Vous êtes sur la page 1sur 18

Continental J.

Pharmaceutical Sciences 3: 1 - 6, 2009


©Wilolud Online Journals, 2009.

EFFECT OF NATURAL BIODEGRADABLE AND SYNTHETIC POLYMER FOR GASTRIC DISEASE BY


FLOATING MICROSPHERES

A. K. Jain1, C. P. Jain1, K. Gaur2, A. Kakde3, M. Meena4, R. K. Nema5


1
Department of Pharmaceutical Sciences, M.L.S. University, Udaipur, Rajasthan, India
2
Geetanjali College of Pharmaceutical Studies, Manwa Kheda, Udaipur, Rajasthan, India
3
Vinayaka mission university, Selam, Tamilnadu, India
4
I C M R, Delhi, India, 5Rishiraj College of Pharmacy, Indore, Madhya Pradesh, India

ABSTRACT
The present study involves preparation and evaluation of floating microspheres with
famotidine as model drug for prolongation of gastric residence time. The microspheres were
prepared by the solvent evaporation method using polymers acrycoat S100 and chitosan. The
shape and surface morphology of prepared microspheres were characterized by optical and
scanning electron microscopy, respectively. In vitro drug release studies were performed and
drug release kinetics was evaluated using the linear regression method. Effects of natural
biodegradable polymer chitosan and another acrycoat S 100 polymer on the size of
microspheres, incorporation efficiency and in-vitro drug release were also observed. The
prepared microspheres exhibited prolonged drug release more than 18 h, incorporation
efficiency was change due to increase concentration of poly vinyl alcohol solution but
remained buoyant for more than 12 h because clumping of polymers on surface of
microspheres. The mean particle size increased and the drug release rate decreased at higher
polymer concentration. In vitro studies demonstrated diffusion-controlled drug release from
the prepared floating microspheres.

KEYWORDS: Floating microspheres, famotidine, in vitro release, bioavailability

INTRODUCTION
Floating drug delivery systems developed to increase the gastric residence time of dosage forms for gastric disease
(Seth, 1984; Moes, 1993; Deshpande et al., 1996). The multiple unit system has been developed to identify the merit
over single unit dosage form because the single-unit floating systems are more popular but have a disadvantage
owing to their ‘all-or-nothing’ emptying process leading to high variability of the gastrointestinal transit time
(Whitehead et al., 1998; Talukder and Fassihi, 2004). Still, the multiple-unit dosage forms may be better suited
because they are claimed to reduce the intersubject variability in absorption and lower the probability of dose
dumping (Rouge et al., 1997). Such a dosage form can be distributed widely throughout the gastrointestinal tract,
affording the possibility of a longer lasting and more reliable release of the drug from the dosage form (Sato et al.,
2003). The synthetic polymers have been used to prepare floating microspheres. Kawashima et al. prepared hollow
microspheres or microballoons of ibuprofen by the emulsion-solvent diffusion method using acrylic polymers
(Kawashima et al., 1992). The microspheres exhibited good in vitro floatability and drug release decreased
drastically with increasing polymer concentration. Floating microspheres of cellulose acetate loaded with three
different drugs were prepared using the solvent diffusion-evaporation method (Soppimath et al., 2002). The
microspheres remained buoyant for more than 12 h. Methylcellulose and chitosan micropellets loaded with
lansoprazole had a lower density than gastric contents and exhibited better encapsulation efficiencies (Muthusamy et
al., 2005). Other polymer solution systems that have been used to prepare floating microspheres are
polycarbonate/dichloromethane (Thanoo et al., 1993; Joseph et al., 2002), cellulose acetate butyrate/Eudragit RL100
mixture in acetone (Stithit et al., 1998) and Eudragit S100/i-propanol (Lee et al., 1999). Famotidine was used as
model drug. It is an antihistaminic drug that has been widely used in treating gastric and duodenal ulceration and
also in zollinger ellison syndrome and reflux esophagitis (Gladiziwa and Klotz, 1993). It is poorly absorbed from the
lower gastrointestinal tract and has a short elimination half life (3 h). The objective of the present study was to
develop floating microspheres of famotidine in order to achieve an extended retention in the upper gastrointestinal
tract, which may result in enhanced absorption and thereby improved bioavailability. The prepared microspheres
were evaluated for particle size, in vitro release, buoyancy and incorporation efficiency. The effect of various
formulation variables on the size of microspheres, incorporation efficiency and drug release was investigated.

1
A. K. Jain et al: Continental J. Pharmaceutical Sciences 3: 1 - 6, 2009

EXPERIMENTAL
Materials and Apparatus
Famotidine was obtained as a gift sample from Intas Pharmaceutical, Ahmedabad. Polyvinyl alcohol was obtained
from S.D. Fine Chemicals Ltd. Mumbai. Dichloromethane, ethyl acetate, acetone, acrycoat S100, chitosan were
obtained from Central Drug House (Pvt) Ltd. Delhi. All other chemicals/reagents were used of analytical grade.

Table 1: Different formulations of floating microspheres


Polyvinyl
Drug: Polymer
S. No. Code Organic solvent system alcohol
ratio
(%w/v)
1. C1* 1:1 Dichloromethane: Ethanol 1:1 0.25
2. C2 1:2 Dichloromethane: Ethanol 1:1 0.25
3. C3 1:3 Dichloromethane: Ethanol 1:1 0.25
3. C4* 1:1 Dichloromethane: Ethanol 1.:1 0.5
3. C5* 1:1 Dichloromethane: Ethanol 1:1 1.0
4. A1 1:1 Dichloromethane: Ethanol 1:1 2.0
5. A2 1:2 Dichloromethane: Ethanol 1:1 2.0
6. A3 1:3 Dichloromethane: Ethanol 1:1 2.0
*Formulation coded with different concentration of polyvinyl alcohol (% w/v) using same drug:
polymer ratio

Table 2: Various formulation parameters for microspheres


Mean Incorporation Buoyancy Husner’s Carr’s index Angle of
Code particle efficiency percentageb ratio repose (θ)
sizea (%)b

375.1±1.818 86.15±0.044 80.6±2.1 1.66±0.023 14.285±0.345 23.41°±0.035


C1*
378.7±2.914 83.15±0.011 79.01±3.3 1.146±0.059 12.79±0.421 22.43°±0.581
C2
389.3±1.984 84.15±0.012 77.56±3.6 1.166±0.039 12.35±0.746 22.15°±0.351
C3
376.1±1.657 85.52±0.065 81.7±2.1 1.56±0.056 13.13±0.345 22.24°±0.028
C4*
379.1±1.546 84.38±0.038 82.8±2.1 1.42±0.073 13.06±0.254 21.85°±0.026
C5*
380.8±2.164 83.15±0.032 76.14±2.1 1.58±0.046 13.58±0.546 22.87°±0.124
A1
383.6±1.854 83.51±0.034 74.85±1.8 1.42±0.028 12.468±0.214 21.85°±0.256
A2
390.6±1.931 81.35±0.016 75.56±1.9 1.35±0.075 11.845±0.587 20.85°±0.428
A3
a
Mean±SD, n=10.bMean±SD, n=3.

Preparation of microspheres
Microspheres were prepared by the solvent evaporation emulsification technique as employed by Struebel et al.
(Struebel et al., 2003). Famotidine as model drug and polymers such as acrycoat S100 (batch A1-A3) or chitosan
(batch C1-C5) was dissolved in a mixture of solvent system at room temperature to be prepare different formulation
systems (Table 1). This dispersed medium was poured into 150 mL 0.1 M acidic solution containing polyvinyl
alcohol as continuous medium at room temperature and subsequently stirred at ranging agitation speed for 2 h to

2
A. K. Jain et al: Continental J. Pharmaceutical Sciences 3: 1 - 6, 2009

allow the volatile solvent evaporate. The prepared microspheres were filtered, washed with distilled water and dried
in vacuum.

Characterization of microspheres
Size and shape of microspheres: The size of microspheres was determined using a microscope (Olympus NWF 10x,
Educational Scientific Stores, India) fitted with an ocular micrometer and stage micrometer. Scanning electron
microscopy (SEM) (Philips-XL-20, The Netherlands) was performed to characterize the surface of formed
microspheres. Microspheres were mounted directly onto the sample stub and coated with gold film (~ 200 nm)
under reduced pressure (0.133 Pa).

Flow properties: It was determined in terms of carr’s index (Ic) and hausner’s ratio (HR) using following equations:
HR=ρt/ ρb
Ic=ρt- ρb/ ρt

Figure 1: Scanning electron microscopy photographs of floating microspheres

The angle of repose (θ) of the microspheres, which measure the resistance to particle flow, was determined by fixed
funnel method by using following equation:
tan θ=2H/D

Where, 2H/D is the surface area of the free standing height of the microspheres heap that formed after making the
microspheres flow from the glass funnel.

Buoyancy percentage: Microspheres (0.3 g) were spread over the surface of a USP XXIV dissolution apparatus
(type II) filled with 150 ml 0.1 mol L–1 hydrochloric acid (USP, 2000). The medium was agitated with a paddle
rotating at 500 rpm for 12 h. The floating and the settled portions of microspheres were recovered separately. The
floated microspheres were collect by decantation and weighed.

Buoyancy % = Floated mass of prepared microspheres / Total mass of prepared microspheres


Incorporation efficiency: To determine the incorporation efficiency, prepared microspheres were taken, thoroughly
triturated in pestle mortar equivalent as drug used in formulation and suspended in a 5 ml of dichloromethane as
dissolving medium. The suspension was suitably diluted with water and filtered to separate shell fragments. Drug
content was analyzed spectrophotometrically at 265 nm.

In-vitro drug release: A USP basket apparatus has been used to study in vitro drug release from microspheres (Singh
and Kim, 2000; Dinarvand et al., 2002; Abrol et al., 2004). In the present study, drug release was studied using a
modified USP XXIV dissolution apparatus type I (basket mesh # 120, equals 125 µm) at 100 rpm in distilled water
and 0.1 mol L–1 hydrochloric acid (pH 1.2) as dissolution fluids (900 ml) maintained at 37 ± 0.5 °C. Withdrawn
samples (10 ml) were analyzed spectrophotometrically as stated above. The volume was replenished with the same

3
A. K. Jain et al: Continental J. Pharmaceutical Sciences 3: 1 - 6, 2009

amount of fresh dissolution fluid each time to maintain the sink condition. All experiments were performed in
triplicate. Linear regression was used to analyze the in vitro release mechanism.

Statistical analysis: Experimental results were expressed as mean ± SD. Student’s t-test and one-way analysis of
variance (ANOVA) were applied to check significant differences in drug release from different formulations.
Differences were considered to be statistically significant at P< 0.05.

RESULTS AND DISCUSSION


Floating microspheres were prepared by the solvent evaporation method by using acrycoat S100 and chitosan (Table
1). The SEM photographs showed that the fabricated microspheres were spherical with a smooth surface and
exhibited a range of sizes within each batch (Fig. 1). The microspheres floated for prolonged time over the surface
of the dissolution medium without any apparent gelation. Buoyancy percentage of the microspheres was shown in
Table 2.

Figure 2: Effect of chitosan polymeric concentration on in vitro drug release from floating microspheres (bars
represents mean±SD; n=3)

Microspheres were prepared using different polymers with gradually increasing polymer concentration and same
polymer with variability in concentration of PVA in combination with a fixed concentration of drug to assess the
effect of polymer concentration on the size of microspheres. The mean particle size of the microspheres significantly
increased with increasing cellulose acetate concentration (p < 0.05) and was in the range 375.1±1.818 to
389.3±1.984 µm (Table II). The viscosity of the medium increases at a higher polymer concentration resulting in
enhanced interfacial tension (Reddy et al., 1990; Ishizaka, 1981). The size of prepared microsphere was change due
to variability in concentration of PVA, as concentration of PVA increase size of microspheres increase but
incorporation efficiency was reduced. The same concentration of polymer with changing concentration of PVA,
buoyancy percentage and period was increase till to 18 h. In vitro FM release studies were performed in 0.1 mol/L
hydrochloric acid for 18 h. The cumulative release of FM significantly decreased with increasing polymer
concentration (p < 0.05, Fig. 2, 3) because of clumping of polymer on surface of microspheres. The increased
density of the polymer matrix at higher concentrations results in an increased diffusional path length. This may
decrease the overall drug release from the polymer matrix because higher gelation of surface. Furthermore, smaller
microspheres are formed at a lower polymer concentration and have a larger surface area exposed to dissolution
medium, giving rise to faster drug release. The data obtained for in vitro release were fitted into equations for the
zero-order, first-order and Higuchi release models (Costa and Lobo, 1987; Wagner, 1969; Schefter, 1963). The
interpretation of data was based on the value of the resulting regression coefficients. The in vitro drug release
showed the highest regression coefficient values for Higuchi’s model, indicating diffusion to be the predominant
mechanism of drug release. in-vivo floating behavior will be discussed in future for in-vitro and in-vivo co-relation.

4
A. K. Jain et al: Continental J. Pharmaceutical Sciences 3: 1 - 6, 2009

CONCLUSIONS
In vitro data obtained for floating microspheres of famotidine showed excellent floatability, good buoyancy and
prolonged drug release. Microspheres of different size and drug content could be obtained by varying the
formulation variables. Diffusion was found to be the main release mechanism. Thus, the prepared floating
microspheres may prove to be potential candidates for multiple-unit delivery devices adaptable to gastric disease.

ACKNOWLEDGEMENTS
The authors acknowledge the Director, Bhupal Noble’s College of Pharmacy, Udaipur, Rajasthan.

Figure 3: Effect of acrycoat S100 polymeric concentration on in vitro drug release from floating microspheres (bars
represents mean±SD; n=3)

REFERENCES
Abrol S, Trehan A, Katare OP. (2004): Formulation, characterization, and in vitro evaluation of silymarin-loaded
lipid microspheres. Drug Deliv., 11: 185-191.

Costa P, Lobo JMS. (1987): Modeling and comparison of dissolution profiles. Eur. J. Pharm. Sci., 39: 39-45.

Deshpande A, Rhodes CT, Shah NH, Malick AW. (1996): Controlled-release drug delivery systems for prolonged
gastric residence: an overview. Drug Dev. Ind. Pharm., 22: 531-539.

Dinarvand R, Mirfattahi S, Atyabi F. (2002): Preparation, characterization and in vitro drug release of isosorbide
dinitrate microspheres. J. Microencaps., 19: 73-81.

Gladiziwa U, Klotz U. (1993): Pharmacokinetics and pharmacodynamics of H2 receptor antagonists in patients with
renal insufficiency. Clin. Pharmacokinet., 24: 319-332.

Ishizaka T. (1981): Preparation of egg albumin microspheres and microcapsules. J. Pharm. Sci., 70: 358-361.

Joseph NJ, Lakshmi S, Jayakrishnan A. (2002): A floating-type oral dosage form for piroxicam based on hollow
polycarbonate microspheres: in vitro and in vivo evaluation in rabbits. J. Control. Rel., 79: 71-79.
Kawashima Y, Niwa T, Takechi H, Hino T, Itoh Y. (1992): Hollow microspheres for use as floating controlled drug
delivery systems in the stomach. J. Pharm. Sci., 81: 135-140.

Lee JH, Park TG, Choi HK. (1999): Development of oral drug delivery system using floating microspheres. J.
Microencaps., 16: 715-729.

Moes AJ. Gastroretentive dosage forms. (1993): Crit. Rev. Ther. Drug Carrier Syst., 10: 143-195.

Muthusamy K, Govindarazan G, Ravi TK. (2005): Preparation and evaluation of lansoprazole floating micropellets.
Ind. J. Pharm. Sci., 67: 75-79.

5
A. K. Jain et al: Continental J. Pharmaceutical Sciences 3: 1 - 6, 2009

Reddy BP, Dorle AK, Krishna DK. (1990): Albumin microspheres: effect of process variables on the distribution
and in vitro release. Drug Dev. Ind. Pharm., 16: 1781-1803.

Rouge N, Leroux JC, Cole ET, Doelker E, Buri P. (1997): Prevention of the sticking tendency of floating minitablets
filled into hard gelatin capsules. Eur. J. Pharm. Biopharm., 43: 165-171.

Sato Y, Kawashima Y, Takeuchi H, Yamamoto H. (2003): In vivo evaluation of riboflavin-containing microballoons


for floating controlled drug delivery system in healthy human volunteers. J. Control. Rel., 93: 39-47.

Schefter, Higuchi T. (1963): Dissolution behavior of crystalline solvated and non-solvated forms of some
pharmaceuticals. J. Pharm. Sci., 25: 781-791.

Seth PR, Tossounian J. (1984): The hydrodynamically balanced system HBSTM: A novel drug delivery system for
oral use, Drug Dev. Ind. Pharm., 10: 313-339.

Singh N, Kim KH. (2000): Floating drug delivery systems: an approach to oral controlled drug delivery via gastric
retention. J. Control. Rel., 63: 235-259.

Soppimath KS, Kulkarni AR, Rudzinski WE, Aminabhavi TM. (2002): Microspheres as floating drug-delivery
systems to increase gastric retention of drugs. Drug Metab. Rev., : 33: 149-160.

Stithit S, Chen W, Price JC. (1998): Development and characterization of buoyant theophylline microspheres with
near zero order release kinetics. J. Microencaps., 15: 725-737.

Struebel A, Siepmann J, Bodmeier R. (2003): Multiple units gastroretentive drug delivery systems: a new
preparation method for low density microspheres. J. Microencaps., 20: 329-347.

Talukder R, Fassihi R. (2004): Gastroretentive delivery systems: a mini review. Drug Dev. Ind. Pharm., 30: 1019-
1028.

Thanoo C, Sunny MC, Jayakrishnan A. (1993): Oral sustained-release drug delivery systems using polycarbonate
microspheres capable of floating on the gastric fluid. J. Pharm. Pharmacol., 45: 21-24.

The United States Pharmacopoeia XXIV, United States Pharmacopoeial Convention, Rockville; 2000. pp. 1941-
1943.

Wagner JG. (1969): Interpretation of percent dissolved-time plots derived from in vitro testing of conventional
tablets and capsules. J. Pharm. Sci., 58: 1253-1257.

Whitehead L, Fell JT, Collett JH, Sharma HL, Smith AM. (1998): Floating dosage forms: an in vivo study
demonstrating prolonged gastric retention. J. Control. Rel., 55: 3-12.

Received for Publication: 27/04/2009


Accepted for Publication: 12/05/2009

Corresponding Author:
Abhishek K. Jain
1
Department of Pharmaceutical Sciences, M.L.S. University, Udaipur, Rajasthan, India
E-mail address: akjainmlsu@gmail.com

6
Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009
©Wilolud Online Journals, 2009.

EVALUATION OF PHYSICO-CHEMICAL STABILITY PARAMETERS OF MEDROXYPROGESTRONE


ACETATE TABLET

A. Kakde1, A. K. Jain2, C. P. Jain2, M. Meena3, K. Gaur4 , R. K. Nema5


1
Plethico Pharmaceutical Pvt. Ltd., Indore, M.P., India.2 Department of pharmaceutical sciences, M.L.S. University,
Udaipur, Rajasthan, India.3 I C M R, Delhi, India.4 G C P S, Manwa Kheda, Udaipur, Rajasthan, India.5 Rishiraj
college of Pharm. Science, Indore, M. P., India.

ABSTRACT
There is a possibility that lower air, moisture and light protection could impact on physico-
chemical stability of medicines, this has not yet been proved. The objectives of the study were
to examine the physico-chemical stability of prepared medroxy progesterone acetate tablets
stored in a compliance aid at different temperature and humidity to simulate practice
conditions. Medroxy progesterone acetate 10 mg tablets in HDPE bottle were stored for six
months at 400C ± 20 and 75% ± 5 % RH / 300C ± 20C and 65% ± 5% RH / 250C and 60% ± 5%
RH. Physical tests were conducted to standards as laid down in the United State
Pharmacopoeia 2005, and dissolution to those of the United States Pharmacopoeia volume 24.
Chemically identified or assessed by a validated high-performance liquid chromatography
method. Tablets at 250C/60% RH room temperature in packaging the same in appearance and
passed physico-chemical tests. Although chemical stability was unaffected, storage in
compliance aids at 40°C with 75% relative humidity change nature of medroxy progesterone
acetate tablets.
KEYWORDS: Stability study, medroxy progesterone acetate, patient compliance, high
performance liquid chromatography.

INTRODUCTION
The Tripartite guideline, which has been developed within the Expert Working Group (Quality) of the International
Conference on Harmonisation (ICH), provides a general indication of the requirements for stability testing. It
primarily addresses the information required in applications for registration for new chemical entities and associated
medicinal products. This guideline is adopted with only minor modifications. It contains aspects relating to testing
conditions, numbers of batches to be tested and the requirements regarding follow-up stability data (Guidance for
Industry Q1A Stability testing of new drug substances and drug products, 2001). The Tripartite guideline latest
additions/ updates appear on the ICH, FDA and EMEA websites. Stability information from accelerated and long-
term testing is to be provided on at least three batches. The long-term testing should cover a minimum of 12 months
duration on at least three batches at the time of submission of the application for registration. The batches,
manufactured to a minimum of pilot plant scale, should be produced by the same synthesis route, and with a method
of manufacture and procedure, which simulates the final process to be used on a manufacturing scale. The length of
the studies and the storage conditions should be sufficient to cover the periods of storage, shipment, and subsequent
use. Application of the same storage conditions as applied to the drug product will facilitate comparative review and
assessment. Other storage conditions are allowed if they can be fully justified by the applicant. In particular,
temperature sensitive APIs should be stored under an alternative, lower temperature condition, which will then
become the designated long-term testing storage temperature.

The six months accelerated testing should then be carried out at a temperature at least 15 °C above this designated
long-term storage temperature (together with appropriate relative humidity conditions for that temperature). The
designated long-term testing conditions will be reflected in the labelling and retest date (CPMP Note for Guidance
on stability testing: Stability testing of new drug substances and products, 2003). Where “significant change” occurs
during six months of storage under conditions of accelerated testing (40 oC ± 2 oC/75 % RH ± 5 %), additional
testing at an intermediate storage condition (such as 30 oC ± 2 oC/65 % ± 5 % RH), should be conducted. This
applies to APIs that will be used in dosage forms which will be subjected to long-term testing at 25 oC/60 % RH.
This information should also be included

7
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

Table 1: storage conditions and minimum time period at 40 oC/75 % RH


Testing parameters Storage conditions Minimum time period at
submission
Long-term testing 25 ± 2 oC/60 ± 5 % RH 12 months
Accelerated testing 40 ± 2 oC/75 ± 5 % RH 6 months

Table 2: storage conditions and minimum time period for stability study at 30 oC/65 % RH
Testing parameters Storage conditions Minimum time period at
submission
Long-term testing 25 ± 2 oC/60 ± 5 % RH 6 months
Accelerated testing 40 ± 2 oC/75 ± 5 % RH 3 months

Table 3: Formulae of medroxyprogestrone actetate tablet 10 mg


QUANTITY MG /
NAME OF INGREDIENT USES
TABLET
1. Dry Mixing/Granulation
Medroxyprogesterone Acetate USP 10.14 Active drug
Dibasic Calcium Phosphate 30 Diluent
Lactose (DCL 15) 31 Diluent
Maize Starch 35.46 Diluent
Sodium Lauryl Sulphate 12 Surfactant
Crosscarmellose Sodium ( Ac-di-Sol) 5 Super disintegrant
Polyplasdone XL ( Crosspovidone XL) 12 Disintegrant
Polyvinyl pyrilidone K-30 16 Binder
Iso Propyl Alcohol q.s. Solvent
2. Blending/Lubrication
Purified Talc 1.2 Lubricant
Magnesium Stearate 1.2 Lubricant
Aerosil( Colloidal silicon Dioxide) 1 Glident
Crosscarmellose Sodium ( Ac-di-Sol) 5 Guper disintegrant
3. Total Weight of Tablet 160 mg

in the application for registration. The initial application should include a minimum of 6 months’ data from a 12-
month study. "Significant change" at 40 oC/75 % RH or 30 oC/65 % RH, is defined as failure to meet the specified
requirements. (Table 1) The long-term testing will be continued for a sufficient period of time beyond 12 months to
cover all appropriate retest periods. The additional data can be submitted to the Council during the assessment
period of the application. The data (from accelerated testing or from testing at an intermediate condition) may be
used to evaluate the impact of short-term excursions outside the label storage conditions, such as may occur during
shipping. Long-term stability studies can also be performed at 30 oC/65 % RH, in which case additional data at
intermediate conditions are not required (Zones III & IV).

The long-term testing should cover at least 6 months duration at the time of submission (Table 2). The containers are
to be used in the long-term, real-time stability evaluation, should be the same as, or closely simulate, the actual
packaging, to be used for storage and distribution. Any evaluation should not only cover the assay of the API, but
also the levels of degradation products and other appropriate attributes. When degradation products are identified in
significant quantities (0.1 % or more) or suspected of toxicity, a concerted effort has to be made to collect the
following additional information about the substance concerned:
1. Chemical structure
2. Cross-reference to any available information about biological effect and significance at the concentrations
likely to be encountered
3. Procedure for isolation and purification
4. Mechanism of formation, including order of reaction

8
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

5. Physical and chemical properties


6. Specifications and directions for testing their presence at the concentration levels expected
7. Indication of pharmacological activity, or inactivity or toxicity profile

Where the route of degradation is not known, suitable screening chromatographic or other tests may be required.
The results of any toxicity studies, which may have been conducted, should be reported. Consideration should be
given to the stereo-chemical and polymorphic integrity of APIs. Stability information gained, should enable the
applicant to institute a routine system whereby re-analysis to validate conformance to specifications of the API, is
conducted to assure the stability of a particular dosage form.

Table 4: HPLC data of standard solution of medroxyprogesterone acetate at dissolution study


S. No. PARAMETERS STD
1. TOTAL 605876
2. MEAN 121175.2
3. STDEV 2291.678
4. RSD 1.691211

Table 5: HPLC data of standard and sample solution of medroxyprogesterone acetate at assay
S. No. PARMETERS STD SAMPLE
1. Total 56672505 21553800
2. Mean 11334501 10776900
3. STDEV 22332.408 173928.469
4. RSD 0.19703 1.6139007
5. Mg found 10 9.62

METHODS
Preparation of tablet:
Medroxy progesterone acetate USP tablet was prepared according to Table-1. All the excipients without magnesium
stearate, purified talc, aerosil and crosscarmellose sodium were mixed at time of blending of mass. The prepared
granules were compressed with single punch compression machine using 7.1 mm circular, concave with breakline
punches. (Table 3)

Storage of medroxy progesterone acetate tablets:


Prepared tablets of medroxy progesterone acetate 10 mg in HDPE bottle were stored for six months at 400C ± 20 and
75% ± 5 % RH / 300C ± 20C and 65% ± 5% RH / 250C and 60% ± 5% RH. Tablets were also stored for four weeks
in Petri dishes under ambient conditions.

Physical stability study:


The physical stability of medroxy progesterone acetate tablets was assessed three month after storage to United State
Pharmacopoeia standards, unless otherwise stated, according to the following methods.

1. Visual inspection of appearance:


This was assessed under standard laboratory lighting. Each of 10 tablets per condition as examined for colour,
texture and appearance.

2. Theoretical average weight:


Each of 10 tablets per condition was weighed separately on an analytical balance, accurate to five decimal places,
and the mass recorded. The weight increase or decrease in relation to that of tablets stored under condition A were
recorded as a percentage and expressed as the mean of 10 tablets.

9
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

3. Disintegration time:
Each of six tablets per condition was placed separately in the six cylinders of the disintegration apparatus with water
(37° ± 0.5°C) as the medium. Disintegration time was recorded as the time point corresponding to the breakdown of
all six tablets.

4. Hardness test:
Each of 10 tablets per condition was placed in turn on the platform of a manually hardness tester (Pfizer).

Table 6: Physico-chemical evaluation parameters of medroxyprogestrone acetate tablet 10 mg at I month


I
Month
S.No. Test Limits Initial
250C/60% 300C/65% 400C/75%
RH RH RH
White
circular
,biconvex
1 Description uncoated Complies complies complies Complies
tablet with
break line on
one side
Identification
2 Identification complies complies complies complies
by HPLC
Theortical
3 ±7.5% 163.09 mg 160.5 165.3 166.9
avg.wt.
85 % to 110
97.55%
Uniformity of %
4 to108.6 % NA NA NA
content RSD: NMT
RSD: 3.27%
6%
Disintegration 5.0 min
5 NMT 15 min 4 min 5 min 6 min
Time
Assay
6 95%-105% 100.69 % 101.72 100.59 96.77
1] 81.57
1] 85.43 1] 88.68 1] 97.06
2] 84.33
2] 91.68 2] 86.12 2] 99.66
3] 86.59
Dissolution 3] 84.28 3] 93.99 3] 86.94
7 NLT 75 % 4] 84.26
4] 89.76 4] 78.87 4] 83.53
5] 86.56
5] 85.42 5] 86.42 5] 87.27
6] 84.27
6] 85.62 6] 87.55 6] 86.69

Figure 1: Chromatogram of assay of medroxyprogesterone acetate tablet 10 mg

10
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

Figure 2: Chromatogram of dissolution profile of medroxyprogesterone acetate tablet 10 mg


Sample I D: Medroxyprogestroneacetate 10 mg 40°C with 75% RH, Injection Volume: 20 l
Detector: 254 nm

Table 7: Physico-chemical evaluation parameters of medroxyprogestrone acetate tablet 10 mg at II month

II
Month
S.no. Test Limits Initial
250C/60% 300C/65% 400C/75%
RH RH RH
White circular
,biconvex
1 Description uncoated tablet Complies complies complies complies
with break line
on one side
Identification
2 Identification complies complies complies complies
by HPLC
Theortical
3 ±7.5% 163.09 mg 164.3 163.9 162.4
avg.wt.
97.55%
85 % to 110 %
Uniformity of to108.6 %
4 RSD: NMT 6 NA NA NA
content RSD:
%
3.27%
Disintegration 5.0 min
5 NMT 15 min 5 min 5 min 5 min
Time
Assay
6 95%-105% 100.69 % 97.85 100.01 99.21
1] 81.57
1] 99.70 1] 97.4 1] 96.95
2] 84.33
2] 98.00 2] 95.80 2] 92.20
3] 86.59
Dissolution 3] 95.20 3] 93.81 3] 97.52
7 NLT 75 % 4] 84.26
4] 98.70 4] 95.20 4]101.94
5] 86.56
5] 99.60 5] 94.41 5] 91.07
6] 84.27
6] 100.4 6] 90.60 6] 96.60

5. Friability test:
The total mass of 10 tablets was determined before and after placing those in the clean drum of a friability
tester (Roche friabilator) operated at 25 rotations per minute for four minutes. (Aulton ME., 2002)

11
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

6. Dissolution test:
The dissolution medium, 900 mL 0.1 N HCl with 0.5 % sodiumlauryle sulphate and thermostated to 37.0 ±
0.5°C, was placed in each of six vessels of a dissolution test apparatus I.P. Type I with paddle speeds of 50

Table 8: Physico-chemical evaluation parameters of medroxyprogestrone acetate tablet 10 mg at III month


III
Month
S.no. Test Limits Initial
250C/60% 300C/65% 400C/75%
RH RH RH
White circular
,biconvex
1 Description uncoated tablet Complies complies complies complies
with break line
on one side
Identification
2 Identification complies complies complies complies
by HPLC
Theortical
3 ±7.5% 163.09 mg 160.8 163.9 160.8
avg.wt.
97.55%
85 % to 110 %
Uniformity of to108.6 %
4 RSD: NMT 6 NA NA NA
content RSD:
%
3.27%
Disintegration 5.0 min
5 NMT 15 min 4 min 4 min 6 min
Time
Assay
6 95%-105% 100.69 % 97.54 101.27 100.54
1] 81.57
1] 95.10 1] 91.30 1] 95.70
2] 84.33
2] 88.70 2] 92.50 2] 98.30
3] 86.59
Dissolution 3] 96.50 3] 95.30 3] 93.80
7 NLT 75 % 4] 84.26
4] 87.10 4] 88.40 4] 90.10
5] 86.56
5] 97.03 5] 94.00 5] 99.20
6] 84.27
6] 85.50 6] 94.30 6] 95.80

rotations per minute (rpm). The dissolution medium was sampled at different time intervals. The samples
were filtered with filter paper (filter pore size 0.45 µm) and assayed by using the HPLC protocol described
below. Dissolution was deemed acceptable where the concentration of medroxy progesterone acetate
released at 45 minutes was NLT 75% of the total labelled content.

High performance liquid chromatography (HPLC) variables:


Column: 150 X 3.95 µm Novapack C18
Mobile phase: Acetonitrile 600 ml + Water 400 ml
Dilution Factor: Std. 70 mg in 140 ml stock solution + dilute with water to 250 ml
Flow rate: 1
Injection volume: 20
Detector: UV 254
Chromatogram:
Retention time: 8.267 (Table 4)

12
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

7. High performance liquid chromatography assay:


Sample preparation: Grind tablets to a fine powder; weigh out amount equivalent to about 10 mg
medroxyprogesterone acetate, add 10 mL acetonitrile sonicate for 5 min, filter with whatman filter paper
(0.45 µm), inject a 20 µL aliquot.

HPLC variables:
Column: 150 X 3.95 µm Novapack C18
Mobile phase: Acetonitrile 600 ml + Water 400 ml
Dilution Factor: Std. 25 mg in 25 ml Acetonitrile
Flow rate: 1
Injection volume: 20
Detector: UV 254
Chromatogram:
Retention time: 8.508
(Fatmi AA et al., 1988) (Table 5)

Formulae: sample Area * potency * Std. wt. * Avg. wt.


Acceptance criteria: chemical stability: The assay of drug concentration was accepted where the
concentration of medroxy progesterone acetate for each condition was ≥95.0% of the concentration of
medroxy progesterone acetate stored in blister packaging at ambient temperature measured by HPLC. This
is in line with the International Conference of Harmonisation definition of ‘significant change’ for a drug
product as a 5% change in assay from its initial value in stability testing.

RESULTS AND DISCUSSION


The physico-chemical stability of medroxy progesterone acetate tablets stored for 03 months under
different conditions was determined (Tables 1 and 2). The temperature in the laboratory remained at 25°C ±
5°C during the study period. Tablets stored in HDPE bottle were stored for six months at 400C ± 20 and
75% ± 5 % RH / 300C ± 20C and 65% ± 5% RH / 250C and 60% ± 5% RH. were similar in appearance
during study period and passed all physico-chemical tests (Tables 4, 5, 6, 7 and 8). Tablets exposed to 40°C
with 75% RH in compliance aids passed the tests for uniformity of weight, friability and chemical stability
during study period but became paler and moist in appearance, significantly softer (p< 0.0001) and passed
the tests for disintegration (>15 minutes) and dissolution (95.80 % medroxy progesterone acetate released
at 30 minutes).

CONCLUSION
Although chemical stability was unaffected, storage in compliance aids at 40°C with 75% RH softened
medroxy progesterone acetate tablets, increase disintegration time and change in dissolution profile after
study period. This formulation of medroxy progesterone acetate tablets could be suitable for storage in
compliance aids at 25°C to 32°C, but not in hotter, humid weather.

REFERENCES
Guidance for Industry Q1A Stability testing of new drug substances and drug products U.S. Department of
Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research
(CDER), August 2001.

CPMP Note for Guidance on stability testing: Stability testing of new drug substances and products
(revision 2) Date for coming into operation August 2003.

Aulton ME. (2002): Pharmaceutics: the science of dosage form design. 2nd ed. Edinburgh: Churchill
Livingstone.

Fatmi AA, Williams GV, hickson EA. (1988): liquid chromatographic determination of
medroxyprogesterone acetate in tablets, J Assoc. Off. Anal Chem.,71: 528-530.

13
Kakde A et al: Continental J. Pharmaceutical Sciences 3: 7 - 14, 2009

Received for Publication: 27/05/2009


Accepted for Publication: 12/06/2009

Corresponding Author:
Abhishek Kumar Jain
Department of Pharmaceutical sciences, M.L.S. University, Udaipur, Rajasthan, India.
E-mail address: akjainmlsu@gmail.com

14
Continental J. Pharmaceutical Sciences 3: 15 - 17, 2009
©Wilolud Online Journals, 2009.

EVALUATION OF THE ACCURACY OF LABEL STRENGTH OF SOME LOCALLY PRODUCED BRANDS


OF PARACETAMOL SYRUPS MARKETED IN PORT HARCOURT, NIGERIA.

K.C. Ugoeze1, N.E. Vaikosen2, J.N. Egwurugwu3 and J.I. Omokhapue1


1
Department of Pharmaceutics and Pharmaceutical Technology, 2Department of Pharmaceutical and Medicinal
Chemistry, Faculty of Pharmacy, Niger Delta University, Wilberforce Island, P.M.B 071 Yenegoa, Bayelsa State,
Nigeria. 3Department of Physiology, College of Medicine, Imo State University, Owerri, Nigeria.

ABSTRACT
The authenticity of the label strength of some locally produced brands of liquid paracetamol
purchased in Port Harcourt, Nigeria has been investigated. Drug content was determined using
the High Performance Liquid Chromatographic method. Results show that 53.33% of the
products evaluated assayed within the acceptable range for total drug content and 73.33% were
within acceptable pH of maximum stability. None of the brands met with its label strength
claim per teaspoonful. It shows that defective and substandard paracetamol syrups are still in
circulation in Nigeria.

KEY WORDS: Paracetamol syrup, Evaluation, Accuracy, Label strength.

INTRODUCTION
The increasing number of commercial brands of a particular drug from multiple sources in Nigeria has become a
great concern of which healthcare providers must be very articulate to ensure that one brand from among the
seemingly equivalent products is recommended. However, many unenlightened users who engage in poly-pharmacy
or self medication/prescription may be endangered due to drug over dose from concurrent use of the same drug with
varying brand names. Paracetamol syrup, one of such drugs with varying brands is often a drug of first choice used
as a non-prescription analgesic and antipyretic agent for children. It is commonly available in any registered or
unregistered drug outlet in urban or remote areas of Nigeria (Ameer et al, 1983). Paracetamol is rapidly absorbed
from oral dosage forms and studies carried out in the U.S.A showed no significant differences between the
bioavailability of various commercial oral dosage forms of it (Mattock et al 1971; WHO 1996).
Since the marketing of various brands of this drug is prevalent in Nigeria, the objective of this study is to evaluate
the pharmaceutical quality of available paracetamol syrups produced and used in Nigeria for the accuracy of their
label strength and stability. Paracetamol syrup was studied because of its ready availability and use in the country.

MATERIALS AND METHODS


Materials
Brands of paracetamol syrup (coded PCM-A - PCM-O), Methanol (M&B), Acetonitrile (Sigma Aldrich, U.S.A) and
Paracetamol powder (GSK).

Methods
Drug sampling
15 brands of paracetamol syrup were purchased from retail pharmacy outlets in Port Harcourt, Nigeria where they
were available.

Standardization of samples
A 10 mg quantity of standard paracetamol was weighed on a Denver analytical balance and transferred into a 10 ml
volumetric flask. About 7 ml of diluent (10% acetonitrile in 90 % methanol) was added and sonicated using
ultrasonic bath at 30 0 C for 15 min and later made up to 10 ml mark. By dilution method, graded concentrations of
the standard paracetamol containing 25, 50, 75, 100, 150 and 200 µg /ml were respectively prepared and injected.
The respective peak area was recorded and integrated by an enhanced integrator. The acquired data (peak area) with
reference to the respective concentrations were used for plotting calibration curve (May Van 2005/2006).

15
K.C. Ugoeze et al.,: Continental J. Pharmaceutical Sciences 3: 15 - 17, 2009

Drug testing
Following purchase of these samples, information on brands, manufacturer, NAFDAC (National Agency for Food,
Drug Administration and Control) status, manufacturing and expiry dates were recorded from the product packages.
Organoleptic properties and pH were tested.

Equivalent of 10 mg of the respective samples of brands of paracetamol syrups was dispensed into a 10ml
volumetric flask. About 7 ml of diluent was added and sonicated for 15 min at 30 0 C. This was made up to volume.
By dilution method, an amount equivalent to 100 µg / ml was prepared, filtered with membrane filter (0.45 µm) and
were analysed using High Performance Liquid Chromatographic (HPLC) method. The HPLC equipment (HPLC
1100 series, Agilent Technologies, Japan) was fitted with Hypersil APS-1 C18 column. The mobile phase was a
mixture of acetonitrile and methanol in the ratio of 10:90. This was ran at a flow rate of 0.25ml / min and maintained
at 35 o C. The samples were read at a wavelength of 270nm (May Van 2005/2006).

RESULTS AND DISCUSSION


Table 1: Description of brands of paracetamol syrup tested.

Brand Code Date of manufacture Expiry date NAFDAC Status


PCM-A 09: 2007 09: 2010 Registered
PCM-B 09: 2007 09: 2010 Registered
PCM-C 11: 2008 10: 2011 Registered
PCM-D 02: 2008 01: 2011 Registered
PCM-E 03: 2009 03: 2012 Registered
PCM-F 03: 2009 02: 2012 Registered
PCM-G 01: 2009 12: 2011 Registered
PCM-H 09: 2007 08: 2010 Registered
PCM-I 10: 2008 10: 2011 Registered
PCM-J 06: 2008 06: 2011 Registered
PCM-K 07: 2008 06: 2011 Registered
PCM-L 01: 2008 12: 2010 Registered
PCM-M 12: 2007 11: 2010 Registered
PCM-N 02: 2009 02: 2012 Registered
PCM-O 02: 2008 01: 2010 Registered

A total of 15 brands of paracetamol syrups were analysed. All samples were within shelf life at the time of the study.
The descriptions of the brands were presented in Table 1. The results of products properties and drug contents are
shown in Table 2. The United States Pharmacopeia (USP 2007) stipulates that paracetamol oral solution should
contain not less than 90.0% and not more than 110.0% of the labelled amount and it also states that for maximum
stability, the pH of such preparations should be within 3.8-6.1. The results presented in Table 2 show that only
53.33% (PCM-A, B, G, H, I & J) of the brands investigated assayed within the acceptable range for total drug
content. Of the 46.67% that failed, a brand, (PCM-D) has total drug content less than 50% and one, (PCM-O)
assayed above 110.0%. In terms of pH, 73.33% were within stable region. None of the brands met with its label
strength claim per teaspoonful.

The above results show that in spite of the concerted efforts by NAFDAC in ensuring that safe and effective drugs
are in circulation, there are still some defects in the pharmaceutical quality of paracetamol syrups sold in Nigeria.
Inadequacy in quality of some of these commercial products may be due to poor quality of formulation ingredients
employed, degradation due to storage conditions and poor packaging, methods of quality control adopted, use of
obsolete and insensitive analytical equipment or acts of deliberate counterfeiting. None of these could be
ascertained. However, these findings serve as signals for the regulatory bodies to be alert and not relax after a
product has been registered. If consistent effort is put in place to monitor and ensure quality of pharmaceutical
products, there will be reduction in therapeutic failure and improvement in the health status of patients who may be
endangered by use of substandard drugs.

16
K.C. Ugoeze et al.,: Continental J. Pharmaceutical Sciences 3: 15 - 17, 2009

Table 2: Properties and drug content of brands of paracetamol syrups tested.


Brand Appearance Taste pH Label strength Total drug Actual content
Code (mg/5ml) content (%) (mg/5ml)

PCM-A Pinkish syrup Slight bitter 3.50 120 94.65 113.58


after taste
PCM-B Pinkish & Slight bitter 3.50 120 94.05 112.86
viscous susp. after taste
PCM-C Slightly pinkish Sweet 4.20 120 61.45 73.74
syrup
PCM-D Yellowish Sweet 5.60 120 34.05 40.86
viscous susp.
PCM-E Pinkish syrup Sweet 5.20 125 61.50 76.88

PCM-F Pinkish syrup Sweet 4.10 125 89.38 107.26


PCM-G Pinkish syrup Slight bitter 4.90 125 96.04 120.05
after taste
PCM-H Pinkish syrup Sweet 4.30 125 92.43 115.54
PCM-I Pinkish syrup Fairly sweet 3.70 125 97.40 121.75
PCM-J Pinkish syrup Sweet 4.00 125 99.35 124.19
PCM-K Pinkish syrup Slightly bitter 5.80 125 67.49 84.36
PCM-L Slight pinkish Slightly bitter 4.30 125 82.24 102.80
syrup
PCM-M Pinkish syrup Sweet 3.00 125 97.91 122.39
PCM-N Pinkish syrup Sweet 5.00 125 100.99 126.24
PCM-O Pinkish syrup Slight bitter 5.40 125 129.68 162.10
after taste

REFERENCES
Ameer, B; Divoll, M; Abernethy, D.R; Green, D.J and Shargel, L. (1983). Absolute and relative bioavailability of
oral acetaminophen preparations. J. Pharm. Sci. 78:955-958

Mattock, G.L; McGilverary, I.J and Mainville, L.A. (1971). Acetaminophen III: Dissolution studies of commercial
tablets of acetaminophen and comparison with in vivo absorption parameters. J Pharm. Sci., 60: 561-564.

Mattock, G.L; McGilveray, I.J and Mainville, L.A.( 1971). Acetaminophen I: A protocol for the comparison of
physiological availabilities of ten different dosage forms. Can J Pharm. Sci. 6:35-38.

May, Van (2005/2006). The Essential Chromatography Catalogue from Agilent Technologies, p.643.

The United States Pharmacopeia, USP/NF (2007). The United States Pharmacopeial Convention, Rockville, p.1267.

World Health Organization Expert Committee on Specifications for Pharmaceutical Preparations (1996). 34th Report
WHO Technical Report Series, No. 863, Geneva, Switzerland, pp. 114-54.

Received for Publication: 07/10/2009


Accepted for Publication: 27/10/2009

Corresponding Author:
K.C. Ugoeze
Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Niger Delta University,
Wilberforce Island, P.M.B 071 Yenegoa, Bayelsa State, Nigeria. E-mail: kcugoeze@yahoo.com

17
18

Vous aimerez peut-être aussi