initial dosage of
conventional or enteric-coated Tablets of diclofenac is 150mg daily in 2 or 3 divided doses
with meals, and in most patients therapeutic control can be maintained on 100mg daily.[33-35]
A sustained release formulation can be administered once daily, and suppositories can be
administered once or twice daily. Intramuscular diclofenac 75mg can be given for the urgent
relief of acute pain such as renal or biliary colic. A further dose may be administered after 30
minutes if necessary, but as with oral administration the daily dosage should not exceed
150mg.[36-42]

2. MATERIAL AND METHOD


2.1 Material
All the chemicals used in this research were of standard analytical or pharmaceutical grade.
Diclofenac Sodium was a gift sample obtained from pharmaceutical industry. Formulation of
patch include- active ingredient, backing agent, plasticizer, penetration enhancer and solvent
used are shown in Table1.

Table 1: Ingredients Table of Diclofenac Sodium Patch.


S. No. INGREDIENTS ACTIVITY
1. Diclofenac Sodium (mg) Active ingredient (Drug)
2. Methyl Cellulose (mg) Backing agent
3. PG, PEG-400 (ml) Plasticizer
4. Dibutyl Phthalate (ml) Penetration enhancer
5. Distilled water: Ethanol (ml) Solvent

2.2 Preparation of Patch by Solvent Casting Method


Transdermal patches were fabricated using methyl cellulose as a polymers containing
diclofenac sodium by solvent casting method[43] shown in Figure 1. According to the formula
methyl cellulose were accurately weighed and dissolved in mixture of ethanol: water (1:2)
used as a solvent. The drug was then dispersed in the polymeric solution and plasticizer of
dibutyl phthalate was added with continuous stirring using a magnetic stirrer to obtain
homogeneous mixture. Lastly, the bottom of petridish were covered with aluminium foil and
the resulting solution was poured into levelled mercury surface in a petridish covered with a

www.wjpr.net Vol 9, Issue 5, 2020. 1145


Sahu et al. World Journal of Pharmaceutical Research

funnel in inverted position. The solvent was allowed to evaporate and left undisturbed at
room temperature for the next 24 hour. The patch was obtained intact by slowly lifting from
the Petri dish and transdermal patches were cut into radius of 2cm2.[44-46] Ingredients to be
used are shown in Table 2.

Figure 1: Preparation of Transdermal Patch by Solvent Casting Method.

Table 2: Composition of Diclofenac Sodium Transdermal Patch in Different Ratio.


S. No. INGREDIENTS F1 F2 F3 F4
1. Diclofenac Sodium (mg) 25 25 25 25
2. Methyl Cellulose(mg) 300 300 400 400
3. PEG-400 - - 1.2 1.2
4. PG (ml) 1.2 1.2 - -
5. Dibutyl Phthalate 1.2 1.2 1.2 1.2
6. Ethanol: Distilled water 1:2 1:2 1:2 1:2

3. Evaluation and Characterization of Medicated Patch


After getting the best formula based on accurate diclofenac sodium, it was further studied for
its physical, mechanical and permeability of the drug. Physical and mechanical properties of
medicated transdermal patch such as thickness, surface pH, content uniformity, folding
endurance, moisture loss, moisture uptake, weight uniformity, and stability studies. Also
medicated patch were evaluated for drug content, in-vitro drug release.

www.wjpr.net Vol 9, Issue 5, 2020. 1146


Sahu et al. World Journal of Pharmaceutical Research

3.1 Physical Appearance


All the transdermal film were organoleptically inspected and all over elegance for colour,
transparency, shape, texture of the surface, homogeneity of thickness, film formation (no
collapse or shrinkage)upon drying.[47]

3.2 Thickness of Patch


The thickness of the drug loaded patch is determined by screw gauge and micrometer at
different point and average of readings were calculated.[48]

3.3 Uniformity of Weight


Weight variation is determined by weighing 5 randomly selected patches and calculating the
average weight. The individual weight should not digress significantly from the average
weight.[49]

3.4 Folding Endurance


This test is performed to determine the elasticity and fragility of transdermal patches.[50] The
test was conducted by folding the patch at the same point n number of times till the patch is
broken. The number of folds is considered to be the value of resistance to folding.[51]

3.5 Surface pH
Each film was allowed to swell by adding 0.5mL of distilled water on the film surface for 1hr
at room temperature. Then, pH was noted by bringing the electrode into contact to the surface
of the film and allowing it to equilibrate for 1 min.[52]

3.6 Swelling Index


The film were weighed (WF) immersed in a beaker containing 25mL phosphate buffer pH
7.4.The beaker were kept at 25℃ using thermo stated water bath. At specific intervals up to
2 hour, the swollen film were weighed (WS) after removal of excess surface water by light
blotting with a filter paper. The experiment was discontinued when the film begin to
dissolve.[53] The swelling index was calculated by.

