Jitesh M-Pharm Thesis
Jitesh M-Pharm Thesis
Jitesh M-Pharm Thesis
EVALUATION OF ORODISPERSIBLE
TABLETS OF PROTON PUMP INHIBITOR
By
MASTER OF PHARMACY
IN
PHARMACEUTICS
Department of Pharmaceutics
Nargund College of Pharmacy
Bangalore
February-2011
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA, BANGALORE
DATE:
PLACE: BANGALORE Mr. Chaudhari Jitesh Jagdish
List of Abbreviations
µg : microgram
Abs : Absorbance
BP : British Pharmacopoeia
CI : Carr‘s Index
Cocn. : Concentration
cm : Centimeter
CP : Crospovidone
FC : Formulation code
Fig. : Figure
gm : Gram
H+ : Hydrogen ion
HR : Hausners Ratio
IP : Indian Pharmacopoeia
IR : Infra-red
K+ : Potassium ions
M : Moles
mins : Minutes
ml : millilitre
mm : millimetre
mg : Milligram
r2 : Correlation factor
RT : Room temperature
RH : Relative humidity
Sec. : Second
UV : Ultra Violet
ABSTRACT
Proton pump inhibitors are benzimidazole substitutions exhibiting potent and long-
lasting inhibition of gastric acid secretion by selective interaction with the gastric proton
Proton pump inhibitors have low bioavailability after oral administration, since they
undergo degradation very rapidly in the stomach followed by first- pass metabolism in the
liver. Thus attempts such as inclusion complex of proton pump inhibitors with
suppository as an alternative dosage form has also been tried; however they yielded
unsatisfactory results.
Most elderly patients, children, patients with dysphagia, have difficulty swallowing
conventional tablets or capsules. Mainly, proton pump inhibitors are available as enteric
coated pellets filled in capsules or compressed into tablets so instant relief is not achieved;
hence a need exists for a relatively fast acting orodispersible tablet form.
METHODS:
natural gums as superdisintegrants. They were then evaluated for pre and post compression
parameters.
RESULTS:
friability, taste evaluation, mouth feel and in vitro dissolution studies. More than 90% of
drug was released from almost all the formulations within 10 min. Further formulations
were subjected to accelerated stability studies for 90 days. Tablets showed no appreciable
performed the best. Crospovidone (3% and 5%) showed good similarity with marketed
preparation. Bicarbonate can be used as buffer for stabilizing the drugs which are degraded
in acidic pH.
KEYWORDS:
Subliming agents.
Table of Contents
CHAPTER PAGE
TABLE OF CONTENTS
NO. NO.
LIST OF TABLES
LIST OF FIGUERS
1 INTRODUCTION 1
1.1 History 1
1.5.4.1 Superdisintegrants 14
1.5.5 Mass-Extrusion 15
1.5.7 Sublimation 16
1.5.8 Nanonization 17
1.8.1 Chemistry 24
1.8.2 Stomach 25
1.10.8 Metabolism 34
2 OBJECTIVE 37
3 REVIEW OF LITERATURE 39
4 METHODOLOGY 56
4.2.2.1 Crospovidone 64
4.2.2.10 Menthol 73
4.2.2.11 Thymol 74
4.2.2.12 Camphor 75
4.2.2.15 Talc 78
4.3 Instruments 80
5 RESULT 103
6 DISCUSSION 126
7 CONCLUSION 139
8 SUMMARY 141
9. BIBLIOGRAPHY 143
4.2.2.10 Menthol 73
4.2.2.11 Thymol 74
4.2.2.12 Camphor 75
4.2.2.14 Aspartame 77
4.2.2.15 Talc 78
1.4 8
Common steps in tablet disintegration
1.4.1.2 10
Disintegration of tablet by wicking and swelling
1.4.1.6 11
Disintegration by deformation and repulsion
1.4.1.7 12
Release of gas during tablet disintegration
1.8.2.4 30
B- Conversion of Omeprazole to a sulfenamide in the acidic
secretory canaliculi of the Parietal cell
1.9 Mode of action of Proton pump Inhibitors 31
5.3.5.3 Cumulative percent drug release of OMEZ INSTA which was 123
taken as standard
5.3.5.4 Cumulative percent drug release of the best formulation F3, F4 124
and standard formulation
6.1.2 Solubility of drug in different medium 127
1. INTRODUCTION
1.1 HISTORY
Pharmaceutics is a branch of pharmacy which deals mainly with the formulation and
indirectly to the growth of medical science. It is rightly said by Isaac Newton that ‘If you
have to see further it’s only by climbing on the shoulder of a giant’; in the recent past
unbelievable advances in the field of medical sciences have occurred, most of which
some might have not been thought of a decade back. This was made possible by some
Medicine is an art that has been practiced since time immemorial whose exact
start cannot be predicted as its history is routed right from the development of human
civilization and is continued till now. The use of herbs and natural medicaments to
relieve pain or to aid the sick in coping with their afflictions has been a part of all
societies. In the Western world, medicine has developed since the time of the Greeks and
Romans. The Hippocratic Oath reminds us of this nearly 2500-year history. Much prior
to the modern day allopathic medicine, others systems like the oldest documented system
of medicine- Ayurveda as well as Unani, and Siddha existed. However, the progress of
medicine has been very different from that of many other arts within society. It has come
successful treatment for a human ailment, medicine is very much a development of the
last 100–150 years. Indeed, the major advances have come in the last 50–75 years.1
thought to the difference between a white powder and a tablet, and think that ―a drug is a
drug is a drug‖. This huge presumption is doubtless, because we do not anymore make
pharmaceutical formulation ourselves, and precious few of us have ever understood that
drug‖ because when administered to a human being, in the general case, it contains
The oral route of drug administration is the most important and convenient
method of administering drug. It is probable that at least 90% of all the drugs used to
produce systemic effect are administered by the oral route. When a new drug is
discovered the pharmaceutical company makes every effort to ensure that the drug can be
by oral route, and should a more complex parentral route be the only alternative, then the
self administration of drugs cannot be achieved, the sales of the drug may constitute only
Tablets and capsules have emerged as the most popular solid oral dosage forms
used today. This includes conventional and controlled-release tablets as well as hard and
soft gelatin capsules.2 Tablet formulation and design may be described as the process
whereby the formulator ensures that the correct amount of the active drug in the right
form is delivered at or over the proper time at the proper rate and in the desired location,
Fig. No. 1.2 Different types of oral drug delivery systems (ODDs)
and elixirs are usually designed to contain one dose of medication in 5 to 30 ml. Such
dosage measurements are typically contributing an error ranging from 20 to 50% when
the drug is self-administered by the patient. However, tablets and capsules have the
advantage of being unit dosage forms in which one usual dose of the drug has been
However most elderly patients, children, and patients with dysphagia have
difficulty in swallowing conventional tablets and hard gelatin capsules, and therefore do
not take medication as prescribed by physicians. It is estimated that 35% of the general
hospitalized for acute neuromuscular disorders and head injuries have dysphagia. The
(AIDS). Thyroid surgery, radiation therapy to head and neck or oral cavity, and other
swallowing tablets and identified the reasons for this difficulty. More than 26 percent of
patients reported problems in swallowing tablets. Prominent complaints were size of the
tablet, surface of the tablet, forms of tablets, taste of the tablets. Twice as many women as
experienced the greatest difficulty in swallowing tablets as well as people who are ill and
supine in bed and those patients who are busy travelling without having asses to water
pertinent to their performance. Recent advances in novel drug delivery (NDDS) aim to
enhance safety and efficacy of drug molecule by formulation and to achieve better patient
compliance. One such approach is orally disintegrating tablets (OrDTs) which has gained
Products of ODT technologies entered the market in the 1980s and have grown
steadily in demand, and their product pipelines are rapidly expanding. New ODT
technologies address many pharmaceutical and patients needs, ranging from enhanced
patients with dysphagia. The mouth dissolving tablets (MDT) or ODTs by overcoming
disperse in saliva within few seconds.5 USFDA has defined OrDTs tablet as ―A solid
dosage form containing medicinal substances, which disintegrate rapidly, usually within a
Recently European Pharmacopoeia also adopted the term ―ODT‖ as a tablet that is
intended to be placed in the mouth where it disperses rapidly before swallowing. These
Fast dissolve, quick dissolve, rapid melt, quick disintegrating, mouth dissolving,
orally disintegrating, orodispersible, melt-in-mouth, tablets etc are some of the terms
which are used to refer to this unique form of drug delivery, which has many advantages
manufacture. The necessary property of such tablets is the ability to disintegrate rapidly
and disperse or dissolve in saliva, thereby obviating the need for water. Various
technologies have been developed that enable ODT to perform this unique function.
1) Ease of administration to the patient who cannot swallow, and patient who refuse
3) Rapid dissolution and absorption of the drug, which will produce quick onset of
action.
5) Have sufficient strength to withstand the rigors of the manufacturing process and
temperature.
machinery.
8) Good mouths feel to change the perception of medication as bitter pill particularly
in paediatric patients.
4) Stability for longer duration of time, since the drug remains in solid dosage form
till it is consumed.
However it should be noted that high drug loading is possible in case of ODTs.
For a drug to be readily absorbable by the body, it has first to be in solution. For
most tablets, the first important step toward solution is breakdown of the tablet into
USP/NF. Before a tablet dissolves, it has to disintegrate first, unless the tablet is
designed for quick surface erosion. The materials used as disintegrants include starches,
agar, amylose, cellulose and its derivatives, gum and its derivatives, gelatin, resins, and
silicone compounds.
Research has established that one should not automatically expect a correlation
between disintegration and dissolution. However, since the dissolution of a drug from the
fragmented tablet appears to control partially or completely the appearance of the drug in
the blood, disintegration is still used as a guide to the formulator in the preparation of an
uniformity.2
In the tablet disintegration process, several factors may affect the disintegration.
They include the rate of water absorption, porosity of the tablet, processing parameters,
and effect of active ingredients, surfactants, binders, and lubricants. Fast disintegration
always requires fast absorption of water into the centre of the tablet. Thus, having open
pore structures inside the tablets is very important for making fast dissolving tablets.6
The tablet breaks to primary particles by one or more of the mechanisms described
below:-
Disintegration by capillary action is always the first step. When tablets are placed
in suitable aqueous medium, the medium penetrates into the tablet and replaces the air
adsorbed on the particles, which weakens the intermolecular bond and breaks the tablet
into fine particles. Water uptake by tablet depends upon hydrophilicity of the drug
of porous structure and low interfacial tension towards aqueous fluid is necessary to help
generated due to capillary air expansion, which helps in disintegration of tablet. This
explanation, however, is limited to only a few types of disintegrants and cannot describe
repulsion theory based on the observation that nonswelling particle also cause
disintegration of tablets. The electric repulsive forces between particles are the
mechanism of disintegration and water is required for it. Researchers found that repulsion
is secondary to wicking.
Perhaps the most widely accepted general mechanism of action for tablet
disintegration is swelling Tablets with high porosity show poor disintegration due to lack
of adequate swelling force. On the other hand, sufficient swelling force is exerted in the
tablet with low porosity. It is worthwhile to note that if the packing fraction is very high,
fluid is unable to penetrate in the tablet and disintegration is again slows down.
binding action of binders some of the enzymes which act on the binders are listed in the
Table no. 1.4.1.5 List of enzymes and their use for the disintegration of tablets
Enzymes Binder
Amylase Starch
Protease Gelatin
Invertase Sucrose
Hess et al. had proved that during tablet compression, disintegrated particles get
deformed and these deformed particles get into their normal structure when they come in
contact with aqueous media or water. Occasionally, the swelling capacity of starch was
improved when granules were extensively deformed during compression. This increase in
size of the deformed particles produces a faster breakup of the tablet. This may be a
bicarbonate or carbonate with citric acid or tartaric acid. The tablet disintegrates due to
generation of pressure within the tablet. This effervescent mixture is used when it is
desired to formulate very rapidly dissolving tablets or fast disintegrating tablet. As these
disintegrants are highly sensitive to small changes in humidity level and temperature,
Freeze drying/ lyophilisation are the process in which water is sublimed from the
product after it is frozen. This technique creates an amorphous porous structure that can
dissolve rapidly. Briefly, the procedure involves dispersion of the active drug and
packs. The trays holding the blister packs are then passed through liquid nitrogen freezing
tunnel to freeze the drug solution or dispersion. The frozen blister packs are next placed
foil backing is applied on a blister-sealing machine. Finally the blisters are packaged and
shipped. The freeze-drying technique has demonstrated improved absorption and increase
expensive and time consuming; fragility makes conventional packaging unsuitable for
Moulding process is of two kinds; solvent method and heat method. Solvent
method involves moistening the powder blend with a hydro alcoholic solvent followed by
moulding). The solvent is then removed by air-drying. The tablets manufactured in this
manner are less compact than compressed tablets and posses a porous structure that
hastens dissolution. The heat moulding process involves preparation of a suspension that
contains a drug, agar and sugar (e.g. mannitol or lactose) and pouring the suspension in
the blister packaging wells, solidifying the agar at the room temperature to form a jelly
and drying at 30°C under vacuum. The mechanical strength of moulded tablets is a matter
of great concern. Binding agents, which increase the mechanical strength of the tablets,
The taste masked drug particles are prepared by spray congealing a molten
and an active ingredient into a lactose based tablet triturate form. Compared to the
lyophillization technique, tablets produced by the moulding technique are easier to scale
manufactured from the spray-dried powder have been reported to disintegrate in less than
20 seconds in aqueous medium. The formulation contained bulking agent like mannitol
and lactose, a superdisintegrant like SSG & CCS and acidic ingredient (citric acid) and/or
Direct compression represents the simplest and most cost effective tablet
based excipients.
1.5.4.2 Sugar Based Excipients: This is another approach to manufacture ODT by direct
compression. The use of sugar based excipients especially bulking agents like
hydrolysate, polydextrose and xylitol, which display high aqueous solubility and
sweetness, and hence impart taste masking property and a pleasing mouth feel.
Mizumito et al. have classified sugar-based excipients into two types on the basis
Type1 Saccharides (lactose & mannitol): low mouldability but high dissolution rate.
Type2 Saccharides (maltose & maltilol): high mouldability and low dissolution rate.
This technology involves softening the active blend using the solvent mixture of
water- soluble polyethylene glycol and methanol and subsequent expulsion of softened
mass through the extruder or syringe to get a cylinder of the product into even segments
using heated blade to form tablet. The dried cylinder can also be used to coat granules for
matrix known as ‗floss‘, made from a combination of excipients, either alone or with
drugs. The floss is a fibrous material similar to cotton-candy fibers, commonly made of
polydextrose can be transformed into fibers at 30–40% lower temperature than sucrose.
This modification permits the safe incorporation of thermo labile drugs into the
formulation. The tablets manufactured by this process are highly porous in nature and
offer very pleasant mouth feel due to fast solubilization of sugars in presence of saliva.
1.5.7 Sublimation
urea, urethane and phthalic anhydride may be compressed along with other excipients
into a tablet. This volatile material is then removed by sublimation leaving behind a
highly porous matrix. Tablets manufactured by this technique have been reported to
usually disintegrate in 10-20 sec. Even solvents like cyclohexane; benzene can be used as
1.5.8 Nanonization
size of drug to nanosize by milling the drug using wet-milling technique. The
selected stabilizers, which are then incorporated into MDTs. This technique is especially
advantageous for poorly water soluble drugs. Other advantages of this technology include
conventional packaging due to exceptional durability and wide range of doses (up to 200
polyvinyl pyrrolidone, polyvinyl alcohol or sodium alginate, etc.), drug and other taste
masking ingredients, which is allowed to form a film after evaporation of solvent. In case
of a bitter drug, resin adsorbate or coated microparticles of the drug can be incorporated
into the film. This film, when placed in mouth, melts or dissolves rapidly, releasing the
drug in solution or suspension form. The features of this system include paper thin films
of size less than 2X2 inches, dissolution in 5 sec, instant drug delivery and flavoured after
taste.6
entrapped or dissolved within the matrix of fast dissolving carrier material. When zydis
units are put into the mouth, the freeze-dried structure disintegrates instantaneously and
does not require water to aid swallowing. The zydis matrix is composed of many material
handling, polymers such as gelatin, dextran or alginates are incorporated. These form a
glossy amorphous structure, which imparts strength. To obtain crystallinity, elegance and
hardness, saccharides such as mannitol or sorbitol are incorporated. Water is used in the
disintegration while various gums are used to prevent sedimentation of dispersed drug
particles in the manufacturing process. Collapse protectants such as glycine prevent the
products are packed in blister packs to protect the formulation from moisture in the
environment.
Durasolv is the patented technology of Cima labs. The tablets made by this
technology consist of drug, filler and a lubricant. Tablets are prepared by using
conventional tabletting equipment and have good rigidity. These can be packaged into
CIMA labs have also developed Orasolv Technology. In this system active
medicament is taste masked. It also contains effervescent disintegrating agent. Tablets are
made by direct compression technique at low compression force in order to minimize oral
dissolution time. Conventional blenders and tablet machine are used to produce the
Flash dose technology has been patented by Fuisz Nurofen meltlet, a new form of
ibuprofen as melt in mouth tablets (MIM) prepared using flash dose technology is the
first commercial product launched by Biovail Corporation. Flash dose tablets consist of
self-binding shear form matrix termed as ―floss‖. Shear form matrices are prepared by
and high mouldability saccharides is used to obtain a rapidly melting strong tablet. The
active ingredient is mixed with a low mouldability saccharide (eg lactose, glucose, and
Prographarm laboratories have patented the Flash tab technology. Tablet prepared
by this system consists of an active ingredient in the form of micro crystals. Drug micro
granules may be prepared by using the conventional techniques like coacervation, micro
tabletting technology.
Table 1.6 Marketed products along with the technology used, their inventors and
the active ingredients present
Patent Basis of Technology developed Active Ingredient
Technology Technology by Company (Brand Name)
Zydia Lyophilization R.P.Scherer. inc Loratidine
Quicksolv Lyophilization Janssen Pharmaceutics Risperidone
Lyoc Lyophilization Farmalyoc Phloroglucinol Hydrate
Flashtab Direct compression Ethypharm Ibuprofen
Orasolv Direct compression Cima Labs, Inc. Paracetamol
Durasolv Direct compression Cima Labs, Inc Zolmitriptan
Wowtab Direct compression Yamanouchi Pharma Famotidine
Ziplets DC Microcaps Eurand International Ibuprofen
Advatab Diffuscap CR Tech. Eurand International Cetrizine
Tramadol
Flashdose Cotton Candy Fuisz Technology
hydrochloride
Micromask taste
Oraquick KV Pharm Hyoscyamine Sulfate
masking
emergence of more novel technologies for MDTs in the days to come. These innovations
dosage forms. It is reasonable to expect that future trends in innovations of drug delivery
systems will continue to bring together different technological disciplines to create novel
technologies.5
conditions in which injuries by gastric acid (and activated pepsin) is thought to play an
important role. These disorders include gastroesophageal reflux disease (GERD), benign
―peptic‖ ulcers of the stomach and duodenum, ulcers secondary to the use of
Mankind has lived with peptic ulcers since ancient times. Perhaps the first
description of this malady is the one inscribed on the pillars of the temple of Aesculapius
of Epidaurus from around the fourth century B.C.: ―A man with an ulcer in his stomach.
He incubated and saw a vision: the god seemed to order his followers to seize and hold
him, that he might incise his stomach. So he fled, but they caught and tied him to the
doorknocker. Then Asklepios opened his stomach, cut out the ulcer, sewed him again,
and loosed his bonds.‖ Many prominent people have suffered from indigestion and
ulcers, including the Roman emperor Marcus Aurtlius, whose death has been attributed
by some to a perforated ulcer and whose physician was none other than Galen himself.
Acid neutralization was recognised as effective treatment more than 12 centuries ago by
Paulus Aeginata, who prescribed a mixture of Samian and Lemnian earths and milk, not
Helicobacter pylori (H. Pylori) and PPIs. We now know that eradication of H. Pylori
effectively promotes healing of peptic ulcers and prevents their recurrence in most cases.
PPIs have become the drugs of choice in prompting healing from erosive esophagitis and
peptic ulcer because of their ability to nearly completely suppress acid production.21
In 1977 George Sachs & John Forte discovered the ―Proton pump‖ in the human
body. After a lot of efforts in 1982 the first published clinical study was on omeprazole.
PPIs were approved for prescription in 1989.21PPIs are widely used for treating acid-
induced inflammation, ulcers of the stomach and duodenum, GERD, erosive esophagitis,
Syndrome. They are also used in combination with antibiotics for eradicating H. pylori
PPIs are the most potent suppressors of gastric acid secretion are inhibitors of the
gastric H+,K+-ATPase proton pump. In typical doses, these drugs diminish the daily
production of acid (basal and stimulated) by 80% to 95%. Five PPIs are available for
clinical use: Omeprazole and its S-isomer, Esomeprazole, Lansoprazole, Rabeprazole and
1.8.1 Chemistry
CH3 CH3
N N
N N
S S
N O N O
H3CO H H
Fig No. 1.8.1.1 Structure of Omeprazole Fig. No. 1.8.1.2 Structure of Lansoprazole
OCH3 O (CH2)3OCH3
OCH3 CH3
N N
F2HCO N
N
S S
N O N O
H H
Fig. No.1.8.1.3 Structure of Pantoprazole Fig No. 1.8.1.3 Structure of Rabeprazole
claims to the contrary, all PPIs have equivalent efficacy at comparable doses.22
selectively interacting with the gastric proton pump (K /H -ATPase) in the parietal cell
secretory membrane.23
1.8.2 Stomach
The stomach is typically a J-shaped enlargement of the GIT directly inferior to the
diaphragm in the epigastric, umbilical, and left hypochondriac regions of the abdomen.
The stomach connects the esophagus to the duodenum, the first part of the small
intestine.24
The stomach has four main regions: the cardia, fundus, body, and pylorus. The
Cardia (CAR-dē-a) surrounds the superior opening of the stomach. The rounded portion
superior to the left of the cardia is the fundus (FUN-dus) inferior to the fundus is the
larger central portion of the stomach, called the body. The region of the stomach that
connects to the duodenum is the pylorus (pī-LOR-us; pyl-= gate; -orus=guard); it has
two parts, the pyloric antrum (AN-trum =cave), which is connects to the body of the
stomach, and the pyloric canal, which leads into duodenum. When the stomach is empty,
the mucosa lies in large folds, called rugae (ROO-gē = wrinkles), that can be seen with
unaided eye. The pylorus communicates with the duodenum of the small intestine via a
sphincter called pyloric sphinter. The concave medial border of the stomach is called the
lesser curvature, and the convex lateral border is called the greater curvature.24
The stomach wall is composed of the same four basic layers as the rest of the
GIT, with certain modifications. The surface of the mucosa is a layer of simple columnar
epithelial cells called surface mucous cells. The mucosa contains a laminal propria
(areolar connective tissue) and muscularis mucosae (smooth muscle). Epithelial cells
extend down into the lamina propria. Where they form columns of secretory cells called
gastric glands that line many narrow channels called gastric pits. Secretions from seeral
gastric glands flow into each gastric pit and then into the lumen of the stomach.24
The gastric glands contain three types of exocrine gland cells that secrete their
products into the stomach lume; mucous neck cells, cheif cells, and parietal cells, both
surface mucous cells and mucous neck cells secrete mucus. Parietal cells produce
intrinsic factor, which is needed for absorption of vitamin B12, and hydrochloric acid. The
chief cells secrete pepsinogen and gastric lipase. The secretions of the mucous, parietal,
and chief cells form gastric juice, which totals 2000-3000 ml/day. In addition, gastric
glands include a type of enteroendocrine cells, the G cell, which is located mainly in the
pyloric antrum and secretes the hormone gastrin into the bloodstream.24
Parietal cells secrete hydrogen ions (H+) and chloride ions (Cl-) separately into the
stomach lumen, the net effect are secretion of hydrochloric acid (HCl). Proton pumps
powered by H+/K+ ATPases actively transport H+ into the lumen while bringing
potassium ions (K+) into the cell. At the same time Cl- and K+ diffuse out through Cl- and
especially plentiful in parietal cells, catalyzes the formation of carbonic acid (H2CO3)
from water (H2O) and carbon dioxide (CO2). As carbonic acid dissociates, it provides a
ready source of H+ for the proton pumps but also generates bicarbonate ions (HCO3-). As
HCO3- builds up in the cytosol, it exits the parietal cell in exchange for Cl - via Cl-/HCO3-
antiporters in the basolateral membrane. HCO3- diffuses into nearby blood capillaries.
This ―alkaline tide‖ of bicarbonate ions entering the bloodstream after a meal may be
large enough to slightly elevate blood pH and make urine more alkaline.
The strongly acidic fluid of the stomach kills many microbes in food, and HCl
partially denatures (unfolds) proteins in food and stimulates the secretion of hormones
that promote the flow of bile and pancreatic juice. Enzymatic digestion of proteins also
begins in the stomach. The only proteolytic (protein-digesting) enzyme in the stomach is
pepsin, which is secreted by chief cells. Because pepsin breaks certain peptide bonds
between the amino acids making up proteins, a protein chain of many amino acid are
broken down into smaller peptide fragments. Pepsin is most effective in the very acidic
factors all regulate acid secretion. Their specific receptors (M3, H2, and CCK2 receptors,
respectively) are on the basolateral membrane of parietal cells in the body and fundus of
AMP-PKA pathway. ACh and gastrin signal through GPCRs that couple to the Gq-PLC-
IP3-Ca2+ pathway in parietal cells. In parietal cells, the cyclic AMP and the Ca2+-
hydrogen and potassium ions across the parietal cell membrane. This pump generates the
largest known ion gradient in vertebrates, with an intracellular pH of about 7.3 and an
The most important structures for CNS stimulation of gastric acid secretion are
the dorsal motor nucleus of the vagal nerve, the hypothalamus, and the solitary tract
nucleus. Efferent fibers originating in the dorsal motor nuclei descend to the stomach via
the vagus nerve and synapse with ganglion cells of the enteric nervous system. ACh
released from postganglionic vagal fibers directly stimulates gastric acid secretion
through muscarinic M3 receptors on the basolateral membrane of parietal cells. The CNS
predominantly modulates the activity of the enteric nervous system via ACh, stimulating
gastric acid secretion in response to the sight, smell, taste, or anticipation of food (the
"cephalic" phase of acid secretion). ACh also indirectly affects parietal cells by
increasing the release of histamine from the enterochromaffin-like (ECL) cells in the
fundus of the stomach and of gastrin from G cells in the gastric antrum.
ECL cells, the source of gastric histamine secretion, usually are in close proximity
to parietal cells. Histamine acts as a paracrine mediator, diffusing from its site of release
to nearby parietal cells, where it activates H2 receptors. The critical role of histamine in
Gastrin, which is produced by antral G cells, is the most potent inducer of acid
secretion. Multiple pathways stimulate gastrin release, including CNS activation, local
distention, and chemical components of the gastric contents. Gastrin stimulates acid
secretion indirectly by inducing the release of histamine by ECL cells; a direct effect on
secretion. Acidification of the gastric luminal pH to <3 stimulates SST release, which in
turn suppresses gastrin release in a negative feedback loop. SST-producing cells are
decreased in patients with H. pylori infection, and the consequent reduction of SST's
PPIs are prodrugs that require activation in an acid environment. After absorption
into the systemic circulation, the prodrug diffuses into the parietal cells of the stomach
The activated form then binds covalently with sulfhydryl groups of cysteines in
the H+/K+-ATPase, irreversibly inactivating the pump molecule. Acid secretion resumes
only after new pump molecules are synthesized and inserted into the luminal membrane,
providing a prolonged (up to 24- to 48-hour) suppression of acid secretion, despite the
much shorter plasma half-lives (0.5 to 2 hours) of the parent compounds. Because they
block the final step in acid production, the PPIs are effective in acid suppression
The recommended adult dose Of PPIs for the oral treatment of duodenal ulcer is
40 mg as PPIs are given once daily in the morning. Healing usually occurs within 2
weeks. For patients not healed after this initial course of therapy, an additional course of
2 weeks is recommended.
The recommended adult oral dose of PPIs for the oral treatment of gastric ulcer is
40 mg given once daily in the morning. Healing usually occurs within 4 weeks. For
patients not healed after this initial course of therapy, an additional course of 4 weeks is
recommended.
dose for H. pylori eradication is treatment for seven days with PPIs 40 mg together with
for H. pylori eradication is treatment for seven days with PPIs 40 mg together with
The recommended adult oral dose for the treatment of symptoms of GERD,
The recommended adult oral dose of PPIs is 40 mg, given once daily in the
morning. In most patients, healing usually occurs within 4 weeks. For patients not healed
For the prevention of relapse in patients with reflux esophagitis, the recommended
adult oral dose is 20 mg PPIs are given once daily in the morning and the dose is
Since an acidic pH in the parietal cell acid canaliculi is required for drug
activation, and since food stimulates acid production, these drugs ideally should be given
about 30 mins before meals. Concurrent administration of food may reduce somewhat the
rate of absorption of PPIs, but this effect is not thought to be clinically significant.
Concomitant use of other drugs that inhibit acid secretion, such as H2-receptor
antagonists, might be predicted to lessen the effectiveness of the PPIs, but the clinical
PPIs are absorbed rapidly in both rat and dog. Peak plasma levels are attained
within 15 to 20 mins in the rat and after about 1 hour in the dog. Oral bioavailability is
33% in the rat and 49 % in the dog. Following absorption, autoradiography and
quantitative tissue distribution experiments have shown that PPIs get rapidly distributed
the blood and most organs is found to be uniform initially. After 16 hours, radiolabelled
PPIs are predominantly detected in the stomach wall. After 48 hours, the entire
barrier by radiolabelled PPIs is very low. Protein binding in the rat and dog is 95% and
86%, respectively.26
1.10.8 Metabolism
PPIs get readily bound to serum proteins (98%) and almost completely
metabolized in the liver. Renal elimination represents the major route of excretion (about
82%) for the metabolites of PPIs. If we consider pantoprazole for e.g. the main
as a sulphate conjugate. The half-life of the main metabolite (about 1.5 hours) is not
much longer than that of pantoprazole sodium (approximately 1 hour).Once in the small
bowel, PPIs are rapidly absorbed, highly protein bound, and extensively metabolized by
hepatic CYPs, particularly CYP2C19 and CYP3A4. Several variants of CYP2C19 have
been identified. Asians are more likely than Caucasians or African-Americans to have the
CYP2C19 genotype that correlates with slow metabolism of Pantoprazole (23% vs. 3%,
toxicity in this ethnic group. Although the CYP2C19 genotype is correlated with the
reflux disease, there is no evidence that the CYP2C19 genotype predicts clinical efficacy
of these. 26
Because not all pumps or all parietal cells are active simultaneously, maximal
suppression of acid secretion requires several doses of the PPIs. For example, it may take
2 to 5 days of therapy with once-daily dosing to achieve the 70% inhibition of proton
pumps that is seen at steady state. More frequent initial dosing (e.g., twice daily) will
reduce the time to achieve full inhibition but is not proven to improve patient outcome.
Since the proton pump inhibition is irreversible, acid secretion will be suppressed for 24
to 48 hours, or more, until new proton pumps are synthesized and incorporated into the
mg enteric coated tablet. Its oral bioavailability compared to the i.v. dosage form is 77%
and does not change upon multiple dosing. Following an oral dose of 40 mg, C max is
Pantoprazole sodium shows linear pharmacokinetics after both i.v. and oral
bioavailability of pantoprazole sodium. Chronic renal failure does not lead to drug
accumulation with once-a-day dosing of the PPIs. Hepatic disease substantially reduces
the clearance of esomeprazole and lansoprazole. Thus, in patients with severe hepatic
disease, dose reduction is recommended for esomeprazole and should be considered for
lansoprazole.25
tablets dissolve only at alkaline pH substantially improve the oral bioavailability of these
acid-labile drugs.
Until recently, the requirement for enteric coating posed a challenge to the
administration of PPIs in patients for whom the oral route of administration is not
available. These patients and those requiring immediate acid suppression now can be
treated powdered drug combined with sodium bicarbonate (pantoprazole) which has less
lansoprazole, both of which are approved for intravenous administration in the United
United States. Thus an orodispersible tablet is a need to be developed which increases the
2. OBJECTIVE
and PPIs by direct compression method and to evaluate their quick disintegration and
immediate release properties. The effect of various formulations and process variables on
the particle morphology, micromeritics properties, in- vitro release behaviour, etc.
Pantoprazole will be used as a representative drug for the PPI in this research project.
3. To make available most of the drug at the site of action thus higher suppression of
acid secretion.
5. To compare the various formulations using different excipients and choosing the
release/dissolved.
PPIs are the most potent suppressors of gastric acid secretion thus inhibiting the
gastric H+/K+-ATPase (PP). In typical doses, these drugs diminish the daily
They are the drugs of choice for the treatment of various gastro intestinal
disorders.
lag time as they are enteric coated and fluctuations in the plasma drug levels,
the drug at the site of action thus instant relief is not achieved; fast dissolution and
absorption of the drug which will result quick onset of action and accumulation of
Most elderly patients, children, and patients with dysphagia have difficulty in
swallowing conventional tablets and hard gelatin capsules, and therefore do not
patients hospitalized for acute neuromuscular disorders and head injuries have
3. REVIEW OF LITERATURE
pantoprazole in comparison with those of other available PPIs. Like other PPIs,
(H+/K+-adenosine triphosphatase) in the parietal cells but, compared with other PPIs, its
binding may be more specific for the proton pump. Pantoprazole is well absorbed when
subsequently undergoes sulfate conjugation. The elimination half-life ranges from 0.9 to
1.9 hours was found to be independent of dose. Pantoprazole has similar efficacy to other
PPIs in the healing of gastric and duodenal ulcers, as well as erosive esophagitis, and as
part of triple-drug regimens for the eradication of Helicobacter pylori from the gastric
mucosa. It is well tolerated, with the most common adverse effects being headache,
diarrhea, flatulence, and abdominal pain. In clinical studies, it has been shown to have no
Bell W, Staar U. et al studied the action of the H+/K+ -ATPase inhibitors such as
gastric membrane vesicle under conditions it shows acidification of the vesicle interior,
pantoprazole and omeprazole inhibited H+/K+-ATPase activity. Their study showed that
both drugs inhibited, with similar potency, papain activity at pH 3.0 inactivated the
than pantoprazole and enzyme inhibition was faster than with pantoprazole. The results
conditions and that it is more stable than omeprazole at a slightly acidic pH such
as pH 5.0.28
Li XQ. et al. compared the potency and specificity of the currently used PPIs,
four cytochrome P450 enzymes (CYP2C9, 2C19, 2D6, and 3A4), they performed in vitro
studies using human liver microsomal preparations and recombinant CYP2C19. They did
mass spectometry. With several systems for CYP2C19 activity (two marker reactions, S-
liver microsomes or recombinant CYP2C19), the five PPIs showed competitive inhibition
rabeprazole. Their data suggest that, although the inhibitory profiles of the five studied
PPIs were similar, lansoprazole and pantoprazole are the most potent in vitro inhibitors of
compared with omeprazole and it‘s R-enantiomer. The inhibitory potency of rabeprazole
was relatively lower than the other PPIs, but its thioether analog showed potent inhibition
and 40 mg, on gastric acidity in healthy volunteers. They selected thirty-six subjects
intragastric pH and distal oesophageal pH were monitored at baseline and on the last day
of each treatment period. The measured endpoints were the median intragastric and
oesophageal pH, the percentage of time the intragastric pH < 4 and oesophageal pH < 4
and the area under the curve for gastric acidity over 24 h. Safety was evaluated by
dose-related effect in the range 10–40 mg once daily. The once-daily dose of 40 mg
provides the highest and most consistent control of gastric pH, especially at night.30
Raghunath AS. et al. reviewed the available preclinical and clinical studies
tolerability was also reviewed. The search terms used were gastroesophageal reflux
disease, reflux esophagitis, and proton pump inhibitor; all comparisons of esomeprazole
and lansoprazole at any dose were considered. The comparative studies that were
identified fell into 4 categories: (1) Intragastric acid suppression studies; (2) Randomized
controlled trials in the healing of erosive esophagitis; (3) Randomized controlled trials in
the maintenance of erosive esophagitis; and (4) Health economic analyses. Based on
once daily) and low doses (20 and 15 mg once daily, respectively) were compared, it was
seen that the data for esomeprazole and lansoprazole indicate clinical and cost-
Numans ME. et al. estimated the diagnostic test characteristics of successful PPI
published literature. They reviled that for a successful short-term treatment with a PPI in
patients suspected of having GERD does not confidently establish the diagnosis when
simulated intestinal fluid (pH 6.8) containing various generally recognized as safe listed
excipients, including Brij 58, Poloxamer 188, Cremophor RH40, Gelucire 44/14 and PEG
6000. After incubation at 37 and 60°C, the amounts of rabeprazole and its degradation
product, thioether rabeprazole, were quantitated by HPLC analysis. The main degradation
followed first-order kinetics. In the absence of any excipients, the rate constants (k)
obtained at 37 and 60°C were 0.75 and 2.78 h−1, respectively. In contrast, the addition of
excipients improved its stability. Among several excipients tested in this study, Brij 58
displayed the greatest stabilizing effect. The stabilizing mechanisms of these hydrophilic
polymeric excipients with optimal HLB values could be partially explained in terms of
conclusion, rabeprazole formulations that contain suitable excipients would improve its
Armstrong D. et al. compared acid suppression (time with pH > 3.0, 4.0, 5.0 and
in water, produces greater acid suppression than pantoprazole 40 mg IV oral dose after 1
Wahbi AAM. et al. showed that the compensation method and other
consuming and need special reagents. The Amax method has been proved to be inaccurate
due to matrix interference. They reported that the other methods are not stability
indicating, while the present methods using difference spectrophotometry eliminate acid
methods are more versatile and easy to apply than the polarographic and voltammetric
methods. The chromatographic methods need special equipment that may not be
that they cannot be applied to biological fluids containing these compounds and their
conjugated forms. They validated these methods; the limits of detection where carried
out. These proposed methods have been applied to the determination of the three drugs in
unaffected by the presence of acid induced degradation products; hence can be used as a
sesquihydrate Two methods were based on charge transfer complexation reaction of these
benzoquinone and with s acceptor as iodine. A third method was also investigated
depending on ternary complex formation with eosin and copper. The coloured products
have the advantages of being simple, accurate, sensitive and suitable for routine analysis
utilize a single step reaction and single solvent. The iodine acceptor method was more
sensitive in the case of lansoprazole than pantoprazole. The ternary complex method did
not require prior extraction procedure and have the advantages of sensitivity, simplicity
and reproducibility. All these methods can be used as general methods for the
in bulk and in pharmaceutical formulations. They are convenient for Q.C. and routine
Ramakrishna NVS. et al. developed a very sensitive and selective HPLC method
(70/30, v/v), the analyte and internal standard (zonisamide) were separated using an
isocratic mobile phase of 10milli moles of phosphate buffer (pH 6.0)/acetonitrile (61/39,
v/v) on reverse phase Waters symmetry C18 column. A linear range of 20–5000 µg/mL
was established. This HPLC method was validated with between-batch and within-batch
pharmacokinetic studies.37
methanol/ammonia 4.0% v/v, where the sufficient spectra resolutions of drug and
corresponding impurity were obtained, using the amplitudes. Method showed good
obtained amplitudes for impurities OMS and NPA in comparison to expected values
considered to have a similar therapeutic efficacy when used under the same therapeutic
conditions. They reported the design, results and some important aspects involved in a
bioequivalence study between two solid oral formulations from different manufacturers.
Some important findings were the high intra-subject variability observed for Cmax and the
variability observed between subject profiles, probably caused by the multi unit type of
formulations studied.39
Choi HG, Jung JH. et al. developed omeprazole buccal adhesive tablets, and
also studied the release and bioavailability of omeprazole delivered by buccal adhesive
and CCS. They concluded that CCS enhanced the release of omeprazole from the tablets.
The analysis of the release mechanism showed that CCS changed the release profile of
omeprazole from first to zero-order release kinetics by forming porous channels in the
tablet matrix. However, it decreased the bioadhesive forces and stability of omeprazole
tablets in human saliva. The tablet where composed of omeprazole: sodium alginate:
which may be attached to the human cheek without collapse and also enhanced the
stability of omeprazole in human saliva for at least 4 hrs, giving a fast release of
min after buccal administration and remained at the high level for 6 hrs. The buccal
the omeprazole buccal adhesive tablet would be useful to deliver omeprazole which
degrades very rapidly in acidic aqueous medium and undergoes hepatic first-pass
Another report by Choi HG, Jung JH. et al. developed an omeprazole buccal
adhesive tablet, they studied the release and bioavailability of omeprazole delivered by
magnesium oxide and CCS. Their studied concluded that CCS enhanced the release of
omeprazole from the tablets. The analysis of the release mechanism showed that CCS
changed the release profile of omeprazole from first to zero-order release kinetics by
forming porous channels in the tablet matrix. However, it decreased the bioadhesive
forces and stability of omeprazole tablets in human saliva. The tablet where composed of
(20:24:6:50:10 mg). It may be attached to the human cheek without collapse and it
enhanced the stability of omeprazole in human saliva for at least 4 h, giving a fast release
of omeprazole. Their results demonstrate that the omeprazole buccal adhesive tablet
would be useful to deliver omeprazole which degrades very rapidly in acidic aqueous
medium and undergoes hepatic first pass metabolism after oral administration.41
evaluate a carbamazepine floating DDS. In this method glass beaker was modified by
adding a side arm at the bottom of the beaker so that the beaker can hold 70 ml of 0.1 N
HCl dissolution mediums and allow collection of samples. The tablet did not stick to the
agitating device in the proposed dissolution method. The drug release followed zero order
kinetics in the proposed method. The proposed test may show good in vitro in vivo
Garcia CV. et al. developed and validated a dissolution test for rabeprazole
sodium coated tablets using a reverse-phase liquid chromategraphic method. After test
sink conditions, dissolution medium and stability of the drug, the best conditions were:
paddle at 75 rotations per minute (rpm) stirring speed, HCl 0.1M and borate buffer pH
9.0 as dissolution medium for acidic and basic steps, respectively, volume of 900ml for
both. The quantitation method was also adapted and validated. Less than 10% of the label
amount was released in the acid step, while more than 95% was achieved over 30min in
the basic one. The dissolution profile for tablets was considered satisfactory. The
dissolution test developed was adequate for its purpose and could be applied for quality
control of rabeprazole tablets, since there is no official monograph. The dissolution test
developed and validated for rabeprazole sodium coated tablets was considered
satisfactory. It was carefully studied in order to guarantee the drug stability during all
analysis time. The conditions that allowed the dissolution profile determination were
borate buffer pH 9.0 medium, paddle (USP apparatus II) and 75 rpm stirring speed. The
suspension for use in patients who have difficulty swallowing. They performed in vitro
and suitability for administration orally or via enteral tubes. It also describes the
existing solid dosage forms (capsules and tablets) in a clinical bioequivalence study. In
these analyses, the packet (sachet) formulation of esomeprazole was chemically stable in
suspension and when administered orally and via enteral tubes. The formulation had a
short reconstitution time, remaining fully dispersed in water for at least 30 mins, and was
dispersed in applesauce, apple juice, or orange juice without compromising its stability or
dispersion characteristics. The packet formulation met the regulatory definition for
friendly and more convenient formulation of lansoprazole which can be taken with or
without water is the first PPI to be made available as an orally disintegrating tablet. It
compressed with an inactive, rapidly dispersing matrix to form a tablet. When the tablet
is placed on the tongue and sucked gently it disintegrates rapidly in the mouth, releasing
the enteric-coated microgranules which are swallowed with the patient‘s saliva without
water. Alternatively, the tablet can be swallowed with a drink of water. Studies have
dosages for lansoprazole fast disintegrating tablet are therefore identical to lansoprazole
capsules. The new formulation may be of particular benefit to those with active life-styles
who do not always have water available, patients who have difficulty in swallowing, and
elderly patients. The new formulation will offer greater flexibility in the prescribing of
lansoprazole, may improve patient compliance with treatment, and may be of particular
benefit to patients who have difficulty in swallowing and elderly patients. Future studies,
comparing the two lansoprazole formulations in different clinical settings, will provide
tablet which comprises of. An inner core, such as, sugar particles or granules, which can
be spherical or any other useful shape, coated with a drug, preferably, an acid sensitive
drug, such as lansoprazole, where the drug layer preferably comprises a film forming
extra fine talc, in addition to the drug. The ―first coating layer,‖ Subcoat I, which is
totally free of both any alkaline stabilizing agent and the drug, where the inert first
coating layer preferably comprises a film forming agent, such as hypromellose, and an
excipient, such as talc, preferably, extra fine talc. Subcoat II which is also called as
magnesium carbonate, and can further comprise a film forming agent, such as
hypromellose. The outermost layer which is also called as ―outer coating,‖ that comprises
an enteric coating where the enteric coating is preferably polymeric, such as methacrylic
Kundu S. and Sahoo PK. in their article noted that OrDTs are formulated to
improve patient compliance and convenience. It offers as a good alternative drug delivery
for geriatric and paediatric patient. OrDTs are prepared by various technologies and so
formulation scientists have more alternatives to choose the best technology according to
the specific drug profile. OrDT administration is easy for patients who have problems of
deglutition or for those persons who would like their treatment without simultaneous
parameters which are equivalent to those obtainable with existing tablets or gel
capsules.47
Kumaresan C. et al. demonstrated different ways for the formulation of the oral
dispersible tablets and how the product performance depends on the drug suitability and
excipients selection in the delivery system. He described the ideal characteristics of the
drug and excipients required to be employed in the formulation of oral dispersible tablets
Hirani JJ. et al. illustrated various different applications of OrDTs and their use
in today‘s pharmaceutical market. They also highlighted the various novel patented
procedures for the manufacturing of OrDTs along with their inventors and their
advantages and disadvantages. They also highlighted the challenges likely to be faced by
the formulator in the formulation of OrDT, along with criteria for the selection of the
drug.49
Fu Fu. et al. showed how the popularity of FDTs has increased tremendously
over the last decade. There are about 40 drugs that have been formulated and marketed as
FDTs using various technologies. The key to FDT formulations is fast disintegration,
dissolution, or melting in the mouth and this can be achieved by producing the porous
FDTs prepared by direct compression usually have good mechanical properties, and the
The clinical studies show FDTs can improve patient compliance, provide a rapid onset of
action, and increase bioavailability. Considering the many benefits of FDTs, it is only a
matter of time until a majority of oral formulations are prepared in FDT forms.50
suspension on contact with the saliva, after ingested in mouth. These are particularly
useful for pediatric or geriatric patients, can be taken without liquids and facilitate
treatment of emergent pain, irrespective of the place and situation where it may arise. The
developed formulations have suitable characteristics that distinguish them from common
solid dosage forms, such as rapid disaggregation, combining advantages of both liquid
makes it possible to produce sufficiently hard tablets that still disaggregate within
seconds and most of the developed tablets can be considered as ‗‗fast dispersible‖. They
can also be programmed not only for oral dispersibility, but also for delayed release, with
dissolution of the drug taking place far from the buccal district. Finally these tablets can
control of the side effects of the drug at gastric level. Formulations containing Kollidon
that offer an alternative to traditional tablets for orally dispersible ibuprofen tablets.51
to be swallowed without the aid of water. A direct compression method was used to
prepare these two types of tablets containing coated ibuprofen as a high dosed model
drug. The properties of the water dispersible tablet, such as porosity, hardness,
disintegration time and increase in viscosity after dispersion, were investigated. The
before the galactomannan starts to swell. These tablets disperse in water within 40 s and
design was applied to predict the effects of the quantitative factors mannitol and CP as
well as compression force on the characteristics of the tablet. Special emphasis was paid
optimum tablet formulation, containing 34% mannitol and 13% CP, provides a short
wetting time of 17 sec and a sufficient crushing strength of 40 N. It was concluded that
fast dispersible tablets with acceptable hardness and desirable taste could be prepared
acceptable salt thereof, a selective norepinephrine reuptake inhibitor indicated for the
atomoxetine in the plasma. The dosage form may comprise one or more populations of
designed rapid release profile after a predesigned lag-time of about 0 to 6 hours following
oral administration.53
products, i.e., mainly appearance, odour and taste are essential factors in assessing the
potential tool to improve patient compliance and commercial success of the product. Ion
exchange resins are solid and suitably insoluble high molecular weight polyelectrolytes
that can exchange their mobile ions of equal charge with the surrounding medium. The
resulting ion-exchange is reversible and stoichiometric with the displacement of one ionic
species by another. They developed Doxilamine ODTs with considerable increase in drug
studied. To prevent bitter taste and unacceptable odour of the drug, the drug was taste
masked with weak cation exchange resins like Indion 234, Indion 204 and Indion 414.
They characterised the drug according to different compendial methods, on the basis of
the three resins, they selected one resin for further studies i.e., Indion 234, because of
high drug loading capacity. Drug–resin complex was prepared using batch method and
effect of various processing parameters viz. drug–resin ratio, pH, temperature and drug
Okuda Y. et al. took the challenge to design a new orally disintegrating tablet
(ODT) that has high tablet hardness and a fast oral disintegration rate using a new
light anhydrous silicic acid, or hydroxypropyl starch was also included in the suspension.
The RDGs obtained possessed extremely large surface areas, narrow particle size
distribution, and numerous micro-pores. When tabletting these RDGs, it was found that
the RDGs increased tablet hardness. The newly developed ODT was found to have
superior properties as an ODT, comparatively high hardness of tablet, and fast oral
disintegration rate. As the method to prepare this ODT (suspension method) is simple and
does not requires the special equipment, this technology is expected to provide better
ODTs for many kinds of drugs that can improve the quality of life of patients.55
Shoukri RA. et al. developed a lyophilized ODT that enhanced the in vitro
dissolution and in vivo absorption of nimesulide, which is a drug with poor solubility and
parameters on the disintegration time and in vitro dissolution of nimesulide from ODTs
along with other tablet characteristics was investigated. Results obtained from
disintegration and dissolution studies showed that lyophilized ODTs disintegrated within
few seconds and showed significantly faster dissolution rate of nimesulide compared to
the plain powder drug and nimesulide in commercially available immediate release tablet.
absorbed into the blood stream with significantly higher bioavailability when compared
to standard immediate releasing oral dosage form. The study suggests that ODT
nimesulide.56
Kuno Y. et al. proposed a new tablet preparation method that employs the phase
powder containing two sugar alcohols with high and low melting points, and subsequent
heating at a temperature between the high and low melting points. A combination of two
sugar alcohols and the heating process is needed to prepare RDT with sufficient hardness.
Tablet hardness is related to the increase of inter-particle bonds or the bonding surface
area in tablets induced by the phase transition of the lower melting point sugar alcohol.57
4. METHODOLOGY
Bulk density
Tapped density
Angle of repose
Carr‘s Inndex
Hausner‘s ratio
Colour
Shape
Hardness
Friability
Tablet thickness
Weight variation
Table No. 4.2 Materials used with their grade and their manufacturers or suppliers.
3. CP A.G K. P. Pharmaceuticals.
Here A.G indicates Analytical grade and L.G indicates Laboratory grade
For this research work Pantoprazole Sodium was used as a representative for the class
of PPIs.
Category: Antiulcer
ide, sesquihydrate.
OCH3
OCH3
F2HCO
N
N O
H
Description: Pantoprazole sodium is a white to off- white power which contains not less
than 98.0 percent and not more than 102.0 percent of C16H14F 2N3 NaO4S,1 H2O
pH: 1% aqueous solution has a pH of 10.05 and 10% aqueous solution has a pH of 10.85.
Storage: Store protected from light and moisture, between 2°C to 8°C.
Clinical pharmacology:
absorption into the systemic circulation, the prodrug diffuses into the parietal cells of the
cannot diffuse back across the canalicular membrane. The activated form then binds
inactivating the pump molecule. Acid secretion resumes only after new pump molecules
are synthesized and inserted into the luminal membrane, providing a prolonged (up to 24-
to 48-hour) suppression of acid secretion, despite the much shorter plasma half-lives (0.5
to 2 hours) of the parent compounds. Because they block the final step in acid production,
the PPIs are effective in acid suppression regardless of other stimulating factors.
Therapeutic Uses:
It is used principally to promote healing of gastric and duodenal ulcers and to treat
continue NSAID use. In addition, it is FDA approved for reducing the risk of duodenal
Adverse effects:
Pantoprazole sodium generally causes remarkably few adverse effects. The most
common side effects are nausea, abdominal pain, constipation, flatulence, and diarrhea.
Subacute myopathy, arthralgias, headaches, and skin rashes also have been reported. As
noted above, all PPIs are metabolized by hepatic CYPs and therefore may interfere with
Contraindications:
Warnings:
before therapy with pantoprazole sodium is instituted since treatment with pantoprazole
It should not be administered to pregnant women unless the expected benefits outweigh
human milk. It should not be given to nursing mothers unless its use is believed to
Use in Children: The safety and effectiveness of pantoprazole sodium in children has not
Precautions:
adenomas and carcinomas in the liver and neoplastic changes in the thyroid.
Pediatric and Geriatric studies: Short-term and long-term treatment with pantoprazole
sodium in a limited number of patients up to 6 years have not resulted in any significant
pathological changes in gastric oxyntic exocrine cells. A slight increase in AUC (12%)
and Cmax (7%) for pantoprazole sodium occurs in elderly volunteers when compared to
younger volunteers. The daily dose used in elderly patients, as a rule, should not exceed
of 5 to 7, and the Cmax increased by a factor of 1.5 in patients with liver cirrhosis
and the Cmax increased by a factor of 1.3 in patients with severe liver cirrhosis compared
between patients who are dialyzed and those who are not is 4%.
Drug drug interactions: Pantoprazole sodium is metabolized in the liver via the CYP
450 system. Pharmacokinetic drug interaction studies in man did not demonstrate the
inhibition of the oxidative metabolism of the drug. Pantoprazole sodium does not interact
Concomitant use of antacids or consumption of food does not affect the pharmacokinetics
of pantoprazole sodium. Changes in absorption should be taken into account when drugs
Others: Generally, daily treatment with any acid blocking medicines over a long time
(e.g. longer than 3 years) may lead to malabsorption of cyanocobalamin caused by hypo-
have been reported in the literature and should be considered if respective clinical
symptom profile was observed after ingestion of high doses of pantoprazole. Doses of up
Structural Formula:
Chemical Name and Cellulose, carboxymethyl ether, sodium salt, cross linked
CAS Registry No.: [74811-65-7].
Structural Formula:
Structural Formula:
Structural Formula:
Solubility and storage L-HPC is a stable, though hygroscopic, material. The powder
conditions: should be stored in a well- closed container.
Structural Formula:
Functional categories: Tablet and capsule diluent, disintegrant and tablet binder.
4.2.2.10 Menthol
Structural Formula:
.
Solubility: Very soluble in ethanol (95%), chloroform, ether, fatty oils
and liquid paraffin; soluble in acetone and benzene; very
slightly soluble in glycerin; practically insoluble in water.
Functional categories: Flavouring agent; therapeutic agent.
4.2.2.11 Thymol
Structural Formula:
.
Solubility: Soluble in chloroform, ethanol (95%), ether, glacial acetic
acid, olive oil, water. Freely soluble in essential oils, fixed
oils, and fats. Sparingly soluble in glycerine.
Functional categories: Flavouring agent; therapeutic agent.
4.2.2.12 Camphor
Structural Formula:
.
Functional categories: Flavourings agent; therapeutic agent.
Structural Formula:
.
Functional Diluent; diluent for lyphilized preparations; sweetening agent;
categories: tablet and capsule diluent; tonicity agent.
4.2.2.14 Aspartame
Structural Formula:
.
Functional Sweetening agent.
categories:
4.2.2.15 Talc
Chemical Name and 2-Butenedioic acid, monooctadecyl ester, sodium salt [4070-
CAS Registry No.: 80-8].
Structural Formula:
4.3 INSTRUMENTS
13. Tablet punching machine, (Rimek mini Karnavati Engineering Ltd, Mehsana ,
press-1) 10- station Gujarat.
The goals of preformulation studies are to choose the correct form of the drug
substance, evaluate its physical and chemical properties, and generate a thorough
understanding of the material‘s stability under the conditions that will lead to the
umbrella for the fingerprinting of a drug substance or product both at the early and latter
critical learning time about candidate drugs. Typically, it begins during the lead
optimization phase and continues through prenomination and into the early phases of
development. Decisions made on the information generated during this phase can have a
substance.
Hence, preformulation studies on the obtained sample of drug include colour, taste,
Melting point of Pantoprazole sodium was set to determine by open cup capillary
method.
The infrared absorption spectrum of Pantoprazole was recorded with a KBr disc
amber coloured volumetric flask and dissolved in small quantity of 0.1 N HCl. To this a
added. The volume was made up with the 0.1 N HCl to get a conc. of 1000µg/ml
From this 1ml was withdrawn and diluted to 100ml to get a conc. of 10µg/ml (SS-
II). From SS-II aliquots of 2ml, 4ml, 6ml, 8ml and 10ml were pipetted into 10ml
volumetric flasks. The volume was made up with 0.1 N HCl to get the final Conc. of 2, 4,
6, 8, 10 µg/ml respectively. When this solution was scanned in the UV range i.e. from
200nm to 800nm λmax was found to be 281.5 nm for Pantoprazole sodium in 0.1N HCl as
The same solution was stored at room temperature and abs of the solutions was
Conc =
amber coloured volumetric flask and dissolved in small quantity of 0.1 N HCl. To this a
added. The volume was made up with the 0.1 N HCl to get a conc. of 1000µg/ml
From this 1ml was withdrawn and diluted to 100ml to get a conc. of 10µg/ml (SS-
II). From SS-II aliquots of 2ml, 4ml, 6ml, 8ml and 10ml were pipetted into 10ml
volumetric flasks. The volume was made up with 0.1 N HCl to get the final conc. of 2, 4,
6, 8, 10 µg/ml respectively. When this solution was scanned in the UV range i.e. from
200nm to 800nm λmax was found to be 301nm for Omeprazole sodium in 0.1N HCl as a
blank in UV-Visible Spectrophotometer (UV-1800 Shimadzu). The abs of each conc. was
The same solution was stored at room temperature and abs of the solutions was
Conc=
used to formulate ODTs where studied by FTIR analysis. FTIR spectral analysis of
pantoprazole sodium and superdisintegrants and excipients were carried out to investigate
the changes in chemical composition of the drug after combining it with the excipients.
were obtained by adding potassium bromide 100 times the quantity of the sample to be
studied.
Compression method.
addition method
were passed through 60# mesh sieve separately and collected. The drug was weighed
along with the other excipients and was mixed in geometrical order. This mixture was
shaken for few mins to ensure proper mixing of all the ingredients. The tablets were
compressed using flat face 16.4 X 8 mm flat oval shaped punch to get tablets of 1300 mg
weight using ten stations Rimek tablet compression machine (Karnavati Engineering Ltd.
Ahmadabad, India).
using a combination of two different superdisintegrants in the ratio of 1:1. All the
ingredients were passed through 60# mesh sieve separately and collected. The drug was
weighed along with the other excipients and was mixed in geometrical order. This
mixture was shaken for few mins to ensure proper mixing of all the ingredients. The
tablets were compressed using flat face 16.4 X 8 mm flat oval punch to get tablets of
1300 mg weight using ten stations Rimek tablet compression machine (Karnavati
were prepared by direct compression.63 All the ingredients were passed through 60# mesh
sieve separately and collected. The drug was weighed along with the other excipients and
was mixed in geometrical order. This mixture was shaken for few mins to ensure proper
mixing of all the ingredients. The tablets were compressed using flat face 16.4 X 8 mm
flat oval size punches to get tablets of 1300 mg weight using ten stations Rimek tablet
compression machine (Karnavati Engineering Ltd. Ahmadabad, India). The tablets were
superdisintegrants and one sublimating agents where used. All the ingredients were
passed through 60# mesh sieve separately and collected. The drug was weighed along
with the other excipients and was mixed in geometrical order. This mixture was shaken
for few mins to ensure proper mixing of all the ingredients. The tablets were compressed
using flat face 16.4 X 8 mm flat oval shaped punch to get tablets of 1300 mg weight
using ten stations Rimek tablet compression machine (Karnavati Engineering Ltd.
Ahmadabad, India).
Treated agar gum (TAG) powder has shown having superdisintegration property.
direct compression. The gums like agar, gaur gum, gum acacia etc where taken.
10gm of the gums was accurately weighed and added to 250 ml beaker containing
100 ml distilled water and then stirred for few minis. This was kept aside for 2 days and
then dried until it form dry powered. This mass was pulverised and pass throw 60# sieve.
All the other ingredients were also passed through 60# mesh sieve separately and
collected. The drug was weighed along with the other excipients and was mixed in
geometrical order. This mixture was shaken for few mins to ensure proper mixing of all
the ingredients. The tablets were compressed using flat face 16.4 X 8 mm flat oval size
punch to get a tablets of 1300 mg weight using ten station Rimek tablet compression
Table No. 4.4.2.1.1 Formulations of Orodispersible tablets using superdisintigrant addition method
Sr. Ingredients Quantity used in mg
No
F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 F12 F13 F14 F15 F16
1. Pantoprazole 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20
2. CP - 13 39 65 - - - - - - - - - - - -
3. SSG - - - - 13 39 65 - - - - - - - - -
4. CCS - - - - - - - 13 39 65 - - - - - -
5. L-HPC - - - - - - - - - - 13 39 65 - - -
6. Pregelatinized Starch - - - - - - - - - - - - - 13 39 65
7. Treated Agar - - - - - - - - - - - - - - - -
8. Treated Gaur - - - - - - - - - - - - - - - -
9. Aspartame 52 52 52 52 52 52 52 52 52 52 52 52 52 52 52 52
10. Mannitol DC 77.5 64.5 38.5 12.5 64.5 38.5 12.5 64.5 38.5 12.5 64.5 38.5 12.5 64.5 38.5 12.5
11. Talc 26 26 26 26 26 26 26 26 26 26 26 26 26 26 26 26
12. SSF 13 13 13 13 13 13 13 13 13 13 13 13 13 13 13 13
13. Sodium Bicarbonate 585 585 585 585 585 585 585 585 585 585 585 585 585 585 585 585
14. Potassium Bicarbonate 520 520 520 520 520 520 520 520 520 520 520 520 520 520 520 520
15. Flavour 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5 6.5
Angle of repose is defined as the maximum angle possible between the surface of
a pile of the powder and horizontal plane. If more material is added to the pile, it slides
down the sides until the mutual friction of the particles, producing a surface at an angle
is in equilibrium with the gravitational force; the tangent of the angle of repose is equal to
the coefficient of friction, µ, between the particles. The frictional force in a loose powder
tan = h / r
= tan-1 (h/r)
h is height of pile
Different ranges of flow ability in terms of angle of repose are given in table no.4.5.1.1
Table No 4.5.1.1 Relationship between Angle of Repose () and flow properties.
<25 Excellent
25=30 Good
30-40 Passable
Method
A funnel was filled to the brim and the test sample was allowed to flow smoothly
through the orifice under gravity. From the cone formed on a graph sheet was taken to
measure the area of pile, thereby evaluating the flowability of the granules. Height of the
Bulk density is defined as the mass of a powder divided by the bulk volume. The
bulk density of a powder depends primarily on particle size distribution, particle shape,
and the tendency of the particles to adhere to one another. The bulk density of a powder
powder is likely to have a greater arch strength then a less consolidated one and may
therefore be more resistant to powder flow. The ease with which a powder consolidates
Method
Both loose bulk density (LBD) and tapped bulk density (TBD) were determined by
tap density tester. A quantity of accurately weighed powder from each formula,
previously shaken to break any agglomerates formed was introduced into a measuring
cylinder. After the initial volume was observed, the cylinder was allowed to fall under its
own weight onto a hard surface from the height of 2.5 cm at 2 seconds interval. The
taping was continued until no further change in volume was noted. LBD and TBD were
compressibility index. Grading of the powders for their flow properties according to CI is
Table No 4.5.1.3 Grading of the powders for their flow properties according to
Carr’s Index
5 – 15 Excellent
12-16 Good
23-35 Poor
Hausner found that the ratio bulk density by tapped density war related to
interparticle friction and, as such, could be used to predict powder flow property. He
showed that powders with low interparticle friction, such as coarse spheres, have ratios of
approximately 1.2. Where more cohesive, less free-flowing powders such as flakes have
HR =
Table No 4.5.1.4 Grading of the powders for their flow properties according to
Hausners Ratio
HR Flow
The tablets were examined under a lens for the shape of the tablet and colour by
The crown thickness of individual tablet may be measured with a vernier caliper,
which permits accurate measurements and provides information on the variation between
in a holding tray, where their total crown thickness may be measured with a sliding
caliper scale. The tablet thickness was measured using vernier caliper.
shipping. The hardness of the tablets was determined using Monsanto Hardness tester. It
is expressed in Kg/cm2. Three tablets were randomly picked from each formulation and
It is the phenomenon whereby tablet surfaces are damaged and/or show evidence
Ten tablets were initially weighed [W (initial)] and transferred into friabilator. The
friabilator was operated at 25 rpm for 4 mins or run up to 100 revolutions. The tablets
were weighed again [W (final)]. The percentage friability was then calculated by,
F=
<1 Acceptable
The tablets were selected randomly from each formulation and weighed
individually to check for weight variation. The U.S Pharmacopoeia allows a little
variation in the weight of a tablet. The percentage deviation in weight variation is shown
in table no 4.5.2.5.
130 mg or less 10
In all the formulations the tablet weight was above 324 mg and, hence 5% maximum
The content uniformity test is used to ensure that every tablet contains the amount
of drug substance intended with little variation among tablets within a batch. Due to
increased awareness of physiological availability, the content uniformity test has been
included in the monographs of all coated and uncoated tablets intended for oral
administration where the range of size of the dosage form available includes 50 mg or
smaller sizes. For content uniformity test, representative samples of 30 tablets are
selected and 10 are assayed individually. At least 9 must assay within ± 15% of the
assay and amount of active ingredient is calculated. Since active ingredient of present
investigation is not official in any pharmacopoeia the following method was used for
pantoprazole sodium was weighed and dissolved in sufficient quantity of 0.1N HCl. The
solution was filtered through Whatmann filter paper (No.41), suitably diluted with 0.1N
HCl and assayed at 281.5 nm, using a UV-Visible double beam spectrophotometer (UV-
1800 Shimadzu).
Method
For a drug to be absorbed from a solid dosage form after oral administration, it
must first be in solution, and the first important step toward this condition is usually the
had to be modified, since the standard glass tube is 21.5 mm in internal diameter and the
tested tablets have, however, a mean diameter of 25 mm.57 The disintegration was carried
temperature between 15 and 25°C. Only one tablet at a time was tested and considered
F
ig. No. 4.5.2.7 Method used for the disintegration of the Orodispersible tablets
In vitro release studies were carried out using a modified USP XXIII dissolution
test apparatus. Two objectives in the development of in vitro dissolution tests was to
show that,
i) Release of the drug from the tablet is as close as possible upto 100%.
ii) Rate of drug release is uniform from batch to batch and is the same as the release
Modification:
The normal USP XXIII dissolution apparatus was chosen in which a beaker was
placed. This beaker is an elongated one generally used for TLC and other purpose.
Another modification was that basket was used in place of paddles because of the narrow
mouth opening of the beaker. Outside this beaker water was putted at a level till the
dissolution fluid in the beaker reaches. The temperature was validated and kept at 40.1°C
and the rotation of the basket was kept at 75 RPM. Only 190 ml dissolution fluid was
used. Summary of general in vitro dissolution conditions employed throughout the study
to determine the in vitro dissolution rate for all the formulation is given in the following
table.
5. max 281.5nm
specific container, to remain within its physical, chemical, therapeutic and toxicological
specifications. Stability studies was conducted as per the specified ICH guidelines the
selected best formulation. The purpose of stability testing is to provide evidence on how
the quality of a drug substance or drug product varies with time under the influence of a
variety of environmental factors such as temperature, humidity and light and enables
In the present study, the ODTs where packed in suitable packaging material and
5%
The tablets were withdrawn after period of 15, 45 and 90days and analyzed for
Moore and Flanner proposed two new indices (f 1 and f 2 ) to compare dissolution
profiles of a test and a reference formulations . The concept of similarity factor (f 2) has
been endorsed by Food and Drug Administration (FDA); therefore, it is widely adopted
in formulation and development and dossier preparation. The equation of similarity factor
This is a widely used factor used to determine the similarity between two
formulations. It is widely used when you have to check your formulation is similar to that
6. RESULTS
The present study was aimed at formulating ODTs of PPIs. This is a novel
approach for increasing the patient compliance with a faster onset of action as compared
FTIR is one of the most widely used methods for checking the compatability
between substances and for the identification of the drug. Pantoprazole sodium,
All the samples were scanned at the resolution of 4 cm-1 over the wave number
region 4000-400 cm-1 using KBr disk method. This KBr disks are formed by taking drug
and KBr in a ratio of 1:100 respectively. Then this mixture was mixed well in mortar for
three to five mins. A very small amount of this mixture was uniformly spread and
sandwich between the pellets and pressed using KBr pellet press at a pressure of 20,000
psi for 1 mins. The pressure was then released and pellet was placed into the pellet holder
The selected formulation shows the characteristics peak similar to that obtained in
the pure pantoprazole sodium indicating that there is no incompatability between the drug
3363.97cm-1 &
N-H 3500-3300 cm-1 1500 cm-1 1450 cm-1
3489.94 cm-1
1585.54 cm-1
C=C 16000-1400cm-1
& 1469.81cm-1
823.63 cm-1&
AromaticC-H 3150-3050 cm-1 900-690 cm-1 3057.27 cm-1
806.37cm-1
1350-1000
C-N 1317.43 cm-1
cm-1
16000-
C=C 1535.39 cm-1
1400cm-1
900-690
AromaticC-H 3150-3050cm-1 3063.40 cm-1 833.28 cm-1
cm-1
Aromatic C-H 3150-3050 cm-1 900-690 cm-1 3084.28 cm-1 833.28 cm-1
1. 0 0
2. 5 0.1535
3. 10 0.32583
4. 15 0.505
5. 20 0.6525
6. 25 0.81783
Omeprazole sodium was used for the determination of the standard curve. The
marketed product, “OMEZ INSTA” label claim states that it contains only omeprazole.
Therefore a correction with respect to the weight of sodium was performed and
Method:
Using the above relationship, the concentration of omeprazole was calculated from the
below.
1 0 µg/ml 0 µg/ml
1. 0 0
2. 1.875 0.05275
3. 3.750 0.10575
4. 5.625 0.16025
5. 7.501 0.21125
6. 9.376 0.261
5.3 EVALUATION
F 22 0.70 1.14
27.35 0.80 12.50
Table No. 5.3.1.5 Evaluation of different powder blend prepared by treated natural
gums used as Superdisintegrants.
FC Angle of repose LBD(gmc/m3) TBD(gm/cm3) CI (%) HR
F 17 0.38±0.20 1307.5±65.38
7.03±0.01 3.3±0.1
F 19 1303.5±65.18
7.05±0.01 3.47±0.06 0.33±0.09
* All the parameters shown above are based on 3 replicates and are expressed as Mean
±S.D
Uniformity of Weight
FC Hardness (kg/cm2) Friability %
Thickness(mm) Variation (mg)
* All the parameters shown above are based on 3 replicates and are expressed as Mean
±S.D
* All the parameters shown above are based on 3 replicates and are expressed as Mean
±S.D
* All the parameters shown above are based on 3 replicates and are expressed as Mean
±S.D
Disintegration plays an important role for ODTs as the drug release and its pattern
purely depends on the disintegration time. For ODTs shortest disintegration time is
desired as the dosage form should be capable of being immediately absorbed with the
disintegrated portion reaching the gastric fluid in seconds without the requirement of
Disintegration time of 3 min (180 sec) was fixed as the upper limit for branding
having disintegration time greater than 180 sec were not selected for further studies such
subliming agents, and treated natural gums to be used as superdisintegrants did not yield
No limits for the drug content are presently available in the pharmacopoeias for
PPI prepared as ODTs. Therefore, IP 2010 limits of 90 to 110% for enteric coated tablets
The drug content in the selected formulation was found to be in the range of
90.94% to 100.94% with a standard deviation of ± 0.18 to ± 0.60. This lies within the IP
Drug dissolution profile is one of the important aspects in development of all the
dosage forms. In this research project dissolution time of 10 min was chosen since the
expected dissolution time for ODT was around 5 min. In addition, IP has prescribed a
dissolution time of 10 min for Ondansetron DTs. Certain modification where done as
explained in the 4.5.2.8 In vitro dissolution studies under the topic called methodology.
All the formulation which passed the in vitro disintegration where subjected to in
vitro dissolution studies. This study also plays an important part in the selection of the
5.3.5.1 Dissolution drug profile of ODT formulations prepared by superdisintegrant addition method
Table No. 5.3.5.1 Cumulative percent drug release from Orodispersible tablets prepared by superdisintegrant addition method
Time
F2 F3 F4 F6 F7 F9 F10 F13 F16
(mins)
0 0±0 0 ±0 0 ±0 0 ±0 0 ±0 0 ±0 0 ±0 0 ±0 0 ±1.523
0.5 33.03 ± 3.18 87.65 ±2.17 95.35 ±0.72 20.69 ±5.73 40.91 ±2.78 37.18 ±1.57 47.00 ±1.78 3.97 ±1.93 8.10 ±1.5
1 44.52 ±1.40 93.59±2.10 102.52±0.23 31.06 ±2.24 75.77 ±3.26 60.45±5.28 67.64 ±4.05 7.60 ±1.70 11.81 ±1.96
1.5 73.11 ±3.35 97.34 ±.084 102.12±0.60 45.14±1.89 87.96 ±2.64 78.42 ±3.78 82.74 ±4.63 18.46 ±1.94 25.22 ±1.01
2 82.14 ±3.06 98.87 ±1.25 101.80±0.70 52.25 ±2.55 98.64 ±1.06 89.76 ±5.01 84.84 ±5.58 24.82 ±3.82 30.80 ±1.10
2.5 87.26 ±1.82 92.99 ±0.91 100.46±0.91 59.28 ±2.99 102.18±3.13 94.51±2.40 84.55 ±1.37 20.62±10.88 40.71 ±2.52
3 90.35 ±1.61 88.14 ±1.55 99.81 ±1.49 69.67 ±3.40 99.71 ±3.21 89.42 ±2.49 85.54 ±2.22 23.57±12.65 46.10 ±2.26
3.5 92.75 ±1.63 83.57 ±2.13 98.39 ±2.06 80.87 ±6.78 98.65 ±1.98 87.58 ±3.50 85.76 ±3.42 34.84 ±3.90 51.36 ±2.70
4 91.93 ±0.58 76.23 ±2.95 97.12 ±3.10 94.17 ±1.48 98.05 ±0.65 86.52 ±2.27 83.47 ±2.57 38.41 ±3.99 59.99 ±3.07
4.5 92.69 ±0.85 71.00 ±1.27 96.30 ±3.27 97.06 ±0.91 97.99 ±1.46 85.75 ±2.15 81.51 ±4.41 42.42 ±3.66 66.20 ±2.96
5 91.60 ±2.51 68.99 ±1.67 93.97 ±3.27 96.42 ±1.95 97.80 ±1.13 85.35 ±1.86 79.53 ±2.82 48.94 ±1.04 82.70 ±8.15
5.5 90.03 ±2.72 66.43 ±1.40 92.41 ±2.75 95.70 ±1.28 97.16 ±0.85 85.71 ±1.66 79.32 ±3.31 54.28 ±2.28 85.37 ±3.37
6 90.73 ±1.95 63.66 ±2.15 89.92 ±2.82 94.99 ±1.19 96.37 ±0.84 84.52 ±1.82 78.98 ±2.83 59.95 ±2.29 84.72 ±4.68
6.5 88.97 ±2.16 61.16 ±1.78 87.61 ±2.46 94.13 ±1.44 97.37 ±1.69 84.64 ±2.31 79.11 ±3.62 66.11 ±0.99 89.73 ±1.08
7 88.52 ±2.18 58.49 ±2.10 84.96 ±1.57 92.95 ±2.40 95.62 ±2.08 85.90 ±1.69 77.94 ±2.36 70.70 ±1.55 92.11 ±1.15
7.5 88.64 ±2.35 55.34 ±3.18 85.06 ±1.34 91.36 ±2.14 95.10 ±1.68 85.53 ±1.43 77.89 ±2.35 75.89 ±1.57 90.38 ±1.78
8 89.20 ±2.81 53.65 ±2.15 84.04 ±1.14 90.33 ±1.60 94.29 ±2.64 84.85 ±1.30 77.21 ±1.79 82.56 ±1.46 92.20 ±2.76
8.5 87.92 ±2.87 51.47 ±2.95 83.30 ±1.56 90.18 ±1.87 93.33 ±2.71 84.36 ±0.98 75.72 ±1.36 84.30 ±2.35 90.04 ±1.95
9 88.68 ±2.36 49.30 ±3.27 82.10 ±1.50 88.63 ±1.80 93.39 ±4.62 85.20 ±1.27 75.80 ±1.78 84.93 ±3.79 90.41 ±3.76
9.5 88.16 ±2.61 47.55 ±3.49 81.14 ±0.86 87.63 ±3.29 92.55 ±5.36 85.46 ±1.59 74.88 ±2.17 84.79 ±3.03 87.39 ±4.20
10 88.224±2.72 45.27 ±3.78 80.56 ±1.21 87.76 ±4.44 90.88 ±4.58 85.06 ±1.68 75.99 ±2 86.52 ±3.12 84.34 ±3.43
* Here n=6 for F3 and F4 formulations and n=4 for rest of all formulations (F2, F6, F7, F9, F10, F13, F16
Fig No. 5.3.5.1 Cumulative percentage drug released from Orodispersible tablet
formulations prepared by superdisintegrant addition method
Table No. 5.3.5.2 Cumulative percent drug released from Orodispersible tablet
formulations prepared by combination of different superdisintegrants
Time
F17 F18 F19 F20
(min)
0 0 ±0 0 ±0 0 ±0 0 ±0
0.5 2.79±1.29 33.09 ±0.97 22.96 ±4.28 36.15 ±2.25
1 34.49 ±2.70 57.63 ±5.58 43.03 ±2.2 50.01 ±1.11
1.5 49.86 ±1.56 79.65 ±3.99 80.96 ±3.39 77.28 ±7.86
2 61.99 ±0.42 85.95 ±2.52 92.70 ±1.95 86.85 ±2.30
2.5 72.60 ±1.57 90.60 ±3.19 98.08 ±2.20 93.134 ±2.50
3 81.64 ±1.73 92.57 ±2.74 98.14 ±0.86 94.53 ±1.68
3.5 88.01 ±1.50 95.17 ±1.09 98.37 ±1.36 95.42 ±1.08
Fig No. 5.3.5.2 Cumulative percentage drug released from Orodispersible tablet
formulations prepared by combination of different superdisintegrants
Based on the disintegration time and the release patterns, formulations coded F4
and F3 were selected as the optimum formulations which fulfil best the criteria for ODTs.
OMEZ INSTA was used as a reference for the evaluation of the similarity in
Table No.5.3.5.4 Comparative table showing cumulative percent drug release of the
selected formulations F4 and F3 with marketed formulation OMEZ INSTA
Standard
Time (mins) F3 F4
Formulation
0 0 ±0 0 ±0 0 ±0
0.5 87.65 ±2.17 89.90 ±3.43 95.35 ±0.72
1 93.59±2.10 98.11 ±2.71 102.52±0.23
1.5 97.34 ±.084 100.27 ±1.29 102.12±0.60
2 98.87 ±1.25 101.95 ±1.92 101.80±0.70
2.5 92.99 ±0.91 101.57 ±1.58 100.46±0.91
3 88.14 ±1.55 101.12 ±0.84 99.81 ±1.49
3.5 83.57 ±2.13 100.36 ±1.57 98.39 ±2.06
4 76.23 ±2.95 101.42 ±0.98 97.12 ±3.10
4.5 71.00 ±1.27 100.36 ±2.14 96.30 ±3.27
5 68.99 ±1.67 100.53 ±2.13 93.97 ±3.27
Fig No. 5.3.5.4 table showing cumulative percent drug release of the selected
formulation F4 and F3 and marketed formulation OMEZ INSTA
as per the USP guidelines, both these formulations are similar to the reference
formulation.
Parameters
FC Time Hardness Drug DT
CPDR Friability Appearance
(Kg/cm2) content (sec)
Thus the selected formulations pass the stability test since none of the examined
parameters are outside the respective acceptance limits prescribed by ICH guidelines..
6 DISCUSSION
In the present work, an attempt was made to prepare an ODTs of PPIs. ODTs have
an advantage over the conventional tablets in elderly, pediatric and patients with
dysphagia. Pantoprazole sodium was used as a model drug to represent the class of PPIs.
The main challenge of this research was to deal with the stability of the drug. All PPIs are
unstable at a pH below 5 and the project involved delivery of the drug into stomach with a
pH of 1.2. The degradation half life of the drug in the stomach is 1-2 min. No UV
The pre and post compression parameters of all the formulation were evaluated.
varying ratios on various pre and post parameters were evaluated using parameters such
specific characteristic of PPIs. A similar observation was also made in this set of
Thus, determination of the melting point of pantaprazole sodium was not possible as
expected.
6.1.2 Solubility
buffer of 6.8 and 7.4. pantoprazole sodium shows a clear solution in stimulated gastric
pH 1.2 but also show a characteristic yellow coloration when PPIs are dissolved in acidic
pH. The coloration intensifies as the pH decreases. Thus this yellow coloration could
tricky and complex procedure for PPIs. Many methods were tried but the main problem
associated with the standard curve preparation was its stability. As stated in the
is just 1 to 2 mins. Therefore, as soon as the drug was added into the acidic medium
degradation sets in. Thus it was contemplated to add buffer along with it but then a new
problem arose. When buffer was dissolved in1.2 pH the drug was not getting solubilized
even after sonication for 1 hr. A different method was adopted by adding of 5 ml of
purified water into 100 ml of solution of HCl, pH 1.2. The pH of the solution remained
unchanged around 1.2; within the expected limits. Thus a method involving dissolution
of the drug in 5ml of the purified water, followed by addition of this solution to the
bicarbonate dissolved in 1.2 pH, and further making up the volume to 100 ml using 1.2
pH HCl (SS-I) was followed. Other stock solutions were prepared by dilution of SS-I
The scanning of drug solution was done in the UV region (200–400 nm) to find
out the wavelength of maximum absorption (λ max) in the gastric fluid pH 1.2. The λ max
was found to be at 281.5 nm. The standard calibration curve of pantoprazole sodium was
developed at this wavelength. The calibration curve was linear between 1–100 µg/ml
conc. ranges. The standard calibration was obtained by plotting absorbance against conc.
at 281.5 nm, and it follows the Beer‘s law. Results were tabulated in Table No.5.1.2 and
plotted in Fig. no.5.1.2 The r2 was found to be 0.9994 and slope was found to be 0.032 in
0.1 N HCl.
The scanning of drug solution was done in the UV region (200–400 nm) to find
out the wavelength of maximum absorption (λ max) in the gastric fluid pH 1.2. The λ max
was found to be at 301 nm. The standard calibration curve of omeprazole sodium was
absorbance against conc. at 301 nm, and it follows the Beer‘s law. Results were tabulated
in Table No.5.1.3 and plotted in Fig. no.5.1.3 The r2 was found to be 0.9998 and slope
Drug- excipient interactions play a crucial role with respect to the stability and-
potency of the drug. FTIR techniques have been used here to study the physical and
spectrum of pure pantoprazole sodium and physical mixture of drug and polymers were
studied. The FTIR scan shows characteristic absorption peaks of pantoprazole sodium at
3363.97 cm-1, 1450.52 cm-1 (N-H stretching and bending vibrations); 1303.92 cm-1 (C-N
stretching vibration); 1589.40 cm-1 and 823.63 cm-1 (C=C stretching vibration);
3057.27cm-1 (Aromatic C-H stretching and bending vibrations), 1041.60 cm-1 (S=O
The FTIR spectrum of pure drug and different excipients and the tablet formulation
The peaks obtained in the spectra of the pure drug have a correlation with the
peaks obtained when the drug and excipients were scanned together, thus indicating that
Based on this study it was concluded that there is no chemical interaction between
drug and the excipients used and thus it can be safely used in the formulations.
LBD of the formulation blend plays an important role in the compression of the
powder the LBD of the formulation was found to be in the range of 0.679 g/cm3 to
0.719 g/cm3.
formulation blend it was found to be in the range of 0.69g/cm3 to 0.83 g/cm3. It was noted
that the TBD of all the formulation where greater than their respective LBD thus
The angle of repose for the formulated blend was carried out and the results were
shown in Table no. 5.2.2.1 to 5.2.2.5. It was concluded that the entire formulations blend
were in the range 230491' to 29⁰511' thus falling in the official limit range of 250 to 300
which indicates that all the formulation blend have good flow property.
CI was calculated on the basis of the LBD and TBD and the results were shown in
table 5.2.2.1 to 5.2.2.5. It was found to be in the range of 11.32% to 14.55% which lies in
the official limits i.e. 5% to 15%, indicating the granules blend has excellent flow
HR was calculated on the basis of the LBD and TBD. It is an ratio between TBD
and LBD and was found to be in the range of 1.127 to 1.17 thus indicating that
formulation blend have free flowing property which is ideal for ODTs.
For the formulation of ODTs the most easy and economical method direct
compression, was selected. This is one of the most widely used methods. Five
approaches were designed to be used for this research work: superdisintegrant addition,
preparation of ODTs. Mannitol DC, SSG and other excipients were chosen based on the
Due to the use of high amount of buffer which is always associated with bad taste,
Orange DC100 was incorporated as a flavour in the concentration of 0.5% to mask this
effect. All the superdisintegrants were used in the concentration of 2%, 3%, and 5%
respectively.
Blending time was carefully controlled since SSF was used. Long mixing times can
result in the formation of hydrophobic powder beds which do not disperse easily.
Therefore, SSF was added and mixed for a period of only 2 to 5 minutes. An increase in
the coefficient of variation of mixing and a decrease in the dissolution rate have been
observed following long mixing periods of SSF with a tablet powder. Tablet dissolution
rate and crushing strength is decreased as the time of blending is increased; and SSF may
The uniform blends of tablet were directly compressed by keeping tablet press
setting constant for all the formulations. Proper lubrication of powder blends was
essential for ease of ejection of compressed tablets as well as for free movement of lower
punch during compression cycle. The tablets were compressed using flat face 16.4 X 8
mm flat oval size punches to get tablets of 1300 mg weight using ten stations Rimek
During the course of this research, a lot of questions arose due to various
perspectives of this research work. First the traditional method was considered for
performing the dissolution in 900 ml of dissolution media, but the results were
unsatisfactory. The quantity of buffer used was then considered. Further review showed
that in IP 2010 in the assay for Ondansetron orally disintegrating tablets dissolution was
use 500 ml of dissolution medium. The percentage of drug released using 500 ml of
dissolution medium was more than that obtained using 900 ml, although the cumulative
percent released was less than 72%. Further review of the literature revealed that a
marketing pamphlet existed for ―OMEZ INSTA‖ which quoted that the marketed product
Thus based on back calculation it was determined that 190 ml of acid should be
present to increase the pH above 6. Further it was seen that good similarity existed
between the dissolution profiles obtained with the test formulations and the marketed
formulation of OMEZ INSTA when both 900 ml and 500 ml of dissolution medium was
used. The temperature was validated and kept at 40.1°C and the rotation of the basket
The photograph shown above (fig. 6.5) of the dissolution study done in 500 ml
dissolution medium for OMEZ INSTA clearly shows the yellow colouration indicating
A combination of two buffers i.e. Sodium and potassium bicarbonate were used
for stabilizing agents so that the level of sodium will be maintained. The quantity of 1.1
gm of the buffer mixture consisting of sodium and potassium bicarbonate was arrived at
by a series of in vitro dissolution tests. The least amount of buffer mixture required to
that the intensity of the yellow colouration is minimal with both the F3-CP 3% and F4-
degradation of PPIs indicating the correct choice of the optimum amount of buffer.
The colour of the tablet was white and formulation prepared by addition of treated
natural gums shows specific brown to black colouration depending on the colour of the
6.8.3 Thickness
The crushing strength of the tablets of each batch ranged between 3.13 to 4.23
The values of friability test were in the range from 0.21 to 0.64% the percent
friability of all the formulation was less than 1% ensuring that the tablets were
mechanically stable.
The percentage weight variations for all formulations were done. All the
formulated tablets passed weight variation test as the percent weight variation was within
the pharmacopoeial limits as the formulation blend of all the formulations have a good
flow thus the percent weight deviation was in between 5% of the average weight. The
weights of all the tablets were found to be uniform with low standard deviation values.
The percentage of drug content for all formulation was found to 91.98% to
100.94% which lies in the IP limit for enteric coated formulation of 90 to 110% which
was taken into consideration as ODTs of pantoprazole sodium is not official in any
pharmacopoeia.
This is the most important test with respect to ODT formulations. Five
superdisintegrants, three subliming agents and, two treated gums were used for these
studies. Among all CP was selected as the best superdisintegrant as it gave the least in
vitro disintegration time. It was noted that no superdisintegrant was able to give the in
vitro disintegration time with in 180 sec at a cocn. of 2% except CP. As the cocn. of CP
was increased in vitro disintegration time reduced; least in vitro disintegration time was
effect on in vitro disintegration time only when CP was used. Sublimation method totally
failed the disintegration test perhaps due to the cocn. used (5%) and the size and weight
of tablet. When superdisintegrants were combined with subliming agents it was strange to
see that even when the concentration of CP used was 2.5% it did not give the in vitro
disintegration time with in the specified limit of 3 mins. At the start of the test some part
of the tablet disintegrated quickly possibly due to the presence of CP on the surface, but
not the whole tablet. Thus we can conclude that 2.5 to 5% of subliming agents are not
Natural gums like Acacia and Gaur were selected as they gave a proper dried
mass after the treatment. However, they failed to give in vitro disintegration time with in
the specified limit thus indicating that 5% of the treated natural gums are not sufficient
All the selected formulations which passed the in vitro disintegration test were
subjected to in vitro release studies using modified USP dissolution apparatus II in 0.1N
HCl pH 1.2.
Depending on the in vitro disintegration test, in vitro dissolution test and the
mins and for the formulation F4 the maximum drug release (102.52 ± 0.23) was found
within 1 minute. These results are in tune with those obtained for the disintegration time
The selected formulation were subjected to accelerated stability studies and the
formulations where evaluated for appearance, hardness, friability, drug content, in vitro
disintegration time and in vitro dissolution test. The formulations where stored at 40°C
±1°C and RH 75 ± 5%. All the formulation where analysed after every 15, 30, 45 and 90
days. All the formulations show no change in all the above parameters thus successfully
passes the accelerated stability study which was conducted for 90 days.
7. CONCLUSION
The in vitro disintegration test revealed that the tablets prepared with CP, and
prepared with rest of superdisintegrants, subliming agent and treated natural gums used
as Superdisintegrants.
Even the dissolution studies confirmed that tablets prepared with CP, and its
mixture show faster drug release as compared to tablets prepared with rest of
superdisintegrants, subliming agent subliming agent and treated natural gums used as
Superdisintegrants.
The flow properties of the formulation powder have good flow property which is
Direct compression method is the best method for the formulation of ODTs. This
method is also very economical and time saving. CP was found to be the best
superdisintegrant among all with 5 percent concentration yielding the best results. It was
also concluded that 1.1 gm of bicarbonate is required for the stability of the PPIs in acidic
conditions.
In vitro disintegration time and in vitro drug release shows that among all the
superdisintegrants used CP gives the least in vitro disintegration time and release the
maximum amount of drug within 1-3 mins. The results show that an increase in CP level
leads to a decreases in the in vitro disintegration time and thus decrease in the in vitro
drug release time. Thus formulation F4 and F3 were selected as best formulations among
those examined.
Stability studies reviled that the formulation F3 and F4 i.e. formulation with 3%
8. SUMMARY
Mankind has lived with peptic ulcers since ancient times. Perhaps the first
description of this malady is the one inscribed on the pillars of the temple of Aesculapius
PPIs are the most potent suppressors of gastric acid secretion and are inhibitors
doses, these drugs diminish the daily production of acid (basal and stimulated) by 80% to
95%. PPIs are widely used for treating acid induced inflammation, ulcers of the stomach
critically ill patients, and Zollinger-Ellison Syndrome. They are also used in combination
longer lag time as they are enteric coated and fluctuations in the plasma drug levels,
Thus ODTs can act as a dosage form which can nullify all the drawbacks and
Present work was an attempt in designing ODTs for a drug which have
The system can be further worked upon to give a more specific release by using
different superdisintegrants.
In-vivo study can be carried out in animals for better prediction of in vivo
behaviour using models such as pyloric ligation method, chemical induced ulcer,
drug by the ODT product as compared to the conventional oral PPIs preparations.
9. BIBLIOGRAPHY
1. Edwards LD, Fletcher AJ, Fox AW. Principles and practice of pharmaceutical
2. Lachman L, Liberman HA, Kaing JL. The theory and practice of industrial pharmacy.
3. Liberman HA, Lachman L, Schwartz JB. Pharmaceutical dosage forms tablet volume
4. Ghosh TK, Pfister WR. Drug delivery to the oral cavity molecules to market. U.S.A:
(2): 9-12.
10. http://www.pharmpedia.com
11. Mishra DN, Bindal M, Singh SK, Kumar SGV. Rapidly disintegrating tablets of
13. