Vous êtes sur la page 1sur 87

International Journal of Current Trends in Pharmaceutical Research

Volume-2, Issue-4, July 2014


ISSN: 23221-3760

About IJCTPR

International Journal of Current Trends in Pharmaceutical Research is an open


access bi-monthly online journal. The primary mission of the journal is to
become the premier source of high quality research from whole of the world.
The aim of the journal is to increase the impact of research both in academia
and industry, with strong emphasis on quality and originality. It is peer-
reviewed and is published in electronic version. The language of the Journal is
English.

The journal is devoted to the promotion of all fields of pharmaceutical sciences


like Pharmaceutics and Industrial pharmacy, Medicinal Chemistry,
Pharmacology and Toxicology, Phytopharmacy, Medicine, Clinical Pharmacy,
Pharmacy Practice. Pharmaceutical Microbiolog and Biotechnolog,
Biopharmaceutics, Pharmacokinetics and Pharmacodynamics, Computational
Chemistry, Molecular modeling and Drug design, Pharmacoinformatics,
Chemo informatics.

It publishes research papers, short communications, dissertation abstracts and


reviews, notes dealing with entire aspects of Medicine and pharmaceutical
sciences. The ultimate vision of the journal is to become the premier source of
high quality research from whole of the world. The aim of the journal is to
increase the impact of research both in academia and industry, with strong
emphasis on quality and originality.
Manuscript Submission
Submitted manuscripts must meet the following criteria:
1. The material must be original.

2. The writing must be clear, concise and unambiguous.

3. The data should be valid

4. The discussions and conclusions drawn by the authors must be reasonable


and supportable.

5. The information must be relevant and interesting.

Authors must read the Author Guide lines carefully before submitting the
manuscript.

The authors must submit their manuscript at: pharmaresearchlibrary@gmail.com

About PRL
The effects of globalization have driven tremendous changes within the
research. Today, change is so rapid that the researchers have to keep itself
aware with the latest discoveries, findings and research. Pharma research
Library Journals are aims to explore the research data, inventions, findings in
the field of Pharmaceutical Sciences and applied sciences as Chemical
Sciences, Biochemistry, Microbiology and Biotechnology, Pharmacology and
Toxicology; Pharmaceutics and Cosmetology, Medicinal Chemistry,
Pharmacognosy and herbal formulations, Pharmaceutical Analysis. Focusing
on information that you can immediately use in your practice, the Pharma
Research Library journals allows you to obtain effective and proven latest
technological information regarding their current uses in the form of news
items.

Pharma research Library Journals are monthly, bi-monthly, quarterly, bi-


annual andannual publishing online Journals, publishes peer-reviewed high-
quality research papers and review papers, short communications and

technical reports in all aspects of Pharmaceutical and Life Sciences. Pharma


Pharma research Library Journals are monthly, bi-monthly, quarterly, bi-
annual andannual publishing online Journals, publishes peer-reviewed
high-quality research papers and review papers, short communications and
technical reports in all aspects of Pharmaceutical and Life Sciences. Pharma
research Library is to provide an expert publishing service to the scientific
persons as authors, editors, librarians, researchers, professionals, teachers
and their students, assisting them improve their knowledge.

Pharma Research Library Journals are International and peer-reviewed


open Access online journals, PRL publishes all departments of
Pharmaceutical, Chemical, Biological, Medical and Life Sciences. Open
Access (OA) is the practice of providing unrestricted access via the Internet
to peer-reviewed scholarly journal articles. Open access publication is
supported by authors’ institutes or research funding agency by payment of a
comparatively low Article Processing Charge for accepted articles.

Pharma Research Library is an open access publisher of its high quality


research publications around the world ensuring that the light of knowledge
is disseminated without any interference and ambiguity to all its users.

Editor-In-Chief
Mr. Ramesh Dhani

Publisher
PHARMA RESEARCH LIBRARY
(An International Scientific Publication Group)
Nellore City, A.P, India-524002
PH: +91-9290623231
International Journal of Current Trends in Pharmaceutical Research

Table of Contents: Volume-2, Issue-4, July 2014


S.No Title Page

1 Formulation and Evaluation of Sustained Release Matrix Tablet 494-501


of a Model Anti-Hypertensive Drug Using Natural Polymers
K. Navaneetha*, Y. Upendar Rao, Dr. B. Venkateswara Reddy, T.
Lavanya, Ch. Divya, K. Revanth Chandra
2 Preliminary Study to Evaluate the Health Benefits of 502-507
Natural(Crude) versus Commercial black seed oil in Sample of
Healthy volunteers
Doa’a Anwar Ibrahim*
3 Development and Evaluation Sustained Release Matrices of 508-515
Lamivudine by using Synthetic Polymer
Supriya Deogire*, Sonali Mahaparale
4 Formulation and In-Vitro Evaluation of Floating Beads of 516-524
Propranolol Hydrochloride Using Foam Technology
Sanjay K. Sharma*, Deepak Sharma,Vaibhav Saxena, Manish
Jaimini, Shailender Mohan
5 525-540
Formulation, Development and Process Optimization of
Immediate Release Tablets of Glimepiride
Vennela K.G*, Ramesh Reddy K, Uma Sankar , Jayachandra
Reddy P
6 A Brief Study on Carica Papaya- A Review 541-550
Asha Roshan*, Navneet Kumar Verma, Anubha Gupta
7 A Devastating Inherited Neurodegenerative Disease: 551-554
Huntington’s Disease: A Clinical Review
Patel Chirag J*, Satyanand Tyagi, Patel Kanu J, Patel Tushar, Patel
Harnish K, Patel Priyanka H
8 A Novel Pharmaceutical Microscopic Vesicle: Liposome 555-561
Patel Chirag J*, Satyanand Tyagi, Patel Kanu J, Patel Tushar, Patel
Harnish K, Patel Priyanka H
9 A Review on Anti- Inflammatory and Anti-Oxidant Activity of 562-565
Polyherbal Plant Extracts
Y. Bhagyasri*, M.S. Divyasree, C.K. Ashok kumar, A. Nagamani
10 Transferomes: Emerging Trends for Drug Delivery 566-574
Krishna moorthy S.B*, Sandeep. J, Jyothirmaie. K, Rajalakshmi.R
K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

ISSN: 2321-3760
Research Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 494-501
www.pharmaresearchlibrary.com/ijctpr

Formulation and Evaluation of Sustained Release Matrix Tablet of a Model


Anti-Hypertensive Drug Using Natural Polymers
K. Navaneetha1*, Y. Upendar Rao2, Dr. B. Venkateswara Reddy1, T. Lavanya1, Ch. Divya1,
K. Revanth Chandra1
1
Department of Pharmaceutics, St. Pauls College of Pharmacy, Turkayamjal (V), Hayathnagar (M),
R.R.Dist, India-501510
2
Department of Pharmaceutics, khammam College of Pharmacy, khammam, India
Received: 15 May 2014, Accepted: 27 June 2014, Published Online: 15 July 2014

Abstract
The objective of the present study is to develop a sustained release matrix tablets of Losartan potassium, an
anti hypertensive drug. The sustained release tablets were prepared by wet granulation and formulated using
different drug and polymer ratios, formulations such as F1 to F9. Natural polymers like Xanthan Gum (XG),
Guar gum, Cellulose were used. Compatibility of the drug with various excipients was studied. The
compressed tablets were evaluated and showed compliance with Pharmacopeial limits. The optimized
formulation is F2 on the basis of acceptable tablet properties and in vitro drug release. The resulting
formulation produced robust tablets with optimum hardness, consistent weight uniformity and friability. In-
Vitro dissolution studies have shown the sustained release for Losartan potassium i.e., 90.88% released at
the end of 12h. A decrease in release kinetics of the drug was observed on increasing polymer ratio.
Keywords: Sustain release; Losartan Potassium; Anti Hypertensive; Wet Granulation; Natural Polymers.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 494
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .497
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . .501
5. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .501

*Corresponding author
K. Navaneetha
Department of Pharmaceutics, St.Pauls
College of Pharmacy, Turkayamjal (V),
Hayathnagar (M), R.R. Dist, India-501510
Manuscript ID: IJCTPR2142 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved

1. Introduction
Oral delivery of drugs is by far the most preferable route of drug delivery due to the ease of administration, patient
compliance and flexibility in formulation, etc. Many of the drug delivery systems, available in the market are oral
drug delivery type systems. Oral drug delivery systems have progressed from immediate release to site-specific

494 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

delivery over a period of time. Every patient would always like to have a ideal drug delivery system possessing the
two main properties that are single dose or less frequent dosing for the whole duration of treatment and the dosage
form must release active drug directly at the site of action. Thus the objective of the pharmacist is to develop
systems that can be as ideal system as possible. Attempts to develop a single- dose therapy for the whole duration of
treatment have focused attention on controlled or sustained release drug delivery systems. The therapy of many
chronic diseases requires a repeated dosing of a drug. Drugs having a short half-life have to be administered up to
several times daily within short intervals. To reduce the application frequently sustained formulations have been
developed [1].

Sustained Release Dosage Forms


Sustained release, sustained action, prolonged action, controlled release, extended action, timed release, depot and
repository dosage forms are terms used to identify drug delivery system that are designed to achieve or prolonged
therapeutic effect by continuously releasing medication over an extended period of time after administration of a
single dose [2]. Basically there are three basic modes of drug delivery i.e. targeted delivery, controlled release and
modulated release. Targeted delivery refers to the systemic administration of a drug carrier with the goal of
delivering the drug to specific cell types; tissues or organs. Controlled release refers to the use of a delivery device
with the objective of releasing the drug into the patient’s body at a predetermined rate, or at specific release profiles.
On the other hand modulated release implies use of as drug delivery device that releases the drug at a variable rate
controlled by environment conditions, biofeedback, sensor input or an external control device [3]. Many times
sustained release and controlled release terms are used interchangeably. However sustained release system deliver
the active agent although at slower than a conventional formulation but the release is substantially affected by
external environment. Sustained release dosage forms are generally administered by four delivery modes, namely
Oral controlled drug delivery, transdermal drug delivery, implantable drug delivery and particulate drug delivery.

Drug Properties Relevant To Sustained Release Formulations [4-7]:


During design of sustained release delivery systems, variables such as the route of drug delivery, the type of delivery
system, the disease being treated, the patient, the length of therapy and the properties of the drug, are considered. Of
particular interest to the scientist designing the system are the constraints imposed by the properties of the drug.
These properties are classified as
(a) Physicochemical
(b) Biological properties
These properties have the greatest effect on the behavior of the drug in the delivery system and in the body. There is
no clear cut distinction between these two categories since the biological properties of a drug are a function of its
physicochemical properties. By definition, physicochemical properties are those that can be determined from in vitro
experiments and biological properties will be those that result from typical Pharmacokinetic studies of the
absorption, distribution, metabolism, and excretion (ADME) characteristics of a drug and those resulting from
pharmacological studies. In the recent years extensive efforts have been made in various pharmaceutical research
laboratories of sustained release drug delivery systems, with an aim of improved patient compliance, better
therapeutic efficacy, less side effects and reduced dosage regimen with less toxicity for treatment for many acute
and chronic diseases [8-10]. Losartan potassium is a potent, highly specific angiotensin II type 1 receptor antagonist
with antihypertensive activity. It is readily absorbed from the gastrointestinal tract with oral bioavailability of about
33% and plasma elimination half-life ranging from1.5 to2.5 hours [11]. Matrix tablets composed of drug and
polymer as release retarding material offer the simplest approach in designing a sustained release system. The present
study aims to develop sustained release matrix tablets using natural polymers along with drug in varying
proportions by wet granulation method [12].

2. Materials and Methods


Materials:
Losartan potassium was obtained from Chandra labs kukatpally. Guar gum and Xanthan gum from wonder herbs,
Hyderabad, India. Cellulose from Lo b a c h e mie P vt. ltd , Mu mb ai . Microcrystalline cellulose,PVP, Magnesium
stearate and talc from S.D. Fine Chem. Ltd, Mumbai, India.
Drug- Polymer Interaction
It was carried out by taking FT-IR Infrared spectra of pure drug, and drug-polymer by KBr pellet technique and was
recorded in the range of 4000 – 400 cm-1 using FT-IR Spectrophotometer.
Methods:
Preparation of Losartan Potassium Tablets by Wet G r a nu la t io n:
The active ingredient, disintegration agents are weighed as mentioned in the table 1 and mixed. The wet granulate is
prepared by adding binder solution i.e., PVP, then damp mass is screened to get the granules. The granules obtained

495 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

are dried in hot air oven at 600C for one hour. After the granules are dried, they are passed through a screen of
smaller size than the one used for the wet mass to select granules of uniform size to allow even fill in the die cavity.
A dry lubricant is added to the granules either by dusting over the spread-out granules or by blending with the
granules. The granules are fed into the die cavity and then compressed between a lower and an upper punch. 7.5mm
size punches were used for punching tablets.

Table 1. Master formulation for Losartan potassium tablets


Ingredients F1 F2 F3 F4 F5 F6 F7 F8 F9
Losartan 50 50 50 50 50 50 50 50 50
potassium
Xanthan gum 50 100 150 - - - - - -
Guar gum - - - 50 100 150 - - -
Cellulose - - - - - - 50 100 150
MCC 225 175 125 225 175 125 225 175 125
PVP 20 20 20 20 20 20 20 20 20
Iso propyl Q.S Q.S Q.S Q.S Q.S Q.S Q.S Q.S Q.S
alcohol
Talc 3 3 3 3 3 3 3 3 3
Magnesium 2 2 2 2 2 2 2 2 2
stearate
Total 350 350 350 350 350 350 350 350 350
weight(mg)

Evaluation Parameters
A) Precompression Parameters
The prepared granules were evaluated for various precompression parameters such as bulk density, tapped density,
angle of repose, hausner’s ratio, compressibility index to determine the flow properties of the powdered blend.
B) Post compression Parameters [13]
Hardness:
The hardness of the tablet was determined using a Monsanto hardness tester. It is expressed in Kg / cm2.
Friability (F):
The friability of the tablet was determined using Roche Friabilator. It is expressed in percentage (%). 10 tablets
were initially weighed (W initial) and transferred into the friabilator. The friabilator was operated at 25 rpm for four
mins. The tablets were weighed again (Wfinal). The percentage friability was then calculated.
Weight Variation:
Ten tablets were selected randomly from the lot and weighed individually to check for weight variation. IP limit for
weight variation in case of tablets weighing more than 325mg is ± 5%.
Thickness:
The thickness of the tablets was measured by screw gauge. It is expressed in mm.
Content uniformity test:
Ten tablets were weighed and powdered, a quantity of powder equivalent to 10 mg of formulation was transferred to a
25 ml volumetric flask and 15 ml water is added. The drug is extracted in water by vigorously shaking the stoppered
flask for 15 minutes. Then the volume is adjusted to the mark with distilled water and the liquid is filtered. The drug
content was determined by measuring the absorbance at 234 nm after appropriate dilution. The drug content was
calculated using the standard calibration curve. The mean percent drug content was calculated.
Invitro dissolution studies:
The Dissolution study was carried out using dissolution test apparatus USP type II (Paddle). The dissolution medium
used was 900 ml of simulated intestinal fluid at 37±0.5º. The paddle speed was kept at 50 rpm throughout the study.
Aliquot of 5 ml was withdrawn at predetermined time interval and equivalent amount of fresh medium was replaced to
maintain a constant volume. After each sampling and suitably diluted with buffer, solutions were analyzed
spectrophotometrically at 234nm against suitable blank using UV-visible spectrophotometer.
Kinetics modeling of drug dissolution profiles [14]:
To analyze the in vitro release data various kinetic models were used to describe the release kinetics. The dissolution
profile of the all formulations were fitted to zero order, first order, Higuchi and Korsmeyer equation / Peppa’s model
to ascertain the kinetic modeling of the drug release.
C = K 0t
Where, K0 = Zero-order rate constant and t = Time

496 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

LogC = LogC0 - Kt / 2.303


Where, C0 = initial concentration of drug and K = First order constant.
Q = K t1/ 2
Where, K = Constant and t = Time.
Mt / Mα = Ktn
Log Mt / Mα = Log K + n Log t
Where, Mt / Mα = the fraction of drug released at time‘t’, K = Constant incorporating the structural and geometrical
characteristics of the drug / polymer system and N = Diffusion exponent related to the mechanism of the release.

3. Results and Discussion


Determination of Interaction between Drug and Polymers:
Compatibility studies were performed using FT-IR spectrophotometer. The peaks obtained in the spectra of each
formulation correlates with the peaks of drug spectrum. This indicates that the drug is compatible with the
formulation components. Pure drug Losartan potassium has exhibited IR spectrum indicating the presence of C-H
group at 3055.35cm -1 and 2943.49cm-1, C=C group at 1575.89cm-1 to 1733cm-1, C-O peak at 1631cm-1. The IR
spectrum of pure drug losartan potassium is shown in Figure 1. In the IR spectra of drug and different polymers
obtained, there is no interaction found in between drug and polymers. The drug functional groups are observed in
each of IR spectra of combination of drug and polymers. IR spectra of pure drug and drug with other excipients are
shown in figures 1, 2, 3, 4.

Figure 1. FTIR for pure drug of losartan potassium

Figure 2. FTIR for losartan potassium + Guar gum


497 | International Journal of Current Trends in Pharmaceutical Research
K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

Figure 3. FTIR for losartan potassium + xanthan gum

Figure 4. FTIR for losartan potassium + cellulose

Precompression Parameters: The method employed for tabletting in this study was wet granulation for which the
drug or the mixture of drug and polymer should possess good flow properties. Granules ready for compression
containing drug and various excipients were subjected for pre-compression parameters (Micromeritic properties) to
study the flow properties of powder blend, to achieve constant uniformity of tablet weight. The data obtained for
angle of repose for all the formulations were tabulated in the table no.2 The values were found to be in the range of
22°.69′ a nd 26°.95′. All the formulations showed the angle of repose less than 30°, which reveals the good flow
property. The results of Carr’s consolidation index or compressibility index (%) for the entire formulation blend
ranged from 14.28 to 16.92 %. the powder blend showed excellent compressibility index values up to 15% result
in good to excellent flow properties.

Table 2. Precompression studies for the granules


Formulation Angle of repose (θ) Carr’s Index (%) Hausner’s ratio Drug uniformity* (%)
F1 26°.95′ 15.15 1.178 98.94±0.40
F2 24°.56′ 14.35 1.182 99.31±0.32
F3 25°.94′ 16.42 1.196 99.75±0.33
F4 23°.96′ 16.92 1.204 96.31±0.41
F5 24°.14′ 16.42 1.196 98.69±0.22
F6 25°.91′ 16.67 1.2 99.75±0.34
F7 22°.69′ 14.28 1.167 99.75±0.21
F8 23°.58′ 16.67 1.2 98.94±0.25
F9 25°.95′ 14.92 1.175 99.75±0.34

498 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

Post Compression Parameters:


Hardness: The hardness of the tablet formulations was found to be in the range of 5.1 to 6.1 kg/cm2.
Friability: The friability values were found to be in the range of 0.141 to 0.191 %.
Uniformity of weight: All the prepared tablets of losartan potassium were evaluated for weight variation. The
weight of all the tablets was found to be in the range of 345 to 358%.
Uniformity of drug content: The low values of standard deviation indicates uniform drug content within the tablets
The percent drug content of all the tablets was found to be in the range of 97 to 99 %. The values are represented in
the table 3.
Table 3. Post formulation studies of all formulations
Formula Diameter* Thickness* Weight variation Hardness* Friability Drug content*
tions (mm) (mm) (mg) (kg /cm2 ) (%) (%)
F1 10.05±0.030 4.45±0.11 351±5 5.1 ± 0.12 0.191 97.00±0.24
F2 10.06±0.040 4.50±0.04 350±5 5.5 ± 0.24 0.149 98.90±0.22
F3 10.04±0.030 4.49±0.05 354±5 5.8 ± 0.21 0.146 97.86±0.34
F4 10.02±0.030 4.45±0.12 345±5 5 .2± 0.23 0.149 98.75±0.32
F5 10.02±0.054 4.46±0.03 347±5 5.9 ± 0.12 0.145 96.26±0.46
F6 10.05±0.064 4.54±0.23 351±5 6.1 ± 0.14 0.191 98.45±0.26
F7 10.07±0.022 4.52 ±0.2 352±5 5.2 ± 0.18 0.193 98.00±0.28
F8 10.05±0.035 4.51±0.12 349±5 5.4 ± 0.22 0.146 99.72±0.30
F9 10.04±0.059 4.53 ±0.3 348±5 6 .1± 0.21 0.191 98.82±0.34

In vitro Drug Release Profile:


In vitro drug release studies were carried out on dissolution test apparatus in 900ml of 0.1N HCl for 2 hours and
phosphate buffer 6.8 pH for the next remaining hours. The release rate of the drug from the matrix tablets decreased
with an increase in polymer proportion because of increase in gel strength as well as the formation of a gel layer
with a longer diffusional path. Results are given in the table 4,5 and represented in the figure 5,6 and 7.
The order of release was found to be: F2>F1>F5> F4>F3>F8 >F7>F9
The formulation F2 has shown better drug release than other formulations. In the formulation F2 having swellable
polymer as guar gum showing better drug release profile than other polymer (xanthan gum, cellulose). Hence the
natural polymer guar gum is better suitable for sustained release delivery than other natural polymers.

Table 4. Dissolution table of formulations in 0.1N HCL Acid stage


Time in hrs 1hr 2hr
F1 19.24 27.42
F2 18.64 26.46
F3 17.44 24.82
F4 18.82 26.72
F5 17.12 24.12
F6 16.41 23.12
F7 17.12 25.12
F8 16.42 22.18
F9 15.12 22.01

Table 5. Dissolution table of formulations in 6.8 Phosphate buffer buffer stage


Time in hrs 4hr 6hr 8hr 10hr 12hr
F1 40.12 65.42 72.12 80.52 87.12
F2 39.73 62.84 74.52 82.12 90.88
F3 36.42 55.42 62.45 74.45 83.52
F4 39.42 61.24 68.62 76.96 84.22
F5 39.92 61.12 72.52 79.89 86.17
F6 33.12 52.42 60.86 72.85 80.33
F7 38.12 50.19 67.62 74.62 80.14
F8 38.80 60.40 69.75 77.75 82.55
F9 31.45 51.09 59.36 70.46 78.26

499 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

Losartan + Guar gum


100

80

% Drug release
60
F1
40
F2
20
F3
0
0 5 10 15
Time(hr)

Figure 5. Dissolution for losartan potassium with guar gum

Losartan + xanthan gum


100
80
% Drug release

60
F4
40
F5
20
F6
0
0 5 10 15
Time(hr)

Figure 6. Dissolution for losartan potassium with xanthan gum

90 Losartan+Cellulose
80
70
60
%Drug Release

50 F7
40 F8
30 F9
20
10
0
0 2 4 6 8 10 12 14
Time(Hr)
Figure 7. Dissolution for losartan potassium with cellulose

500 | International Journal of Current Trends in Pharmaceutical Research


K. Navaneetha et al, IJCTPR, 2014, Vol.2(4): 494-501

Release Kinetics:
The data obtained from in-vitro release of best formulation F2 were fitted into equations for the zero order and first
order, Higuchi and Krosmeyer release models; the interpretation of the data was based on the values (Table 6) of the
resulting regression co-efficient. Result obtained from the release kinetics shows that the formulation F2 follows the
first order and Krosmeyer Peppas plots were found to be fairly linear and the ‘r’ coefficient value for pure drug
losartan potassium and its formulations with guar gum (1:2). So the regression data of first order and Krosmeyer
Peppas plots indicates that the drug was released by first order kinetics and non fickian.

Table 6. Release kinetics of formulation F2


Formulation Zero order First order Higuchi Krosmeyer Peppas
R2 value R2 value R2 value
R2 value ‘n’value
F-2 0.964 0.981 0.976 0.987 0.670

4. Conclusion
Sustained release matrix tablets of losartan potassium were successfully formulated and evaluated. Sustained release
matrix tablets of losartan potassium were successfully prepared by wet granulation using guar gum, xanthan gum,
cellulose, CMC as polymers and PVP as binder. The isopropyl alcohol was used as granulating fluid for binder.
Sustained release tablets were evaluated for pharmacopeial and non-pharmacopeial (industry specified) tests. The
drug content was uniform in all the formulations of tablets prepared. The low values of standard deviation indicate
uniform distribution of drug. Based on the results, F-2 was identified as better formulation among the developed
formulations. In F-2 formulation drug release was found to be 90.83% at the end of 12hrs. The drug release profile
of F2 (guar gum) is higher than rest of the formulations made by using xanthan gum and cellulose. In the
formulation 2 having swellable polymer as guar gum showing better drug release profile than other polymer
(xanthan gum and cellulose). Hence the natural polymer guar gum is better suitable for sustained release delivery
than other polymers.

5. References
1. Encyclopedia Of Pharmaceutical Technology, 2edition, Vol.1, pp. 850- 851.
2. Chang RK, Robinson JR. Tablets. In: Lieberman, HA, Lachman L. Pharmaceutical Dosage Forms. New
York, Vol.3, Marcel Dekker, 1990, pp. 200.
3. Robinson M., Sustained Action Dosage Forms, In: Lachman L., Lieberman H, Kanig J. The Theory and
Practice of Industrial Pharmacy. Philadelphia, 2nd edition, Lea and Febiger, 1970, pp. 666.
4. Lee VHL, Robinson JR, In: Robinson, JR. Sustained and Controlled Release Drug Delivery Systems 1978,
pp. 6.
5. Lee P, Good W. “Overview of Controlled Release Drug Delivery” In Controlled Release Technology:
Pharmaceutical Applications. ACS Symposium Series 348, American Chemical Society, Washington D.C.,
1987, pp. 1-13.
6. Chien Y W, Novel drug delivery system, 2nd edition, Marcel Dekker, INC., 1992, pp.1-43, 44-139, 140-
197.
7. Banker GS, Rhodes CT. Modern Pharmaceutics. 2nd edition, Marcel Dekker, INC., 1996, pp. 635- 671.
8. Chang, R.K., Robinson, J.R., Tablets. In: Lieberman HA, Lachman L. Pharmaceutical Dosage Forms. New
York, Vol.3, Marcel Dekker, 1990, pp. 206.
9. Kydonieus Agis, Treatise on Controlled drug delivery system, Marcel Dekker, INC., 1992, pp. 199- 224.
10. Shanmugam S, Chakrahari R, Sundaramoorthy K, Ayyappan T, Vetrichelvan T. Formulation and
Evaluation of Sustained release matrix tablets of Losartan Potassium. International Journal PharmTech
Research, 2011, 3(1): 526-234.
11. Subramaniam K, Manivannan R, Kugalur GP, Kakkatummal N, Natesan SK. Formulation and Evaluation of
sustained release tablets of Aceclofenac using hydrophilic matrix system. International Journal of
PharmaTech Research, 2010, 2: 1775-1780.
12. Lachman L, Liberman HA, Kang JL. The theory and practice of industrial pharmacy. 3rd ed. Mumbai:
Varghese Publishing house, 1987, pp. 296-303.
13. Suvakanta Dash, Padala narasimha Murthy, Lilakantha Nath and Prasanta Chowdhury., Kinetic modeling
on drug release from controlled drug delivery systems- A review. Acta Poloniae Pharmaceutica- Drug
Research, 2010, 67(3): 217-23.

501 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

ISSN: 2321-3760
Research Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 502-507
www.pharmaresearchlibrary.com/ijctpr

Preliminary Study to Evaluate the Health Benefits of Natural(Crude) versus


Commercial black seed oil in Sample of Healthy volunteers
Doa’a Anwar Ibrahim*
Department of Pharmacology, University of Science and Technology, Sana’a, Yemen
Received: 28 May 2014, Accepted: 30 June 2014, Published Online: 15 July 2014

Abstract
Plants had been used for medicinal purposes long before recorded history. The seeds of Nigella sativa
Linn. (Ranunculaceae), commonly known as black seed or black cumin, are used in folk (herbal)
medicine all over the world for the treatment and prevention of a number of diseases.Black seed is
recommended by the Prophet Muhammad (SWS): The beard hair of the Holy Prophet (peace be upon
him). "Hold on to the use of the black seed for indeed it has a remedy for every disease except death."
This study is aimed to compare between natural (crude) versus commercial black seed oil through
measuring some parameters: liver function enzymes (SGOT and SGPT), CBC as well as
FBS.Methodology: 20 healthy volunteers were divided randomly to 3 groups (I, II and III). Group I
(n=8) was allowed to take natural (crude) black seed oil (2 teaspoonful/d), Group II (n=8) was allowed
to take commercial ANSO-SYP black seed oil (Nigoil) (2 teaspoonful/d), group III (n=6) was kept as
placebo only taken (2 teaspoonful/d of sun flower oil) for two weeks. Washout period was done for all
groups for two weeks. Cross over between group I and II was done in this part, where group I (n=8) was
allowed to take natural (crude) black seed oil (2 teaspoonful/d), group II (n=8) was allowed to take
commercial ANSO3 Phenomenalsoft gelatin capsule (2 capsules/d), group III (n=6) was kept as
placebo only taken (2 teaspoonful/d of sun flower oil) for two weeks.Blood sample was taken before
(baseline) starting this study and at the end of study. The following parameters were measured complete
blood count CBC hepatic function enzymes (SGOT and SGPT) and fasting blood sugar. Results:
commercial black seed oil especially(soft gelatin capsule) showed significant increase (p<0.05) fasting
blood sugar as compared with natural (crude) black seed oil and placebo, while natural oil showed
reduction in FBS.Conclusion: commercial black seed oil especially soft gelatin capsule showed
significant increase in fasting blood sugar. This may referred to the presence of impurities.
Key words: Natural(crude) black seed; Commercial black seed; Liver function enzymes; Complete
blood count; Fasting blood sugar.

*Corresponding author
Doa’a Anwar Ibrahim
Department of Pharmacology,
University of Science and Technology
Sana’a, Yemen
Manuscript ID: IJCTPR2143 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved

502 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 503
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 503
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .504
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . .506
5. Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . 506
6. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 506

1. Introduction
The last two decades witnessed an enormous research rush to reveal the pharmacological actions of an annual spicy
delicate and beautiful herb known by the Latin name Nigella sativa Linnaeus variety hispidula (brachyloba [1]
.Nigella sativa is a herbal plant which belongs to Ranunculaceae family. It is a native of southern Europe. It is found
all over India and is specially seen in the eastern region and also in Arab countries [2].

The seeds contain a brown colored volatile oil 0.5 % to 1.6 % mainly thymoquinone and nigellone and red colored
stable oil which is 31 %. Besides this it contains albumin, sugar, carbonic acid, seponin, melanthin, Arabic acid, a
bitter compound named nigellin, resins, tennins and ash 7 %. It contains a volatile oil carvone 45 to 60 %, D-
lymonine and cymine[3]. They are acrid, bitter, thermogenic, aromatic, carminative, diuretic, antibacterial,
deodorant, appetizing, digestive, anthelmintic, constipating, febrifuge, stimulant, galactagogue, and expectorant.
They are useful in skin diseases, hemorrhoids, jaundice, inflammation (through Inhibition of thromboxane A2 and
LTB4 and leukocytes ), fever, paralysis, ophthalmia, halitosis, anorexia, dyspepsia, flatulence, diarrhea, dysentery,
cough, amenorrhea, dysmenorrhea and helminthiasis, especially tapeworm. Moreover, seeds have benefits on heart
[4], respiratory system[5,6, 7],improve and protect gastric ulcer [8,9], insulin secretagouge [10, 11, 12 ]and
hepatoprotective effect [13, 14]. Moreover, black seed oil particularly thymoquinone (TQ) has protective effect
against cisplatin- induced nephrotoxicity [15].

In 2005, Rooney and Ryan [16] found that although TQ exerted cytotoxicity against lungs, larynx, colon and
pancreas carcinomas yet it was more potent against the larynx ones. In a later investigation, the authors delineated
the TQ-induced cytotoxicity being due to the depletion of cellular glutathione and the activation of caspase-3
enzyme [17]. The aim of this study is to evaluate the health benefits of natural (crude) compared with that of
commercial black seed oil through measuring some parameters in healthy volunteers including : Complete blood
count ( CBC), hepatic function enzymes including (SGOT and SGPT) and fasting blood sugar (FBS).

2. Materials and Methods


2.1.Materials
2.1.1.Participants: 20 Healthy volunteers with age range from (22-24 years old) were participated in this study.
They were kept under standard situation and excluded intake herbs, drinks or medications that may interfere with the
outcomes of this study.
2.1.2.Drugs: Natural Nigella sativa(Black seeds) was purchased from special Herb stores , Sana’a City and taken to
squeezing stores to get the pure crude oil after identification by botanist in Pharmacognosy department –UST,
ANSO3 Phenomenalsoft gelatin capsule was purchased from BLUE BIOTECH INT. Germany and ANSO-SYP
black seed oil (Nigoil) was purchased from Refal Co. with A.U.F-GMBH- Germany.
2.2. Study design
20 healthy volunteers were divided randomly to 3 groups (I, II and III). Group I (n=8) was allowed to take natural
(crude)black seed oil (2 teaspoonful/d), Group II (n=8) was allowed to take commercial ANSO-SYP black seed oil
(Nigoil) (2 teaspoonful/d), group III (n=6) was kept as placebo only taken (2 teaspoonful/d of sun flower oil).
Washout period was done for all groups for two weeks. Cross over between group I and II was done in this part,
where group I (n=8) was allowed to take natural (crude) black seed oil (2 teaspoonful/d), group II (n=8) was allowed
to take commercial ANSO3 Phenomenalsoft gelatin capsule (2 capsules/d), group III (n=6) was kept as placebo
only taken (2 teaspoonful/d of sun flower oil) for two weeks. Blood samples were taken before (baseline) starting
this study and after two weeks of experiment and washout period. The following parameters were measured
complete blood count CBC (AXSYM XT-2000-ROCH), hepatic function enzymes (SGOT [18] and SGPT [19]) and
fasting blood sugar[20]. All this cross over study’s procedure was in accordance with the guidelines for the human,
and approval from the Institutional Research and Ethics Committee, UST was received prior to the experiments.
Statistical analysis
Results were expressed as mean ± s.e.m. (standard error of mean). Statistical difference was calculated by the
students’ t-test and p<0.05 was considered as statistically significant.

503 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

3. Results and Discussion


Table 1. Effect of natural black seed oil and commercial oil and soft gelatin capsule on average (M±SE)
hepatic function
Parameters Natural (crude oil) Commercial ANSO-SYP Placebo
black seed oil (Nigoil)
Groups Before After Before After Before After
SGOT (U/L) 18.83±1.60 18.0±1.61 18.5±1.05 17.7±1.38 17.5±0.64 17.6±1.20
SGPT (U/L) 15.16±2.27 14.66±2.3 14.33±1.49 13.5±2.07 14.1±1.66 15.0±2.30
Washout period for 2 weeks
Parameters Natural (crude oil) ANSO3 Phenomenal soft Placebo
gelatin capsule
Groups Before After Before After Before After
SGOT (U/L) 17.8±0.98 18.0±1.23 15.1±2.27 16.66±2.70 15.9±3.2 16.44±0.78
SGPT (U/L) 14.8±1.77 13.5±2.07 14.66±1.20 14.66±1.20 13.00±1.21 14.27±1.08
Insignificant change with P-value > 0. 05

Table 2. Effect of natural black seed oil and commercial oil and soft gelatin capsule on average (M±SE)
fasting blood sugar in healthy volunteers (n=20)
Parameters Natural (crude oil) Commercial ANSO-SYP black Placebo
seed oil (Nigoil)
Groups Before After Before After Before After
FBS(mg/dl) 90.0±1.46 80.3.±4.84* 94.6±1.52 91.5±2.8 87.2±1.25 89.5±1.61
p-value 0.015 0.26 0.40
Washout period for 2 weeks
Parameters Natural (crude oil) ANSO3 Phenomenal soft Placebo
gelatin capsule
Groups Before After Before After Before After
FBS(mg/dl) 91.2±1.44 79.1±0.90* 93.0±2.08 102.0±1.57*† 89.8±1.39 89.0±2.68
p-value 0.01 0.04 0.33
*Significant as compared with control (Before) at P<0.05, †Significant as compared with Natural NS oil at P<0.05

Table 3. Effect of natural black seed oil and commercial oil and soft gelatin capsule on average (M±SE)
complete blood count (CBC) in healthy volunteers (n=20)
Parameters Natural (crude oil) Commercial ANSO-SYP Placebo
black seed oil (Nigoil)
Groups Before After Before After Before After
Hb(g/dl) 14.58±0.41 14.87±0.54 15.33±0.45 15.25±0.47 13.5±0.311 13.8±0.25
PCV% 43.5±1.23 44.3±1.49 46.0±1.21 45.8±1.44 41.5±0.64 42.6±0.61
RBC X10×12/L 5.27±0.061 5.26±0.098 5.28±0.08 5.55±0.05 5.12±0.1 5.31±0.17
MCV (Femto liters) 83.33±1.40 83.83±1.27 85.66±1.90 85.00±1.31 81.0±0.91 80.1±2.8
MCH (pg) 27.3±0.80 27.5±0.80 29.3±0.98 27.5±0.80 26.7±0.47 26.1±1.24
MCHC (g/dl) 32.67±0.42 32.5±0.43 34.3±0.55 32.3±0.49 32.7±0.48 32.4±0.50
T.WBCX10×9/L 7.12±1.12 6.40±1.37 7.4±0.95 6.53±1.23 5.46±0.62 5.66±0.67
PlateletsX10ᶺ9/L 263.3±20.3 268.0±19.29 250.8±13.5 261.0±16.4 307.8±29.7 305.8±44.0
Washout period for 2 weeks
Parameters Natural (crude oil) ANSO3 Phenomenal soft Placebo
gelatin capsule
Groups Before After Before After Before After
Hb(g/dl) 15.0±0.36 15.3±0.36 14.38±0.48 14.83±0.45 14.68±0.322 14.75±0.41
PCV% 43.4±0.649 43.28±0.83 43.5±0.84 44.5±1.31 46.0±1.21 45.8±1.44
RBC X10×12/L 5.28±0.09 5.25±0.12 5.13±0.033 5.37±0.076 5.28±0.08 5.55±0.05
MCV(Femtoliters) 85.66±1.90 85.00±1.31 84.83±1.40 84.16±1.51 85.66±1.90 85.00±1.31
MCH (pg) 27.6±0.76 27.5±0.84 28.1±0.87 27.6±0.76 27.85±0.67 28.0±0.72
MCHC (g/dl) 32.8±0.47 32.6±0.42 33.1±0.40 32.6±0.33 33.85±0.40 34.0±0.37
T.WBCX10×9/L 6.13±0.97 6.21±0.73 6.33±1.10 5.81±0.96 5.64±0.63 5.90±0.54
PlateletsX10ᶺ9/L 264.1±26.8 317.8±37.3 6.33±1.10 5.81±0.96 5.64±0.63 5.90±0.54

504 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

Discussion
The oil of crude nigella sativa is so beneficial due to its content of over a hundred components such as aromatic oils,
trace elements, vitamins and enzymes. It contains 58% of essential fatty acids including omega 6 and omega 3.
These are necessary for the forming of Prostaglandin E1 which balances and strengthens the immune system giving
it the power to prevent infections and allergies and control chronic illnesses. Healthy cells are protected from viruses
thus inhibiting tumors. Blackseed oil also contains about 0.5 – 1.5% volatile oils including nigellone and
thymoqinone which are responsible for its anti-histamine, anti-oxidant, anti-infective and gastroprotetive effect[4,8].
Outcomes of this study showed that neither natural (crude) nor commercial black seed oil have any change on liver
function enzymes as well as complete blood count.

Ali BH, Blunden G, 2003 disagreed with our findings. He found that treatment of rats with the seed extract for up to
12 weeks induce changes in the haemogram that include an increase in both the packed cell volume (PCV) and
haemoglobin (Hb), and a decrease in plasma concentrations of cholesterol, triglycerides and glucose [21]. Moreover,
preliminary evidence suggests that oil may help minimize chemotherapy-induced decreases in hemoglobin and
leukocyte counts. It helps with the process of thrombosis inside and external side of the body, ensuring that clotting
of blood happens quickly to prevent further loss of blood from the injured parts of the body. It also controls fibrin to
prevent excessive coagulation of blood and to ensure smooth flow of blood in the arteries and veins [22]. In
addition, Enomoto et al., 2001showed that some aromatic compounds present in the extract were found to be more
potent than aspirin, which is well known as a remedy for thrombosis [23].

In many Arab countries N. sativa and its derived products are consumed abusively for traditional treatment of blood
homeostasis abnormalities and as a treatment for dyslipidemia [24].Our results were supported by other studies.
They referred the hepatoprotective of NS to the presence of thymoquinone (TQ)as an active constituent in NS. In
fact, the antioxidant and free radical scavenging properties of many144 Lead molecules from natural products:
discovery and new trends plants have been found to play an important role in their hepatoprotective activity [25, 26,
27, 28].Oxidant stress can increase the susceptibility to irreversible injury by oxidative intoxication and by free
radicals that can result in lipid peroxidation, protein oxidation,protein inactivation, disturbance in calcium
homeostasis and consequent loss of cell viability [29, 30].

In this study, crude oil showed significant reduction in fasting blood sugar. Controversially, commercial NS capsule
showed unexpected significant elevation in the level of fasting blood sugar compared with baseline measurement
(before), natural (crude) as well as placebo group. There is no clear or exact explanation of this effect. It may refer
to the presence of traces amount of impurities. Traditionally N. sativa plant has been used in many Middle Eastern
countries as a natural remedy for diabetes. Significant reduction in blood glucose and cholesterol levels in humans
following the use of the plant was reported [31, 32, 33] to elucidate the mechanism of this antidiabetic action, the
rate of gluconeogenesis in isolated hepatocytes as well as the activity of pyruvate carboxylase and
phosphoenolpyruvate carboxykinase in rat liver homogenates was examined. Extracts of this plant showed
significant decreased in hepatic gluconeogenesis. Similar insulinotropic effects of NSO were recently observed
instreptozotocin plus nicotinamide-induced diabetes mellitus in hamsters (a model of type 2 diabetes) orally fed with
the oil (Fararh et al., 2002) [34].

In this study, positive immunoreactivity for the presence of insulin was observed in the pancreases from oil-treated
vs. non-treated hamsters using immunohistochemical staining, suggestingthat the hypoglycemic effect of NSO
resulted, partly, from a stimulatory effect onbeta cell function with consequent increase in serum insulin level. The
ability of NSO to lower blood glucose concentrations was later confirmed in streptozotocin diabetic. [35], in
contrast, Meral et al, 2001indicated that the anti-hyperglycaemic effect of N. sativa is independent of its
insulinotropic action [36].

However, the hepatic glucose production through gluconeogenesisis known to contribute significantly to
Hyperglycaemia in diabetic patients [37]. Studies on isolated hepatocytes demonstrated a significant decrease in
glucose output from gluconeogenic precursors (alanine,glycerol and lactate) in N. sativa oil treated compared to
untreated animals [38, 39]. This significant decrease in liver glucose output suggests that the observed antidiabetic
action of N. sativa oil is at leastpartially mediated through a decrease in hepatic gluconeogenesis through the
inhibition of the enzymatic activities of phosphoenol pyruvate carboxykinase(PEPCK) and of pyruvate carboxylase
(PC) [38]. Increased glycolysis in peripheral tissues and/or inhibition of the release of counter-regulatory hormones
(e.g., glucagon, cortisol and growth hormone)are possible contributory mechanisms that may be considered and
require further investigation [39].

505 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

4. Conclusion
From this study we can conclude that crude natural herbs contain several hundred constituents and some of them are
present at very low concentrations each one may have benefits or no effect. Commercial products may use only the
major constituents; some of them are composed from volatile oil. During formulation process or long standing
storage may lose their activity. In our study commercial NS capsules showed significant elevation in fasting blood
sugar with no effect on other parameters. Further study should be focused on the efficacy of commercial products
after their storage through quality control assessment.

5. Acknowledgement
This study was partially funded by College of Pharmacy. Author would like to thank all who contributed to the
success of this study, especially graduated students.

6. References
1. Zubaida A. Hawsawi; Basil A. Ali; Abdullah O. Bamosa. Effect of nigella sativa (black seed) and
thymoquinone on blood glucose in albino rats. Annals of Saudi Medicine, 2001, 21: 242-244.
2. Anna Wajs, Radoslaw Bonikowski and Danuta Kalemba. Composition of essential oil from seeds of
Nigella sativa L. cultivated in Poland. Flavor and Fragrance Journal Flavour Fragr., 2008, 23: 126–132.
3. Menounos P, Staphylakins K and Gegiou D, The sterols of Nigella sativa seed oil. Phytochemistry, 1986,
25: 761-763
4. Gali-muhtasib H, El-najjar N, Regine Schneider S. The medicinal potential of black seed (Nigella sativa)
and its componentsLead Molecules from Natural Products, 2006, pp.133-153
5. Shahzad M1, Yang X, RazaAsim MB, Sun Q, Han Y, Zhang F, Cao Y, Lu S (). Black seed oil ameliorates
allergic airway inflammation by inhibiting T-cell proliferation in rats. Pulm Pharmacol Ther., 2009,
22(1):37-43
6. Marozzi FJ Jr, Kocialski AB and Malne M.H. Studies on the antihistaminic effects of thymoquinone and
Quercetin. Arzneim Forsceh (Drug Res), 1970, 10: 1574-1577
7. ElTahir KEH, Ashour MM and Al-Harbi MM (1993). The respiratory effects of the volatile oil the
blackseed (Nigella sativa) in guinea-pigs; elucidation of the mechanism of action. Gen Pharmacol;24:
1115-1122
8. Kanter M, Demir H, Karakaya C and Ozbek H. Gastroprotective activity of Nigella sativaL. oil and its
constituent, thymoquinone against acute alcohol-induced gastric mucosal injury in rats.World J
Gastroenterol, 2005, 11: 6662-6
9. Kanter M, Meral I, Yener Z, Ozbek H and Demir H. Partial Regeneration / Proliferation of the B-cells in
the islets of Langerhans by N. sativa L in Streptozotocin-induced diabetic rats. Tohoku J. Exp Med; 2003,
201: 213-9
10. Mutabagani A and El-Mahdy SAM. A study of the anti-inflammatory activity of Nigella sativa L. and
thymoquinone in rats. Saudi Pharm J, 1997, 5(2-3): 110-113
11. Mohtashami R, Amini M, FallahHuseini H, Ghamarchehre M, Sadeqhi Z , Hajiagaee R , FallahHuseini A
(2011). Blood Glucose Lowering Effects of Nigella Sativa L. Seeds Oil in Healthy Volunteers: a
Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Journal of Medicinal Plants, 2011, 10(39):
90-94.
12. Alzuhair HH, El-Sayed, MI and Sudek MA. Hypoglycemic effect of the volatile oil of Nigellasativa and
Allium sativum and their interactions with glipizide on alloxan-diabetic rats. Bull Faculty of Pharmacy
(Cairo), 1996, pp. 9
13. Al Gharably NM, Badry O, Nagi M. Protective effect of thymoquinone against CCl4-inducedhepatotoxicity
in mice. Res. Comm. Pharmacol Toxicol, 1997, 2: 41-50
14. Nagi MN, Alam K, Badary OA, Al-Shabanah OA, Al-Sawaf HA and Al-Behairy AM. Thymoquinone
protects against CCl4-hepatotoxicity in mice via an antioxidant mechanism.BiochemMolBiolInt, 1999, 47:
153-9J
15. Badary GA, Nagi MN, Al-Shabanah OA, Al-Sawaf HA, Al-Sohaibani MO and Al-Bekairi AM.
Thymoquinone ameliorates the nephrotoxicity induced by cisplatin in rodents and potentiates itsanticancer
activity. Can J PhysiolPharmacol, 1997, 75: 1356-61
16. Rooney S and Ryan MF. Effects of alpha-hederin and thymoquinone - a constituent of Nigellasativa, on
human cancer cell lines. Anticancer Res, 2005, 25(38): 2197-2204
17. Bergmeyer HU, Hørder M, Rej R. Approved recommendation on IFCC methods for the measurement of
catalytic concentration of enzymes. Part2. IFCC method for aspartate aminotransferase. J. ClinChemClin
Biochem, 1985, 24: 497-510.

506 | International Journal of Current Trends in Pharmaceutical Research


Doa’a Anwar Ibrahim, IJCTPR, 2014, Vol.2(4): 502-507

18. Rooney S and Ryan M. F. Modes of Action of alpha-hederin and thymoquinone, active constituentsof
Nigella sativa against HEP-2 cancer cells. Anticancer Res., 2005, 25(36): 4255-9
19. Schumann G et al (1986). IFCC Primary Reference Procedures for the Measurement of Catalytic Activity
Concentrations of Enzymes at 37 °C – Part 4. Reference Procedure for the Measurement of Catalytic
Activity Concentrations of Alanine Aminotransferase.
20. Knudson PE, Weinstock RS. Carbohydrates. In: Henry JB, ed.ClinicalDiagnosis and Management by
Laboratory Methods. 20th ed.Philadelphia: WB Saunders, 2001, pp. 211-223.
21. Ali BH, Blunden G (2003). Pharmacological and toxicological properties of Nigella sativa. Phytother Res.,
2003, 17(4): 299-305
22. Breyer S, Effenberger K and Schobert R (2009). Effect of thymoquinone-fatty acid conjugates on cancer
cells, Chem Med Chem, 2009, 4(5): 761-768.
23. Enomoto S, Asano R, Iwahori Y, Narui T, Okada Y, Singab AN, Okuyama T. Hematological studies on
black cumin oil from the seeds of Nigella sativa L. BiolPharm, Bull., 2001 24: 307–10.
24. Zaoui A, Cherrah Y, Alaoui K, Mahassine N, Amarouch H, Hassar M. Effects ofNigella sativa fixed oil on
blood homeostasis in rat. J Ethnopharmacol, 2002, 79: 23–6
25. Kiso Y, Tohkin M, Hikine H, Hattori M, Sakamoto T, Namba T. Mechanism of antihepatotoxic activity of
glycerrhizin. Effect of free radical generation and lipid peroxidation. Planta Med, 1984, 50: 298–302.
26. Valenzuela A, Guera R, Videla LA. Antioxidant properties of the flavonoids silybinand +X_ cyanidanol-3:
comparison with butylatedhydroxy anisole and butylatedhydroxy toluene. Planta Med, 1986, 52: 438–40.
27. Navaro MC, Montilla MP, Martin A, Jimenez J, Utrilla MP. Free radical scavenger and antihepatotoxic
activity of Rosemarinustomentosus. Planta Med., 1993, 53: 312–4.
28. Thabrew MI, Gove CD, Hughes RD, McFarlane IG, Williams R. Protective effects of Osbeckiaoctandra
against galactosamine and tert-butyl hydroperoxide-induced hepatocytedamage. J Ethnopharm, 1995, 49:
69–76.
29. Masaki N, Kyle ME, Farber JL. Tert-butyl hydroperoxide kills cultured hepatocytes byperoxidizing
membrane lipids. Arch BiochemBiophys, 1989, 269: 390–9.
30. Shertzer HG, Bannenberg H, Zhu RM, Moldeus P. The role of thiols in mitochondrial susceptibility to iron
and tert-butyl hydroperoxide-mediated toxicity in cultured hepatocytes. Chem Res Toxicol, 1994, 7: 358–
66.
31. Padmaa M (2010). Nigella sativa Linn-A comprehensive review. Indian Journal of Natural Products and
Resources., 2010, 1(4): 409-429
32. Al-Hader AA, Aqel MB, Hasan ZA. Hypoglycemic effects of the volatile oil of Nigellasativa seeds. Int J
Pharmacogn, 1993, 31: 96–100
33. Haq A, Lobo PI, Al-Tufail M, Rama NR, Al-Sedairy ST. Immunomodulatory effect ofNigella sativa
proteins fractionated by ion exchange chromatography. Int J Immunopharmacol, 1999, 21: 283–95.
34. Fararh KM, Atoji Y, Shimizu Y, Takewaki T. Insulinotropic properties of Nigellasativa oil in
streptozotocin plus nicotinamide diabetic hamster. Res Vet Sci, 2002, 73: 279–82.
35. El Dakhakhny M, Mady N, Lembert N, Ammon HP. The hypoglycemic effect ofNigella sativa oil is
mediated by extrapancreatic actions. Planta Med., 2002, 68:465–6.
36. Meral I, Yener Z, Kahraman T, Mert N. Effect ofNigella sativa on glucose concentration, lipid
peroxidation,anti-oxidant defence system and liver damaga in experimentally-induced diabetic rabbits.J Vet
Med A, 2001, 48: 593-599
37. Ishikawa Y, Watanabe K, Takeno H, Tani T. Effectof the novel oral antidiabtic agent HQL-975 on
oralglucose and lipid metabolism in diabetic db/dbmice. Drug Res., 1998, 48: 245-250.
38. Al-Awadi FM, Fatania H, Shamte U. The effect ofa plant mixture extract on liver gluconeogenesis
instreptozotocin induced diabetic rats. Diabetes Res., 1991, 18: 163-168.
39. Fararh KM, Atoji Y, Shimizu Y, Shiina T, NikamiH, Takewaki T. Mechanisms of the hypoglycaemic and
immuopotentiating effects of Nigella sativa L. oil in streptozotocin-induced diabetichamsters. Res Vet Sci.,
2004, 77: 123-129.

507 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

ISSN: 2321-3760
Research Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 508-515
www.pharmaresearchlibrary.com/ijctpr

Development and Evaluation Sustained Release Matrices of Lamivudine by


using Synthetic Polymer
Supriya Deogire*, Sonali Mahaparale
Padm. Dr. D.Y. Patil College of Pharmacy Akurdi, Pune–44, India.
Received: 28 May 2014, Accepted: 30 June 2014, Published Online: 15 July 2014

Abstract
The present work is aimed at preparing and evaluating sustained release matrix tablets of Lamivudine using
Synthetic polymer i.e. Glyceryl behenate (Compritol 888) with different polymers concentration by direct
compression and Hot melt granulation techniques. Lamivudine is a potent hydrophilic anti viral agent
indicated for treatment of AIDS (Acquired Immunodeficiency Syndrome). It was found that the cumulative
percent drug release decreased with increasing concentration of Polymer. Matrix tablets were prepared by
direct compression method and hot melt granulation technique taking Glyceryl behenate (Compritol 888) as
different concentration in increasing order and F001 to F008 total eight formulation were prepared. The
powders are evaluated for flow properties and tablets were evaluated for hardness, friability, dissolution
rate, kinetics studies etc. No chemical interaction between Drug and the polymer were seen as confirmed by
FT-IR studies. Among all the formulation made by direct compression and hot melt granulation it was
found that the formulation containing Glyceryl behenate (Compritol 888) in higher concentration using Hot
melt granulation showing best sustained release or retard the release of drug up to 24 hours.
Keywords: Lamivudine; Glyceryl behenate (Compritol 888); hot melt granulation; Sustained release.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 508
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 509
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .511
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . .514
5. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .514

*Corresponding author
Supriya Deogire
Padm. Dr. D.Y. Patil College of
Pharmacy Akurdi, Pune–44, India.
Manuscript ID: IJCTPR2152 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved

1. Introduction
Lamivudine is a potent hydrophilic anti viral agent indicated for treatment of AIDS (Acquired Immunodeficiency
Syndrome). It belongs to class III of the BCS Classification with High solubility and low permeability.
Pharmaceutical research since 1950 turned to a new era towards optimizing the efficacy of the drug by designing the
drug in different dosage forms posing challenges to the pharmaceutical technologists. The oral conventional types of
drug delivery systems are known to provide a prompt release of drug [1-2]. Therefore, to achieve as well as to

508 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

maintain the drug concentration within the therapeutically effective range needed for treatment, it is often necessary
to take this type of drug delivery systems several times a day. This results in a significant fluctuation in drug levels
often with sub-therapeutic and/or toxic levels and wastage of drug. In recent years, various modified-release drug
products have been developed to control the release rate of the drug and/or the time for drug release. [3-7].
Lamivudine is a potent nucleoside analog reverse transcriptase inhibitor (nRTI) and it is the (-) enantiomer of a
dideoxy analogue of cytidine. Lamivudine is rapidly absorbed with a bioavailability of over 80% following oral
ingestion. The drug half-life in plasma is approximately 5-7 hours. It is bound to plasma proteins less than 36%. It
can inhibit both types (1 and 2) of HIV reverse transcriptase and also the reverse transcriptase of hepatitis B [8]. The
aim of present investigation was to design and evaluate sustained release matrix tablet with compritol 888 by direct
compression and melt granulation technique and effect of polymer concentration on release kinetics of drug release,
using distinct formulations.
2. Materials and Methods
2.1 Materials:
Lamivudine (LAM), Glyceryl behenate (Compritol 888), Lactose monohydrate, Aerosil, Magnesium stearate were
obtained as a gift sample from Cipla Pharma Pvt Ltd (Mumbai, India).
2.2 Preparation of Lamivudine Standard graph
Stock solution of Lamivudine was prepared by using 100 ml volumetric flask.10 mg of lamivudine dissolved in 25
ml methanol and volume was make up by distilled water. 1 to 12 mcg/mL concentrations were prepared of solutions.
The absorbance of above solutions was recorded at λmax (271 nm) using double beam UV-Visible
spectrophotometer. Standard graph was plotted between the concentration (on X-axis) and absorbance (on Y axis).
2.3 Compatibility of Lamivudine with drug-polymer
The pure drug and drug-polymer combinations of various physical mixtures were subjected to IR Spectroscopy
using Fourier Transform Infrared spectrophotometer (Bruker, Germany). Their spectra were obtained over the wave
number range of 4000 – 400 cm-1.
2.4 Preparation of Lamivudine Matrix Tablets
All the matrix tablets, each containing 150 mg of Lamivudine, were prepared by direct compression and Hot melt
granulation technique.
2.4.1 Direct compression method:
The distinct formulations of the matrix tablets analyzed along this study are provided in Table 1. The drug is
weighed as per the formula and passed through the sieve no.25 and excipients were weighed as per formula and
passed through Sieve no. 40 mesh separately and collected. Ingredients were mixed in geometrical order and
thoroughly mixed in a polythene bag for 15 minutes to get a uniform mixture. Magnesium stearate were added to
the powder mixture and compressed on a 16- station (single punch, semi automatic, model 999, Shimadzu
Corporation, Kyoto, Japan) tablet compression machine using 9.8 mm X 10mm round flat face punch.(Batch no.
F001, F002, F003, F004). The drug polymer ratio was developed to adjust drug release as per theoretical release
profile and to keep total weight of tablet constant for all the fabricated batches under experimental conditions of
preparations. The total weight of the matrix tablets was 325 mg with different drug polymer ratios like 1:0.25,
1:0.50, and 1:0.75.
2.4.2 Melt granulation method:
The distinct formulations of the matrix tablets analyzed along this study are provided in Table 1. The drug is
weighed as per the formula and passed through the sieve no.25 and excipients were weighed as per formula and
passed through Sieve no. 40 mesh separately and collected. Glyceryl behenate melted in a porcelain dish over a
water bath maintained at 75 °C for 3 min and Lamivudine was gradually added with continuous stirring until
uniformly mixed. The molten mixture was allowed to cool and solidify at room temperature crushed in a mortar and
passed through a sieve no. 25. The granules were lubricated by magnesium stearate as per quantity in formula and
compressed on 16 stations (single punch, semi automatic, model 999, Shimadzu Corporation, Kyoto, Japan) tablet
compression machine using 9.8 mm X 10mm round flat face punch at a force of 1 ton.(Batch no. F005, F006, F007,
F008) [9, 10].

Table 1. Composition of different formulations


Formulation Code F1 F2 F3 F4 F5 F6 F7 F8
Lamivudine 150 150 150 150 150 150 150 150
Glyceryl behenate 37.5 75 75 112.5 37.5 37.5 75.00 112.5
MCC 126.50 _ _ _ _ _ _ _
Lactose monohydrate _ 89.0 87.00 49.50 126.50 125.00 87.50 50.00
Aerosil 5.00 5.00 5.00 5.00 5.00 6.5 6.5 6.5
Magnesium stearate 6.00 6.00 8.00 8.00 6.00 6.00 6.00 6.00
Total weight of tablet 325.00 325.00 325.00 325.00 325.00 325.00 325.00 325.00

509 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

2.5 Evaluation of Precompression Blend


Prior to compression, granules were evaluated for their micromeritic parameters. Angle of repose was determined by
funnel method. Bulk density (BD) and tapped density (TD) were determined by cylinder method, and Carr’s index
(CI) was calculated using the following equation
CI= (TD-BD)/TD ×100.......................................(1)
Hausner’s ratio (HR) was calculated by the following equation
HR=TD/BD..........................................................(2)
2.6 Physicochemical Evaluation of Matrix Tablets
2.6.1. Friability
Friability test is performed to assess the effect of friction and shocks, which may often cause tablet to chip, cap or
break. Preweighed randomly selected twenty tablets were placed in a Roche friability tester and operated for 4 min
at 25 rpm. Compressed tablets should not lose more than 1% of their weigh. [11]
2.6.2. Thickness and diameter
Tablet thickness and diameter were measured by Vernier calipers (Mitatoyo, Japan). [12 ]
2.6.3. Weight variation
A weight variation test was performed according to USP30 NF25 on 20 tablets by taking samples from a batch after
production of every 100 tablets and randomly from a total batch of 300 tablets using an electronic balance (Contech
Instruments CA 224, India). [13]
2.6.4. Hardness
Tablet hardness was measured by using Monsanto hardness tester. From each batch six tablets were measured for
the hardness and average of six values was noted along with standard deviations.
2.6.5. Drug Content Uniformity
The drug content of the matrix tablets was determined according to in-house standards and it
meets the requirements if the amount of the active ingredient in each of the 10 tested tablets lies within the range of
90% to 110% of the standard amount.Ten tablets were weighed and taken into a mortar and crushed into fine
powder. An accurately weighed portion of the powder equivalent to about 150 mg of lamivudine matrix tablet was
transferred to a conical flask containing 100ml of pH 0.1N HCl. It was shaken by mechanical means for 24h.Then it
was filtered through a Whatman filter paper (No. 1) and appropriate dilutions were made and the absorbance was
measured at 280 nm by using double beam UV-VIS spectrophotometer. [14]
2.6.6 In -Vitro Drug Release Characteristics
Drug release was assessed by dissolution test under the following conditions: n = 3, USP type II dissolution
apparatus (Paddle method) at 75 rpm in 900 mL of 0.1N HCl throughout the dissolution up to 24 hours, maintained
at 37°C ± 0.5°C. An aliquot (5mL) was withdrawn at specific time intervals and replaced with the same volume of
pre warmed (37°C ± 0.5°C) fresh dissolution medium. The samples withdrawn were filtered through Whatman filter
paper (No.1) and drug content in each sample was analyzed by UV-visible spectrophotometer at 280 nm. [15]
2.6.7 Drug release kinetics
To determine the mechanism of drug release from the formulations, the data were subjected to

Zero-order------ (Eq 1),


First order------- (Eq2) and
Highuchi -------- (Eq 3) release kinetics

Mt = M0 + k0t ….…………………………(1)
In Mt = In M0 + k1t ………….…………..(2)
Mt = M0 + kHt1/2 ………………….…….(3)

Where Mt is the cumulative amount of drug released at any time, t, and M0 is the dose of the drug incorporated in
the delivery system. k0, k1 and kH are rate constants for zero-order, first order and Higuchi models, respectively.
The dissolution data were also fitted according to the well-known exponential equation of Peppas as in Eq 4, which
is often used to describe drug release behavior from polymeric systems.

Mt/M_ = ktn ……………….……………….. (4)

Where, Mt/M_ is the fraction of drug released at time, t, k is the kinetic constant, and n is the diffusional exponent
for drug release. The diffusional exponent, n, is dependent on the geometry of the device as well as the physical
mechanism of release. Zero order release describes a release rate independent of drug concentration while the
Higuchi square root kinetic model describes a time dependent release process. The value of n indicates the drug
release mechanism; if 0.1 < n < 0.5, Fickian diffusion is indicated while 0.5 < n < 1 indicates non-Fickian diffusion.
[16].
510 | International Journal of Current Trends in Pharmaceutical Research
Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

3. Results and Discussion


3.1 Lamivudine calibration curve
The standard graph of Lamivudine has shown good linearity with R2 value 0.993 in
pH 6.8 buffer (Fig-1), which suggests that it obeys the “Beer-Lambert’s law”.

Figure 1. Standard graph of Lamivudine in Methanol

3.2 Fourier Transmission Infra Red (FTIR) Studies


FTIR studies were performed on drug and the optimized formulation using Shimadzu FTIR
(Shimadzu Corp., India). The samples were analyzed between wave numbers 4000 and 400 cm-1. (Figure-2, 3)

Figure 2. IR Spectra of Lamivudine

45

%T

40

35

30
2098.62

1888.37

25
1963.60
2343.59

459.07
545.87
852.56
916.22

636.53

20
725.26
2671.50

1388.79

796.63
1732.13

1350.22

1282.71

1039.67
3319.60
3273.31

3080.42

1168.90
3130.57

1637.62

1502.60
3200.01

2845.10

15
2908.75

10

4000 3600 3200 2800 2400 2000 1800 1600 1400 1200 1000 800 600 400
Lamov udine 1/cm

Figure 3. IR Spectra of Lamivudine with Excipients

511 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

3.3 .Micromeretic properties of granules of different formulations.


The physical mixture for matrix tablets were characterized with respect to angle of repose, bulk density, tapped
density, Carr’s index, and drug content (Table 2). Angle of repose was less than25° and Carr’s index values were
greater than 25 for the powder of all the batches indicating excellent to poor flowability and compressibility.
Hausner’s ratio was found to be between 1.4 to1.7 for all the batches indicating that passable to poor flow
properties. The drug content was more than 95 % for all the different formulations.

Table 2. Micromeretic properties of granules of different formulations


Formulation Bulk Tapped Hausner’s Compressibility
Angle of repose
Code Density Density Ratio Index
F001 0.495 0.625 1.26 20.8 35 ± 0.65
F002 0.523 0.689 1.31 24 40 ± 0.72
F003 0.512 0.625 1.22 18 37 ± 0.77
F004 0.521 0.671 1.28 22 28 ± 0.29
F005 0.515 0.662 1.28 22 33 ± 0.81
F006 0.588 0.659 1.12 10.77 30 ± 0.72
F007 0.595 0.675 1.13 13.44 38 ±0.65
F008 0.591 0.681 1.11 9.00 38 ±0.65
*All values are expressed as mean ± S.D,

3.4 Physiochemical characterization of Tablets


The results of the uniformity of weight, hardness, thickness, friability, and drug content of the Tablets are given in
(Table 3). All the tablets of different batches complied with the official requirements of uniformity of weight as their
weights varied between 308.75 mg and 341.2mg (±5%) The hardness of the tablets ranged from 7.00 to
10.00kg/cm2 and the friability values were less than 0.8% indicating that the matrix tablets were compact and hard.
The thickness of the tablets ranged from 6.80 to 7.25 mm. All the formulations satisfied the content of the drug as
they contained 95 to 99 % of Lamivudine and good uniformity in drug content was observed. Thus all the physical
attributes of the prepared tablets were found be practically within control.

Table 3. Physicochemical characterization of Tablets


Formulation Code Average Weight (mg) Thickness (mm) Hardness (Kg/cm2) Friability(%)
F001 324.854.53 6.790.05 90.458 0.10%
F002 328.235.31 7.120.04 80.435 0.18%
F003 322.562.56 6.890.07 8.10.519 0.15%
F004 324.343.43 6.770.02 7.7660.65 0.14%
F005 324.273.57 7.210.06 8.350.535 0.16%
F006 324.632.45 6.840.09 70.5 0.19%
F007 324.524.12 6.870.07 9.1330.50 0.19%
F008 325.734.23 6.910.13 8.740.329 0.12%
*All values are expressed as mean ± S.D.

3.5 In vitro drug release


The drug release data are shown in Table 4. Expectedly, drug release from the tablets Prepared by melt granulation
were 78.85±0.98%, 72.25±0.19% and 66.78±1.02 after 12 h for formulations F006, F007, and F008 respectively,
thus indicating that drug release fall as the glyceryl behenate content of the tablets increased. The difference in
release rate between the batches was statistically significant (p < 0.05). Furthermore, comparison of the two methods
of formulation used indicate that cumulative release from the tablets prepared by direct compression (F002,86.80 %)
was significantly higher (p < 0.05) than from the equivalent formulation made by melt granulation (F008, 66.78 %)

512 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

Table 4. Dissolution Profile (% drug release) of different formulations at different time points.
Time % of Drug Release
(hr) F001 F002 F003 F004 F005 F006 F007 F008
0 0 0 0 0 0 0 0 0
2 28.66±1.25 25.42±1.04 24.22±1.17 21.34±1.15 26.49±1.02 25.69±1.13 20.42±1.11 19.44±0.99
4 40.63±1.30 35.63±1.23 34.83±1.11 30.65±1.08 36.59±1.07 35.85±1.15 31.65±1.05 29.39±1.10
6 52.49±0.92 45.72±0.92 45.43±0.95 40.72±0.93 47.41±0.93 48.70±0.97 40.45±0.81 39.46±0.83
8 64.37±1.15 58.45±0.87 56.56±0.80 49.73±0.90 57.68±1.11 58.43±1.06 49.35±0.92 49.50±1.10
10 76.42±0.87 74.12±0.80 73.14±0.78 60.14±0.74 68.61±0.81 69.52±0.97 61.45±0.97 57.51±0.85
12 86.80±0.45 80.45±1.11 78.12±0.98 74.14±0.16 80.67±0.15 78.85±0.98 72.25±0.19 66.78±1.02
*All values are expressed as mean ± S.D

3.6 Drug release kinetics


Table 5 shows that the best-fit release kinetic data with the highest values of regression
Coefficient (R2) was shown by zero order and Higuchi models. Zero order release describes the release rate
independent of drug concentration.Higuchi square root kinetic model describes, release drug from the insoluble
matrix as square of time dependant process. It describes release of drug by simple diffusion mechanism. The values
of n were in the range of 0.5531 to 0.7166 (i.e., more than 0.5) indicating non- Fickian release (diffusion
controlled).R2 data indicate that Higuchi and Peppas models also suitably described the release of Lamivudine from
the matrix tablets.

Table 5. Kinetic Values Obtained from Different Plots of Formulations


Formulation Zero order First order Higuchi Korsmeyer and Peppas
Code R2 R2 R2 n R2
F001 0.9580±0.012 0.8580±0.005 0.9580±0.0033 0.5679 0.9780±0.064
F002 0..9967±0.022 0..897±0.0 0..9468±0.081 0.5903 0..9680±0.0115
F003 0.9985±0.007 0.9085±0.004 0.95185±0.0040 0.6670 0.9804±0.0087
F004 0.9952±0.003 0.9152±0.004 0.9752±0.0026 0.5639 0.9801±0.0046
F005 0.9675±0.004 0.8975±0.006 0.9875±0.0036 0.5616 0.9747±0.0038
F006 0.9635±0.001 0.9154±0.0034 0.9834±0.0070 0.6203 0.9818±0.0038
F007 0.9876±0.003 0.9276±0.004 0.9843±0.007 0.7075 0.9762±0.0070
F008 0.9996±0.001 0.9446±0.008 0.9916±0.008 0.7164 0.9815±0.0054

Figure 4. Comparative Drug release profile of all Batches

3.7 Effect of concentration of the glyceryl behenate matrix on drug release


During preliminary studies (not reported here), it was observed that at low concentrations of the glyceryl behenate
the matrices of the tablets readily disintegrated during dissolution test. Disintegration properties of these matrices
were depending on content of matrix forming agent. Hence different ratios of drug: Lipophilic binders were to
prepare matrices and drug release retardation. This was not, however, the case when the content of the matrix former
was increased, thus indicating that a minimum of level of the glyceryl behenate is required to form a proper matrix
that would not readily disintegrate. This may be due to slower penetration of the dissolution medium into the waxy
513 | International Journal of Current Trends in Pharmaceutical Research
Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

matrices i.e. increasing the ratio of drug: lipophilic binder from 1:0.25, 1:0.50, 1:0.75 resulting decrease release of
drug. In all formulation F008 shows maximum drug release retardation because of high concentration of polymer in
the ratio of 1:0.75 i.e. 66.78±1.02%. From figure 4 it can be observed that for all the matrices drug release is
inversely proportional to level of rate retarding matrix former present in the matrix system i.e. the rate and extent of
drug release decrease with increase in total lipid content of matrix. Lamivudinne release occurred by different
mechanism diffusion or erosion depending on lipid binder used. Increasing the ratio of glyceryl behenate in granules
resulted in decreasing the release of drug. As stated earlier, based on kinetic analysis of release data, Lamivudine
release occurred by a non- Fickian diffusion mechanism. The initial drug release (i.e., in the 1st hour) of
15.44±1.21% and after 12 h 66.78±1.02 respectively of batch F008.
3.8 Effect of method of tablet preparation on drug release
Figure 5 compares the release profiles of tablets prepared by melt granulation and direct compression methods,
respectively. Both cumulative drug release and drug release rates were higher for the matrix tablets made by direct
compression of physical mixtures (F001) than for the tablets obtained by the compression of granules made by melt
granulation (F008) This can be attributed to the formation of a more uniform and, therefore, more effective coating
of the glyceryl behenate matrix around the drug particles in the tablets prepared by melt granulation technique than
in those made by direct compression. Thus the tablets made by the former technique are likely to show greater
integrity. Consequently, while the probable mechanism of drug release from the direct compressed-matrix tablets is
erosion control, drug release from melt granulated tablets would likely be diffusion-controlled as confirmed by the
kinetic data in Table 5. All the parameters were run 3 times (n=3).The difference in the mean of % Cumulative drug
release between batch F008 and F001, F002,F003 was significant (p< 0.05)
Melt granulation > physical mixture

Figure 5. Comparative Drug release profile of batches showing effect of method preparation of matrix tablet

4. Conclusion
From preformulation studies, it was found that the sample of lamivudine is pure and suitable drug candidate for
formulation of lipophilic matrix tablets. From compatibility studies it was no evidence of interaction between drug
and polymer and polymer is suitable for preparation of matrices of highly water soluble drug. From pre-compression
studies it was concluded that all the parameters passes the standards and the granules are suitable for preparation of
lamivudine matrix. From post-compression studies it was concluded that all the parameters passes the standards and
the melt granulation method was suitable for preparation of lamivudine matrices. From the in-vitro release study it
was concluded that as the concentration of compritol 888 is increased, the drug release rate was decreased. Both
cumulative drug release and drug release rates were higher for the matrix tablets made by direct compression of
physical mixtures than for the tablets obtained by the compression of granules made by melt granulation. Overall the
curves fitting into various kinetic models confirmed that in-vitro release kinetics of all formulations was best fitted
into Zero order model and Higuchi model. The n values more than 0.5 indicates that the mechanism in which the
drug release from matrices follow non-fickian diffusion mechanism. The developed Lamivudine matrix tablets
prepared by melt granulation may be used clinically for prolonged drug release for at least 24 h.

5. References
1. Balyan JC. Encyclopedia of Pharmaceutical Technology. New York: Marcel Dekker. 281-313, 1990.
2. Y.W. Chien. Novel drug delivery systems. 2nd ed. New York, Marcel Dekker, Inc. 1992.
3. J.R.Robinson, S.P. Eriksen. J Pharm Sci. 1966, 55: 1254-1263.

514 | International Journal of Current Trends in Pharmaceutical Research


Supriya Deogire, IJCTPR, 2014, Vol.2(4): 508-515

4. R.W. Korsmeyer, R.Gurny, E. Doelker, P.Buri, N.A Peppas.. Int J Pharm. 1983, 15: 25-35.
5. L. Lachman, H.A.Lieberman, J.L.Kanig. The Theory and Practice of Industrial Pharmacy. Philadelphia,
PA: Lea and Febiger. 1987, pp. 317-318.
6. P.N. Krishnan, S. Sangeetha, D.N. Venkatesh, R. Saraswathi. Asian J Pharmaceutics. 2007; 1(4):213-216.
7. V.Pillay, R.Fassihi. J Pharm Sci. 1999, 88(11): 1140-1148.
8. P.R.Ravi, U.K.Kotreka, R.N. Saha. AAPS PharmSciTech., 2008, 9(1): 302-313.
9. Aulton E.M., Pharmaceutics: The science of dosage form design, 1990, 610 – 612.
10. L. Lachman, H.A.Lieberman, J.L.Kanig. The Theory and Practice of Industrial Pharmacy. Philadelphia,
PA: Lea and Febiger., 1987, 317-318.
11. Gohel MC, Parikh RK, Brahmbhatt BK, Shah AR. Preparation and assessment of novel coprocessed
superdisintegrant consisting of crospovidone and sodium starch glycolate: a technical note. AAPS
PharmSciTech., 2007, 8: 9.
12. Fukami J, Yonemochi B, Yoshihashi Y, Terada K. Evaluation of rapidly disintegrating tablets containing
glycine and carboxymethylcellulose. Int J Pharm., 2006, 310: 101-9.
13. Madgulkar AR, Bhalekar MR, Padalkar RR. Formulation design and optimization of novel taste masked
mouth-dissolving tablets of tramadol having adequate mechanical strength. AAPS Pharm Sci Tech., 2009,
10: 574-81.
14. Hamdani J, Moës AJ, Amighi K. Physical and thermal characterisation of Precirol and Compritol as
lipophilic glycerides used for the preparation of controlled release matrix pellets. Int J Pharm., 2003, 260:
47-57.
15. Ritger PL, Peppas NA. A simple equation for description of solute release, II: Fickian and anomalous
release from swellable devices. J Control Rel., 1987, 5: 37-42.
16. Higuchi T. Mechanism of sustained-action medication. Theoretical analysis of rate of release of solid drugs
dispersed in solid matrices. J Pharm Sci., 1963, 52: 1145–1149.

515 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

ISSN: 2321-3760
Research Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 516-524
www.pharmaresearchlibrary.com/ijctpr

Formulation and In-Vitro Evaluation of Floating Beads of Propranolol


Hydrochloride Using Foam Technology
Sanjay K. Sharma*, Deepak Sharma,Vaibhav Saxena, Manish Jaimini, Shailender Mohan
Department of Pharmaceutics, Jaipur College of Pharmacy, ISI-15, RIICO, Sitapura, Jaipur (Raj) - 302022, India
Received: 12 May 2014, Accepted: 18 June 2014, Published Online: 15 July 2014

Abstract
An attempt was made to formulate the floating alginate beads containing Propranolol hydrochloride, using
foam solution. Propranolol hydrochloride is a sympatholytic non-selective beta blocker with its absorption
site at stomach. propranolol hydrochloride is more soluble in acidic pH and slightly soluble in neutral or
alkaline pH conditions. Therefore development of a sustained release formulation of propranolol
hydrochloride is advantageous, if the system can remain in stomach for prolonged period of time. Three
polymers sodium alginate, poloxamer188, poloxamer407 were used for the present study. Poloxamer were
used to impart buoyancy to the beads. Total 14 formulations were prepared with different polymer
concentrations. Formulated beads were evaluated for production yield, particle size, swelling index,
buoyancy, drug entrapment efficiency, morphology, in vitro release characteristics. Formulated beads
showed satisfactory floating characteristics and subjected for in vitro release study using 0.1N HCl (pH 1.2)
in USP apparatus II (paddle type). The results indicated that sodium alginate with poloxamer 188 sustain
the drug release better up to 12 hr. To analyze the mechanism of drug release from the beads, the in vitro
release data was fitted into various release models.
Keywords: Floating Beads, Propranolol hydrochloride, HPMC, Buoyancy Test, Poloxamer.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .519
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . .523
5. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 524

*Corresponding author
Sanjay K. Sharma
Department of Pharmaceutics, Jaipur
College of Pharmacy, ISI-15, RIICO,
Sitapura, Jaipur (Raj) - 302022, India
Manuscript ID: IJCTPR2169 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved

516 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

1. Introduction
The oral route represents nowadays the predominant and most preferable route for drug delivery. Oral drug delivery
systems (DDS) are divided into immediate release and modified release systems. Immediate release DDS are
intended to disintegrate rapidly, and exhibit instant drug release. Modified release systems, on the other hand, have
been developed to improve the pharmacokinetic profiles of active pharmaceutical ingredients (APIs) and patient
compliance, as well as reducing side effects (1). Extended, sustained or prolonged release drug delivery systems are
terms used synonymously to describe this group of controlled drug delivery devices, with predictability and
reproducibility in the drug release kinetics. The retention of oral dosage forms in the upper GIT causes prolonged
contact time of drug with the GI mucosa, leading to higher bioavailability, and hence therapeutic efficacy, reduced
time intervals for drug administration, potentially reduced dose size and thus improved patient compliance.
Therefore, extended release DDS possessing gastric retention properties may be potentially useful (2). The concept
of floating drug delivery was reported in the literature as early as 1968, where Davis discovered a method for
overcoming the difficult experienced by some persons of gagging or choking while swallowing medicinal pills.
Some other suggested that such difficulty could be overcome by proving pills having a density of less than
1.004gm/cm3 so they float on water surface. The various buoyant preparation include hollow microsphere
(microballoons), granules powder, capsule, tablet (pills) laminated films (3).
Advantages of Floating Drug Delivery Systems (4):
 Controlled Drug Delivery: These systems can remain in the stomach for longer period of time and hence can
release the drug over an extended period. The problem of short gastric residence time, encountered with an oral
CR formulation can hence be overcome with these systems. These systems have a bulk density of less than 1, as
a result of which they can float on the gastric contents.
 Site-Specific Drug Delivery: These systems are particularly advantageous for drugs that are specifically
absorbed from stomach or the proximal part of the small intestine. Eg. Riboflavin, Furosemide, Ciprofloxacin,
etc.
 Absorption Enhancement: Drugs that have poor bioavailability because of site-specific absorption from the
upper part of the gastrointestinal tract are potential candidates to be formulated as floating drug delivery
systems, thereby maximizing their absorption.
 Reduction of dose: By formulating FDDS of a drug, the dose of the drug can be reduced along with the
frequency of administration.

2. Materials and Methods


Propanalol HCl was gifted from Oniosome Healthcare Pvt. Ltd., Mohali. Sodium alginate and calcium chloride was
purchased from Thomas Baker Pvt. Ltd. Poloxamer 188 was obtained from Signet chemical corporation Pvt. Ltd.
Poloxamer 407 was purchased from Ludwigshfen/BASF Company. All the ingredients used were of analytical
grade.
Preparation of Floating Beads:
Sodium alginate and poloxamer (Alg) was dissolved in distilled water and then poloxamer was then added into the
sodium alginate solution and agitated vigorously by using mechanical stirrer for 20 min. Then drug was added into
the foam solution under vigorous stirring condition continuously. The foam solution was introduced using a 21
gauge syringe into the CaCl2 solution under gentle stirring condition. The distance between the edge of the needle
and the surface of the CaCl2 medium was about 10 cm. The beads formed were left in the solution with gentle
stirring for 10 min at room temperature to be cured. The beads were collected, washed with distilled water twice and
oven-dried subsequently (40 ◦C) (5).

Figure 1. Method of preparation of floating beads using foam solution


517 | International Journal of Current Trends in Pharmaceutical Research
Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

Formula design: 14 batches of floating beads of Propranolol hydrochloride were formulated. Firstly, we optimize
the concentration of Poloxamer for preparation of floating beads by formability and foam stability study.
Foamability and foam stability: Foamability refers to the “ability” of the system to form foam. Foam stability is a
parameter describing variations of the foam properties (mostly as changes of height or volume) with time,
immediately after the foam was generated. Foams were prepared using magnetic stirring. Different amount of
Alginate, poloxamer and Propranolol hydrochloride was added into water and agitated for 20 min at 2600 rpm;
foams were immediately transferred into a graduated cylinder for continued observation. The initial foam volume
after preparation is used to evaluate the foamability. Foamability (FD) was characterized as the physical density of
the foam (ratio of volume of foam/volume of liquid used).
FD = V (foam)/ V (liquid)
Foam stability is characterized as the time interval after which 10 percent of the original amount of liquid has
drained from the foam.

Table 1. Fomability and foam stability


S. No. Poloxamer Poloxamer 188 Poloxamer 407
188, 407 % Foamability Foam Stability Foamability Foam Stability
(Density) (Min) (Density) (Min)
1. 0.0125 1.22 5 1.6 16
2. 0.025 1.6 10 2.2 17
3. 0.05 1.7 18 2.1 22
4. 0.075 2.4 30 1.22 24
5. 0.1 2.5 40 1.8 42
6. 0.2 1.96 10 2.2 13
7. 0.4 2.2 11 2.22 18
8. 0.5 1.92 21 2.2 15

So it was found that as the concentration of poloxamer was increased, foamability and foam stability also got
increased up to 0.1% and then decreased on further increasing the concentration of poloxamer, therefore the
optimize conc. of poloxamer was taken as 0.1% which has highest foam stability for preparation of floating beads.

Table 2. Formula development


Formulation Drug Sodium Poloxamer Poloxamer Calcium
code (mg) Alginate 188 (mg) 407 (mg) Chloride
F-1 80 1% 200 - 1%
F-2 80 1% - 200 1%
F-3 80 1.5% 200 - 1%
F-4 80 1.5% - 200 1%
F-5 80 2% 200 - 1%
F-6 80 2% - 200 1%
F-7 80 3% 200 - 1%
F-8 80 3% - 200 1%
F-9 80 4% 200 - 1%
F-10 80 4% - 200 1%
F-11 80 4% - 200 2%
F-12 80 4% - 200 3%
F-13 80 4% - 200 4%
F-14 80 4% - 200 5%

Evaluation parameters for floating beads:


1. Surface Morphology and Particle Size: The surface morphology and particle size of prepared floating beads
determined by scanning electron microscopy. (5)
2. Percentage Yield: The obtained Beads of each formulation were collected and weighed to determine
production yield (PY) using following equation (6).
Practical yield/Theoritical yield X100
3. Buoyancy study: Floating properties of beads were evaluated using USP dissolution apparatus containing SGF
(pH 1.2). Beads (one hundred) of each batch were placed in 100 ml of 0.1 N HCl agitated at 100 rpm,

518 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

temperature was maintained at 37±0.50c. The number of sinking beads was observed visually. The percentage
of floating beads was calculated according to the following equation:
F (%) = NF/NT X 100
F: floating percent; NF: number of floating beads; NT: total number of the beads. (7).
4. Drug entrapment and entrapment efficiency: Accurately weighed quantities of approximately 50 mg beads
were dissolved in 5 ml 0.1 N HCL (simulated gastric fluid, pH 1.2) and then boil for 20 minute. The solution
was centrifuged at 5000 rpm for 30 min and drug concentration was assayed at 289 nm using a
spectrophotometer. The drug concentration in the sample was used to calculate the percentage drug loading by
dividing the weight of beads initially dissolved and the encapsulation efficiency was calculated as
DL (%) = WD/WT ×100
DL: drug loading; WD: the weight of the drug loaded in the beads; WT: the total weight of the beads.
EE (%) = WA/WT × 100
EE: encapsulation efficiency; WA: actual drug content; WT: theoretical drug content (5).
5. Swelling study: Swelling studies of the beads were carried out by taking Known weight of the Beads and
immersed in excess of 0.1NHCL for definite time interval and then beads were removed and weighed
immediately at regular time interval. The percentage swelling (Ps) of the beads was calculated as:
Ps = [Ws –Wd]/ Wd X 100
Where Ws is the weight of swollen beads and Wd is the weight of dried beads. (8)
6. In vitro drug release study: In vitro dissolution studies were performed for all the formulations using USP
apparatus II (paddle type). An accurately weighed floating alginate beads were taken into 900 ml 0.1N HCL
(pH 1.2). The temperature was maintained at 37±0.50c and stirred at a speed of 50 rpm. At definite time
intervals, a 5-ml aliquot of the sample was withdrawn and the volume was replaced with an equivalent amount
of plain dissolution medium kept at 37±0.50c. The collected samples were filtered and analyzed at 289 nm using
UV- visible spectrophotometer against 0.1N HCl (pH 1.2) taken as blank (9).
7. Drug release kinetics: The release kinetic was studied by various kinetic models as zero order plot, first order
plot, higuchi plot and korsmeyer-peppas. In order to identify a particular release mechanism, experimental data
of statistical significance are compared to a solution of the theoretical model. It is therefore clear that only a
combination of accurate and precise data with models accurately depicting the physical situation will provide an
insight into the actual mechanism of release. To analyse the mechanism for the drug release and drug release
rate kinetics of the dosage form, the data obtained was fitted into Zero order, First order, Higuchi matrix,
Korsemeyer-Peppas. By comparing the R2-values obtained from the above equations, the best-fit model was
selected (10).

3. Results and Discussion


FT-IR Spectra of drug: The samples were scanned in 400-4000 wave number range, using KBr pellet technique.

Figure 2. FT-IR Spectra of Propranolol hydrochloride

Drug-Excipient Interaction Study: This study was carried out by using FT-IR spectrophotometer to find out if
there is any possible chemical interaction of Propranolol hydrochloride with sodium alginate, poloxamer188,
poloxamer 407 and Calcium chloride. No significant interaction is found owing to IR range of drug.
519 | International Journal of Current Trends in Pharmaceutical Research
Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

Figure 3. Drug-excipients compatibility study

Evaluation parameters for floating beads:


Production yield of floating beads: The Production yield of all formulation is tabulated below

Table 3. Production yield of floating beads


Formulation Percentage yield (%)
F3 60.67
F4 60.97
F5 68.64
F6 69.87
F7 81.25
F8 82.81
F9 79.68
F10 88
F11 85
F12 90
F13 82
F14 87

Micromeritic Properties of floating beads of propranolol hydrochloride:

Table 4. Micromeritic Properties of floating beads


Formulation Angle of Bulk Tap Carrs Hausners
code repose density density index ratio
F3 18.85 0.695 0.805 13.65 1.15
F4 19.10 0.712 0.810 12.10 1.13
F5 19.85 0.745 0.855 12.86 1.14
F6 20.55 0.695 0.807 13.87 1.16
F7 23.15 0.625 0.758 17.62 1.20
F8 28.16 0.566 0.675 16.14 1.19
F9 22.65 0.665 0.782 14.96 1.17
F10 25.15 0.595 0.735 19.05 1.23
F11 27.75 0.565 0.697 18.95 1.22
F12 24.66 0.588 0.724 18.80 1.24
F13 30.65 0.515 0.665 22.55 1.29
F14 26.25 0.635 0.753 17.64 1.21

520 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

Surface morphology and Particle size distribution analysis: The morphology of the floating beads was examined
by SEM the view of the microspheres showed a spherical shape with a smooth surface morphology.

Figure 4. SEM of optimized batch (F-14)

Figure 5. SEM of optimized batch (F-14)

Figure 6. SEM of optimized batch (F-14)

521 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

Particle size: It’s determined by an optical microscope fitted with an ocular and stage micrometer and particle size
distribution was calculated. The instrument was calibrated at 1unit of eyepiece micrometer was equal to 1/30mm
(33.33μm).
Table 5. Particle Size
Formulation code Mean Particle Size (µm)
F3 334.78
F4 335.12
F5 321.11
F6 326.25
F7 422.16
F8 456.36
F9 482.22
F10 512.34
F11 516.14
F12 515.12
F13 525.17
F14 544.22
Floating beads of Propranolol hydrochloride prepared in this study were well rounded spheres with the size range
from 321.11 to 544.22 µm.

Drug entrapment efficiency:


The entrapment efficiency varied from 18.06% to 87.15%. The formulation F14 is having high encapsulation
efficiency of 87.15% and F3 is having low encapsulation efficiency of 18.06%. The results of drug loading and drug
entrapment efficiency of all formulation are given below.

Figure 7. % Drug entrapment efficiency

Buoyancy of floating beads: The results indicate that poloxamer is good floating agent which is an important
parameter for floating drug delivery system. Total floating time was found to be greater than 12 hrs and % floating
found in range 80-100%.

Table 6. Buoyancy study of floating beads


Formulation Total floating time (hrs.) % Floating (F (%) = NF/NTX100)
F3 24 90
F4 24 80
F5 24 95
F6 24 92
F7 24 100
F8 24 100
F9 24 100
F10 24 100
F11 24 100
F12 24 100
F13 24 100
F14 24 100

522 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

Swelling study of floating beads:


All formulation show good swelling index, on increasing concentration of sodium alginate swelling index increases
while conc. of calcium chloride have no significant effect on swelling index. Swelling index is directly proportional
to concentration of sodium alginate. The results of swelling studies are given below.

Figure 8. % swelling index of floating beads formulation (F-3 to F-13)

All formulation show good swelling index, on increasing concentration of sodium alginate swelling index increases
while conc. of calcium chloride have no significant effect on swelling index. Swellling index is directly proportional
to concentration of sodium alginate.

In-vitro drug release study: The results of in vitro drug release are given below of optimized formulation F-14.

Figure 9. Drug release pattern of optimized batch F-14

4. Conclusion
The aim of the study was to develop and physically characterize the floating alginate beads of propranolol
hydrochloride using foam solution. Propranolol hydrochloride is a sympatholytic non-selective beta blocker with its
absorption site at stomach. propranolol hydrochloride is more soluble in acidic pH and slightly soluble in neutral or
alkaline pH conditions. Therefore development of a sustained release formulation of propranolol hydrochloride is
advantageous, if the system can remain in stomach for prolonged period of time. Three polymers sodium alginate,
poloxamer188, poloxamer 407 were used for the present study. Poloxamer were used to impart buoyancy to the
beads. Total fourteen formulations were prepared with different polymer concentrations. In the present work, it was
concluded that formulation F14 shows the best release that follows zero order kinetics with anomalous diffusion
method. Therefore, it can be concluded that the Propranolol hydrochloride Floatin Beads have an advantages of
lowering the dose frequency and improve the patient compliance by providing the better management of
hypertension.

523 | International Journal of Current Trends in Pharmaceutical Research


Sanjay K. Sharma et al, IJCTPR, 2014, Vol.2(4): 516-524

5. References
1. Eisen, S.A., Miller, D.K., Woodward, R.S., Spitznagel, E., Przybeck, T.R. The effect of prescribed daily dose
frequency on patient medication compliance. Arch. Intern. Med. 1990, 150: 1881-1884.
2. Shah Chainesh, Aundhia Chintan, Ramani Vinod, Shah Nirmal, et al. “An Overview on Gastro Retentive
Floating Microspheres” Int J Pharm Res Tech. 2012, 2(2): 1-8.
3. Singh, B.N., Kim, K.H.; Floating drug delivery system; An approach to oral controlled drug delivery via gastric
retention. J. Control Release, 2000, 63 (3): 235-259.
4. Kumar Mukesh; Floating drug delivery system: a innovative approach, J Drug Del & Ther, 2012, 2(6): 117-123.
5. Yao Huimin, et.al. Preparation and evaluation of a novel gastric floating alginate/poloxamer inner-porous beads
using foam solution, Int J Pharm. 2011, 422: 211-219.
6. Thomas M., et.al. Preparation and Evaluation of Atenolol Floating Beads as a Controlled Delivery System ,
Iraqi J Pharm Sci, 2010, 20(1): 70-79.
7. Nimse. PK. Preparation and evaluation of floating calcium alginate beads of clarithromycin, Int J Pharm Res
Dev, 2010, (9): 139–45.
8. Pasparakis George, Bouropoulos Nikolaos., Swelling studies and in vitro release of verapamil from calcium
alginate and calcium alginate–chitosan beads, Int J Pharm. 2006, 323: 34–42.
9. Khan AD, Bajpai M. Formulation and Evaluation of Floating beads of Verapamil hydrochloride. Int J Pharm
Tech Res, 2011, 3(3): 1537–46.
10. Patel L Yagnesh, et al., The effect of Drug concentration and curing time on processing and properties of
calcium alginate beads Containing Metronidazole by R; AAPS Pharm Sci Tech, 2006, 7(4).

524 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

ISSN: 2321-3760
Research Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 525-540
www.pharmaresearchlibrary.com/ijctpr

Formulation, Development and Process Optimization of Immediate Release


Tablets of Glimepiride
Vennela K.G*1, Ramesh Reddy K1, Uma Sankar K1, Jayachandra Reddy P2
1
Department of Pharmaceutics, Krishna Teja Pharmacy College, Chadalawada Nagar, Reniguntla Road,
Tirupathi-517 506, Chittoor (dist), A.P., India
2
Department of Pharmaceutical Analysis, Krishna Teja pharmacy College, Chadalawada Nagar, Reniguntla Road,
Tirupathi-517 506, Chittoor (dist), A.P., India
Received: 15 May 2014, Accepted: 7 July 2014, Published Online: 15 July 2014

Abstract
The objective of this study was to design Glimepiride Immediate Release tablets. It is mainly used for the
treatment of type 2 diabetics, sulphonylureas plays important role as therapeutic as well as maintenance
therapy. Glimepiride is Anti diabetic drug. It is used for type 2 diabetic disorders, very low solubility in
aqueous media & oral bioavailability is 100%.its half life is 5-8 hrs & very low clearance 48 ml/min.
Tablets were prepared by wet granulation technique using different polymers such as Lactose
Monohydrate, Starch, Magnesium Stearate, Povidone K 30 as release rate retardant , Colloidal silicon
dioxide, Sodium starch glycolate, Cross carmelose sodium, Crossprovidone, Povidone k-90, Avicel PH
102, as release rate retardant and tablets were evaluated for hardness, friability, weight variation, thickness
and drug content uniformity. In vitro release studies were performed using USP type II apparatus (Basket
method). In vitro release studies revealed that the release rate decreased with increase of polymer loading.
The Drug release was analyzed using zero-order, first order and Higuchi and Korsmeyer-Peppas equations
to explore and explain the mechanism of drug release from the Immediate Release tablets. Mathematical
analysis of the release kinetics indicated that release from the immediate tablets followed diffusion. So the
Immediate tablets could be a potential dosage form for delivering Glimepiride .
Keywords: Glimepiride, Immediate Release tablets, Hydrophilic polymers, Wet granulation.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 526
2. Experimental . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 527
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .535
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . .540
5. Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . .540
6. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .540

*Corresponding author
Vennela K.G
Department of Pharmaceutics, Krishna
Teja Pharmacy College, Tirupathi,
Chittoor, A.P., India-517 506,
Manuscript ID: IJCTPR2189 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved


525 | International Journal of Current Trends in Pharmaceutical Research
Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

1. Introduction
The Product development usually begins when the active chemical entity has been shown to process the necessary
attributes for a commercial product. Generally product development activities can be sub divided into formulation
development and process development1.
Formulation development
Formulation development provides the basic information on the active chemical, the formula and the impact of raw
materials or excipients on the product. A typical supportive data generated during these activities may include:
1. Preformulation profile, which includes all the basic physical or chemical information about the chemical
entity.
2. Effect of formulation variable on the bioavailability of the product.
3. Specific test methods.
4. Key product attributes and specification
5. Optimum formulation
Formulation development should not be considered complete until all those factors which could significantly alter
the formulation have been studied. Subsequent minor changes to the formulation, however, may be acceptable,
provide they are thoroughly tested and as shown to have no adverse effect on product characteristics. In case of drug
development process, compound tested is only one. A variety of studies must be performed for this single drug, each
designed to characterize its efficacy, safety, selectivity or purity. Much of the data generation is driven by strict and
extensive regulatory control and in this most of the studies are interdependent.
Process development can be divided into several stages2:
1. Design
2. Ranging
3. Characterization
4. Verification
1. Design
This is the initial planning stage of process development. During this stage, technical operation in both the
manufacturing and quality-control departments should be consulted. The practically and the reality of the
manufacturing operation should be kept in perspective.
Key documents for the technical definition of the process are the flow diagram, the cause and effect diagram and the
influence matrix.
The flow diagram provides a convenient basic on which to develop a detailed list of variables and responses.
Preliminary working documents are critical, but they should never be “cast in stone”, since new experimental data
may drastically alter them. The final version will eventually be an essential part of the process characterization and
technical transfer documents.
Ranging
Process-ranging studies will test whether identified parameter are critical to the product and process being
developed. These studies determined the:
a. Feasibility of the design process
b. Criticality of the parameter
c. Failure limits for each of the critical variable
d. Validity of the test methods
This is usually a transition stage between the laboratory and the projected final process.
Characterization
Process characterization provides a systematic examination of critical variables found during process ranging.
Advantages
i. The transfer of technology from R & D (sending unit) to manufacturing (Receiving unit) is the first key steps
to getting a high quality product to the market place.
ii. It also useful to make a timeframe of the process for that particular product.
iii. Hold time studies is useful for the planning of the product with other batches.
Objectives
The objective of the technology transfer guide in two-fold.
1. To describe the appropriate information set that needs to be complied to support the transfer of the
information and provide regulatory filing documents.
2. To provide guidance on effective approaches for ensuring this information is available at “print of use”
where guidance on specific topic already exists this will be referred.
Process optimization: In the environment of increasing international competition where counters with lower
production cost luckily catch up technologically, new thinking is required in order to meeting the competition is to
focus on maximizing the utilization of exiting technology. This means much more than just investing in new

526 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

equipment. The ability to optimize or improve a process is dependent upon the ability to control the process. The
ability to control the process is dependent upon the access to reliable and valid management.
Optimization technology: There are two type optimization problems. They are:
Constrained optimization: Constrains are those restricted placed on the system due to physical limitation.
Unconstrained optimization: In unconstrained optimization problems there are no restriction (such as tablet
hardness and disintegration).
An additional complication in pharmacy is that formulations are not usually simple system. They often contain many
ingredients and variables, which may interact with one another to produce unexpected.
The most commonly used member of biguanides is Metformin. Biguanides [Metformin] is an Antihyperglycemic
and not Hypoglycemic agent. It does not stimulate pancreas to secrete insulin and does not cause hypoglycemia (as a
side effect) even in large doses. Also it has no effect on secretion of Glucagon or Somatostatin
Objectives:
The objective of the study was to develop a formulation for Glimepiride tablets by using lactose monohydrate &
starch as diluent, binder such as povidone K-30, and disintegrants such as sodium starch glycolate, croscarmellose
sodium; and then evaluating Glimepiride tablets. Preformulation testing is the first step in rational development of
dosage form of drug substance. In this study, characterization of API is most important study. Hygroscopicity,
solubility study, bulk density, tapped density, compressibility& particle size analysis by Malvern analyzer of API
done for characterization of API3. For development of Glimepiride formulation know about the details of innovator
product so done the characterization of innovator product. For selection of excipients, drug compatibility study was
required. The storage condition used to examine compatibility can vary widely in terms of temperature and
humidity.

2. Materials and Methods


Wet granulation:
Wet granules are formed by binding the powder together with the adhesion, instead of by compaction. This process
employs a slurry containing a binder , which is usually added to mixture of powder but binder is added to powder
mix & liquid is added itself. Liquid will form bridges are developed between particles. During addition of liquid
uniform mixing is done. During wet granulation chopper is kept on for kneading so that the lumps of the granules
that form will break soon4. After this it will keep for drying in dryer and loss on drying is checked .LOD is kept in
certain limits. This process is required in all wet granulation procedures to remove the solvent that was required in
forming the agglomerate & to reduce the moisture content to an optimum level.
Manufacturing steps[5]:
Dispensing: Dispense and check the weight along with the analytical report nos. of all the ingredients as per the
material requisition note.
Sifting: Geometrically mix calculated quantity of Glimepiride USP* and approximately 5 Kg of Lactose IP * from
calculated qty in polybag and sift through 60# using vibratory sifter and collect in a suitable container. Sift
remaining quantity of Lactose IP through 60# using vibratory sifter. Mix the above materials and resift through 60#
and collect in a suitable container. Sift 7.560 Kg of Starch IP through 60# using vibratory sifter and collect in a
suitable container. Sift separately 1.050 Kg of Sodium starch glycollate IP through 40# on a vibratory sifter and
collect in a suitable container.
Dry Mixing: Load the materials into a clean dry Rapid Mixer Granulator and Mix for 5 minutes with Impeller speed
slow & chopper off. Record the time of mixing in the BMR & send the sample to QC for Analysis.
Binder Solution Preparation: Add 1.050 Kg of Povidone K-30 Ph.Eur. in to 2.625 Kg of Purified water
USP/Ph.Eur/BP/IP/IH under constant stirring to get a clear solution.
Granulation:
Add binder solution to the powder mix in Rapid mixer granulator within 2 to 3 minutes with impeller at slow speed
and chopper off. Knead it for 2-3 minute with impeller and chopper at fast speed. Add extra quantity of Purified
water if required. Record the ampere load, additional quantity of Purified Water USP/Ph. Eur/ BP/IP/IH and time of
mixing in the BMR.
Unload wet mass into the bowl of FBD through co mill fitted with 0.8 mm SS Screen.
Drying: Dry the wet mass in Fluidized Bed dryer at inlet temperature 65 °C ± 5 °C till to get LOD between 1.5 to
3.0 % w/w at 105 °C.
Sizing: Pass the dried granules through oscillating granulator fitted with 0.8 mm perforated SS screen and collect it
in clean dry Conta blender.
Weigh and record the weight of the granules in BMR.
Blending & Lubrication [6]:
Blend the granules with sifted material for 7 minutes in Conta blender. Record the time of blending in the BMR.
Add the sifted materials in to the blended material and lubricate for 3 minutes. Record the time of lubrication in the

527 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

BMR. Send the sample of lubricated granules along with sample test request slip to QC for analysis. Weigh &
record the weight of lubricated granules in the BMR.
Compression7: After getting QC/QA approval, compress the lubricated granules into tablets on Rotary Tablet
Compression Machine using 8/32” (6.35mm) round shape standard concave punches with plain upper punches, plain
lower punches and suitable dies. Check Description, weight of 20 tablets, Average weight, Uniformity of weight,
Hardness, Thickness, disintegration time & Friability and record in the BMR. Send the sample of compressed tablet
along with Sample test request slip to QC. Weigh and record the weight of compressed tablets in BMR.
Stability Studies [8]:
From the prepared formulation and development of glimepride which showed appropriate balance between the
buoyancy and the percentage release was selected for stability studies. The prepared formulation were placed in
borosilicate screw capped glass containers and stored at three different temperature (27±2 °C, 65% RH), Oven
temperature (40±2°C, 65% RH) and in freezing temperature (5 – 8°C, 65% RH) in stability chamber for a period of
90 days. The samples were evaluated for cumulative percentage drug release at regular intervals of two week.

Table 1. Composition of Glimepiride tablet


Qty / tablet % In tablet
Sr. No Ingredients
(mg) (w/w)
1 Glimepiride 4.00 4.44
2 DCP 61.00 67.24
3 Starch 20.00 22.22
4 Sodium Starch Glycolate 3.00 3.33
5 Magnesium stearate 1.00 1.11
6 Colloidal silicon dioxide 1.00 1.11
Total Weight in mg 90.0 99.98

Table 2. Comparative physical parameters


Punch Used Weight Disintegration Hardness Friability Thickness
Variation time
Lower Higher (N)
8/32”round 12.0 9.0 10 – 20 seconds 40-60 N 1.25% 2.9 – 3.1

Table 3. Characterization of blend


Bulk Density Tapped Density Carr’s Index Hausner’s Angle of
(g/ml) (g/ml) (%) Ratio Repose

0.498 0.682 26.97 1.369 36.57°

Table 4. Comparative dissolution profiles


Glimepiride Dissolution profile in 500 ml 0.2 % SLS In pH 3.0 buffer.
Tablet @ 40 rpm
5 min 10 min 15 min 30 min 45 min 60min
Batch No: 001 91.3 95.6 101.0 101.2 100.8 100.4

Table 5. Composition of Glimepiride tablet


Qty / tablet % In tablet
S. No Ingredients
(Mg) (W/w)
1 Glimepiride 4.00 4.44
2 Mannitol 61.00 67.24
3 Starch 20.00 22.22
4 Sodium Starch Glycolate 3.00 3.33
5 Magnesium stearate 1.00 1.11
6 Colloidal silicon dioxide 1.00 1.11
Total Weight in mg 90.0 99.98

528 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Table 6. Characterization of blend

Bulk Density Tapped Density Carr’s Index (%) Hausner’s Angle of


(g/ml) (g/ml) Ratio Repose

0.468 0.652 28 1.39 39.26°

Table 7. Comparative physical parameters


Punch Weight Disintegration Hardness Friability Thickness
Used Variation time
Lower Higher (N)
8/32”
2.0 3.0 30- 50 seconds 40-60 N 0.65% 2.9 – 3.1
round

Table 8. Comparative dissolution profiles


Glimepiride Dissolution profile in 500 ml 0.2 % SLS In pH 3.0 buffer.
Tablet @ 40 rpm
5 min 10 min 15 min 30 min 45 min 60min
Batch No: 002 81.3 91.6 95.2 98.2 100.2 101.4

Selection of dissolution medium:


The primary product development of Glimepiride Tablets 4 mg has been initiated in bench scale to match the
dissolution profile with the innovator product in the following medias [9]. The two media that are selected for the
comparison based on the solubility and pharmacokinetics of the drug are:
a) PH3.0 Buffer with 0.2% SLS
b) Water + 0.5 % SLS
Dissolution testing has emerged in the pharmaceutical field as a very important tool to characterize drug product
performance. Dissolution of drug depends upon solubility of the drug in the medium. Solubility is the one of the key
determinants for oral bioavailability of drug. Development of dissolution medium for poorly water soluble drug is a
critical issue. Glimepiride is a anti-diabetic drug having poor water solubility.

Table 9. Comparative dissolution profiles of Amaryl 4 mg tablets in different media


Amaryl Tablet 4 mg % drug dissolved
B.No. 303154 5 min 10 min 15min 20 min 30 min 45 min 60 min
5 % IPA in water 50.0 62.9 65.2 64.2 67.1 69.8 69.8
0.2 % SLS in pH 3.0 43.4 64.7 72.3 77.8 81.5 84.6 87.4
0.5 % twin in water 24.0 32.4 35.7 35.9 37.5 37.7 39.4
0.1% SLS in pH 3.0 42.9 47.1 49.8 50.3 52.8 55.2 56.7
0.2 twin 80 in 6.8 pH 29.3 38.5 40.2 42.6 44.7 41.3 44.5
0.3 % SLS in pH 3.0 68.1 81.1 85.8 90.4 92.1 95.0 96.1
0.1 % twin 80in pH 7.4 68.4 72.0 90.3 92.6 95.4 95.5 96.5
0.1% twin 80 in pH 6.8 23.5 28.0 30.8 32.9 32.7 34.3 34.0
0.05% SLS in water 36.8 41.5 42.3 40.8 44.8 48.2 50.2
1% SLS in water 46.2 50.9 56.2 57.7 57.9 60.4 61.1
0.5 % SLS in water 85.4 99.0 99.9 100.5 100.6 101.6 101.9
0.1% SLS in pH 4.5 39.3 47.3 51.5 52.6 53.7 56.4 58.5

The composition of the medium was determined on the basis of solubility data of the drug in different Medias.
Saturation solubility of drug was found to be more in pH 3. The effects of surfactant (Sodium lauryl sulphate and
Tween 80) in the different concentrations were studied of solubility of drug in pH 3. The study reveled that pH 3
with 0.2% SLS showed higher solubility, hence, was considered to be a suitable dissolution medium. Dissolution
can be used as a test to reflect the bioavailability of the product in humans. Dissolution testing has emerged in the
pharmaceutical field as a important tool to characterize drug product performance. The composition of the medium
was determined on the basis of solubility data and dissolution profile of drug in different medias [10]. In 0.2 % SLS
in pH 3.0 buffer at 50 RPM the drug is completely released at 45 minutes. Besides, since this is an immediate
release formulation, this media is bio-relevant media because drug shall be completely released and will be available
for absorption in-vivo at this pH. Hence this media was selected for routine and QC analysis.

529 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Comparative dissolution profiles of Amaryl 4 mg Tablets


in different media

120 5 % IPA in water


100 0.2 % SLS in pH 3.0

% drug release
0.5 % twin in water
80
0.1% SLS in pH 3.0
60 0.2 twin 80 in 6.8 pH
40 0.3 % SLS in pH 3.0
0.1 % twin 80in pH 7.4
20
0.1% twin 80 in pH 6.8
0 0.05% SLS in water
0 5 10 15 20 30 45 60 1% SLS in water
min min min min min min min 0.5 % SLS in water
Time 0.1% SLS in pH 4.5

Figure 1. Comparative dissolution profiles of Amaryl 4 mg tablets in different media

Table 10. Composition of Glimepiride tablet


Qty/tablet in mg
Sr.No Raw material
B.No:003 B.No:004 B.No:005
1.0 Glimepiride 4 4 4
2.0 Lactose 58.00 56.50 55.00
3.0 Starch 20.00 20.00 20.00
4.0 PVP K 30 3.0 (3.35%) 4.5 (5.0%) 6.0 (6.65%)
5.0 Sod. Starch glycolate 3.00 3.00 3.00
6.0 Mg. Stearate 1.00 1.00 1.00
7.0 Aerosil 200 1.00 1.00 1.00
8.0 Purified Water q.s q.s q.s
Tablet Weight 90.00 90.00 90.00

Table 11. Comparative physical parameters


B. No. Thickness (mm) Hardness (N) D.T.
003 2.82-2.98 60 – 80 2.45
004 3.85-3.10 70 – 100 8.30
005 3.82-3.10 75 - 110 9.10

Table 12. Comparative dissolution profiles


Dissolution profile in 500ml pH 3.0 with 0.2 % SLS @
% of
Product B.No. 40rpm (Type II)
Binder
5 min 10 min 15 min 30 min 45 min
003 3.35 54.0 81.5 89.8 93.7 94.0
Glimepiride 4
004 5.0 30.9 55.5 78.2 98.9 98.8
Mg Tabs
005 6.65 28.1 49.6 66.4 85.3 86.9

Table 13. Composition of Glimepiride tablet


Qty/tablet in mg
Sr. No Raw material
B.No:006 B.No:007 B.No:008
1.0 Glimepiride 4 4 4
2.0 Lactose 61.60 58.90 58.90
3.0 Starch 20.00 20.00 20.00
4.0 PVPK 30 3.00 3.00 3.00
5.0 Sod. Starch glycolate - 2.7(3. 0%) -
6.0 Croscarmellose Sodium - - 2.7(3. 0%)
7.0 Mg. Stearate 1.00 1.00 1.00
8.0 Aerosil 200 1.00 1.00 1.00
9.0 Purified Water q.s q.s q.s
Total Weight 90.00 90.00 90.00

530 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

120

100

80

% Release
60

40
3

20 5

4
0
0 5 1 1 3 4

Figure 2. Comparative dissolution profiles

Table 14. Comparative physical parameters


B. No. Thickness (mm) Hardness (N) D.T.(mins)
006 2.85-2.97 45 – 58 6.45
007 3.89-3.10 55 – 90 3.14
008 3.80-3.10 60 – 85 3.58

Table 15. Comparative dissolution profiles


Dissolution profile in 500ml pH 3.0 with 0.2 % SLS @
Product B.No. 40rpm (Type II)
5 min 10 min 15 min 30 min 45 min
006 28.4 42.5 67.0 83.90 90.70
Glimepiride
007 48.9 75.5 83.10 88.90 89.60
4 Mg Tabs
008 45.1 69.6 75.60 82.60 84.10

100

90

80

70
% Release

60

50

40

30 6
20 7

10 8

0
0 5 min 10 min 15 min 30 min 45 min

Figure 3. Comparative dissolution profiles

531 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Table 16. Composition of Glimepiride tablet


Raw Qty/tablet in mg
Sr.No
material B.No:009 B.No:010 B.No:011 B.No:012 B.No:013 B.No:014
1.0 Glimepiride 4 4 4 4 4 4
2.0 Lactose 61.00 57.40 56.50 55.60 54.70 53.80
3.0 Starch 20.00 20.00 20.00 20.00 20.00 20.00
4.0 PVP K 30 3.00 3.00 3.00 3.00 3.00 3.00
Sod. Starch -- 3.6(4.0%) 4.5(5.0%) 5.4(6.0%) 6.3(7.0%) 7.2(8.0%)
5.0
glycolate
6.0 Mg. Stearate 1.00 1.00 1.00 1.00 1.00 1.00
7.0 Aerosil 200 1.00 1.00 1.00 1.00 1.00 1.00
Purified
8.0 q.s q.s q.s q.s q.s q.s
Water

Table 17. Comparative physical parameters


% of Thickness Hardness Friability
B. No. D.T.
Disintegrant (mm) (N) (%)
009 0 2.81-3.07 50 - 58 0.14 6 min 58 sec
010 4 2.79-3.00 76-86 0.24 2 min 37 sec
011 5 2.90-3.19 39-53 0.23 2 min 25 sec
012 6 3.15-3.27 32– 43 0.32 1 min 20sec
013 7 3.92-3.11 40-52 0.35 1 min 15 sec
014 8 3.82-3.14 45-64 0.30 65 seconds

Table 18. Comparative dissolution profiles


Dissolution profile in 500ml pH 3.0 with 0.2 % SLS @
Product B.No. 40rpm (Type II)
5 min 10 min 15 min 30 min 45 min
009 28.4 42.5 67.0 83.90 90.70
010 49.7 76.5 83.60 91.0 91.80
Glimepiride4 011 52.6 82.5 84.30 88.20 88.20
Mg Tabs 012 79.4 83.5 89.10 93.90 97.30
013 71.5 78.9 79.40 85.50 86.50
014 64.8 76.4 80.80 86.70 87.10

120

100

80
% Release

9
60 10
11
40 12
13
20 14

0
0 5 min 10 min 15 min 30 min 45 min

Figure 4. Comparative dissolution profiles

532 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Table 19. Composition of Glimepiride tablet


Qty per tablet in mg
Sr.No. Ingredients
015 016 017 018**
1. Glimepiride 4 4 4 4
2. Lactose 59.00 58.50 58.00 57.00
3. Starch 10.00 20.00 30.00 30.00
4. PVPK 30 3.00 3.00 3.00 3.00
5. Sod. Starch glycolate 3.00 3.00 3.00 3.00
7. Purified water -
10. Silica, Colloidal Anhydrous 1.00 1.00 1.00 1.00
11. Magnesium Stearate 1.00 1.00 1.00 1.00
Tablet Weight 230.00 230.00 230.00 230.00
018** in this batch 15 mg intragranular & 15 mg extragranular of starch.

Table 20. Comparative physical parameters


B. No. Thickness (mm) Hardness (N) D.T.(mins) Angle of repose
015 2.88 – 3.05 34 – 97 3.41 23.25
016 2.81 – 3.10 50 – 82 2.58 25.48
017 2.80 – 3.20 54 – 84 1.42 28.24
018** 2.90 – 3.10 56 – 89 2.18 29.45

Table 21. Comparative dissolution profiles


Dissolution profile in 500ml pH 3.0 with 0.2 % SLS @
Product B.No. 40rpm (Type II)
5 min 10 min 15 min 30 min 45 min
015 38.9 78.4 84.9 89.5 97.4
Glimepiride4m 016 42.9 74.8 85.9 92.8 98.4
g tablets 017 45.9 72.9 87.4 93.4 102.5
018** 39.8 70.4 82.6 90.7 101.7

120

100

80
% Release

15
60
16

40
17

18**
20

0
0 5 min 10 min 15 min 30 min 45 min

Figure 5. Comparative dissolution profiles

533 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Table 22. Composition of Glimepiride tablet


Qty per tablet in mg
Sr.No. Ingredients 019 020 021 022
0% 0.55% 1.10% 2.20%
1. Glimepiride 4 4 4 4
2. Lactose 59.00 58.50 58.00 57.00
3. Starch 20.00 20.00 20.00 20.00
4. PVP K 30 3.00 3.00 3.00 3.00
5. Sod. Starch Glycolate 3.00 3.00 3.00 3.00
6. Purified water -----
7. Silica, Colloidal Anhydrous 0 0.50 1.00 2.00
8. Magnesium Stearate 1.00 1.00 1.00 1.00
Tablet Weight 230.00 230.00 230.00 230.00

Table 23. Effect of glidant on physical parameters


Batch % of Hardness Friability Angle of
Glidant Repose D.T.
No. (N) (%)
019 0 47 - 64 0.16 23.05o 2 min 30 sec
o
020 0.55 65-81 0.14 22.32 2 min 20 sec
021 1.10 74 - 93 0.18 20.20 o 2 min 20 sec
022 2.20 68-89 0.24 20.67 o 2 min

Table 24. Composition of Glimepiride tablet


Sr. Qty/tablet in mg
Raw material
No. B.No:023 B.No:024 B.No:025
1.0 Glimepiride 4 4 4
2.0 Lactose 61.60 58.90 58.90
3.0 Starch 20.00 20.00 20.00
4.0 PVP K 30 3.00 3.00 3.00

5.0 Sod. Starch glycolate 3.00 3.00 3.00


7.0 Mg. Stearate 1.00(1.11%) 2.00(2.20%) 3.00(3.30%)
8.0 Aerosil 200 1.00 1.00 1.00
9.0 Purified Water q.s q.s q.s
Tablet Weight 90.00 90.00 90.00

Table 25. Comparative physical parameters


% of Thickness Hardness Friability
Batch No. Lubricant D.T.
(mm) (N) (%)
023 1.11 2.85 – 3.26 33 - 76 0.22 2 min 36 sec
024 2.20 2.89 – 3.27 46 - 73 0.18 2 min 50 sec
025 3.30 3.10 – 3.30 33 - 60 0.17 3 min 20 sec

Table 26 Effect of different concentrations of lubricant on dissolution


Dissolution profile in 500ml pH 3.0 with 0.2 % SLS @
Product B.No. 40rpm (Type II)
5 min 10 min 15 min 30 min 45 min
023 58.9 78.4 89.10 93.90 97.30
Glimepiride 4
024 32.9 54.8 69.80 78.70 85.20
Mg Tabs
025 35.9 52.9 75.20 83.30 86.60

534 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

120

100

80

% Release
60

23
40
24

20 25

0
0 5 min 10 min 15 min 30 min 45 min

Figure 6. Comparative dissolution profiles

3. Results and Discussion


Flow properties of the powder can be judged from the angle of repose. The powder flow depends on 3 general areas:
1) The physical properties of the particle (E.g: shape, size, compressibility) 2)the bulks powder properties (e.g. size,
distribution compaction): and 3)the processing environment (e.g. storage, humidity) The angle of response of batch
no 1&2 >35° so, very poor flow of the powder blend so, during compression weight variation observed11. Poor CI
(%) & poor HR so, hardness was not achieved of higher side & friability was 1.25%. DT of tablets was 10-20 sec. In
5 min 91.3% of drug release was observed. These in process parameters & dissolution were not match with the
innovator product. In batch no.2 replace the DCP by the Mannitol. Mannitol has direct compressible property. But
the flow property of the powder blend was very poor. During compression sticking was observed. In 5 min 81.3% of
drug released. So, to formulate the tablets of Glimperide using wet granulation. The tablet formulation were
prepared by compression method using punch size 8/32” round shaped, upper lower side flat using Cadmach 16-
station compression machine. A rapid mixer granulator was used for wet granulation in the study. Suitable selection
of binder concentration was done[12].
Manufacturing Process:
Table 27. Product Composition
Qty / Tab. (In mg) Qty./ batch
Ingredient Specification# Function
Sr.No Theoretical Actual (In Kg)
01 Glimepiride USP Active 3.000 3.000 1.050
02 Lactose * IP Diluent 59.000 59.000 20.650
03 Starch** IP Diluent 20.000 21.600 7.560
04 Povidone (K-30) Ph.Eur Binder 3.000 3.000 1.050
05 USP /Ph. Eur/
PurifiedWater @ Vehicle q.s. q.s. 2.625
BP/IP/IH
06 Sodium starch
IP Disintegrant 3.000 3.000 1.050
glycollate
07 Magnesium
IP Lubricant 1.000 1.000 0.350
Stearate
08 Colloidal Silicon
IP Glidant 1.000 1.000 0.350
dioxide(Aerosil)
Total weight 90.00 90.00 31.500

*Potency of Glimepiride USP should be adjusted to 100 % and simultaneously quantity of Lactose IP should be
adjusted as per calculation .
**8 % extra starch added to compensate moisture loss on drying.
Formula Calculation:
Calculation for Glimepiride USP:
(Depending on how the assay of Glimepiride USP is reported, use the appropriate factor) Assay on anhydrous or
such basis Basis Qty of Glimepiride USP

535 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Required per batch of = 1.050x 100 x 100 or 1.050 x 100


B
350,000 Tablets Q (Kg) A x (100 – W)

Where A = Assay of Glimepiride USP on anhydrous basis in % w/w


B= Assay of Glimepiride USP on as such basis in % w/w
W= Water content in % w/w

Facility required during manufacturing process

Table 28. Facility required


Area Temperature Relative Humidity
Sifting NMT 27 °C NMT 60 %
Granulation & Drying NMT 27 °C NMT 60 %
Blending NMT 27 °C NMT 60 %
Compression NMT 27 °C NMT 60 %

Table 29. In process test parameters


STAGE TEST ACCEPTANCE CRITERIA
Dry mixing Blend Uniformity NLT 90.0 % and NMT 110.0%
Analysis (RSD: NMT 5.0 %).
Drying Loss on Drying at 105°C 1.5 % to 3.0%w/w.
Description White c olored free flowing granular powder.

Lubrication Assay NLT 95.0% and NMT 105.0% of labeled amount.


Blend Uniformity NLT 9NLT 90.0% and NMT 110.0% of Labeled
Analysis amount and RSD is not more than 5.0 %.
Description White coloured round shaped biconvex uncoated tablets plain on
both sides.
Theoretical weight of 90.0 mg
individual tablets
Average weight 90.0 mg ± 5 % ( 85.5 mg to 94.5 mg)
Weight of 20 Tablets 1.80 g ± 3 % (1.746 mg to 1.854 mg)
Compression Uniformity of weight Average weight  7.5 % (83.25 mg to 96.75 mg)
Hardness 40 to 100 N (4 to 10 Kg/cm2 )
Thickness 2.6 ±0.3 mm
Disintegration time NMT 15 min.
Friability NMT 1.0 % w/w
Uniformity of content Out of 10 units, not more than one of the individual values should
be outside the limits of 85.0 to 115.0% of the average value and
none is outside the limits of 75.0 to 125.0% of the average value.

Sampling protocol:

Table 30. Details of operating parameter of horizontal oscillating granulator(Sizing)


Sr.No Time taken Parameter Operating condition

1 10 min Screen 0.8 mm

Table 31. Details of the physical characteristics of blend after sizing


Sr.No Parameter Result
1 Description White colored free flowing granules

536 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Table 32. Sampling to be done at various stages


Parameter to be Sampling
Sample container Sample Quantity
Stage monitored Interval
Glass vials, rubber 10 samples in triplicate
Blend Uniformity
Dry mixing After mixing closures & (Each approximately
Analysis
aluminum seals 82.0 to 246.0 mg)
Glass vials, rubber 10 samples in triplicate
Blend Uniformity
closures & (Each approximately
Analysis
aluminum seals 90.0 to 270.0 mg)
Description, Bulk
Lubrication Density, Tapped After Lubrication
Density, Sieve Approximately 200 mg
LDPE Bag
Analysis, LOD (Composite)
(For reference only)
Assay
Initial, Middle & Approximately
Content uniformity LDPE Bag
End 100 Tablets
Compression
At low, Optimum Approximately
Dissolution. LDPE Bag
& High Hardness 100 Tablets

Table 33. Process steps


Process Stage Process Control Step Expected response Observed Response Remarks
Granulation: 1.Binder Addition Time 2-3 minutes 3 minutes 100 gm extra
2.Kneading Time 2-3 minutes 2 minutes water was used
Drying LOD@105°C 1.5-3.0%w/w 2.85 % w/w ------
Lubrication BUA 90.0 %to 110.0% S1:96.9
and RSD not more S2:94.5
than 5.0%. S3:97.0
S4:97.4
S5:95.8
S6:96.8 ------
S7:97.5
S8:95.5
S9:94.4
S10:97.0
Mean: 96.3, RSD: 1.2
Compression Average weight 90.0 mg ± 5 %Av. Passes
Uniformity of weight Av.weight ± 7.5% Passes
Hardness 50 to 100 N 43-69 N(opt.) Hardness limit
Friability NMT 1% 0.25% to be changed
Thickness 2.60 mm ± 0.2 mm 2.67-2.73mm
DT NMT 30 min. 1 min 30 sec

Table 34. Details of the monitoring and results of uniformity blend


Dry mix Blending Time:5 min Blending Time:7 min RFC
S1:96.8 S1:97.4 S1:96.9
S2:96.5 S2:93.2 S2:94.5
S3:94.0 S3:91.9 S3:97.0
S4:96.2 S4:96.0 S4:97.4
S1:96.2 S5:97.3 S5:97.3 S5:95.8
S2:96.1 S6:91.8 S6:95.2 S6:96.8
S3:95.5 S7:96.5 S7:97.9 S7:97.5
S4:95.7 S8:93.0 S8:98.0 S8:95.5
S5:96.8 S9:93.0 S9:94.1 S9:94.4
S6:96.8 S10:96.1 S10:98.1 S10:97.0
Mean 96.2 95.1 95.9 96.3
RSD 0.58 2.06 2.31 1.2

537 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Blending without Magnesium stearate: 7 min


Lubrication with Magnesium stearate: 3 min

Blend Uniformity Analysis during different stage:


A) Stage: Dry mixing

Table 35. Blend Uniformity Analysis:


Dry mixing time Min (%) Max (%) Average (%) RSD
3 min 98.66 101.41 100.22 0.8
4 min 99.08 101.05 100.17 0.66
5 min 99.38 101.43 100.19 0.67

B) Stage: Lubrication
1. Composite assay – 100.95%
2. Blend Uniformity Analysis:
Table.36
Lubrication time Min (%) Max (%) Average (%) RSD
1 min 99.56 101.58 100.36 0.75
2 min 99.19 102.09 100.73 0.85
3 min 99.09 102.74 100.47 1.08

C) Stage: Precompression
1. Assay- 98.37%
2. Dissolution:
Table.37
Hardness Min (%) Max (%) Average (%) RSD
Low (4-5 Kg) 89.84 95.59 92.47 2.37
Optimum (6-7 Kg) 91.73 95.03 93.46 1.24
High (9-10 Kg) 89.37 95.12 92.26 2.50

Table 38. Details of the physical characteristics of final blend


Sr.no. Parameter Results
1 Description White colored granular powder
2 Assay 96.3 %
3 Untapped density 0.63 g/cc
4 Tapped density 0.81 g/cc
5 Particle size distribution/Sieve % Cumulative
analysis:
6 Retention over #40 24.93
7 Retention over #60 30.58
8 Retention over #80 32.51
9 Retention over #100 35.80
10 Retention over #200 52.93
11 Pass through # 200 99.98

Compression details:
Details of the monitoring & observations during compression which was operated at optimum speed:
Type of M/C used: 20 station machine.
Type of tooling: B tooling
Punch size: 8/32” round SC punches with plain on the both sides.
No of punches: 4 sets
Weight of 20 tablets: 1.800 gm
Machine Optimum RPM: 25 RPM
Average wt: 88.65 mg
Hardness: 43-69 N
Thickness: 2.67-2.73 mm
538 | International Journal of Current Trends in Pharmaceutical Research
Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

DT: 2 min 30 sec


Friability: 0.25 % w/w

Table 39. Content uniformity analysis of tablets:


Sr. No Initial (%) Middle (%) End (%)
1 99.20 97.79 99.75
2 99.06 98.00 97.72
3 98.10 97.24 97.06
4 96.82 97.49 97.43
5 97.98 97.69 97.74
6 98.49 99.95 97.09
7 98.72 98.24 95.94
8 99.16 100.12 97.01
9 99.02 97.08 97.06
10 98.60 97.61 99.00
Min 96.82 97.08 95.94
Max 99.20 100.12 99.75
Avg 98.52 98.12 97.58
RSD 0.74 1.08 1.11

Analytical method:
Analyte : Glimepiride
Bio analytical technique : LC-MS/MS
Instrument : LC-MS/MS (TSQ Quantum Finnigan)
Internal standard : Imipramine hydrochloride
Type of extraction : Solid phase extraction
Number of samples : A series (17x6 ml) of venous blood sample were Collected over a period of 24 hrs in each
period.
Sampling time : Predose ,0.5,1.0,1.5,2.0,2.5,3.0,3.5, 4.0,5.0, 6.0,8.0,10.0,12.0,15.0,18.0 & 24.0hrs.
Washout period : 10days

Table 40. Pharmacokinetic parameters.


Parameters Test formulation Reference formulation
Mean ±SD CV (%) Mean ±SD CV (%)
AUC0-t 2340.419±2474.57 105.73 2343.414±2411.36 102.9
(ng.h/ml)
AUC0-inf 3489.873±5722.7 164.84 3223.938±4865.01 150.9
(ng.h/ml)
Cmax (ng/ml) 205.063±124.59 60.76 228.193±136.12 59.65

Residual Area (%) 10.124±14.95 147.70 08.857±12.87 145.36


Tmax (h) 6.00±5.00 ---- 6.0 ± 4.5 ----

Kel h-1 0.149±0.05 35.46 0.155±0.05 32.68

T1/2 el (h) 06.310±5.84 92.58 5.662±4.35 76.91

MRT0-t (h) 9.033±1.98 21.90 8.891±2.27 25.49

MRT0-inf (h) 12.408±8.45 67.77 11.357±6.48 57.02

539 | International Journal of Current Trends in Pharmaceutical Research


Vennela K.G et al, IJCTPR, 2014, Vol.2(4): 525-540

Stability study:
Table 41. Stability data of Glimepiride tablet
Condition/ Physical Assay D.T. Dissolution
Period Observation (%) (45 min)

Specification White colored, round shaped


uncoated tablet plain on the 90-110 NMT 15 NLT 70
both side. min
Initial White colored round shaped 98.6 1-33 95.1
tablet
60°C 1 M Same as initial 98.5 3-04 92.6
400C 75%RH 1 M Same as initial 100.74 3-18 92.2
400C 75%RH 2 M Same as initial 99.62 - -

4. Conclusion
The present study seems to be routine and of course Glimepiride tablets of various manufacturers are available in the
Indian Market. But one view on excipients are highly illustrative, which should not be restricted to the knowledge of
use in Glimepiride tablet formulations only. It is study for any a new conventional tablet dosage form in which
selection, quantity and combination of excipients play a major role. Hence present effects should be observed as a
highly fruitful study and may be helpful for new entrepreneur in establishing tablet as a dosage form.

5. Acknowledgement
The authors are thankful to Hetero drugs Ltd, Hyderabad for providing the all facilities for carried out this research
work

6. References
1. Food and Drug Administration, Guidance for Industry: Waiver of In Vivo Bioavailability and
Bioequivalence Studies for Immediate Release Solid Oral Dosage Forms Based on a Biopharmaceutics
Classification System (FDA, Rockville,MD, August 2000).
2. David Fortunato,”Dissolution method development for immediate release oral solid dosage form”
Dissolution technology, August 2005.
3. Banker GS, Anderson NR. Tablets. In: Lachman L, Lieberman HA, Kanig JL, eds. The Theory and
Practice of Industrial Pharmacy. 3rd ed. Philadelphia, PA: Lea & Febiger; 1986: 293Y345.
4. Gohel MC, Jogani PD. A review of co-processed directly compressible excipients. J Pharm Pharm Sci.
2005, 8: 76Y93.
5. The United States Pharmacopoeia. The National Formulary, USP 22, NF 17, United States Pharmacopoeial
Convention, Inc., Rockville, M.D., 1990, 1528.
6. Amidon, G. E.; Augsburger, L. L.; "Physical test methods for powder flow characterization of
pharmaceutical materials: a review of methods"Pharmacopeial Forum, 1999, 25: 8298-8308
7. Kibbe, A. H. "Handbook of pharmaceutical excipients" 3rd edition, American Pharmaceutical Association
and Pharmaceutical Press, Washington London, 2000, 102-106.
8. David e. storey, the role of dissolution testing in the design of immediate release dosage forms, Drug
Information Journal, 1996, 30: 1039–1044,
9. FIP guidelines for dissolution testing of solid oral products [Duplicate publication of FIP guidelines for
dissolution testing of solid oral products. Pharm Ind. 1997, 59: 760-766
10. Qureshi SA, McGilveray IJ. Typical variability in drug dissolution testing: study with USP and FDA
calibrator tablets and a marketed drug (glibenclamide) product. Eur J Pharm Sci., 1999, 7: 249-258.
11. Food and Drug Administration, Guidance for Industry: Dissolution Testing of Immediate Release Solid
Oral Dosage Forms (FDA, Rockville, MD, August 1997).
12. General Chapters , “Dissolution” and & “Drug Release,” in The United States Pharmacopeia 27–National
Formulary 22 (United States Pharmacopeial Convention, Inc., Rockville, MD, 2004), pp.2303-2304 and
2305–2312

540 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

ISSN: 2321-3760
Review Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 541-550
www.pharmaresearchlibrary.com/ijctpr

A Brief Study on Carica Papaya- A Review


Asha Roshan*1, Navneet Kumar Verma2, Anubha Gupta2
1
R.K. College of Pharmacy, Azamgarh, U.P, India
2
Department of Pharmacy, Rameshwaram Institute of Technology and Management, Lucknow, U.P, India
Received: 4 May 2014, Accepted: 18 June 2014, Published Online: 15 July 2014

Abstract
Carica papaya belongs to caricaceae family and it is commonly known as ‘papaya’. Carica papaya is used
in ayurvedic medicines from very long time. It is used as anti-inflammatory, antioxidant, diuretic,
antibacterial, abortifacient, vermifuge, hypoglycemic, antifungal activity, antihelmenthic and
immunomodulatory etc. Scientific evidences suggest their versatile biological function that supports its
traditional use in different diseases. Phytochemical studies shows that plant carica papaya contains mainly
alkaloids carpaine, pseudocarpaine, tannins, flavnoids, carcin, gamma terpine, glycoside carposides, sugars
etc. The plant has effective pharmacological activity such as anti-inflammatory, antioxidant, diuretic,
antibacterial, abortifacient, hypoglycemic, antifungal, antihelmenthic and immunomodulatory,
hepatoprotective and anticonvulsant activity.
Keywords: Carica papaya, caricaceae, abortifacient, pharmacological activity.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .541
2. Cultivation and Description . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .542
3. Chemical constituents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 543
4. Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .548
5. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . 548

*Corresponding author
Asha Roshan
R.K. College of Pharmacy,
Azamgarh, U.P, India
Manuscript ID: IJCTPR2101
PAPER-QR CODE
Copyright © 2013, IJCTPR All Rights Reserved

1. Introduction
Ayurveda, the Indian system of medicine, is attainment superior attention and popularity in many parts of the world.
The disease protective and health promotive approach of Ayurveda, which takes into consideration the entire body,
mind and spirit while dealing with the maintenance of health promotions, now enjoys increasing acceptability [1].
Ayurveda had developed certain dietary and therapeutic measures to delay ageing and rejuvenating whole functional
dynamics of the body organs. This revitalization and rejuvenation is known as the ‘Rasayana chikitsa’ [2]. Herbs are
staging a comeback and herbal ‘renaissance’ is happening all over the globe. The herbal products today symbolise
safety in contrast to the synthetics that are regarded as unsafe to human and environment. Although herbs had been
priced for their medicinal, flavoring and aromatic qualities for centuries, the synthetic products of the modern age

541 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

surpassed their importance, for a while. However, the blind dependence on synthetics is over and people are
returning to the naturals with hope of safety and security [3].Ancient pharmacopoeias from different regions of the
world have recorded numerous herbal medicines purported to have psychotropic potential. These offer a vast
repertory of potential substances that can be developed into modern psychiatric pharmaceuticals. Indeed, nearly 25%
of today’s conventional drugs originated directly or indirectly from plants; many valuable psychoactive drugs, such
as yohimbine, ephedrine, tubocurarine, and galanthamine, were discovered through the study of indigenous remedies
[4].

2. Cultivation and Description


Formerly from southern Mexico (particularly Chiapas and Veracruz), Central America, and northern South America,
the papaya is now cultivated in most tropical countries. In cultivation, it grows rapidly, fruiting within three years. It
is, however, highly frost-sensitive, limiting its production to tropical climates. Temperatures below 29° Fahrenheit
are greatly harmful if not fatal. In Florida, growth is generally limited to southern parts of the state. It also prefers
sandy, well-drained soil as standing water will kill the plant within 24 hours [5]. The papaya has a moderately
complicated means of reproduction. The plants are male, hermaphrodite, or female [6].The male trees are
uncommon, but sometimes occur when homeowners collect their own seeds. Hermaphrodite trees are the
commercial standard, producing a pear shaped fruit. These plants are self pollinated [7].

Papaya exhibits strong apical dominance rarely branching unless the apical meristem is removed, or damaged.
Palmately-lobed leaves, usually large, are arranged spirally and clustered at the crown, although some differences in
the structure and arrangement of leaves have been reported with Malaysian cultivars [8]. Generally, papaya cultivars
are differentiated by the number of leaf main veins, the number of lobes at the leaf margins, leaf shape, stomata
type, and wax structures on the leaf surface, as well as the colour of the leaf petiole. Papaya fruits are borne by both
female and hermaphrodite trees, but their shapes differ. Fruits from female trees are round whereas fruits from
hermaphrodite trees are elongated. The fruit is a berry that can range from 5 cm in diameter and 50 g in weight to 50
cm or longer, weighing 10 kg or more [9]. Papaya fruits are covered with a smooth thin green skin that turns to
yellow or red when ripe. The flesh is succulent, varying in texture and colour ranging from yellow to orange to red.

Taxonomical Classification:

Kingdom : Plantae Varnacular Names:


Subkingdom : Tracheobionta Hindi : Papita
Division : Magnoliophyta English : Papaya
Class : Magnoliospida Eclectics : Papaw
Family : Caricaceae Brazil : Mamao
Genus : Carica L. Caribbean : Ababaï
Species : Carica papaya L. Cuba : Fruta de bomba

Morphology:
Papaya is a polygamous species and it is difficult to identify a plant whether it is male, female or hermaphrodite. It is
a tree reaching 3-10 m in height, with the habit of a palm; the fleshy stem marked by scars where leaves have fallen
off, is surmounted by a terminal panache of leaves on long petioles and with 5-7 lobes. Flowers fragrant,
trimorphous, usually unisexual-dioecious, male flowers in lax many-flowered, densely pubescent cymes at the tips
of the pendulous, fistular rachis; female flowers large, solitary or in few flowered racemes, with a short thick rachis,
fruit a large berry, varying widely in size, elongate to globose with a large central cavity, seeds black, tuberculous
and enclosed in a transparent aril. The fruit bearing trees are less than 18 month old. The leaves and unripe fruit contain milky
juice in which the protein ferment papain is present.

The papaya is a large tree-like plant, with a single stem growing with spirally arranged leaves confined to the top of the
trunk. The lower trunk is conspicuously scarred where leaves and fruit were borne. The leaves are large, 50–70
centimeters (20–28 in) diameter. The tree is usually unbranched, unless lopped. The flowers are similar in shape to the
flowers of the Plumeria, but are much smaller and wax-like. They appear on the axils of the leaves, maturing into the
large 15–45 centimeters (5.9–18 in) long, 10–30 centimeters (3.9–12 in) diameter fruit. The fruit is ripe when it feels
soft (like a ripe avocado or a bit softer) and its skin has attained amber to orange hue. The melon-like fruit varies in
size and shape, and hangs from short, thick peduncles at the leaf axil. Its flowers are mostly dioecious and resemble
each other until they start to develop sexual organs. The species is polygamous and can be classified into three sex
types: male staminate, hermaphroditic (bisexual) and female pistillate. In addition, some plants can produce more
than one kind of flowers [10].

542 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

Figure 1. Papaya plant

The pollination mechanism of the plant is not very well known but researchers 'Baker' and `Bawa' suggested that
"pollination is performed by mimicry of the pistillate flowers to the staminate nectar-producing flowers." Another
theory is that oxyalate packages in the anthers of the papaya play a role in pollination as an enrichment of the nectar.
Whatever the case, we do know that the fruit is of great economic importance to tropical America where it is widely
grown for its luscious fruit. The fruit which is orange-yellow when ripen, is a popular breakfast staple that is also
used in jellies, preserves, fruit juices and as a beverage in certain Latin countries. In addition, the leaves and root of
the plant are also used in a variety of dishes. The bark can also be used for rope making and the leaves as a soap
substitute, is an excellent stain remover. Finally, in Java, even the flowers are eaten. [10]

3. Chemical constituents
Fruits:
Protein, fat, fibre, carbohydrates, minerals: calcium, phosphorous, iron, vitamin C, thiamine, riboflavin, niacin, and
carotene, amino acids, citric and malic acids (green fruits), volatile compounds: linalool, benzyl isothiocyanate, cis
and trans 2, 6-dimethyl-3,6 epoxy-7 octen-2-ol, Alkaloid, α; carpaine, benzyl-β-D glucoside, 2-phenylethyl -β-D-
glucoside, 4-hydroxy-phenyl-2 ethyl-β-D-glucoside and four isomeric malonated benzyl-β-D-glucosides.[10,11]
Juice:
N-butyric, n-hexanoic and n-octanoic acids, lipids; myristic, palmitic, stearic, linoleic, linolenic and cis-vaccenic and
oleic acids.[11]
Seed:
Fatty acids, crude protein, crude fibre, papaya oil, sinigrin, Carpaine, benzylisothiocyanate, benzyl glucosinolate,
glucotropacolin, benzylthiourea, hentriacontane, β-sitosterol, caricin and an enzyme myrosin, leaves related
alkaloids, flavonoids, saponins, tannins, cardiac glycoside, anthraquinones and cardinolodes are present.[11]

H3C CH3
H3C
H3C CH3
H

CH3 H3C H3C CH3


Figure 2. Beta- carotene

O
Figure 3. Anthraquinone

543 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

O
H H O

CH3
H
H
O
H OH
H
HO O O
H OH
H H
H H
OH H
Figure 4. Cardiac glycoside

H2C
HO
N S O
O O

O S

HO

HO OH
OH
Figure 5. Sinigrin

Root:
Carposide and enzyme myrosin.
Leaves:
Alkaloids carpain, pseudocarpain and dehydrocarpaine I and II, choline, carposide, vitamin C and E.
Bark:
β-Sitosterol, glucose, fructose, sucrose, galactose and xylitol.
Latex
Proteolytic enzymes, papain and chemopapain, glutamine cyclotransferase, chymopapains A, B and C, peptidase A
and B and lysozymes.[11]

CH3

H 3C CH3

CH3 CH3

CH3

HO
Figure 6. Avenasterol

544 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

O O

N H
H 3 C

H N
O

Figure 7. Carpaine

O
O

OH
H3C
Figure 8. Glutaric acid

O OH
O
O

OH OH
HO
Figure 9. Citric Acid

OH
O
HO O
OH
OH
OH
Figure 10. Galactouronic acid

Nutrient contents of the papaya


Papaya is a major fruit crop worldwide that is primarily consumed as fresh fruit. Papaya fruits consist mostly of
water and carbohydrate, low in calories and rich in natural vitamins and minerals, particularly in vitamins A and C,
ascorbic acid and potassium [12].

Nutritional value per 100 g (3.5 oz)


Energy 179 kJ (43 kcal)
Carbohydrates 10.82 g
Sugars 7.82 g
Dietary fiber 1.7 g
Fat 0.26 g
Protein 0.47 g
Vitamin A equiv. 47 μg (6%)

545 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

beta-carotene 274 μg (3%)


lutein and zeaxanthin 89 μg
Thiamine (vit. B1) 0.023 mg (2%)
Riboflavin (vit. B2) 0.027 mg (2%)
Niacin (vit. B3) 0.357 mg (2%)
Pantothenic acid (B5) 0.191 mg (4%)
Vitamin B6 0.038 mg (3%)
Folate (vit. B9) 38 μg (10%)
Vitamin C 62 mg (75%)
Vitamin E 0.3 mg (2%)
Vitamin K 2.6 μg (2%)
Calcium 20 mg (2%)
Iron 0.25 mg (2%)
Magnesium 21 mg (6%)
Manganese 0.04 mg (2%)
Phosphorus 10 mg (1%)
Potassium 182 mg (4%)
Sodium 8 mg (1%)
Zinc 0.08 mg (1%)
Lycopene 1828 µg
Link to USDA Database entry
Percentages are roughly approximated
using US recommendations for adults.
Source: USDA Nutrient Database
(www.wikipedia.com)

A green papaya fruit has been reported for its nutrient content, which (per 100 g) provides 26 calories, 92.1 g H2O,
1.0 g protein, 0.1 g fat, 6.2 g total carbohydrate, 0.9 g fiber and 0.6 g ash. USFDA National Nutrient database
recorded an orange-freshed papaya (per 100 g) contained 39 calories, 88.8 g H2O, 0.61 g protein, 0.14 g fat, 9.81 g
total carbohydrate, 1.8 g fiber, 0.61 g ash [13]. Oyoyede tested the chemical profile of unripe pulp of carica papaya
and reported papaya fruit was very rich in carbohydrate (42.28% starch, 15.15% sugar) but low levels of fat [14].
Papaya fruit also contains high levels of vitamin C (51.2 mg/100g), vitamin A precursors including β-carotene
(232.3 μg/100g), and β-cryptoxanthin (594.3 μg/100g), as well as magnesium (19.2-32.7 mg/100g), which has been
reported by Wall [15]. The papaya seeds contain balance-nutrients which consist of protein (24.3%), fatty oil
(25.3%) and total carbohydrate (32.5%). Although it contains significantly high level of unsaturated fatty acids,
papaya seeds seem not to be good oil seeds. In some tropical countries, papaya leaves are used as food sources,
which can be cooked by stir fry. The papaya leaves (per 100 g), were reported by Duke, contains 74 calories, 77.5 g
H2O, 7.0 g. protein, 2.0g fat, 11.3 g total carbohydrate, 1. 8 g fiber, 2.2 g ash, 344 mg Ca, 142 mg P, 0.8 mg Fe, 16
mg Na, 652 mg K, 11,565 ug β-carotene equivalent, 0.09 mg thiamine, 0.48 mg riboflavin, 2.1 mg niacin, and 140
mg ascorbic acid, as well 136 mg vitamin E[13].

Medicinal Uses of Different Parts [16, 17, 18]


Latex: It is used as Anthelmintic, relieves dyspepsia, cure diarrhoea, pain of burns and topical use, bleeding
haemorrhoids, stomachic, whooping cough.
Fruits: Ripe fruits can be used as stomachic, digestive, carminative, diuretic, dysentery and chronic diarrhea,
expectorant, sedative and tonic relieves obesity, bleeding piles, wounds of the urinary tract, ringworm and skin
546 | International Journal of Current Trends in Pharmaceutical Research
Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

disease psoriasis. Unripe fruits are used as diuretic, laxative, dried fruit reduces enlarge spleen and liver, used in
snake bite to remove poison, abortificient and anti implantation activity, anti bacterial activity.
Seeds: Carminative, emmenagogue, vermifuge, abortificient, counterirritant, as paste in ringworm disease, psoriasis,
antifertility agent in males.
Seed juice: Bleeding piles and in large liver and spleen.
Root: Abortificient, diuretic, is checking irregular bleeding from uterus and anti fungal activity, piles.
Leaves:
Young leaves used as vegetables, jaundice, urinary complains, urinary tract infection and gonorrhea, dressing
wounds, anti bacterial activity, vermifuge in colic, fever, beriberi, abortion, asthma.
Flowers: Emmengogue, jaundice, febrifuge and pectoral properties.
Stem bark: Jaundice, antifungal activity, antihelmantic activity.
General uses
Papaya can be used as a diuretic (the roots and leaves), anthelmintic (the Leave and seed) and to treat bilious
conditions (the fruit). Parts of the plant are also used to combat dyspepsia and other digestive disorders (papaya
contains a proteolytic enzyme which soothes the stomach and aides in digestion) and a liquid portion has been used
to reduce enlarged tonsils. The juice is used for warts, cancers, tumors, corns and skin defects while the root is said
to help tumors of the uterus. In Africa a root infusion is also used for syphilis and the leaf is smoked to relieve
asthma attacks. The Javanese believes that eating papaya prevents rheumatism and in Cuba the latex is used for
psoriasis, ringworm and the removal of cancerous growth.
Medicinal and Pharmacological properties
Many biologically active substances have been isolated from papaya and studied for their pharmacological action.
An antifungal chitinase has been gene cloned and characterized from papaya fruit. The chitinase is classified as class
IV chitinase based on its amino acid sequence homology with other plant chitinases. The recombinant papaya
chitinase also has antibacterial activity [19]. The purified chemopapain from commercially available spray dried
latex of the fruits has shown immunological properties [20].

The anthelmintic activity of papaya seed has been variously ascribed to carpaine (an alkaloid) and carpasemine
(later identified as benzyl thiourea) and benzylisothiocyanate [21], cysteine proteinases from papaya fruit have also
been reported [22]. Carpaine, an alkaloid with an intensively bitter taste and a strong depressant action on the heart,
has been obtained from the fruit and seed, but especially from the leaves. Biological activities of papaya are reported
with the crude extracts and different fractions from latex, seed, leaf, root, stem bark and fruit. However, crude
extracts of different parts of papaya have been used as traditional medicine for the treatment of various diseases.
However, apart from these, there are several reports on the therapeutic properties and pharmacological actions of
papaya based on modern scientific investigations. Some have been discussed below.
Antifungal
The latex of papaya and Fluconazole has synergistic action on the inhibition of Candida albicans growth. This
synergistic effect results in partial cell wall degradation (as indicated by transmission electron microscopy
observations) [23]. Latex alone is statically effective on C. albicans when added to a culture during the exponential
growth phase and approximately 60% was achieved. This fungistatic effect is the result of cell wall degradation due
to a lack of polysaccharides constituents in the outermost layers of the fungal cell wall and release of cell debris into
the culture medium [24].
Antimalarial Activity
The petroleum ether extract of the rind of raw papaya fruit exhibits significant antimalarial activity. There may be
significant commercial potential in extracting the active element from this plant, which grows abundantly
throughout the tropics and the rind of which is discarded as waste, can be exploited for antimalarial activity [25].
Antihelmenthic activity
The latex of papaya has anthelmintic efficacy against Heligmosomoides polygyrus in experimentally infected mice,
which suggests its potential role as an anthelmintic against potent intestinal nematodes of mammalian hosts [26]. It
also has anthelmintic activity against natural infection of Ascaris suum in pigs and found to be 100% effective at the
dose of 8g/kg body weight [27]. The plant extracts of papaya possesses a dose dependent significant effect on the
egg, infective larvae and adult worms of Trichostrongylus colubriformis [28]. Alcoholic extracts of papaya shows
potential in vitro anti-parasitic action, which affects eggs, infective larvae and adult Haemonchus contortus [29].
Antimicrobial activity
The seed of papaya has antimicrobial activity against Trichomonas vaginalis trophozoites. The report suggests the
use of papaya seed in urinogenital disorder like trichomoniasis with care to avoid toxicity [30]. The seed and pulp
of papaya was shown to be bacteriostatic against several enteropathogens such as Bacillus subtilis, Enterobacter
cloacae, Escherichia coli, Salmonella typhi, Staphylococcus aureus, Proteus vulgaris, Pseudomonas aeruginosa and
Klebsiella pneumoniae by the agar cup plate method [31]. Purified extracts from ripe and unripe fruits also produces
very significant antibacterial activity on S. aureus, Bacillus cereus, E. coli, P. aeruginosa and Shigella flexneri [32].
547 | International Journal of Current Trends in Pharmaceutical Research
Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

Anti-amoebic actitvity
The cold macerated aqueous extract of matured papaya seeds has shown anti-amoebic activity against Entamoeba
histolytica [33].
Male antifertility
Seed extract showed pronounced hypertrophy and hyperplasia of pituitary gonadotrophs. Whereas the male rats
treated with seed extract revealed gradual degeneration of Germ, Sertoli and Leydig cells as well as germinal
epithelium, which confirmed its antifertility activity [34]. Aqueous extract of papaya seeds, 3 weeks after
commencement of administration showed that the lumina of the somniferous tubules were more prominent and
empty in the experimental animals with no evidence of spermatids and spermatozoa [35].
Diuretic
Aqueous root extract of papaya when given orally at a dose of 10 mg/kg to rats produces significant increase in
urine output and shows similar profiles of urinary electrolyte excretion to that of Hydrochlorothiazide [36].
Female antifertility
Sharma and Mahanta have reported that the composite root extract containing papaya root extract as one of the
constituent, induces morphological changes in the endometrial surface epithelium in albino rat uterus. The
characteristic smooth regular pattern of normal epithelium appears to have changed at places by haphazardly
oriented groups of cells and loss of microvilli indicating a disorganized picture [37].Where as seeds aqueous extract
has shown abortifacient properties on female Sprague Dawley rats [38] and the petroleum ether, alcoholic and aqueous
extracts inhibits ovulation in rabbits [39].
Immunomodulatory activity
Fermented papaya preparation exerts both immunomodulatory and antioxidant activity in the macrophage cell line
RAW 264 and it is a macrophage activator, which augments nitric oxide synthesis and TNF-alpha secretion
independently of lipopolysaccharides [40]. The antioxidant cocktail derived from fermentation of unpolished rice,
papaya and sea weeds with effective microorganisms of lactic acid bacteria, yeast and photosynthetic bacteria has
shown inhibition of lipid peroxidation in vivo, a point dependent on the concentrations of bioactive flavonoids [41].
Nephroprotective activity
Maximum nephroprotection was offered by the extract at 400 mg/kg/day CPE which lasted up to 3 hours post-CCl4
exposure and these biochemical evidences were corroborated by improvements in the renal histological lesions
induced by CCl4 intoxication. Studies showed that CPE has nephroprotective effect on CCl4 renal injured rats, an
effect which could be mediated by any of the phyto components present in it via either antioxidant and/or free
radical scavenging mechanisms. Carica papaya plant has the nephroprotective activity [42].
Antisickling activity
The antisickling properties of fermented dried unripe fruit pulp of Carica papaya have been reported. The extract
got from the materials incubated for 5 days indicated as SP5, was found to have the highest antisickling properties
with 93% inhibitory and 84% reversal activities. The concentration of the day 5 extract was further varied. 0.2 ml
was found to be the optimum volume of the test extracts [43].
Anti-tumor activity
Aqueous extract of Carica papaya leaves exhibits anti-tumor activity and it has been reported. In PBMC, the
production of IL-2 and IL-4 was reduced following the addition of Carica papaya extract, where as that of IL-
12p40, IL-12p70, IFN-γ and TNF-α was enhanced without growth inhibition [44].

4. Conclusion
Papaya plant is mainly used as the food ingredient throughout the world because of its fruits and its nutritive values.
From the above studies about the papaya plant shows that the plant’s leaves, stem, fruits and seeds also contains
different chemical constituents such as Alkaloids carpain, pseudocarpain, dehydrocarpaine I and II, choline,
carposide, vitamin C and E. Carposide and an enzyme myrosin , sinigrin, Carpaine, benzylisothiocyanate, benzyl
glucosinolate, glucotropacolin, benzylthiourea, hentriacontane, β-sitosterol, caricin, leaves related alkaloids,
flavonoids, saponins, tannins, cardiac glycoside, anthraquinones and cardinolodes etc. many of the pharmacological
activities has been done on the papaya plants. But hence extensive investigations on its pharmacodynamics, kinetics,
proper standardization and clinical trials are needed to exploit their therapeutics utility to cure many diseases.

5. References
1. Oh MS, Huh Y, Bae H, Ahn DK and Park SK. The multi-herbal formula Guibitang enhances memory and
increases cell proliferation in the rat hippocampus, Neurosciene Lettrs. 2005, 379: 205–8.
2. Govindarajan R, Vijayakumar M and Pushpangadan P. Antioxidant approach to disease management and
the role of ‘Rasayana’ herbs of Ayurveda. Journal of Ethnopharmacology. 2005, 99: 165-78.
3. Indian Materia Medica by K M Nadkarni, 1st Edn by A. K. Nadkarni, Popular Prakashan Pvt. Ltd, Bombay,
1954, pp.273-277.

548 | International Journal of Current Trends in Pharmaceutical Research


Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

4. Carlini, E.A. Plants and the central nervous system. Pharmacology, Biochemistry and Behavior. 2003, 75:
501–512.
5. Boning, Charles R. (2006). Florida's Best Fruiting Plants: Native and Exotic Trees, Shrubs, and Vines.
Sarasota, Florida: Pineapple Press, Inc. pp. 166–167.
6. Dick Gross (2003). ”Papaya”: A tantalising taste of the Tropics. Maricopa County Master Gardener
Volunteer information, University of Arizona Cooperative Extension. www.papaya Maricopa-
hort@ag.arizo.edu.
7. Jari, S. Papayas are yummy easy to grow. University of Hawaii- Manoa College of Tropical Agric. &
Human Resources. 2009.
8. Chan L-K, Teo C-KH. Micropropagation of Eksotika, a Malaysia papaya cultivar, and the field
performance of the tissue culture derived clones. Acta Horticulturae, 2002, 575: 99-105
9. Storey WB . Papaya. In: Ferwerda FP, Wit F (Eds) Outline of PerennialCrop Breeding in the Tropics, H.
Veenman & Zonen N.V. Wageningen Publishers. 1969, pp. 389-408
10. http://www.ethnoleaflets.com/leaflets/papaya.htm
11. Indian Materia Medica by K M Nadkarni, 1st Edn by A. K. Nadkarni, Popular Prakashan Pvt. Ltd, Bombay,
1954, pp.273-277.
12. Chan H-T, Tang C-S. The chemistry and biochemistry of papaya. In Inglett GE, Charolambous G (Eds)
Tropical Foods (Vol I), Academic Press, New York. 1979
13. Duke, J. A. (1996, July 3). Carica papaya L. Retrieved from http://www.hort.purdue.edu/ newcrop/
duke_energy/Carica_papaya.html
14. Oyoyede, O. I. Chemical Profile of Unripe Pulp of Carica papaya. Pakistan Journal of Nutrition. 2005,
4(6): 379-381.
15. Wall, M. Ascorbic acid, vitamin A, and mineral composition of banana (Musa sp.) and papaya (Carica
papaya) cultivars grown in Hawaii. Journal of Food Composition and Analysis. 2006, 19(5): 434-445.
16. Marotta F, Weksler M, Naito Y, Yoshida C, Yoshioka M and Marandola P, Nutraceutical supplementation,
effect of a fermented papaya preparation on redox status and DNA damage in healthy elderly individuals
and relationship with GSTM1 genotype, a randomized, placebo-controlled, cross-over study, Ann N Y
Acad Sci, 2006, 1067(1): 400-407.
17. Indian Medicinal Plants by KR Kirtikar and BD Basu, Reprint, 2 nd Edn, International Book Distributors,
Dehra Dun, Vol. II, 1998, pp.1097-1099.
18. Arya Vaidya Sala, Carica papaya, In: Indian Medicinal Plants & Compendium of 500 species, 1 st Edn, Vol
I, Orient Longman Pvt Ltd, Hyderabad, 2005, pp. 383-384.
19. Chen YT, Hsu LH, Huang IP, Tsai TC, Lee GC and Shaw JF, Gene cloning and characterization of a
novel recombinant antifungal chitinase from papaya (Carica papaya), J Agric Food Chem, 2007, 55(3),
714-722.
20. Buttle DJ and Barrett AJ, Chymopapains, Chromatographic purification and immunological
characterization, Biochem J, 1984, 223(1); 81-88
21. Kermanshai R, McCarry BE, Rosenfeld J, Summers PS, Weretilnyk EA and Sorger GJ, Benzyl
isothiocyanate is the chief or sole anthelmintic in papaya seed extracts, Phytochemistry, 2001, 57(3): 427-
435.
22. Stepek G, Buttle DJ, Duce IR, Lowe A and Behnke JM, Assessment of the anthelmintic effect of natural
plant cysteine proteinases against the gastrointestinal nematode, Heligmosomoides polygyrus, in vitro,
Parasitology, 2005, 130(2): 203-211.
23. Giordani R, Gachon C, Moulin-Traffort J and Regli P, A synergistic effect of Carica papaya latex sap and
fluconazole on Candida albicans growth, Mycoses, 1997, 40(11-12): 429-437.
24. Giordani R, Cardenas ML, Moulin-Traffort J and Regli P, Fungicidal activity of latex sap from Carica
papaya and Antifungal effect of D (+)-glucosamine on Candida albicans growth, Mycoses, 1996, 39(3-4),
103-110.
25. Bhat Praveen G and Surolia Namita, In vitro Antimalarial activity of extracts of three plants used in the
traditional medicine of India, Am J Trop Med Hyg, 2001, 65(4): 304-308.
26. Satrija F, Nansen P, Murtini S and He S, Anthelmintic activity of papaya latex against Heligmosomoides
polygyrus infections in mice, J Ethnopharmacol, 1995, 48(3): 161-164.
27. Satrija F, Nansen P, Bjorn H, Murtini S and He S, Effect of papaya latex against Ascaris suum in naturally
infected pigs, J Helminthol, 1994, 68(4): 343-346.
28. Hounzangbe-Adote S, Fouraste I, Moutairou K and Hoste H, In vitro effects of four tropical plants on the
activity and development of the parasitic nematode, Trichostrongylus colubriformis, J Helminthol, 2005,
79(1): 29-33.
29. Hounzangbe-Adote MS, Paolini V, FourasteI, Moutairon K and Hoste H, In vitro effects of four tropical
plants on three life-cycle stages of the parasitic nematode, Haemonchus contortus, Res Vet Sci, 2005, 78
549 | International Journal of Current Trends in Pharmaceutical Research
Asha Roshan et al, IJCTPR, 2014, Vol.2(4): 541-550

(2): 155-160.
30. Calzada F, Yepez-Mulia L and Tapia-Contreras A, Effect of Mexican medicinal plant used to treat
trichomoniasis on Trichomonas vaginalis trophozoites, Journal of Ethnopharmacology, 2007,
113(2): 248-251.
31. Osato JA, Santiago LA, Remo GM, Cuadra MS and Mori A, Antimicrobial and antioxidant activities of
unripe papaya, Life Science, 1993, 53(17): 1383-1389.
32. Emeruwa AC, Antibacterial substance from Carica papaya fruit extract, Journal of Natural Product, 1982,
45(2): 123-127.
33. Tona L, Kambu K, Ngimbi N, Cimanga K and Vlietinck AJ, Antiamoebic and phytochemical screening of
some Congolese medicinal plants, J Ethnopharmacol, 1998, 61(1): 57-65.
34. Udoh P, Essien I and Udoh F, Effects of Carica papaya (paw paw) seeds extract on the morphology of
pituitary-gonadal axis of male Wistar rats, Phytother Res, 2005, 19(12): 1065-1068.
35. Uche-N wachi EO, Ezeokoli DC, Adogwa AO and Offiah VN, Effect of water extract of Carica papaya
seed on the germinal epithelium of the seminiferous tubules of Sprague Dawley rats, Kaibogaku Zasshi,
2001,76(6): 517-521.
36. Bungorn Sripanidkulchai, Varima Wongpanich, Pisamai Laupattarakasem, Jamsai Suwansaksri and
Dusit Jirakulsomchok, Diuretic effects of selected Thai indigenous medicinal plants in rats, J
Ethnopharmacol, 2001, 75(2-3): 185-190.
37. Sharma HN and Mahanta HC, Modulation of morphological changes of endometrial surface epithelium by
administration of composite root extract in albino rat, Contraception, 2000, 62(1): 51-54.
38. Oderinde O, Noronha C, Oremosu A, Kusemiju T and Okanlawon OA, Abortifacient properties of aqueous
extract of Carica papaya Linn. Seeds on female Sprague-Dawley rats, Niger Postgrad Med J, 2002,
9(2): 95-98.
39. Kapoor M, Garg SK and Mathur VS, Antiovulatory activity of five indigenous plants in rabbits, Indian J
Med Res, 1974, 62(8): 1225-1227.
40. Rimbach G, Park YC, Guo Q, Moini H, Qureshi N, Saliou C, Takayama K, Virgili F and Packer L, Nitric
oxide synthesis and TNF-alpha secretion in RAW 264.7 macrophages, mode of action of a fermented
papaya preparation, Life Sci, 2000, 67(6): 679-694.
41. Aruoma OI, Deiana M, Rosa A, Casu V, Piga R, Peccagnini S, Dessi MA, Ke B, Liang YF and Higa T,
Assessment of the ability of the antioxidant cocktail-derived from fermentation of plants with effective
microorganisms (EM-X) to modulate oxidative damage in the kidney and liver of rats in vivo, studies upon
the profile of poly and mono-unsaturated fatty acids. Toxicol Lett, 2002, 135(3): 209-217.
42. JA Olagunjua, AA Adeneyeb, BS Fagbohunkac, NA Bisugac, AO Ketikuc, AS Benebod, OM Olufowobic,
AG Adeoyec, MA Alimic, AG Adelekec. Nephroprotective activities of the aqueous seed extract of Carica
papaya Linn. in carbon tetrachloride induced renal injured Wistar rats: a dose- and time-dependent study.
Biology and Medicine, 2009 , 1(1): 11-19
43. Oduala T., Adeniyi F.A.A., Ogunyemi E.O., Bello I.S. and Idowu T.O. Antisickling agent in an extract of
unripe pawpaw (Carica papaya): Is it real?. African Journal of Biotechnology., 2006, 5(20), pp. 1947-1949
44. Noriko Otsuki, Nam H. Dang, Emi Kumagai, Akira Kondo, Satoshi Iwata, Chikao Morimoto. Aqueous
extract of Carica papaya leaves exhibits anti-tumor activity and immunomodulatory effects. Journal of
Ethnopharmacology, 2010, 127( 3): 760-767

550 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 551-554

ISSN: 2321-3760
Review Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 551-554
www.pharmaresearchlibrary.com/ijctpr

A Devastating Inherited Neurodegenerative Disease: Huntington’s Disease: A


Clinical Review
Patel Chirag J*1, Satyanand Tyagi2, Patel Kanu J3, Patel Tushar4, Patel Harnish K5,
Patel Priyanka H6
1
Maharishi Arvind Institute of Pharmacy, Mansarovar, Jaipur, Rajasthan, India-302020.
2
Founder, President & CEO, Tyagi Pharmacy Association, Chattarpur, New Delhi, India-110074.
3
Sharda School of Pharmacy, Pethapur, Gandhinagar, Gujarat, India.
4
Aditya Bangalore Institute for Pharmacy Education & Research, Bangalore, Karnataka, India.
5
Editor-In-Chief, IJPRBS Journal, Gujarat, India.
6
Director, Research Scholar Hub, Gujarat, India.
Received: 15 March 2014, Accepted: 10 June 2014, Published Online: 15 July 2014

Abstract
Huntington’s disease (HD) is an inherited disease of the central nervous system that usually has its onset
between 30 and 50 years of age. The disease occurs in all racial groups but is most common in people of
northern European origin. Although no therapy is currently available to delay the onset of symptoms or
prevent the progression of the disease, symptomatic treatment of patients with Huntington disease (HD)
may improve the quality of life and prevent complications. As is the case with other neurological diseases,
HD makes individuals more vulnerable to side effects from medications, particularly cognitive adverse
effects. Symptomatic treatment for HD can be divided into drugs to treat the movement disorder and drugs
to treat psychiatric or behavioral problems. Symptomatic treatment of Huntington’s disease involves use of
Dopamine antagonists, presynaptic dopamine depleters, Antidepressants, Tranquillizers, Anxiolytic
Benzodiazepines, Anticonvulsants and Antibiotics. Several medications including baclofen, idebenone and
vitamin E have studied in clinical trials with limited samples.
Key words: Huntington’s disease, Chorea, Neurodegenerative, Brain

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .552
2. Motor signs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 553
3. Psychiatric signs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . .553
4. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . 554

*Corresponding author
Patel Chirag J
Department of Pharmaceutics,
Maharishi Arvind Institute of Pharmacy,
Mansarovar, Jaipur, Rajasthan, India
Manuscript ID: IJCTPR2146
PAPER-QR CODE
Copyright © 2013, IJCTPR All Rights Reserved

551 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 551-554

1. Introduction
The disease was originally named Huntington’s chorea after George Huntington, who wrote the first detailed
description in 1872. More recently, however, the name has changed to Huntington’s disease to reflect the fact that
chorea is not the only important manifestation of the disease. Many non-motor symptoms may be more disabling
and distressing than the motor symptoms. One study assessed the effect of cognitive and motor symptoms on the
ability of 67 people with early Huntington’s disease to carry out activities of daily living, and found that cognitive
impairment was associated with reduced functional ability independent of motor impairment [1, 2].

Huntington’s disease is a devastating inherited neurodegenerative disease characterized by progressive motor,


cognitive, and psychiatric symptoms. Patients may present with any of these symptoms, and familiarity with the
phenotype is therefore important. Chorea and loss of balance are early symptoms that patients notice, although
families often notice cognitive or personality changes before this [3, 4].

The mean age of onset of symptoms is 40 years, but juvenile onset (<20 years) and older onset (>70 years) forms are
well recognized. The Huntington’s disease Association (HDA) has records of 6161 adults with symptomatic
Huntington’s disease and 541 children with juvenile Huntington’s disease [2, 5].

Huntington’s disease is characterized by prominent neuronal loss in the caudate/putamen of the brain. The brain in
Huntington’s disease is usually small, offer weighing less than 1100 gm. The areas of the brain such as fewer
neurons in cerebral cortex, hypothalamus and thalamus are affected in the Huntington’s disease. Interneurons and
afferent terminals are largely spared, while the striatal projection neurons are severely affected. This leads to large
decreases in striatal gamma amino butyric acid concentrations, but somatostatin and dopamine concentrations are
relatively preserved [3, 5, 6].

Common Symptoms of Huntington’s Disease


1. Motor symptoms: Chorea, dystonia, loss of postural reflexes, bradykinesia, rigidity.
2. Psychiatric: Depression, obsessive-compulsive disorders, anxiety, irritability, apathy, hyper sexuality
(uncommon), psychosis (uncommon)
3. Metabolic: Weight loss, sleep disturbance.
4. Cognitive symptoms: Disorganization as a result of difficulties with planning, initiating, and organizing
thoughts, activities, and communication; perseveration; impulsivity; perceptual distortions; lack of insight;
distractibility; difficulty in learning new information.
5. Others: Dysphasia (combination of motor and language difficulties), dysphagia (combination of motor
problems, impulsivity, and distractibility) [7, 8].

Diagnosis:
DNA analysis can be used to confirm the diagnosis. Tests are available to identify whether someone has the faulty
gene. Genetic testing can diagnose Huntington’s disease at every stage of the life cycle. There are three categories
for testing such as: antenatal or prenatal, pre-symptomatic and confirmatory testing.
Antenatal or Prenatal Testing:
Either amniocentesis (a sample of fluid from around the fetus), or chorionic villus sampling (CVS)-a sample of fetal
cells from the placenta will indicate whether the body has inherited the gene for Huntington’s disease. Antenatal
tests are carried out early in pregnancy on the unborn children of couples from families affected by Huntington’s
disease. They can be used to calculate the risk of that child going on to develop the disease in their adult life. Again,
the implications of positive results are serious and couples need advice and support from a specialist doctor or
counselor to help them in their decisions [9, 10].
Pre-symptomatic Testing:
These are available to the people who are at risk of inheriting Huntington’s disease from a parent, but do not have
symptoms and don’t know whether or not they carry the gene. Pre-symptomatic tests are carried out in people who
are not showing symptoms of Huntington’s disease, but have a family history of it. The decision to take a test is a
serious one: a positive result can be devastating since it tells the individual that they will one day become severely
mentally ill. There are also issues surrounding testing when the individual parents have themselves not been tested,
since a positive result indicates that one parent also has the faulty gene. Advice from a genetic counselor about the
implications of taking the test is needed before going ahead [11, 12].
Confirmatory Testing:
This determines whether a person showing what appear to be the symptoms of Huntington’s disease, actually has the
disease. Neurological and psychological tests are also conducted to arrive at a conclusive diagnosis of Huntington’s
disease [9, 13].

552 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 551-554

Treatment And Management of Huntington’s Disease


Despite the fact that the pathogenesis of HD has still not been resolved and a cure is not available, many therapeutic
options are available for treating symptoms and signs with a view to improving quality of life. Although many signs
and symptoms can be treated, it is not always necessary to do so. The patient’s limitations in daily life determine
whether or not drugs are required. Very little evidence is available about the drug or the dosage to prescribe for any
signs and symptoms. Drug treatment is, therefore, individualized and based on expert opinion and daily practice 14, 15.
Treatment consists of drug prescription and non-medication advice. Surgical treatment does not play an important
role in HD and will be addressed only briefly.

2. Motor signs
Hyperkinesia, or chorea, is treated with dopamine receptor blocking or depleting agents. Most commonly used drugs
for chorea (Table 1) are typical or atypical neuroleptics (dopamine receptor blocking) and tetrabenazine (dopamine
depleting). The drugs prescribed differ per country. An extensive review of all medication is given by Bonelli. The
most commonly used drugs for depression and aggression are listed in Table 2. Clozapine and olanzapine are
atypical neuroleptics. Both have an antichoreatic effect. Clozapine requires white cell control in the blood and is,
therefore, less practical, making olanzapine the preferred drug. The most frequently reported side effects are weight
increase and anti-depressive effects. From small case studies some support can be found for prescribing quetiapine,
zotepine, ziprasidone, and risperidon. However, only tetrabenazine, a dopamine depleting drug, has been shown in a
controlled trial to significantly reduce chorea [16, 17].

The most common side-effects are depression and sedation. There is a long list of drugs without or with only a very
limited result, mostly in open case studies: a-tocopherol amantadine, baclofen, cannabidiol, chlordiazepoxide,
choline, clonazepam, creatine, deanol, dextromethorphan, fluoxetine, idebenone, ketamine, lamotrigine,
levatiracetam, milacemide, minocycline, muscimol, OPC 14117, PUFA, remacemide, riluzole. Drug treatment for
hypokinesia has been tried using antiparkinsonian drugs, but almost always with very disappointing results. In
practice, therefore, dopaminergic drug are not prescribed. To date, despite several claims, no drug is available with
any neuroprotective or disease-delaying effect. Disease modifying drugs are developed, but not available. Also
embryonic cell implants, still under study, are not proven treatment options at the moment. Surgical intervention to
treat chorea has been described in a few cases. Deep brain stimulation has a place in other movement disorders such
as Parkinson disease. In Alzheimer’s disease, anticholinesterase drugs are in use. In Huntington’s disease no clinical
trials with Rivastigmine or Donepezil are available. In short-term, open studies, no effect was found [18, 19].

Table 1. Drug Treatment for Chorea


Drug Maximum Dose
Tetrabenazine 200 mg
Olanzapine 20 mg
Tiapride 600 mg
Fluphenazine 10 mg
Pimozide 6 mg
Risperidone 16 mg

Table 2. Drug Treatment for Depression (A) And Aggression (B)


A. Depression B. Aggression
Drug Maximum Dose Drug Maximum Dose
Fluoxetine 60 mg Olanzapine 20 mg
Citalopram 60 mg Citalopram 60 mg
Mirtazapine 45 mg Sertraline 200 mg
Carbamazepine 1600 mg Haloperidol 10 mg
Valproinezuur 2000 mg Dipiperon 360 mg

3. Psychiatric signs
As depression and aggressive behavior are the most devastating to family life, the majority of drugs are prescribed
for these signs. All advice is based on open studies and expert opinion. (Table 2) Besides medication, many other
care measures are available. It is important to find the right therapy for the right person at the right time. Non-
medical interventions available are: physiotherapy, occupational therapy, speech therapy, dietician, psychologist,
social worker, and nurse. During the course of the disease, the patient requires more care, which can also help

553 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 551-554

his/her partner, for example by having a nurse at home to help with showering. The burden for the caregiver can
become too heavy and so help must be found in day-care institutions, usually connected to nursing home facilities.
In the period that follows the patient moves into a transition phase and eventually a 24-hour care situation.
Throughout the whole process of increasing dependency, psychological help is often needed for the caregiver, who
has to deal with increasing responsibilities while losing contact with his or her former partner[19, 20].

Partner groups can be very useful. In general, lay organizations play an enormous role in educating caregivers,
patients and families. Medical and non-medical treatment must be individually tailored, as the symptoms and signs
differ by person and over time tremendously. Ideally treatment of patients and their families should be organised by
a multidisciplinary team. Treatment is intended to improve quality of life. To date, no cure is available
unfortunately[21].

4. References
1. Bohanna I, Georgiou-Karistianis N, Hannan AJ, Egan GF. Magnetic resonance imaging as an approach
towards identifying neuropathological biomarkers for Huntington’s disease. Brain Res Rev, 2008, 58: 209-
25.
2. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr. Neuropathological
classification of Huntington’s disease. J Neuropathol Exp Neurol, 1985, 44: 559-77.
3. Rothlind JC, Bylsma FW, Peyser C, Folstein SE, Brandt J. Cognitive and motor correlates of everyday
functioning in early Huntington’s disease. J Nerv Ment Dis, 1993, 181: 194-9.
4. Walker FO. Huntington’s disease. Lancet, 2007, 369: 218-28.
5. Nance MA. Comprehensive care in Huntington’s disease: a physician’s perspective. Brain Res Bull, 2007,
72: 175-8.
6. Craufurd D, Tyler A. Predictive testing for Huntington’s disease: protocol of the UK Huntington’s
Prediction Consortium. J Med Genet, 1992, 29: 915-8.
7. Robert MF. Antibiotic Minocycline May Treat Huntington’s Disease 2000. Nature Medicine, 6: 797-801.
8. Phillips W, Shannon KM, Barker RA. The current clinical management of Huntington’s disease. Mov
Disord, 2008, 23: 1491-504.
9. Adam OR, Jankovic J. Symptomatic treatment of Huntington disease. Neurotherapeutics 2008; 5: 181-97.
10. Harper PS. The Epidemiology of Huntington’s Disease. Hum Genet, 1992, 89: 365-376.
11. Wild EJ, Tabrizi SJ: Huntington’s disease phenocopy syndromes. Curr Opin Neurol, 2007, 20: 681-7.
12. Rubinsztein DC: Lessons from animal models of Huntington’s disease. Trends Genet, 2002, 18: 202-9.
13. Huntington Study Group: Unified Huntington’s disease Rating Scale: reliability and consistency. Mov
Disorders, 1996, 11: 136-42.
14. Henley SM, Wild EJ, Hobbs NZ, Frost C, MacManus DG, Barker RA, Fox NC, Tabrizi SJ: Whole-brain
atrophy as a measure of progression in premanifest and early Huntington’s disease. Mov Disord, 2009, 24:
932-6.
15. Vonsattel JP: Huntington disease models and human neuropathology: similarities and differences. Acta
Neuropathol, 2008, 115: 55-69.
16. Shoulson I, Fahn S: Huntington disease: clinical care and evaluation. Neurology, 1979, 29: 1-3.
17. Huntington Study Group: Tetrabenazine as antichorea therapy in Huntington disease: a randomized
controlled trial. Neurology, 2006, 66: 366-72.
18. Bonelli RM, Hofmann P: A systematic review of the treatment studies in Huntington’s disease since 1990.
Expert Opin Pharmacother, 2007, 8: 141-53.
19. Bonelli RM, Wenning GK: Pharmacological management of Huntington’s disease: an evidence-based
review. Curr Pharm Des, 2006, 12: 2701-20.
20. Gusella JF, Wexler NS, Conneally PM, Naylor SL, Anderson MA, Tanzi RE, Watkins PC, Ottina K,
Wallace MR, Sakaguchi AY: A polymorphic DNA marker genetically linked to Huntington’s disease.
Nature, 1983, 306: 234-8.
21. Decruyenaere M, Evers-Kiebooms G, Boogaerts A, Philippe K, Demyttenaere K, Dom R, Vandenberghe
W, Fryns JP: The complexity of reproductive decision-making in asymptomatic carriers of the Huntington
mutation. Eur J Hum Genet, 2007, 15: 453-62.
22. International Huntington Association (IHA) and the World Federation of Neurology (WFN) Research
Group on Huntington’s chorea: Guidelines for the molecular genetics predictive test in Huntington’s
disease. Neurology, 1994, 44: 1533-6.

554 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

ISSN: 2321-3760
Review Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 555-561
www.pharmaresearchlibrary.com/ijctpr

A Novel Pharmaceutical Microscopic Vesicle: Liposome


Patel Chirag J*1, Satyanand Tyagi2, Patel Kanu J3, Patel Tushar4, Patel Harnish K5,
Patel Priyanka H6
1
Maharishi Arvind Institute of Pharmacy, Mansarovar, Jaipur, Rajasthan, India-302020.
2
Founder, President & CEO, Tyagi Pharmacy Association, Chattarpur, New Delhi, India-110074.
3
Sharda School of Pharmacy, Pethapur, Gandhinagar, Gujarat, India.
4
Aditya Bangalore Institute for Pharmacy Education & Research, Bangalore, Karnataka, India.
5
Editor-In-Chief, IJPRBS Journal, Gujarat, India.
6
Director, Research Scholar Hub, Gujarat, India.
Received: 15 March 2014, Accepted: 10 June 2014, Published Online: 15 July 2014

Abstract
Liposomes are simple microscopic vesicles in which an aqueous volume is entirely enclosed by a membrane
composed of lipid bilayers. Liposomes are defined as structure consisting of one or more concentric spheres
of lipid bilayers separated by water or aqueous buffer compartments. The liposomes have emerged as most
practically useful carriers for in-vivo drug delivery as majority of reports has concentrated on the use of
phospholipid vesicles or liposomes as potential drug carrier systems. The water soluble compounds/drugs are
present in aqueous compartments while lipid soluble compounds/drugs and amphiphilic compounds/drugs
insert themselves in phospholipid bilayers. The present review focuses upon the advantages, disadvantages,
mechanism, classification, method of preparation, characterization and application of liposomes.
Key words: Liposomes, Phospholipid, Stability, Lipid bilayers

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .555
2. Mechanism of Vesicle Formation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 557
3. Classification. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . . . . . . . . 557
4. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . 561

*Corresponding author
Patel Chirag J
Department of Pharmaceutics,
Maharishi Arvind Institute of Pharmacy,
Mansarovar, Jaipur, Rajasthan, India
Manuscript ID: IJCTPR2147
PAPER-QR CODE
Copyright © 2013, IJCTPR All Rights Reserved

1. Introduction
The rising number of complications associated with drugs from varied chemical and biological background not only
made scientists worldwide to search for newer molecules but also to discover the new ways and means for the
proper delivery of molecules. With the help of new delivery systems known as novel drug delivery systems (NDDS)
both old and new molecules can be delivered to the site in demand in a defined manner [1, 2].
555 | International Journal of Current Trends in Pharmaceutical Research
Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

The name liposome is derived from two Greek words: 'Lipos' meaning fat and 'Soma' meaning body. A liposome
can be formed at a variety of sizes as unilamellar or multi-lamellar construction, and its name relates to its structural
building blocks, phospholipids, and not to its size. Liposomes were first described by British haematologist Dr Alec
D Bangham in 1961, at the Babraham Institute, in Cambridge. They were discovered when Bangham and R. W.
Horne were testing the institute's new electron microscope by adding negative stain to dry phospholipids [3, 4].

Liposomes are now used to deliver certain vaccines, enzymes and drugs to the body. When used in the delivery of
certain cancer drugs, liposomes help to shield healthy cells from the drugs toxicity and prevent their concentration in
vulnerable tissues (e.g., kidney, liver), lessening or eliminating the common side effects of nausea, fatigue and hair
lose [1, 5, 6].
Advantages of Liposomes [1,4,7]
a. Provide controlled and sustained release
b. Liposomes are biocompatible, completely biodegradable, non-toxic, flexible and nonimmunogenic for
systemic and non-systemic administrations.
c. Stabilization of entrapped drug from hostile environment
d. Can carry both water and lipid soluble drugs
e. Drugs can be stabilized from oxidation
f. Can incorporate micro and macro molecules
g. Flexibility to couple with site-specific ligands to achieve active targeting
h. Alter pharmacokinetics and pharmacodynamics of drugs
i. Therapeutic index of drugs is increased
j. Controlled hydration
k. Help to reduce exposure of sensitive tissues to toxic drugs.
l. Targeted drug delivery or site specific drug delivery
m. Can be administered through various routes
n. Act as reservoir of drugs
o. Can modulate the distribution of drug
Disadvantages [5, 6, 8]
1. Problem to targeting to various tissue due to their large size
2. Short half life
3. Less stability
4. Leakage and fusion of encapsulated drug / molecules
5. High production cost
6. Low solubility
7. Quick uptake by cells of R.E.S
8. Sometimes phospholipid undergoes oxidation and hydrolysis like reaction.
9. High production cost
10. Allergic reactions may occur to liposomal constituents
List of Materials used for Liposomes Formulation
1. Membrane forming components
Phospholipids that are the major components of the biological membranes are the building blocks of the
liposomes. The phospholipids have tubular shape owning to the presence of two acyl chains attached to a
polar head and on hydration, results into a bilayered membrane. Two types of phospholipids are there i.e.
phosphodiglycerides and sphingolipids along with their corresponding hydrolysis products.
Classification of phospholipids
A. Neutral phospholipids e.g. Sphingomyelin, Phosphatidylethanolamine and Phosphatidylcholine.
B. Negatively charged phospholipids e.g. Dipalmitoyl phosphatidylcholine, Dipalmitoyl phosphatidyl
acid (DDPA), Distearoyl phosphatidyl choline (DSPC), Dioleoyl phosphatidyl choline (DOPC)
etc.
C. Positively charged phospholipids e.g. 1, 2-dihexadecyl-N, N-dimethyl–N-trimethyl amine methyl
ethanol amine etc [8, 9].
2. Membrane Additives (Sterols)
Cholesterol is the most commonly used sterol, which is included in the liposomal membranes. It has been
called as the ‘motar’ of bilayers because by virtue of its molecular shape and solubility properties, it fills in
empty spaces among the phospholipid molecules, anchoring them more strongly into the structure.
Cholesterol is an amphipathic molecule and inserts itself into the membrane with its hydroxyl groups
oriented towards the aqueous phase and aliphatic chain aligned parallel to acyl chains of the phospholipid
molecules. In other words, cholesterol increases the transition temperature of the system by making the

556 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

membrane more ordered. Cholesterol reduces this type of interaction to a great extent and provides both
physical and biological stability[9, 10].
3. Charge inducers and Steric stabilizers
Stearylamine, dicetylphosphate, solulan C-24 and diacylglycerol are commonly used to impart either a
negative or a positive surface charge. Since it is a well-known fact that negatively charged and positively
charged liposomes are more rapidly uptaken by the reticulo-endothelial system as compared to neutral
liposomes, charge inducers are used to overcome this problem. Also they proved to be useful in reducing
aggregation as neutral liposomes show higher tendency to undergo aggregation [2, 11].
4. Other substances
In case, the drug is very prone to oxidation, antioxidants e.g. tocopherol, butylated hydroxy toluene and
stabilizers are used. The use of preservatives is very common to increase the shelf-life of liposomal
formulations [8, 12].

2. Mechanism of Vesicle Formation


It has been proved that phospholipids spontaneously form closed structures when hydrated in aqueous media.
Because phospholipids are amphipathic (both hydrophilic and hydrophobic) in nature, their thermodynamic phase
properties and self-assembling characteristics evoke entropically driven sequestration of hydrophobic regions into
spherical bilayers. In other words, unfavorable interactions come into play between lipid molecules and water
molecules. The self-assembling action of phospholipid molecules into bilayered sheets leads to lowering of
unfavorable interaction between the solvent and long hydrocarbon fatty chains thus acquiring a state of lower energy
and almost maximum stability. Well known amphiphiles include soaps, detergents and polar lipids (lecithins,
kephalins). Further, to gain a completely stable state, bilayer sheets start folding or curl-on itself to form closed
sealed bilayered vesicles enclosing a central aqueous core.

This phenomenon can be understood in quantitative terms by considering the critical micelle concentration (CMC)
of phosphatidylcholine in water. The CMC is defined as the concentration of the lipid in water (usually expressed as
moles per liter) above which the lipid forms either micelles or bilayer structures rather than remaining in solution as
monomers. The CMC of dipalmitoyl phosphatidylcholine has been measured by Smith and Tenford and found to be
4.6x10-10 M in water. This value is in agreement with those obtained for similar amphiphiles. Clearly, this is a very
small number indicating the overwhelming preference of this molecule for a hydrophobic environment such as that
found in the core of a micelle or bilayer [5,11,13].
Mechanism of Transportation Through Liposomes [3,5,14]
Liposome can interact with cells by four different mechanisms:
1. Endocytosis by phagocytic cells of the reticulo endothelial system such as macrophages and neutrophils.
2. Adsorption to the cell surface either by nonspecific weak hydrophobic or electrostatic forces or by specific
interactions with cell-surface components.
3. Fusion with the plasma cell membrane by insertion of the lipid bilayer of the liposome into the plasma
membrane, with simultaneous release of liposomal content into the cytoplasm
4. Transfer of liposomal lipids to cellular or subcellular membranes, or vice versa, without any association of
the liposome contents. It often is difficult to determine what mechanism is operative and more than one
may operate at the same time.
5.
3. Classification
Based on composition and mode of drug delivery
A. Conventional liposomes
These types of liposomes are composed of neutral or negatively charged phospholipids and cholesterol.
It is useful for E.E.S targeting; rapid and saturable uptake by R.E.S; short circulation half life, dose
dependent pharmacokinetics.
B. Cationic Liposomes
These types of liposomes are composed of cationic lipids. These are mainly suitable for delivery of
negatively charged macromolecules (DNA, RNA); ease of formation, structurally unstable; toxic at
high dose, mainly restricted to local administration
C. Temperature or heat sensitive liposomes
These types of liposomes are composed of dipalmitoyl phosphotidyl choline. These are vesicles
showed maximum release at 41˚C, the phase transition temperature of dipalmitoyl phosphotidyl
choline. Liposomes release the entrapped content at the target cell surface upon a brief heating to the
phase transition temperature of the liposome membrane.
D. pH sensitive liposomes

557 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

These types of liposomes are composed of phospholipids such as phosphatidyl ethanolamine, dioleoyl
phosphatidyl ethanolamine. These are subjected to coated pit endocytosis at low pH, fuse with cell or
endosomes membrane and release their contents in cytoplasm; suitable for intra cellular delivery of
weak base and macromolecules. Biodistribution and pharmacokinetics similar to conventional
liposomes.
E. Long circulating or stealth liposomes
These types of liposomes are composed of neutral high transition temperature lipid, cholesterol and 5-
10% of PEG-DSPE. These are subjected to hydrophilic surface coating, low opsonisation and thus low
rate of uptake by R.E.S. So, it has long circulating half life (40 hrs) and dose independent
Pharmacokinetics.
F. Immuno liposomes
These are conventional or stealth liposomes with attached antibody or recognition sequence. These are
subjected to receptor mediated endocytosis. It has cell specific binding (targeting) and can release
contents extra cellularly near the target tissue and drugs diffuse through plasma membrane to produce
their effects.
G. Magnetic Liposomes
These types of liposomes are composed of phosphotidyl choline, cholesterol and small amount of a
linear chain aldehyde and colloidal particles of magnetic iron oxide.These are liposomes that
indigenously contain binding sites for attaching other molecules like antibodies on their exterior
surface. These can be made use by an external vibrating magnetic field on their deliberate, on site,
rapture and immediate release of their components.
1. Based on Size and Number of Lamellae
A. Small Unilamellar Vesicles (S.U.V.)
Single bilayer, homogeneous in size, thermodynamically unstable, susceptible to aggregation and
fusion at low or no charge, limited capture of macro molecules, low aqueous volume to lipid ratio (0.2
: 1.5 : 1 mole lipid) prepared by reducing the size of M.L.V. or L.U.V. using probe sonicator or gas
extruder or by active loading or solvent injection technique.
B. Large Unilamellar Vesicles (L.U.V.)
Large unilamellar vesicles have single bilayer, high aqueous volume to lipid ratio (7: 1 mole lipid),
useful for hydrophilic drugs, high capture of macro molecules; rapidly cleared by R.E.S. Prepared by
detergent dialysis, ether injection, reverse phase evaporation or active loading methods.
C. Multi Lamellar Vesicles (M.L.V)
Multi lamellar vesicles have more than one bilayer; moderate aqueous volume to lipid ratio 4: 1 mole
lipid. Greater encapsulation of lipophilic drug, mechanically stable upon long term storage, rapidly
cleared by R.E.S, useful for targeting the cells of R.E.S, simplest to prepare by thin film hydration of
lipids in presence of an organic solvent.
a. Oligo lamellar vesicles or Paucilamellar vesicles: Intermediate between L.U.V. & M.L.V.
b. Multi vesicular liposomes: Separate compartments are present in a single M.L.V.
c. Stable Pluri lamellar vesicles: Have unique physical and biological properties due to osmotic
compression.

Figure 1. Schematic Representation of Different Liposomes

558 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

Method of Preparation [3,4,8,17]


The preparation of all types of vesicular systems requires the input of energy. Generally all the methods of liposome
preparation involve three basic stages:
1. Dissolved cholesterol and lecithin in organic solvent.
2. Dispersion of lipids in aqueous media.
3. Separation and purification of resultant liposomes.
Various methods used for the preparation of liposome:
1. Passive loading techniques
Passive loading techniques include three different methods:
I. Mechanical dispersion method
a. Lipid film hydration by hand shaking, non-hand shaking or freeze drying
b. Micro-emulsification
c. Sonication
d. French pressure cell
e. Membrane extrusion
f. Dried reconstituted vesicles
g. Freeze-thawed liposomes
II. Solvent dispersion method
a. Ether injection
b. Ethanol injection
c. Double emulsion vesicles
d. Reverse phase evaporation vesicles
e. Stable plurilamellar vesicles
III. Detergent removal method
a. Detergent (cholate, alkylglycoside, Triton X-100) removal form mixed micelles
b. Dialysis
c. Column chromatography
d. Dilution
2. Active loading
Characterization of Liposomes [13,15,17]
Liposome prepared by one of the preceding method must be characterized. The most important parameters of
liposome characterization include visual appearance, turbidity, size distribution, lamellarity, concentration,
composition, presence of degradation products, and stability.
1. Visual Appearance
Liposome suspension can range from translucent to milky, depending on the composition and particle size.
If the turbidity has a bluish shade this means that particles in the sample are homogeneous; a flat, gray
color indicates that presence of nonliposomal dispersion and is most likely disperse inverse hexagonal
phase or dispersed microcrystallites. An optical microscope (phase contrast) can detect liposome > 0.3 μm
and contamination with larger particles.
2. Determination of Lamillarity
The lamellarity of liposomes is measured by electron microscopy or by spectroscopic techniques. Most
frequently the nuclear magnetic resonance spectrum of liposome is recorded with and without the addition
of a paramagnetic agent that shifts or bleaches the signal of the observed nuclei on the outer surface of
liposome. Encapsulation efficiency is measured by encapsulating a hydrophilic marker.
3. Entrapped Volume
The entrapped volume of a population of liposome (in μL/ mg phospholipid) can often be deduced from
measurements of the total quantity of solute entrapped inside liposome assuring that the concentration of
solute in the aqueous medium inside liposomes is the same after separation from unentrapped material. For
example, in two phase method of preparation, water can be lost from the internal compartment during the
drying down step to remove organic solvent.
4. Determination of Liposomal Size Distribution
Size distribution is normally measured by dynamic light scattering. This method is reliable for liposomes
with relatively homogeneous size distribution. A simple but powerful method is gel exclusion
chromatography, in which a truly hydrodynamic radius can be detected. Sephacryl-S100 can separate
liposome in size range of 30-300nm. Sepharose -4B and -2B columns can separate SUV from micelles.
5. Surface Charge
Liposomes are usually prepared using charge imparting constituting lipids and hence it is imparting to
study the charge on the vesicle surface. In general two method are used to assess the charge, namely

559 | International Journal of Current Trends in Pharmaceutical Research


Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

freeflow electrophoresis and zeta potential measurement. From the mobility of the liposomal dispersion in a
suitable buffer, the surface charge on the vesicles.
6. Liposome Stability
Liposome stability is a complex issue, and consists of physical, chemical, and biological stability. In the
pharmaceutical industry and in drug delivery, shelf life stability is also important. Physical stability
indicates mostly the constancy of the size and the ratio of lipid to active agent. The cationic liposomes can
be stable at 4°C for a long period of time, if properly sterilized.
Applications [13,14,18]
1. Liposome in antimicrobial, antifungal and antiviral therapy
A. Liposomal drugs
B. Liposomal biological response modifiers
2. Liposome in tumor therapy
A. Carrier of small cytotoxic molecules
B. Vehicle for macromolecules as cytokines or genes
3. Liposome as drug/protein delivery vehicles
A. Controlled and sustained drug release
B. Altered pharmacokinetics and biodistribution
C. Enhanced drug solubilization
D. Enzyme replacement therapy and biodistribution
E. Altered pharmacokinetics and biodistribution
4. Liposome in immunology
A. Immunoadjuvant
B. Immunodiagnosis
C. Immunomodulator
5. Liposome in gene delivery
A. Gene and antisense therapy
B. Genetic (DNA) vaccination
6. Liposome in enzyme immobilization and bioreactor technology
7. Liposome as radiopharmaceutical and radio diagnostic carriers
8. Liposome in cosmetics and dermatology
9. Liposome as artificial blood surrogates
Future Challenges for Liposomes [2, 17, 18]
Although liposomes have proved their potential as drug delivery vehicles, only few products have come up with the
stage of commercial production. Some to mention are Daunoxome, Ambisome, Doxil, Epaxel, etc. There are
basically three major problems that we come across with the liposomal delivery systems i.e. uptake by reticulo-
endothelial system, large-scale production and instability of phospholipids that pose as a hurdle in their commercial
development.
1. Large scale production of liposomes
Preparation of liposomes involves various steps like evaporation of solvent system under reduced pressure,
preparation of thin lipid film, sonication etc. These steps are difficult to carry out at large scale level
especially the preparation of thin film. So, it is difficult to scale up liposome production from laboratory
level to large-scale production level. Also adding organic solvents such as chloroform, methanol etc to
solubilize and mix lipids is not recommended in such a high concentrations as per the regulatory norms.
2. Uptake of liposomes by Reticulo-endothelial system
For drug delivery, liposomes can be formulated as a suspension, as an aerosol, in a semisolid form such as
a cream, gel or a dry powder and these can be administered .After systemic administration, which seems to
be the most promising route for these carrier systems, liposomes are typically recognized as foreign
particles and consequently endocytosed by cells of the mononuclear phagocyte system (MPS), mostly fixed
Kupffer cells in the liver and spleen. This fate is very useful for delivering drugs to these cells but, in
general, excludes other applications, including site-specific drug delivery by using ligands expressed on the
liposome surface in order to bind to receptors over-expressed on the diseased cells. For this reason, a search
for liposomes that could evade rapid uptake by the MPS started and few lipid compositions that prolonged
liposome blood-circulation times have been discovered. PEG-coated or sterically stabilized liposomes are
good examples in this regard.
3. Stability of liposomes
a. Physical stability
The stability of a pharmaceutical product usually is defined as the capacity of the delivery system
to remain within defined or pre-established limits during the shelf life of the product. There is no
established protocol for either accelerated or long–term stability studies for the liposomal
560 | International Journal of Current Trends in Pharmaceutical Research
Patel Chirag J et al, IJCTPR, 2014, Vol.2(4): 555-561

formulation. Classical models from colloidal science can be used to describe liposome stability.
Colloidal systems are stabilized electrostatically, sterically or electrosterically. In addition the
selfassembling colloids can undergoes fusion or phase change after aggregation. Liposome exhibit
both physical and chemical stability characteristics. Generally, the physical characteristic
describes the preservation of liposome structure and the chemical characteristic refers to molecular
structure of liposomal components. (Hydrolysis and oxidation of phospholipid) Physically stable
formulations preserve both liposome size distribution and the amount of material encapsulated.
The stability problem overcomes by using appropriate techniques like freezing, lyophilization and
osmification.
b. Plasma Stability
Although liposomes resemble biomembranes, they still are foreign objects for the host. Therefore,
liposomes are recognized by the mononuclear phagocytic system (MPS) after interaction with
plasma proteins. As a result, liposomes are cleared from the blood stream. These stability
problems solve by using synthetic phospholipids, gangliosides, polymerization, coating liposomes
with chitin derivatives, freeze drying, microencapsulation and particle coated with amphipathic
polyethylene glycol.

4. References
1. Jain N. K., “Controlled and novel drug delivery”, CBS publishers and distributors, 2007, pp. 304.
2. Allen T.M., “Liposomes: Opportunities in drug delivery”, Drugs, 1997, Vol. 54: 8-14.
3. Kamble R., Pokharkar V. B. and Badde S. and Kumar A., “Development and characterization of
liposomaldrug delivery system for nimesulide”, Int. J. Pharm. Pharm. Sci., 2010, Vol. 2(4): 87-89.
4. Woodle M.C. and Papahadjopoulos D., “Liposome preparation and size characterization”, Methods
Enzymol., 1989, Vol. 171: 193-217.
5. Patel S.S., “Liposome: A versatile platform for targeted delivery of drugs”, Pharmainfo.net, 2006, Vol.
4(5): 1-5.
6. Allen T.M. and Moase E.H., “Therapeutic opportunities for targeted liposomal drug delivery”, Adv Drug
Deliv Rev, 1996, Vol. 21: 117-133.
7. Vyas S.P. and Khar R.K. “Targeted And Controlled Drug Delivery: Novel Carrier Systems”, CBS
Publishers & Distributor, 1st Edition, 2006: 421-427.
8. Bally M., Nayar R., Masin D., Hope M.J., Cullis P.R. and Mayer L.D., “Liposomes with entrapped
doxorubicin exhibit extended blood residence times”, Biochim Biophys Acta, 1990, Vol. 1023: 133-139.
9. Sharma A. and Sharma U.S., “Liposomes in drug delivery: Progress and limitations”, Int. J. Pharm., 1997,
Vol. 154: 123-140.
10. Mayer, L.D., Hope, M.J., Cullis, P.R., Janoff, A.S., 1985. Solute distributions and trapping efficiencies
observed in freeze-thawed multilmellar vesicles. Biochim. Biophys. Acta 817, 193-196.
11. Szoke F.J. and Papahadjopoulos D., “Comparative properties and methods of preparation of lipid vesicles
(liposomes)”, Ann. Rev. Biophys. Bioeng., 1980, Vol. 9: 467-508.
12. Sharma V.K., Mishra D.N., Sharma A.K. and Srivastava B., “Liposomes: Present Prospective and Future
Challenges”, IJCPR, 2010; Vol. 1(2): 7-16.
13. Weiner, N., Martin, F., Riaz, M., Lipoomes as drug delivery system. Drug Develop. Ind. Pharm., 1989, 15,
1523-1554.
14. Mezei, M., Liposomes and the skin. In: Gregoriadis, G., Florence, A.T., Patel, H.M., (Eds.), Liposomes in
Drug Delivery, Harwood Academic Publishers, 1993, pp. 125-135.
15. Chrai S.S., Murari R. and Ahmad I., “Liposomes (a review) part one: manufacturing issues”, Bio. Pharm.,
2001, Vol. 11: 10-14.
16. Hope M.J. and Kitson C.N., “Liposomes: A perspective for dermatologists”, Dermatol. Clin., 1993, Vol.
11: 143-154.
17. Gregoriadis, G., Florence, A.T., Liposomes for drug deliver. Drugs, 1993, 45: 15-28.
18. Lasic, D.D., Novel application of liposomes. Tibitech., 1998, 16: 307-321.

561 | International Journal of Current Trends in Pharmaceutical Research


Y. Bhagyasri et al, IJCTPR, 2014, Vol.2(4): 562-565

ISSN: 2321-3760
Review Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 562-565
www.pharmaresearchlibrary.com/ijctpr

A Review on Anti- Inflammatory and Anti-Oxidant Activity of Polyherbal


Plant Extracts
Y. Bhagyasri*, M.S. Divyasree, C.K. Ashok kumar, A. Nagamani
Department of Pharmacognosy, Sree Vidyanikethan College of pharmacy, Rangampet, Tirupati-517102,
Andhra Pradesh, Iindia.
Received: 15 April 2014, Accepted: 28 June 2014, Published Online: 15 July 2014

Abstract
A lot of medicinal plants, traditionally used for thousands of years, are present in a group of herbal
preparations of the Indian traditional health care system named Rasayana proposed for their interesting
antioxidant activities. Our country is a rich source of both biological and chemical diversity, which may
be useful as a source of novel chemical structures. The timber value of Tectona grandis has been well
known from decades. In The present study was meant to characterize pharmacological potential of
different extracts from leaf, bark and stem of (Curcuma longa L., Abutilan indica L., Momordica
charantia L., Solanum nigrum L., Tinospora cordifolia L., Abutilon indicum L.,) are viewed for their
historical, etymological, morphological, Phytochemical and pharmacological aspects. The plants
described contain antioxidant and anti-inflammatory principles. A comparative phytochemical analysis
was carried to prove that the amount of phyto constituents varied with the fresh and dry stages of plants
contributing to the activity of the extract. Total phenolic and saponnins content of both the extracts was
estimated and was found to be more in the leaf, bark and stem giving positive results and the presence of
steroids and Terpenoids also showed in leaf and stem. Antioxidant activity of extracts was carried out
using ferulic acid and 2, 2-diphenyl-1- picrylhydrazyl (DPPH) assay and anti-inflammatory activity of the
extract were also made. The anti-inflammatory activity was done by in-vivo model i.e. the carrageenan
induced rat paw edema model, this extract reduced carrageenan induced rat paw edema in a dose
dependent manner. This work stimulates the researchers for further research on the potential use of
medicinal plant barks having anti-oxidant and anti-inflammatory property.
Key words: medicinal plant, anti-oxidant and anti-inflammatory activity, herbal extract

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 563
2. Anti-Inflammatory Activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..564
3. Anti-Oxidant Activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . .. . . . . . .564
4. Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 564
5. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . .564

*Corresponding author
Y. Bhagyasri
Department of Pharmacognosy,
Sree Vidyanikethan College of Pharmacy,
Rangampet, Tirupati-517102, A.P, Iindia.
Manuscript ID: IJCTPR2156 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved


562 | International Journal of Current Trends in Pharmaceutical Research
Y. Bhagyasri et al, IJCTPR, 2014, Vol.2(4): 562-565

1. Introduction
The tribal and rural population of India largely depends on medicinal plants for their health care as well as for their
livestock. This attracted the attention of several botanists that lead to an array of reports on ethnomedicine.1The
evaluation of these drugs is primarily based on Phytochemical, pharmacological and allied approaches including
various instrumental techniques such as chromatography, microscopy and others. With the emerging worldwide
interest in adopting and studying traditional systems and exploiting their potential based on different health care
systems, the evaluation of the rich heritage of traditional medicine is essential. Drugs which are used presently for
the management of pain and inflammatory conditions are either steroidal like corticosteroids or non steroidal like
aspirin. All of these drugs possess more or less side and toxic effects like renal failure, allergic reactions, hearing
loss or they may increase the risk of hemorrhages by affecting platelet function. On the contrary many medicines of
plant origin had been used since ages without any adverse effects. It is therefore essential that efforts should be
made to introduce new medicinal plants to develop more effective and cheaper drugs. Plants represent a large
natural source of useful compounds that might serve as lead for the development of novel drugs. It is very important
that profound research with ethno botanical plants possessing anti-inflammatory and analgesic properties can
definitely open up new vistas in inflammatory disorders. Purified natural compounds from plants can serve as
template for the synthesis of new generation anti-inflammatory drugs with low toxicity and higher therapeutic value.
This article reviews such medicinal plants with anti-inflammatory and anti-oxidant properties which have been used
by our ancestors to cure many of their ailments.

Table 1. Plants having Anti-oxidant and Anti-Inflammatory activity


Botanical Part
S.No Family Chemical Constituent Activity Ref
Name used
Solanum Solanidine, solanine,beta Anti-Oxidant And
1 Solanaceae leave 4
nigrum 2- solamargine Anti-Inflammatory
Tinospora Tinosporine, tinosporide, Anti-Oxidant And
2 Menispermaceae Stem 5
cordifolia tinosporidine, Anti-Inflammatory
Abutilon Anti-Oxidant And
3 Malvaceae leave β-Sitosterol, quinque 6
indicum Anti-Inflammatory
Mitragyna pyroligneous acid,methyl
Anti-inflammatory,
4 parvifolia Rubiaceae fruits acetate, ketones and 8
Analgesics
aldehydes
Tectona quinones, steroids,
5 grandis Vervenaceae Leaves glycosides, flavonoids, Anti-inflammatory 9
alkaloids, saponin
Emblica
punigluconin (12%) and
6 officinalis Euphorbiaceae Fruit Anti-oxidant 11
pedunculagin (14%
Gaertn.
Momordica
7 cucurbitacae Fruit Glycolipids,Phospolipids Anti-oxidant 14
charantia L.;

I. Inflammation and Non-steroidal anti-inflammatory drugs (NSAIDs)


The inflammatory process may be defined as a sequence of events that occurs in response to noxious stimuli,
infection or trauma. The classic signs of inflammation are local redness, swelling, pain, heat and loss of function.
The events of inflammation that underline these manifestations are induced and regulated by a large number of
chemical mediators, including kinins, eicosanoids, complement proteins histamine and monokines. NSAIDs are
among the most commonly used drugs worldwide. They are prescribed for orthopedics conditions such as
osteoarthritis, soft-tissue injuries and fractures etc. NSAIDs are one of the best classes of drug to prevent and treat
postoperative pain. The greatest disadvantage in presently available potent synthetic drugs lies in their toxicity and
reappearance of symptoms after discontinuation. Therefore, the screening and development of drugs for their anti-
inflammatory activity is the need of hour and there are many efforts for finding anti-inflammatory drugs from
indigenous medicinal plants. The use of NSAIDs is associated with many side effects, but their unwanted effects on
the gastrointestinal tract, the kidney and the cardiovascular system are considered as major issues with the use of
these drugs. The mechanism of inflammation is attributed, to release of ROS from activated neutrophils and
macrophages. ROS over production results tissue injury by damaging macromolecules and lipid per oxidation of
membranes. In addition, it propagate inflammation by stimulating release of cytokines such as IL-1, TNF- α and
interferon-γ which are responsible for the recruitment of additional neutrophils and macrophages. Thus free radicals
are important mediators that provoke or sustain inflammatory responses and their neutralization by antioxidants and
radical scavengers can reduce inflammation (Filomena Conforti, 2008).

563 | International Journal of Current Trends in Pharmaceutical Research


Y. Bhagyasri et al, IJCTPR, 2014, Vol.2(4): 562-565

2. Anti-Inflammatory Activity
Carrageenan-induced rat paw edema
Four group of six rat were taken. Group I (control) was treated with carboxymethylcellulose (10ml/kg) which is
used as a vehicle. Group II (Standard) was treated with 10mg/kg of Indomethacin by oral route. Group III and Group
IV (test) was treated with 200 mg/kg and 100mg/kg of Plant extract by oral route. After 30 mins of test drug
administration, 0.1% of carrageenan (0.1ml) was administered into the sub plantar tissue of right hind paw (Raji
Y,2002). After a duration of 30min, 1 hr, 2hr, 3hr and 4hr, the paw volume was measured by the digital
plethylsmograph. The percentage of edema inhibition was calculated by the formula.

3. Anti-Oxidant Activity
As plants produce a lot of antioxidants to control the oxidative stress caused by sunbeams and oxygen, they can
represent a source of new compounds with antioxidant activity. Ayurveda, the Indian traditional health care system
(ayus- Life, veda-knowledge, meaning science of life), is the oldest medical system in the world and is being revived
in its complete form under the name of Maharishi Ayurved (Glaser, 1988). These preparations contain a wide
number of plants. Among the different plants, seven of them have been specifically investigated for their well-
demonstrated antioxidant activity:
a. Emblica officinalis L.
b. Curcuma longa L.
c. Mangifera indica L.
d. Momordica charantia L.
e. Santalum album L.
f. Swertia chirata Buch-Ham
g. Withania somnifera L.
h. Abutilaan Indicum L.

Antioxidant Activity
DPPH Free Radical Scavenging Method
A stock solution of 100μg/ml was prepared of Plant extracts as well as of standard ascorbic acid. Different
concentrations were made of 10, 20, 30, 40, 50 and 100 μg/ml from stock solutions using methanol and 0.1mM
solution of DPPH in methanol was prepared in a volumetric flask which was completely kept away from light. Then
1ml of all concentration of test and standards were mixed with 1ml of DPPH solution. This solution was kept for 30
min in dark. Only methanol with DPPH was used as control. Absorbance of all the samples was taken on UV
spectrophotometer.
(% Inhibition = [{Acont - Atest/std} / Acont] × 100)
Atest/std = Absorbance of test or standard
Acont = Absorbance of control

IC50 values were calculated from linear regression by plotting the graph between concentration and % inhibitions
(Bandgar, B.P, 2009). IC50 resembles that it was that concentration of the sample which is required to scavenge
50% of DPPH free radicals.

4. Conclusion
Many studies have been performed to identify antioxidant and anti-inflammatory compounds with
pharmacologically activity and a limited toxicity. In this context, ethnopharmacology represents the most important
way possible of finding interesting and therapeutically helpful molecules. The Phytochemical analysis of extract has
revealed a large number of compounds including tannic acid, flavonoids, terpenoids, carotenoids, glycosides,
alkaloids, polyphenols, etc. which have been shown to have potent antioxidant properties. The herbal mixture
preparations of Indian traditional medicine may have an antioxidant activity arising from their content of plants with
antioxidant and anti-inflammatory activity principles, that act probably in a synergistic way.

5. References
1. Abraham, A., Constituents of Withania somnifera Dun-XIII: the withanolides of chemotype III.
Tetrahedron, 1973, 29: 1353–1364.
2. Agarwal, K., Dhir, H., Sharma, A., Talukder, G., The efficacy of two species of Phyllanthus in
counteracting nickel clastogenicity. Fitoterapia , 1992, 1 63 (1): 49–54.
3. Akthar, M.S., Athar, M.A., Yaqub, M., Effect of Momordica charantia on blood glucose level of normal
and alloxan-diabetic rabbits. Planta Medica, 1981, 42: 205–212.

564 | International Journal of Current Trends in Pharmaceutical Research


Y. Bhagyasri et al, IJCTPR, 2014, Vol.2(4): 562-565

4. Al-Hindawi, M.K., Al-Khafaji, S.H., Abdul-Nabi, M.H.,Anti-granuloma activity of Iraqui Withania


somnifera.Journal of Ethnopharmacology, 1992,37: 113–116.
5. Ammon, H.P.T., Wahl, M.A., Pharmacology of Curcuma longa. Planta Medica, 1991, 57: 17.
6. Aslam, M., Stockley, I.H.,. Interaction between curry ingredient (Karela) and drug (Chlorpropamide). The
Lancet, 1979, 17: 607.
7. Asmawi, M.Z., Kankaanranta, H., Moilanen, E., Vapaatalo, H., Anti-inflammatory activities of Emblica
officinalis Gaertn leaf extracts. Journal Pharmacy and Pharmacology, 1993, 45: 581–584.
8. Azuine, M.A., Kayal, J.J., Bhide, S.V., Protective role of aqueous turmeric extract agaist mutagenicity of
direct-acting carcinogens as well as benzo(a)pyrene-induced genotoxicity and carcinogenicity. Journal of
Cancer Reserch and Clinical Oncology, 1992, 118: 447–452.
9. Ba¨hr, V., Ha¨nsel, R., Immunomodulating properties of 5,20a(R)dihydroxy-6a,7a-epoxy-1-oxo-(5a)-witha-
2,24-dienolide and solasodine. Planta Medica, 1982, 44: 32–33.
10. Banerjee, S., Ecavade, A., Rao, A.R., Modulatory influence of sandalwood oil on mouse hepatic
glutathione-Stransferase activity and acid soluble sulphydryl level. Cancer Letters, 1993, 68: 105–109.
11. Barthakur, N., Arnold, N.P., Chemical analysis of the emblic (Phyllanthus emblica L.) and its potential as a
food source. Scientific Horticulture, 1991, 47 (1–2): 99–105.
12. Basa, S.C., Srinivasulu, C, Costituents of leaves of Phyllanthus emblica L. Chemical Abstract, 1988, 108:
128-517.
13. Blasdell, K.S., Sharma, H.M., Tomlinson, P.F. Jr, Wallace, R.K., Subjective survey, blood chemistry and
complete blood profile of subjects taking Maharishi Amrit Kalash (MAK). Faseb Journal, 1991, 5: A1317.
14. Sakat SS, Juvekar AR And Gambhire MN, InvitroAntioxidant And Antiinflammatoryactivity Of Methanol
Extract of Oxalis Corniculata Linn, International Journal of Pharmacy and Pharmaceutical Sciences, 2010,
2(1): 146-155.
15. Nagore DH, Ghosh VK, Patil MJ, Wahile AM, In Vitro Antioxidant And In Vivo anti-inflammatory
Activity of Cassia sophera Linn, International Journal Of Pharmacy And Pharmaceutical Sciences, 2(1), ,
114-121.
16. Filomena Conforti , Silvio Sosa , Mariangela Marrelli.et al., In vivo anti-inflammatory and in vitro
antioxidant activities of Mediterranean dietary plants. Journal of Ethnopharmacology, 2008, 116: 144–151.
17. Urmilesh Jha, Prites J. Chhajed, Rajesh J. Oswal.et al.: Skeletal muscle relaxant activity of methanolic
extract of Parthenium hysterophorus L. leaves in swiss albino mice. International Journal of Pharmacy and
life science, 2011, 2: 1211-1213
18. Raji Y , Udoh U.S, Oluwadara O. et al : Anti-inflammatory And Analgesic Properties Of The Rhizome
Extract Of Zingiber Officinale African Journal of Biomedicine, 2002, 5: 121 – 124
19. Aguilar AL, Rojas P, Marcelo A, Plaza A, Bauer R, Reininger E, Klaas CA, Merfort I. ‘‘Antiinflammatory
activity of two different extracts of Uncaria tomentosa (Rubiaceae)’’. Journal of Ethnopharmacology,
2002, 81: 271-276.

565 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

ISSN: 2321-3760
Review Article
International Journal of Current Trends in
Pharmaceutical Research
IJCTPR, 2014, Vol. 2(4): 566-574
www.pharmaresearchlibrary.com/ijctpr

Transferomes: Emerging Trends for Drug Delivery


Krishna moorthy S.B*1, Sandeep. J2, Jyothirmaie. K3, Rajalakshmi.R1
1
Department of Pharmaceutics, Sree Vidyanikethan College of Pharmacy, A. Rangampet,
Tirupathi, Andhra Pradesh, India-517102.
2
Department of Pharmaceutics, Sri Padmavathi School of Pharmacy, Tiruchanoor, Tirupathi,
Andhra Pradesh, India-517503.
3
Department of Pharmaceutics, Jagan’s College of Pharmacy, Muthukur, Nellore,
Andhra Pradesh, India- 524002.
Received: 10 May 2014, Accepted: 29 June 2014, Published Online: 15 July 2014

Abstract
Transferosomes have become increasingly apparent that vesicular drug delivery elicits modest possessions
in drug targeting. Transfersomes are a form of elastic or deformable vesicle, which were first introduced in
the early 1990s. Elasticity can be achieved by using an edge activator in the lipid bilayer structure.
Transfersomes are applied in a non-occluded method to the skin and have been shown to permeate through
the stratum corneum lipid lamellar regions as a result of the hydration or osmotic force in the skin.
Transfersomes are made up of a phospholipids component along with a surfactant mixture. The ratio of
individual surfactants and total amount of surfactants control the flexibility of the vesicle. The uniqueness
of this type of drug carrier system lies in the fact that it can accommodate hydrophilic, lipophilic as well as
amphiphilic drugs. These drugs find place in different places in the elastic vesicle before they get delivered
beneath the skin. They can act as a carrier for low as well as high molecular weight drugs e.g. analgesic,
anesthetic, corticosteroids, sex hormone, anticancer, insulin, gap junction protein, and albumin. Peripheral
drug targeting, transdermal immunization can also be achieved with this type of drug delivery system.
Keywords: Transferosomes, stratum corneum, phospholipids, elasticity, transdermal.

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .567
2. Description. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .567
3. Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .572
4. References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . 572

*Corresponding author
Krishna moorthy S.B
Department of Pharmaceutics,
Sree Vidyanikethan College of Pharmacy,
Tirupathi, Andhra Pradesh, India-517102.
Manuscript ID: IJCTPR2161 PAPER-QR CODE

Copyright © 2013, IJCTPR All Rights Reserved

566 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

1. Introduction
In the last few years, the vesicular systems have been promoted as a mean of sustained or controlled release of
drugs. These vesicles are preferred over other formulations because of their specific characteristics such as lack of
toxicity, biodegradation, capacity of encapsulating both hydrophilic and lipophilic molecules, capacity of prolonging
the existence of the drug in the systemic circulation by encapsulation in vesicular structures, capacity of targeting
the organs and tissues, capacity of reducing the drug toxicity and increasing its bioavailability. Vesicular systems
such as transferosomes formulations have been used as drug delivery vehicles for sustained release of proteins and
peptides. These formulations have been used as carriers of cytotoxic drugs with the strategy based on reduction of
toxicity and passive delivery to tumors [1-6].

Transfersome is a term registered as a trademark by the German company IDEA AG, and used by it to refer to its
drug delivery technology. The name means “carrying body”, and is derived from the Latin word 'transferre',
meaning ‘to carry across’, and the Greek word ‘soma’, for a ‘body’. A Transfersome carrier is an artificial vesicle
designed to be like a cell vesicle or a cell engaged in exocytosis, and thus suitable for controlled and, potentially
targeted, drug delivery [7, 8].

Transfersomes (transfersomes) offer a versatile delivery concept for improving the stability as well as the potential
to be used with a wide range of active compounds. They are (quasi) metastable, which makes the vesicle membrane
ultra-flexible, and thus, the vesicles are highly deformable that squeeze through pores in the stratum corneum less
than one-tenth of their own diameter when applied under non-occlusive conditions. Thus, even sizes up to 200–300
nm can penetrate intact skin. It is primarily due to the remarkable strong membrane adaptability that allows the
Transfersomes vesicles to lodge in a confining pore, and thus permeate that pore [9-11].

2. Description
A. Mechanism of Penetration
Transferosomes when applied under suitable condition can transfer 0.1 mg of lipid per hour and cm 2 area across the
intact skin. This value is substantially higher than that which is typically driven by the transdermal concentration
gradients. The reason for this high flux rate is naturally occurring "transdermal osmotic gradients" i.e. another much
more prominent gradient is available across the skin.(12) This osmotic gradient is developed due to the skin
penetration barrier, prevents water loss through the skin and maintains a water activity difference in the viable part
of the epidermis (75% water content) and nearly completely dry stratum corneum, near to the skin surface (15%
water content).(13) The mechanism for penetration is the generation of “osmotic gradient” due to evaporation of
water while applying the lipid suspension (transfersomes) on the skin surface Consequently all lipid vesicles made
from the polar lipid vesicles move from the rather dry location to the sites with a sufficiently high water
concentration, So when lipid suspension (transfersomes) is placed on the skin surface, that is partly dehydrated by
the water evaporation loss and then the lipid vesicles feel this "osmotic gradient" and try to escape complete drying
by moving along this gradient [14, 15]. A Transfersomes vesicle applied on an open biological surface, such as non-
occluded skin, tends to penetrate its barrier and migrate into the water-rich deeper strata to secure its adequate
hydration. During penetration through the stratum corneum, reversible deformation of the bilayer occurs. But it
should be noted that while this deformation is occurring, vesicle integrity, gradient and barrier properties for the
underlying hydration affinity should not be compromised. Since it is too large to diffuse through the skin, the
Transfersome needs to find and enforce its own route through the organ.

Figure 1. Schematic diagram of the two micro routes of penetration

567 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

Transferosomes when placed on skin surface



Dehydrated by water evaporation loss

Lipid vesicle feels osmotic gradient

Move along this gradient, deform to pass to pass through pores in skin

Figure 2. Schematic diagram of the penetration mechanism.

A. Transcellular mechanism: In this pathway hair follicles and sweat ducts offers pores that by pass the
stratum corneum.
B. Intercellular mechanism: In this type of route drug directly goes to the systemic circulation via lipid matrix
between keratocytes.
C.
Intracellular mechanism: In this route drug molecule penetrates directly across the stratum corneum by
diffusion method. Stratum corneum is the main barrier for drug molecules from transdermal drug delivery
system. These are three main pathways for drug molecules to penetrate stratum corneum. Hydrophilic
drugs permeate by Intercellular pathway and lipophilic drugs permeate by Intracellular (Transcellular)
mechanism [16].

Pathways of drug penetration [17,18]

Figure 3. Drug penetration pathways.

1. Through stratum corneum , 2. Transfollicular, 3. Through sweat gland

B. Structure of A Transferosome
Transfersomes possess an infrastructure consisting of hydrophobic and hydrophilic moieties together and as a result
can accommodate drug molecules with wide range of solubility Vesicles are water-filled colloidal particles. The
walls of these capsules consist of amphiphilic molecules (lipids and surfactants) in a bilayer conformation. In an
excess of water these amphiphilic molecules can form one (unilamellar vesicles) or more (multilamellar vesicles)
concentric bilayers. Hydrophilic drugs can be entrapped into the internal aqueous compartment, whereas
amphiphilic, lipophilic and charged hydrophilic drugs can be associated with the vesicle bilayer by hydrophobic
and/or electrostatic interactions [19].

568 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

Figure 4. Structure of trasferosomes

C. Preparation of Transferosomes
Thin film hydration technique
1. A thin film is prepared from the mixture of vesicles forming ingredients that is phospholipids and
surfactant by dissolving in volatile organic solvent (chloroform, methanol). Organic solvent is then
evaporated above the lipid transition temperature (room temp. for pure PC vesicles, or 50°C for
dipalmitoyl, phosphatidyl choline) using rotary evaporator. Final traces of solvent were removed under
vacuum for overnight.
2. A prepared thin film is hydrated with buffer (pH 6.5) by rotation at 60 rpm for 1 hr at the corresponding
temperature. The resulting vesicles were swollen for 2 hr at room temperature.
3. To prepare small vesicles, resulting vesicles were sonicated at room temperature or 50°C for 30 min. using
a bath sonicator or probe sonicated at 4°C for 30 min. The sonicated vesicles were homogenized by manual
extrusion 10 times through a sandwich of 200 and 100 nm polycarbonate membranes.

D. Modified hand shaking, lipid film hydration technique


1. Drug, lecithin (PC) and edge activator were dissolved in ethanol: chloroform (1:1) mixture. Organic solvent was
removed by evaporation while hand shaking above lipid transition temperature (43°C). A thin lipid film was formed
inside the flask wall with rotation. The thin film was kept overnight for complete evaporation of solvent.
2. The film was then hydrated with phosphate buffer (pH 7.4) with gentle shaking for 15 minute at corresponding
temperature. The transfersome suspension further hydrated up to 1 hour at 2-8°C [20-23]

E. Optimization of Formulation Containing Transfersomes


There are various process variables which could affect the preparation and properties of the Transfersomes. The
preparation procedure was accordingly optimized and validated. The process variables are depending upon the
procedure involved for manufacturing of formulation. The preparation of transfersomes involves various process
variables such as,
a. Lecithin: surfactant ratio
b. Effect of various solvents
c. Effect of various surfactants
d. Hydration medium
Optimization was done by selecting entrapment efficiency of drug. During the preparation of a particular system, the
other variables were kept constant.[24, 25].

Table 1. Different Additives Used in Formulation of Transfersome.


Class Example Uses
Soya phosphatidyl Choline, Dipalmitoyl Vesicles
Phospholipids Phosphatidyl choline, Distearoyl phoshatidyl forming
choline. component
Sod. Cholate, Sod.deoxycholate Providing
Surfactant
tween-80 Span-80 flexibility
Alcohol Ethanol solvent
Rhodamine-123, Rhodamine-DHPE, CSLM
Dye
Fluorescein-DHPE Nilered study
Buffering Saline phosphate buffer (pH 6.4) hydrating
Agent medium

569 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

F. Characterisation of Transferosomes
I. Vesicle size, size distribution and vesicle Diameter [26]
Transfersomes can be visualized by transmission electron microscopy (TEM) and vesicle size and size distribution
can be determined by dynamic light scattering (DLS) technique. Samples were prepared in distilled water, filtered
through a 0.2 mm membrane filter and diluted with filtered saline and then size measurement done by using photon
correlation spectroscopy or dynamic light scattering measurements.
II. Vesicle shape and type
Transfersomes vesicles can be visualized by using TEM, with an accelerating voltage of 100 kv. These vesicles can
be visualized without sonication by phase contrast microscopy by using an optical microscope. Dynamic light
scattering is also used for determining vesicle shape.
III. Number of vesicle per cubic mm [27]
This is an essential parameter for optimizing the composition and other process variables. Transfersome
formulations (without sonication) can be diluted five times with 0.9% of sodium chloride solution and studied with
optical microscopy by using haemocytometer.

total number of transferosomes counted × dilution factor


Total number of vesicles per cubic mm = × 4000
total number of squares counted
IV. Degree of Deformability or Permeability Measurement
Degree of deformability is an important and unique parameter of transfersomal formulations because it differentiates
transfersomes from other vesicular carriers like liposomes that are unable to cross the stratum corneum intact. The
deformability study is done against the pure water as standard. Transfersomes preparation is passed through a large
number of pores of known size (through a sandwich of different microporous filters, with pore diameter between 50
nm and 400 nm, depending on the starting transfersomes suspension). Particle size and size distributions are noted
after each pass by dynamic light scattering (DLS) measurements. The degree of deformability was calculated by
using the following formula, as reported by Berge vanden et al. [28]
D=J×(rv/rp)2
where,
D = deformability of vesicle where,
D = deformability of vesicle membrane,
J = amount of suspension, which was extruded during 5 min
rv = size of vesicles (after passes) and
rp = pore size of the barrier.
V. Propensity of penetration
The magnitude of the transport driving force plays an important role:
Flow = Area x (Barrier) Permeability x (Trans-barrier) force

Therefore, the chemically driven lipid flow across the skin always decreases dramatically when lipid solution is
replaced by the same amount of lipids in a suspension.
VI. Confocal Scanning Laser Microscopy (CSLM) study
Conventional light microscopy and electron microscopy both face problem of fixation, sectioning and staining of the
skin samples. Often the structures to be examined are actually incompatible with the corresponding processing
techniques; these give rise to misinterpretation, but can be minimized by Confocal Scanning Laser Microscopy
(CSLM). In this technique lipophilic fluorescence markers are incorporated into the transfersomes and the light
emitted by these markers used for following purpose:
1. For investigating the mechanism of penetration of transfersomes across the skin,
2. For determining histological organization of the skin (epidermal columns, interdigitation), shapes and architecture
of the skin penetration pathways. For comparison and differentiation of the mechanism of penetration of
transfersomes with liposomes, niosomes and micelles. Different fluorescence markers used in CSLM study are
a. Fluorescein-DHPE(1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine-N-(5-fluores
denthiocarbamoyl), triethylammonium salt)
b. Rhodamine-DHPE (1,2- dihexadecanoyl-sn-glycero- 3ogisogietgabikanube-N-LissamineTmrhodamine B
sulfonyl), triethanolaminesalt)
c. NBD-PE (1, 2-dihexadecanoylsn- glycero-3- phosphoethanolamine-N-(7-nitro-Benz-2- oxa-1, 3-diazol- 4-
yl) triethanolamine salt)
d. Nile red
VII. Drug Content
The drug content can be determined using a modified high performance liquid chromatography method (HPLC)
method using a UV detector, column oven, auto sample, pump, and computerized analysis program.

570 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

VIII. Entrapment efficiency


Entrapment efficiency was determined by first separation of un-entrapped drug by the use of minicolumn
centrifugation method. After centrifugation, the vesicle was disrupted using 0.1% Triton X-100 or 50% n-propanol
and then followed by suitable analytical technique to determine the entrapped drugs.
IX. In vitro Drug Release (29, 30)
In vitro drug release study is performed for determining the permeation rate. Time needed to attain steady state
permeation and the permeation flux at steady state and the information from in-vitro studies are used to optimize the
formulation before more expensive in vivo studies are performed. For determining drug release, transfersomes
suspension is incubated at 32°C and samples are taken at different times and the free drug is separated by mini
column centrifugation. The amount of drug released is then calculated indirectly from the amount of drug entrapped
at zero times as the initial amount (100% entrapped and 0% released).
X. Surface charge and charge density, Turbidity measurement:
Surface charge and charge density of transferosomes can be determined using zetasizer. Turbidity of drug in
aqueous solution can be measured using nephelometer.
XI. IN-VIVO
1. Tape stripping method
Tape stripping technique is a widely accepted and it is used to examine the localization and distribution of
substances within the stratum corneum. It is the simplest method for reducing the barrier imposed by the stratum
corneum is to remove it. In this technique, an adhesive tape removes a layer of corneocytes. In vivo, removal of the
stratum corneum by tape stripping is performed by the repeated application of adhesive tapes to the skin’s surface.
This can be used to investigate stratum corneum cohesion in vivo by quantifying the amount of stratum corneum
removed. Tape stripping method can be studied in combination with electron microscopy and FT-IR. There are
different parameters that can affect the quantity of stratum corneum removed by a piece of tape, and these include
tape stripping mode, skin hydration, cohesion between cells, the body site and inter-individual differences.
2. Occlusion Effect
Occlusion of skin is considered to be helpful for permeation of drug in case of traditional topical preparations. But
the same proves to be detrimental for elastic vesicles. Hydrotaxis (movement in the direction) of water is the major
driving force for permeation of vesicles through the skin, from its relatively dry surface to water rich deeper regions.
(28)
Occlusion affects hydration forces as it prevents evaporation of water from skin.
XII. Stability Studies
Transfersomes stability was determined at 4°C and 37°C by TEM visualization and DLS size Measurement at
different time intervals (30, 45, and 60 days), following vesicles preparation.

G. Limitations of Transfersomes
a. Like liposomes, transfersomes have certain limitations:
b. Transfersomes are chemically unstable because of their predisposition to oxidative degradation.
c. Lack of purity of the natural phospholipids comes in the way of adoption of transfersomes as drug delivery
vehicles.
d. Transfersomes formulations are expensive to prepare.

H. Application of Transfersomes
1. Delivery of insulin [31]
Transfersomes is the successful means of non invasive therapeutic use of such large molecular weight drugs on the
skin. Transfersulin application on the intact skin, the first sign of systemic hypoglycaemia are observed after 90 to
180 min, depending on the specific carrier composition.
2. Delivery of proteins and peptides [32, 33]
The transfersomal preparations of protein induced strong immune response after the repeated
epicutaneous application, for example the adjuvant immunogenic bovine serum albumin in transfersomes, after
several dermal challenges is as active immunologically as is the corresponding injected proteo-transfersomes
preparations.
3. Delivery of steroidal hormones and peptides [34]
Transfersomes based cortiosteroids are biologically active at dose several times lower than the currently used
formulation for the treatment of skin diseases. Flexible vesicles of ethinylestradiol showed significant anti-ovulatory
effects as compared to plain drug given orally and traditional liposomes given topically. Extensive work has been
done on other drugs like hormones and peptides viz Estradiol, low molecular-weight Heparin, Retinol, Melatonin.
4. Delivery of Anticancer Drugs [35]
Anti cancer drugs like methotrexate were tried for transdermal delivery using transfersome technology for treatment
of skin cancer.
5. Delivery of Herbal Drugs [36]
571 | International Journal of Current Trends in Pharmaceutical Research
Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

Transfersomes of Capsaicin has been prepared by Xiao-Ying et al. which shows the better topical absorption in
comparison to pure capsaicin.
6. Delivery of NSAIDS [37]
Ketoprofen in a Transfersome formulation gained marketing approval by the Swiss regulatory agency (SwissMedic)
in 2007; the product is expected to be marketed under the trademark Diractin.
7. Delivery of Anesthetics
Transfersome based formulations of local anesthetics- lidocaine and tetracaine showed permeation equivalent to
subcutaneous injections.

I. Future Aspects [38]


This relates to potential use of self-regulating, ultradeformable carriers in devices (patches; electrically controlled
epicutaneous reserviors), and in design of formulation with additional special features, allowing, e.g., targeting of
cellular subsets. The nearest term goal that remains to be reached is expansion of the positive experiences with
NSAID targeting into peripheral tissues to other drugs with similar therapeutic demands.

Table 2. List of drugs used for transfersomes


S.No Drug Inference
1 Oestradiol Improved transdermal flux
2 Norgesterol Improved transdermal flux
Biologically active at dose several times lower than currently
3 Hydrocortosone
used formulation.
Antibody titter is similar or even slightly higher than
4 Human serum albumin
subcutaneous injection.
Controlled release, Overcome stability problem.
5 Interferon-α
High encapsulation efficiency.
Transfer across the skin with an efficiency of >50%.
6 Insulin
Provide non-invasive means of therapeutic use

3. Conclusion
The transdermal route of drug administration has been a route of choice since ancient times because of its merits.
But it itself limits its use as it is unable to transport larger molecules, penetration through the stratum corneum is the
rate limiting step, physicochemical properties of drugs hinder their own transport through skin. The development of
novel approaches like transferosomes have immensely contributed in overcoming these problems. These elastic
vesicles can squeeze themselves through skin pores many times smaller than their own size and can transport larger
molecules. Since a properly designed ultradeformable may even claim the transport of drug equivalent to the
subcutaneous injection, this technology can provide effective tool for non-invasive therapy When tested in artificial
systems transfersomes can pass through even tiny pores (100 mm) nearly as efficiently as water, which is 1500 times
smaller This carrier system does not depend upon the concentration gradient and mainly works on the principle of
hydrotaxis and elasto-mechanics.

Ultra deformable vesicles hold great prospective in delivery of huge range of drug substances which includes large
molecules like peptides, hormones and antibiotics, drugs with poor penetration due to unfavorable physicochemical
characters, drugs for quicker and targeted action, etc.. The bio-distribution of radioactively labelled phospholipids
applied in the form of transfersomes after 24 h is essentially the same after an epicutaneous application or
subcutaneous injection of the preparations. The use of transfersomes carrier result in delivery of high concentration
of active agents to/through the skin, regulated by system composition and their physical characteristics.

4. References
1. Arulsudar N., Subramanian N., Mishra P., Chuttani K., Sharma R.K., Murthy R.S.R., 2004. Preparation,
characterization, and biodistribution study of Technetium-99m– labeled leuprolide acetate-loaded
liposomes in ehrlich ascites tumor-bearing mice. AAPS Pharm Sci. 6 (1).
2. Bhatia A., Kumar R., Tamoxifen in topical liposomes: development, characterization and in vitro
evaluation. J. Pharm. Sci., 2004, 7 (2): 252-259.
3. Petit-Frère C., Clingen P.H., Grewe M., Krutmann J., Roza L., Arlett C.F., Induction of interleukin-6
production by ultraviolet radiation in normal human epidermal keratinocytes and in a human keratinocyte
cell line is mediated by DNA damage. J. Invest. Dermatol., 1998, 111 (3): 354-359.
4. Jain N.K., Advances in controlled and novel drug delivery. CBS Publ. Distrib. New Delhi, 2001, pp. 426-
451.

572 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

5. Popovici A., Lipozomii – vectori medicamentosi. Farmacia, 1986, 34 (1): 1-22.


6. Porfi re A.S., Parvu A.E., Daicoviciu D., Leucuţa S.E., Evaluation of antiinfl amatory activity of liposome
encapsulated superoxide dismutase in rats peritonitis. Farmacia, 2009, 57 (4): 412-423.
7. Jain.N.K, Controlled and novel drug delivery, first edition, CBS publishers and distributors, New
Delhi.1997
8. Vyas SP, Khar RK. Controlled drug delivery system. Pitampura, Delhi, Vallabh prakashan, 2005, pp. 417-
426
9. Vijaya R, Ruckmani K. In vitro and In vivo characterization of the transdermal delivery of sertraline
hydrochloride Films. Daru. 2011, 19: 424–432.
10. Mishra D, Garg M, Dubey V, Jain S, Jain NK. Elastic liposomes mediated transdermal delivery of an anti-
hypertensive agent: Propranolol hydrochloride. J Pharm Sci., 2007, 96: 145–155.
11. Garg T, Jain S, Singh HP, Sharma A, Tiwary AK. Elastic liposomal formulation for sustained delivery of
antimigraine drug: in vitro characterization and biological evaluation. Drug Dev Ind Pharm., 2008, 34:
1100–1110.
12. Cevc G. Isothermal Lipid Phase. Transitions Chemistry and Physics of Lipids., 1991, 57: 293-299.
13. Schatzlein A et al, Cevc G. Skin Penetration by Phospholipids Vesicles, Transfersomes as visualized by
means of the Confocal Scanning Laser Microscopy, in characterization, Metabolisim, and Novel Biological
Applications. Champaign. AOCS Press., 1995, pp. 191-209.
14. Cevc G et al, Blume G, Sehatzlein A, Gebauer D andPaul A. The skin a Pathway for Systemic Treatment
with Patches and Lipid-Based Agent Carriers Advance Drug Delivery Reviews, 1996, 18: 349- 378
15. Chapman SJ et al, Walsh A. Arch. Dermatol Res., 1998, 282: 304-320.
16. chein YW, Chi SL. Transdermal Drug Delivery System with Enhanced skin Permeability. In: Ping IL and
William RG (eds). Controlled Released Technology- Pharmaceutical Application. Washington DC:
American Chemical Society, 1987, pp. 81-299.
17. Schatzlein A and Cevc G: Non-uniform cellular packing of the stratum corneum and permeability barrier
function of intact skin: a high-resolution confocal laser scanning microscopy study using highly deformable
vesicles (Transfersomes). British Journal of Dermatology, 1998, 138: 583-98.
18. Chapman SJ and Walsh A: Arch. Dermatol Res, 1998, 282: 304-20.
19. Hofl and H.E.J., Bouwstra J.A., Spies F., Gooris G., Nagelkerke J.F., Interaction of liposomes and
niosomes with human skin. J. Pharm. Sci., 1994, 83: 1192-1196.
20. Modi CD, Bharadia PD, “Transfersomes: New Dominants for Transdermal Drug Delivery”, Am. J.
PharmTech Res., 2012, 2 (3): 71-91.
21. Pandey S, Goyani M, Devmurari V, Fakir J, “Transferosomes: A Novel Approach for Transdermal Drug
Delivery”, Der Pharmacia Letter, 2009, 1 (2): 143-150.
22. Jain CP, Vyas SP, Dixit VK, “Niosomal system for delivery of rifampicin to lymphatics”, Int J Pharma,
2006, 68: 5758.
23. Elsayed MMS, Abdallah OY, Nagar VF. Deformable liposomes and ethosomes: Mechanism of enhanced
skin delivery. Int J Pharma, 2006, 322: 60-66.
24. Patel R, Singh SK, Singh S, Sheth NR, Gendle R,“Development and Characterization of Curcumin Loaded
Transfersome for Transdermal Delivery” J.Pharm Sci. Res., 2009, 1 (4): 71-80.
25. Sheo DM, Shweta A, Vijay KT, Ram CD, Aklavya S,Ghanshyam M, “Enhanced Transdermal delivery of
indinavir sulfate via transfersomes”, Pharmacie Globale (IJCP), 2010, 1 (6): 1-7.
26. Gamal M., Maghraby M., Williams A.C., Barry B.W., Gamal M., Maghraby M., Williams A.C., Barry
B.W., Skin delivery of Oestradiol from deformable and traditional liposomes. J. Pharm. Pharmacol, 1999,
51: 1123-1134.
27. Berge vanden BAI and Wertz PW, Junginger HE and Bouwstra JA. Elasticity of Vesicles Assessed by
Electron Spin Resonance. Electron Microscopy and Extrusion Measurement. Int J Pharm., 2001, pp.
217:13–24.
28. Berge vanden BAI and Wertz PW, Junginger HE and Bouwstra JA. Elasticity of Vesicles Assessed by
Electron Spin Resonance. Electron Microscopy and Extrusion Measurement. Int J Pharm., 2001, 217: 13-
24.
29. Fry DW, White JC and Goldman ID. Rapid Separation of Low Molecular Weight Solutes from Liposomes
without Dilution. Journal of Analytical Biochemistry., 1978, 90: 809-815.
30. Garg M., Mishra D., Agashe H., and Jain N.K.: Ethinylestradiol loaded ultraflexible liposomes:
Pharmacokinetics and pharmacodynamics. Journal of Pharmacy and Pharmacology, 2006, 58: 459-468.
31. Gregor C, Dieter G, Juliane S, Andreas S, Gabriele B, “Ultra-flexible vesicles, Transfersomes, have an
extremely low pore penetration resistance and transport therapeutic amounts of insulin across the intact
mammalian skin”, Biophysica Acta, 1998, 1368: 201-215.

573 | International Journal of Current Trends in Pharmaceutical Research


Krishna moorthy S.B et al, IJCTPR, 2014, Vol.2(4): 566-574

32. Maghraby EI, Williams GM, Barry BW, “Skin delivery of oestradiol from lipid vesicles: importance of
liposome structure”, Int. J. Pharma, 2000, 204 (1-2): 159-69.
33. Trotta M, Peira E, Carlotti ME, Gallarate M, “Deformable liposomes for dermal administration of
methotrexate”, Int. J. Pharma, 2004, 270: 119.
34. G. Cevc, G. Blume and A. Schatzlein., Journal of Controlled Release., 1997, 45, 211.
35. Modi CD, Bharadia PD, “Transfersomes: New Dominants for Transdermal Drug Delivery”, Am. J.
PharmTech Res., 2012, 2 (3): 71-91.
36. Benson HA, “Transfersomes for transdermal drug delivery”, Expert Opin. Drug Deliv., 2006, 3 (6), 727-37.
37. Dubey V, Mishra D, Asthana A, Jain NK, “Transdermal delivery of a pineal hormone: melatonin via elastic
liposomes”, Biomaterials, 2006, 27 (18): 3491-6.
38. G. Cevc., Modified-Release Drug Delivery Technology. 2nd edition. New York: Informa healthcare.,
2008: 311.

574 | International Journal of Current Trends in Pharmaceutical Research


Related Journals Published by Our Group

 International Journal of Chemistry and Pharmaceutical


Sciences

 International Journal of Current Trends in Pharmaceutical


Research

 International Journal of Medicine and Pharmaceutical


Research

 Journal of Pharmaceutical and Biomedical Analysis Letters

 Asian Journal of Chemical and Pharmaceutical Research

 International Journal of Research in Pharmacy and Life


Sciences

 Asian Journal of Medical and Pharmaceutical Sciences

 Journal of Pharmaceutical and Biological Research

 International Journal of Pharmacy and Natural Medicines

Publisher
PHARMA RESEARCH LIBRARY
(An International Scientific Publication Group)
Nellore City, A.P, India-524002

Vous aimerez peut-être aussi