Swelling index =

Where, WF = weight of the dried polymer film.


WS = weight after swelling.

www.wjpr.net Vol 9, Issue 5, 2020. 1147


Sahu et al. World Journal of Pharmaceutical Research

3.7 Moisture Uptake


The films were placed in a desiccator containing activated silica gel for 24 hr. Then, they
were weighed (WD) and then transfer to another desiccator containing saturated sodium
chloride (75%). The films were weighed daily until they showed constant weight (WU).[54]
The percentage of moisture uptake was calculated by.

Moisture uptake capacity% =

3.8 Moisture Loss


The film were weighed (WF), kept in a desiccator containing silica gel at 25℃ and weighed
daily until they showed constant weight (WD).[55] The percentage moisture loss was
calculated by.

Moisture loss % =

3.9 Content Uniformity Test


Randomly patches are selected and content in each patch is determined individually. If 9 out
of 10 patches have content between 85% to115% of the specified value and one has content
not less than 75% to 125% of the specified value, then transdermal patches passes the test of
content uniformity. But if 3 patches have content in the range of 75% to 125%, then
additional 20 patches are tested for drug content. If these 20 patches have range from 85% to
115%, then the transdermal patches passes the test.[56, 57]

3.10 Drug Content


A specified area of patch is to be dissolved in phosphate buffer solution (pH 7.4). The content
was allowed to dissolve in solution. Then the solution is to be filtered through a filter medium
and absorbance were measured with the help of UV at wavelength 320nm. Each value
represents average of three different samples.[58-60]

3.11 Stability Study


The developed transdermal patch were sealed in polyethylene coated aluminium foils and
kept at 40±0.5℃ and 75±5%RH for 6 months. The samples are withdrawn at different
interval of 0, 30, 60, 90 and 180 days and analysed suitably for the drug content and any
physical changes brought about on storage.[61-64]

www.wjpr.net Vol 9, Issue 5, 2020. 1148


Sahu et al. World Journal of Pharmaceutical Research

3.12 In-Vitro Drug Diffusion Study


The in-vitro release study was performed using a modified Franz diffusion cell is the most
common technique for measuring dermal absorption.[65-69] In this research drug diffusion
were studied with the help of goat skin. After hair were shaven using razor, without
damaging skin. The skin membranes were first hydrated for 30 minutes in the buffer solution
(pH 7.4) at room temperature to remove extraneous debris or blood vessels.[70] The donor
portion contains a transdermal patch of diclofenac sodium. The donor and receptor
compartment separator membrane is the goat skin. The membrane is placed between the
donor compartment and the receptor compartment with the dermis side facing the receptor
compartment of the diffusion cell. The receptor compartment of the diffusion cell was filled
with phosphate buffer (pH 7.4). The whole assembly was fixed on a magnetic stirrer with
constantly and continuous stirred using magnetic bead and temperature was maintained at
32℃. The sample were withdrawn at different time interval and analysed for drug content
spectophotometrically. The receptor phase was replenished with an equal volume of
phosphate buffer at each sample withdrawal to maintain sink condition.[71-73]

4. RESULT AND DISCUSSION


4.1 Standard Graph of Diclofenac Sodium
The lambda max of the Diclofenac sodium was found to be 320nm. After the determination
of lambda max the calibration curve and absorption are to be evaluated by the UV
spectroscopy. The results of the absorption and concentration were given below in the Table
3. Standard graph of diclofenac sodium are shown in Figure 2.

Table 3: Concentration and Absorption of Diclofenac Sodium.


Concentratio
S. No. Absorption
n(µg/ml)
1. 2 0.160
2. 4 0.310
3. 6 0.463
4. 8 0.623
5. 10 0.790
6. 12 0.925

www.wjpr.net Vol 9, Issue 5, 2020. 1149


Sahu et al. World Journal of Pharmaceutical Research

Figure 2: Standard Graph of Diclofenac Sodium.

4.2 Result of Evaluation Studies


4.2.1 Physical Appearance
The patch was visually inspected for colour, surface texture, shape. Figure 3 and Table 4.
Helps to explain the physical appearance of patch. Transdermal patch cut in 2cm2.

Table 4: Physical Appearance of Patch.


Physical
S. No. Result
appearance
1. Color Transparent
2. Surface texture Smooth
3. Shape Round

Figure 3: Transdermal Patch.

4.2.2 Thickness of Patch


The thickness of the prepared patch was measured by Vernier Caliper. The mean thickness
was measured at different point of the film were given in Table 5.

www.wjpr.net Vol 9, Issue 5, 2020. 1150


Sahu et al. World Journal of Pharmaceutical Research

Table 5: Determination of Thickness of Patch.


S. No. Sample Thickness (mm)
1. F1 0.295±0.012
2. F2 0.245±0.09
3. F3 0.259±0.011
4. F4 0.254±0.016

The thickness of patch was determined. It was found that F2 (0.2452mm) show less thickness
whereas F1 (0.295mm) shows more thickness.

4.2.3 Uniformity of Weight


The quantified area of 2cm2 radius is to be cut at different parts of the patch and weigh in
digital balance. The average weight calculated from individual weight are shown in Table 6.

Table 6: Determination of Uniformity of Weight.


Weight
S. No. Sample
variation (mg)
1. F1 590±0.025
2. F2 598±0.016
3. F3 593±0.017
4. F4 587±0.026

Uniformity of weight was measured. It was found that F2 (598mg) shows more weight
whereas F4 (587mg) shows less weight.

4.2.4 Folding Endurance


The folding endurance of the Patches are given below in the Table 7.

Table 7: Determination of Folding Endurance.


Folding
S. No. Sample
Endurance
1. F1 26±8
2. F2 30±7
3. F3 27±4
4. F4 27±5

Folding endurance of prepared transdermal patches were noted. It was found that more
folding endurance value is seen in F2 (30) and less folding endurance value in F4 (27).

4.2.5 Surface pH
Surface pH of F1 to F4 were determined. The results were tabulated in Table 8.

www.wjpr.net Vol 9, Issue 5, 2020. 1151


Sahu et al. World Journal of Pharmaceutical Research

Table 8: Determination of Surface pH.


S. No. Sample pH
1. F1 6.6±1
2. F2 6.8±1
3. F3 5.9±2
4. F4 6.3±1
Surface pH was determined and found to be that F2 (6.8) has more pH as compare to other
samples.

4.2.6 Swelling Index


The swelling index at different time interval are given in Table 9 and Table 10.

Table 9: Determination of Swelling Index after 1 hour.


Swelling
S. No. Sample
index (%)
1. F1 1.86±0.046
2. F2 2.85±0.032
3. F3 2.33±0.049
4. F4 1.75±0.043

Table 10: Determination of Swelling Index after 2 hour.


Swelling
S. No. Sample
Index (%)
1. F1 2.97±0.083
2. F2 2.23±0.107
3. F3 3.54±0.053
4. F4 2.96±0.056

Swelling index after completion of 2 hour was found that F3 has more percentage of swelling
index in couple of hour and also film gets erode firstly as compare to other formulated
patches.

4.2.7 Moisture Uptake


Moisture uptake of prepared transdermal patch F1 to F4 were determined. The results were
tabulated in Table 11.

Table 11: Determination of Moisture Uptake.


S. No. Sample Moisture uptake
1. F1 2.98%
2. F2 2.11%
3. F3 2.75%
4. F4 2.56%

www.wjpr.net Vol 9, Issue 5, 2020. 1152


Sahu et al. World Journal of Pharmaceutical Research

Moisture contents in various formulated patch were determined. It shows that F1 (2.98%) has
more moisture content and F2 (2.11%) shows less moisture content.

4.2.8 Moisture Loss


Moisture loss of prepared transdermal patch F1 to F4 were determined. The results are
tabulated in Table 12.

Table 12: Determination of Moisture Loss.


S. No Sample Moisture Loss
1. F1 0.82%
2. F2 0.79%
3. F3 0.98%
4. F4 0.95%

Moisture loss were determined and found that F3 (0.98%) shows more moisture loss and F2
(0.79%) shows less loss in moisture.

4.2.9 Content Uniformity Test


The Content uniformity of the samples are given below in the Table 13.

Table 13: Determination of Content Uniformity Test.


Content
S. No Sample
Uniformity
1 F1 96.4%
2 F2 98%
3 F3 95.0%
4. F4 97%

4.2.10. Drug Content


Drug content determination of F1 to F4 formulations were measured spectrophotometrically
at 284 nm. The drug content is calculated and results are tabulated in Table 14.

Table 14: Determination of Drug Content.


S. No Sample Drug content
1. F1 91%
2. F2 98%
3. F3 97%
4. F4 94%

Drug content were determined. It shows that F2 with (98%) drug content and F1 (91%)
shows less drug content.

www.wjpr.net Vol 9, Issue 5, 2020. 1153


Sahu et al. World Journal of Pharmaceutical Research

4.2.11. Stability Studies


The Stability of the various formulations at different period of time and in different
temperature are tabulated below in the Table.15- 18.

Table 15: Stability data after 7 days.


S. Temperature ℃
Sample
No. 2-4℃ 20-25℃ 35-40℃
1. F1 Stable Stable Stable
2. F2 Stable Stable Stable
3. F3 Stable Unstable Stable
4. F4 Stable Stable Unstable

Table 16: Stability data after 14 days.


S. Temperature ℃
Sample
No. 2-4℃ 20-25℃ 35-40℃
1. F1 Stable Stable Unstable
2. F2 Stable Stable Stable
3. F3 Stable Unstable Unstable
4. F4 Stable Unstable Stable

Table 17: Stability data after 21 days.


S. Temperature ℃
Sample
No. 2-4℃ 20-25℃ 35-40℃
1. F1 Stable Stable Unstable
2. F2 Stable Stable Stable
3. F3 Unstable Unstable Unstable
4. F4 Stable Unstable Unstable

Table 18: Stability data after 28 days.


S. Temperature ℃
Sample
No. 2-4℃ 20-25℃ 35-40℃
1. F1 Stable Stable Unstable
2. F2 Stable Stable Stable
3. F3 Stable Unstable Unstable
4. F4 Unstable Unstable Unstable

4.2.12 In-Vitro Drug Diffusion Study


In-vitro drug diffusion studies are performed Franz diffusion cell. Figure 4. Shows the
working process of Franz diffusion cell. Sample are withdrawn at different time interval
shown in Table 19. And analysed spectrophotometrically at 320 nm against blank. Figure 5.
Shows graphical representation of In-vitro study.

www.wjpr.net Vol 9, Issue 5, 2020. 1154


Sahu et al. World Journal of Pharmaceutical Research

Figure 4: Working of Franz Diffusion Cell.

Table 19: Drug absorption range determined spectrophotometrically.


Absorption Range
Time in
F1 F2 F3 F4
minutes
05min. 0.110 0.119 0.105 0.157
10min. 0.201 0.239 0.215 0.208
15min. 0.371 0.371 0.308 0.367
20min. 0.458 0.481 0.399 0.467
25min. 0.458 0.615 0.399 0.547
30min. 0.458 0.730 0.399 0.632

Figure 5: Graphical representation data of drug absorption by the Franz diffusion cell.

www.wjpr.net Vol 9, Issue 5, 2020. 1155


Sahu et al. World Journal of Pharmaceutical Research

5. CONCLUSION
Transdermal application is one of the most promising method over the conventional form.
Patch of Diclofenac sodium was successfully prepared with different polymers by solvent
casting method. The present studies were helped in understanding the effect of formulation
process variables especially the concentration of different polymers on the drug release
profile. This study is further aimed to perform in-vivo studies for the concentration of
Diclofenac sodium reaching into the skin and to study its effect, which will help to avoid the
first pass metabolism and to make novel transdermal dosage form.

Evaluation parameters like physical appearance, uniformity of weight, thickness, folding


endurance, moisture content, drug content and diffusion study of formulations F1-F4 were
found to be acceptable. The evaluation studies shows that the patch formulation F2 having
less thickness, high folding endurance, less moisture content, and have finest uniformity of
weight characteristic as compared to other formulations. Parallel it also consumes more drug
content than other formulations.

6. ACKNOWLEDGMENT
Authors want to acknowledge the facilities provided by the Shri Shankracharya Technical
Campus, Faculty of Pharmaceutical Sciences, Junwani, Bhilai, and Chhattisgarh. Special
thanks to Dr. A. k. Jha Sir for providing the lab to perform work.

List of Tables
Table 1. Ingredients Table of Diclofenac Sodium Patch.
Table 2.Composition of Diclofenac Sodium Transdermal Patch in Different Ratio.
Table 3. Concentration and Absorption of Diclofenac Sodium.
Table 4. Physical Appearance of Patch.
Table 5. Determination of Thickness of Patch.
Table 6. Determination of Uniformity of Weight.
Table 7. Determination of Folding Endurance.
Table 8. Determination of Surface pH.
Table 9. Determination of Swelling Index after 1 hour.
Table 10. Determination of Swelling Index after 2 hour.
Table 11. Determination of Moisture Uptake.
Table 12. Determination of Moisture Loss.
Table 13. Determination of Content Uniformity Test.

www.wjpr.net Vol 9, Issue 5, 2020. 1156


Sahu et al. World Journal of Pharmaceutical Research

Table 14. Determination of Drug Content.


Table 15. Stability data after 7 days.
Table 16. Stability data after 14 days.
Table 17. Stability data after 21 days.
Table 18. Stability data after 28 days.
Table 19: Drug absorption range determined spectrophotometrically.

List of Figures
Figure 1. Preparation of Transdermal Patch by Solvent Casting Method.
Figure 2. Standard Graph of Diclofenac Sodium.
Figure 3. Transdermal Patch.
Figure 4. Working of Franz Diffusion Cell.
Fig 5. Graphical representation data of drug absorption by the Franz diffusion cell.

REFERENCES
1. Andrew GP, Laverty TP, Jones DS. Mucoadhesive polymeric platforms for controlled
drug delivery. European Journal of Pharmaceutics and Biopharmaceutics, 2009; 71(3):
505-1.
2. Brown MB, Martin GP, Jones SA, Akomeah FK. Dermal and transdermal drug delivery
system: current and future prospects. Drug delivery, 2006 Jan 1; 13: 175-87.
3. Patel RP, Patel G, Baria A. Formulation and evaluation of transdermal patch of
aceclofenac. Research Journal of Pharmaceutical Dosage FORM and Technology, 2009;
(2): 108-15.
4. Kusum Devi V, Saisivam S, Maria GR, Deepti PU. Design and evaluation of matrix
diffusion controlled transdermal patch of verapamil hydrochloride: Drug Development
and industrial pharmacy, 2003 Jan 1; 29(5): 495-503.
5. Pokala M, Pawarkar K, Tammira KR, Mamidi P, Racha U, Samudrala L. Formulation
and evaluation of transdermal patches of salbutamol. World Journal of Pharmacy and
Pharmaceutical Sciences, 2016; 5(6): 1358-73.
6. Latheeshjlal L, Phanitejaswini P, Soujanya Y, Swapna U, Sarika V, Moulika G.
Transdermal drug delivery systems: an Overview. Int. J. of Pharm Tech. Res, 2011; 3(4):
2140-8.

www.wjpr.net Vol 9, Issue 5, 2020. 1157


Sahu et al. World Journal of Pharmaceutical Research

7. Rathore B, Mahdi AA, Paul BN, Saxena PN, Das SK. Indian herbal medicines: Possible
potent therapeutic agents for rheumatoid arthritis. Journal of clinical biochemistry and
nutrition, 2007 Jul; 41(1): 12.
8. Park YG, Ha CW, Han CD, Bin SI, Kim HC, Jung YB, Lim HC. A prospective,
randomized, double-blind, multicenter comparative study on the safety and efficacy of
Celecoxib and GCSB-5, dried extracts of six herbs, for the treatment of osteoarthritis of
knee joint. Journal of ethnopharmacology, 2013 Oct 7; 149(3): 816-24.
9. Prausnitz M, Mitragotri S, Langer R. Current status and future potential of transdermal
drug delivery. Nature Reviews Drugs discovery, 2004 Feb; 3(2): 115-24.
10. Díaz‐González F, Sánchez‐Madrid F. NSAIDs: learning new tricks from old drugs.
European journal of immunology, 2015 Mar; 45(3): 679-86.
11. Newton RF, Roberts SM, editors. Prostaglandins and Thromboxanes: Butterworths
Monographs in Chemistry. Butterworth-Heinemann, 2016 Jan 21.
12. Murthy SN, Shobha R, Hiremath, Paranjothy KL. Evaluation of carboxymethyl guar
films for the formulation of transdermal therapeutic systems. International Journal of
pharmaceuticals, 2004 Mar 19; 272(1-2): 11–8.
13. Keith AD. Polymer matrix consideration for transdermal devices. Drug development and
industrial pharmacy, 1983 Jan 1; 9(4): 605-25.
14. Bertocchi P, Antoniella E, Valvo L, Alimonti S, Memoli A. Diclofenac sodium
multisource prolonged release tablets- a comparative study on the dissolution profiles.
Journal of pharmaceuticals and biomedical analysis, 2005 Apr 1; 37(4): 679-85.
15. Buritova J, Besson JM. Dose-related antiinflammatory/analgesic effects of lornoxicama: a
spinal c-Fos protein study in the rat: Inflammation Research, 1998 Jan 1; 47(1): 18-25.
16. Begum AS, Sravya AH, Deepak BG, Manasa NG, Srujana M, Uma V, Tejeswari LS,
Padmalatha K. Formulation and Evaluation of Fexofenadine Buccal Mucoadhesive Patch
of Captopril. Research Journal of Pharmacy and Technology, 2018 Nov 1; 11(11):
4892-8.
17. Bhilegaonkar SP, Volvoikar SG, Naik AG. Formulation and Evaluation of Bilayer Gastric
Mucoadhesive Patch of Captopril. Research Journal of Pharmacy and Technology, 2018
Jun 1; 11(6): 2444-53.
18. Manoga K, Shilpa TN. Development of Medicated Topical Patches in Transdermal drug
delivery system. Research Journal of Pharmacy and Technology, 2018; 11(1): 397-404.

www.wjpr.net Vol 9, Issue 5, 2020. 1158


Sahu et al. World Journal of Pharmaceutical Research

19. Samanthula KS, Satla SR, Bairi AG. Development, In-Vitro and Ex-Vivo Evaluation of
Muco-adhesive Buccal patches of Candesartan cilexetil. Research Journal of Pharmacy
and Technology, 2019; 12(6): 3038-44.
20. Lal N, Verma N. Development and Evaluation of Transdermal Patches containing
Carvedilol and Effect of Penetration Enhancer on Drug Release. Research Journal of
Pharmacy and Technology, 2018 Feb 1; 11(2): 745-52.
21. Roy A, Wahane A, Karankal S, Sharma P, Khutel D, Singh O, Shardul V, Pitamber,
Sabha N, Dewangan J, Dewangan A, Jangde A, Rani C, Sahu T, Tripathi D. K., Agrawal
M, Ajazuddin, Sahu G, Alexander A. REVIEW ARTICLE Pharmaceutical Aspects on the
Formulations of Hydrogel: An Update. Research Journal of Pharmaceutical Dosage
Forms and Technology. January- March, 2018; 1-6.
22. Kokima M, Kojima T, Suzuki S, Oguchi T, Oba M, Tsuchiya H, et al. Depression,
inflammation, and pain in patients with rheumatoid arthritis. Arthritis Rheum, 2009; 61:
1018–24.
23. Rathbun A, Harrold R, Reed G. A description of patient and rheumatologist-reported
depression symptoms in an American rheumatoid arthritis registry population. Clin Exp
Rheumatol, 2014; 32: 523–32.
24. Pincus T, Sokka T, Wolfe F. Premature mortality in patients with rheumatoid arthritis.
Arthritis Rheum, 2001; 44: 1234–6.
25. Dickens C, McGowan L, Clark D, Creed F. Depression in rheumatoid arthritis: a
systematic review of the literature with meta-analysis. Psychosom Med, 2002; 64: 52–60.
26. Van der Helm-van Mil AHM, Verpoort KN, Breedveld FC, Toes REM, Huizinga TWJ.
Antibodies to citrullinated proteins and diff erences in clinical progression of rheumatoid
arthritis. Arthritis Res Ther, 2005; 7: R949–58.
27. Van der Woude D, Houwing-Duistermaat JJ, Toes RE, et al. Quantitative heritability of
anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-
negative rheumatoid arthritis. Arthritis Rheum, 2009; 60: 916–23.
28. Barton A, Worthington J. Genetic susceptibility to rheumatoid arthritis: an emerging
picture. Arthritis Rheum, 2009; 61: 1441–46.
29. Orozco G, Eyre S, Hinks A, et al. Association of CD40 with rheumatoid arthritis
confirmed in a large UK case-control study. Ann Rheum Dis, 2010; 69: 813–16.
30. Stahl EA, Raychaudhuri S, Remmers EF, et al. Genome-wide association study meta-
analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet, 2010; 42: 508–14.

www.wjpr.net Vol 9, Issue 5, 2020. 1159


Sahu et al. World Journal of Pharmaceutical Research

31. Plenge RM. Recent progress in rheumatoid arthritis genetics: one step towards improved
patient care. Curr Opin Rheumatol, 2009; 21: 262–71.
32. Sahu G, Sharma H, Gupta A, Kaur C. Review Article Advancements in Microemulsion
Based Drug Delivery Systems for Better Therapeutic Effects. International Journal of
Pharmaceutical Sciences and Developmental Research, 31 August, 2015; 1-8.
33. Tanner T, Marks R. Delivering drugs by the transdermal route: review and comment.
Skin Research and Technology, 2008 Aug; 14(3): 249-60.
34. Irfan M, Verma S, Ram A. Preparation and characterization of ibuprofen loaded
transferosome as a novel carrier for transdermal drug delivery system. Asian journal of
pharmaceutical and clinical research, 2012; 5(3): 162-5.
35. Saroha K, Yadav B, Sharma B. Transdermal patch: a discrete dosage form. Int J Curr
Pharm Res, 2011; 3(3): 98-108.
36. Graf BA, Milbury PE, Blumberg JB. Flavonols, flavones, flavanones, and human health:
epidemiological evidence. Journal of medicinal food, 2005 Sep 1; 8(3): 281-90.
37. Darwhekar G, Jain DK, Patidar VK. Formulation and evaluation of transdermal drug
delivery system of clopidogrel bisulfate. Asian J. Pharm. Life Sci. ISSN, 2011; 2231:
4423.
38. Karthik V, Saravanan K, Bharathi P, Dharanya V, Meiaraj C. An overview of treatments
for the removal of textile dyes. Journal of Chemical and Pharmaceutical Sciences, 2014;
7(4): 301-7.
39. Selvam RP, Singh AK, Sivakumar T. Transdermal drug delivery systems for
antihypertensive drugs-A review. Int J Pharm Biomed Res, 2010 Feb; 1(1): 1-8.
40. Shah SS, Rahul J, Prabhakar P. Formulation and evaluation of transdermal patches of
papaverine hydrochloride. Asian Journal of Pharmaceutics (AJP): Free full text articles
from Asian J Pharm, 2014 Sep 24; 4(1).
41. Chen X, Peng L, Gao J. Novel topical drug delivery systems and their potential use in
scars treatment. Asian Journal of Pharmaceutical Sciences, 2012 Jun 1; 7(3).
42. Pamornpathomkul B, Duangjit S, Laohapatarapant S, Rojanarata T, Opanasopit P,
Ngawhirunpat T. Transdermal delivery of fluorescein isothiocyanate-dextrans using the
combination of microneedles and low-frequency sonophoresis. asian journal of
pharmaceutical sciences, 2015 Oct 1; 10(5): 415-24.
43. David, SR, Rajabalaya R, Zhia ES. Development and in vitro evaluation of self-adhesive
matrix type transdermal delivery system of ondansetron hydrochloride. Tropical Journal
of Pharmaceutical Research, 2015; 14(2): 211-8.

www.wjpr.net Vol 9, Issue 5, 2020. 1160


Sahu et al. World Journal of Pharmaceutical Research

44. Gowda DV, Rajesh N, Somashekhara CN. Siddaramaiah. Development and evaluation of
aceclofenac loaded transdermal film. Int. J. Pharm Tech Res, 2010; 2: 2224-33.
45. Patel RP, Patel G, Baria A. Formulation and evaluation of transdermal patch of
Aceclofenac. International Journal of Drug Delivery, 2010; 1: 41-51.
46. Gaikwad AK. Transdermal drug delivery system: Formulation aspects and evaluation.
Comprehensive Journal of Pharmaceutical Science, 2013 Feb; 1(1): 1-10.
47. Sanap GS, Dama GY, Hande AS, Karpe SP, Nalawade SV, Kakade RS, et al. Preparation
of transdermal monolithic systems of indapamide by solvent casting method and the use
of vegetable oils as permeation enhancer: Int J Green Pharm, 2008; 2: 129-33.
48. Murthy TEGK, Kishore VS. Effect of casting solvent on permeability of antihypertensive
drugs through ethyl cellulose films: J Sci Ind Res, 2008; 67: 147-50.
49. Kumar SS, Behury B, Sachinkumar P. Formulation and evaluation of transdermal patch
of Stavudine. Dhake University Journal of Pharmaceutical Sciences, 2013 Sep 2; 12(1):
63-69.
50. Barhate SD, Potdar MB. Formulation of transdermal patch of Carvedilol by using novel
polymers. Pharmacia, 2012; 2: 185-9.
51. Bhavani PP, Kumar PR, Shankar RK, Santosh T. Formulation and evaluation studies on
transdermal dosage forms of diclofenac sodium. World J Pharm Pharm Sci, 2015; 4:
1043-63.
52. Patel VM, Prajapati BG, Patel MM. Effect of hydrophilic polymer on buccoadhesive
Eudragit patchs of propranolol hydrochloride using factorial design. AAPS Pharm Sci
Tech, 2007 jun 1; 8(2): E119-26.
53. Ammar HO, Ghorab M, El-Nahhas SA, Kamel R. Polymeric matrix system for prolonged
delivery of tramadol hydrochloride, part Ⅰ: physicochemical evaluation. Aaps Pharma
Sci Tech, 2009 Mar 1; 10(1): 7-20.
54. Pichayakorn W, Suksaeree J, Boonme P, Amnuaikit T, Taweepreda W, Ritthidej GC.
Nicotine transdermal patches using polymeric natural rubber as the matrix controlling
system: effects of polymer and plasticizer blends. Journal of membrane science, 2012 Sep
1; 411: 81-90.
55. Ubaidulla U, Reddy MV, Ruckmani K, Ahmad FJ, Khar RK. Transdermal therapeutic
system of carvedilol: effect of hydrophilic and hydrophobic matrix on in vitro and in vivo
characteristics. AAPS Pharm Sci Tech, 2007 Mar 1; 8(1): E13-20.

www.wjpr.net Vol 9, Issue 5, 2020. 1161


Sahu et al. World Journal of Pharmaceutical Research

56. Keleb E, Sharma RK, Mosa EB, Aljahwi A-AZ. Transdermal drug delivery system-
design and evaluation. International Journal of Advances in Pharmaceutical Sciences,
2010; 1: 201-211.
57. Aggarwal G, Dhawan S. Development, fabrication and evaluation of transdermal drug
delivery system –A review. Pharmainfo.net, 2009; 7(5): 1-28.
58. Prajapati ST, Patel CG, Patel CN. Formulation and evaluation of transdermal patch of
repaglinide. ISRN pharmaceutics, 2011 Jul 20; 2011.
59. Vyas SP, Khar RK. Targetted and controlled drug delivery novel carrier system. 1st ed.
CBS Publisher and distributors New Delhi, 2002; 411-447.
60. Vyas SP, Khar RK. Targetted and controlled drug delivery novel carrier system. 1st ed.
CBS Publisher and distributors New Delhi, 2002; 411-447.
61. Santoyo S, Arellano A, Ygartua P, Martin C. Penetration enhancer effects on the in vitro
percutaneous absorption of piroxicam through rat skin. International Journal of
Pharmaceuticals, 1995 Apr 18; 117(2): 219-24.
62. Dureja H, Tiwary AK, Gupta S. Simulation of skin permeability in chitosan membranes.
International Journal of Pharmaceuticals, 2001 Feb 1; 213(1-2): 193-8.
63. Ellaithy HM, El-Shaboury KM. The development of Cutina lipogels and gels
microemulsion for topical administration of fluconazole. Aaps Pharmscitech, 2002 Feb 1;
3(4): 77-85.
64. Ansari N, Bharti R, N. Abdul Kader N.S, Mandavi N, Verma S, Kaur C.D, Sahu G and
Sharma H. Review article skin penetration enhancement in novel drug delivery system.
World journal of pharmacy and pharmaceutical sciences, 2018; 6: 1-19.
65. Tripathi S, Sahu U, Meshram J, Kumari R, Tripathi D.K , Ajazuddin, Alexander A,
Sharma H, Sahu G. Research article Formulation and characterization of virgin coconut
oil emulsion (vcoe) for treatment of alzheimer’s disease. Research Journal of
Pharmaceutical Dosage Forms and Technology, 2018; (02): 1-6.
66. Gupta S, Sahu G, Sharma M, Chandrakar S, Sahu V, Sharma G, Dewangan K Solanki H,
Majumdar M, Tripathi D.K, Alexander A, Ajazuddin. Preparation and Optimization of
floating microbeads of ciprofloxacin HCl. Research J. Pharm. and Tech, 2016; 9(7): 1-5.
67. Dapurkar V, Sahu G, Sharma H, Meshram S, Rai G. Research Article Anti-Arthritic
Activity of Roots Extract of Boerhaavia Diffusa in Adjuvant Induced Arthritis Rats.
Scholars Academic Journal of Pharmacy (SAJP). Sch. Acad. J. Pharm, 2013; 2(2):
107-109.

www.wjpr.net Vol 9, Issue 5, 2020. 1162


Sahu et al. World Journal of Pharmaceutical Research

68. Sahu G, Sharma H, Gupta A, Kaur C. Review Article Advancements in Microemulsion


Based Drug Delivery Systems for Better Therapeutic Effects. International Journal of
Pharmaceutical Sciences and Developmental Research, 2015; 1-8.
69. Kader N S A, Ansari N, Bharti R, Mandavi N, Sahu G, Jha A K, Sharma H; Review
article; Novel Approaches for Colloidal Drug Delivery System: Nanoemulsion; Research
Journal of Pharmaceutical Dosage Forms and Technology, 2018; 10(4).
70. Sharma H, Dapurkar V K, Rai G, Sahu G K; Research article; Microemulsions for the
Topical Administration of 5-Fluorouracil: Preparation and Evaluation Research J. Pharm.
and Tech, 2012; 5(8).
71. Mandavi N, Ansari N, Barti R, Kader S A, Sahu G, Sharma H; Review article,
Microemulsion: A Potential Novel Drug Delivery System; Research Journal of
Pharmaceutical Dosage Forms and Technology, 2018; 10(4).
72. Sahu S k, Ajazuddin, Banjare T, Gupta S, Bhandarkar A, Sahu H, Diwedi S.D, Sahu P,
Agrawal P, Yadav P, Bhatt A, Sahu K, Dewangan D, Thapa H, Deepika, Sahu G, Sharma
M, Tripathi D.K., Alexander A; Research article; Formulation and evaluation of
orodispersible tablet of montelukast sodium; Research J. Pharm. and Tech, March 2018;
11(3).
73. Sahu, G., Sharma H, Dapurkar V, Rai G; Research article; Development And Evaluation
of Methotraxate Loaded BSA Microspheres; Int. Res J Pharm. App Sci, 2012; 2(5): 9-12.
74. Sahu G, Sharma H, Kaur C.D., Review article; A Novel Approach of Magnetic
Modulated Microspheres; Asian J. Pharm. Res, 2013; 3(4): 220-224.

www.wjpr.net Vol 9, Issue 5, 2020. 1163

You might also like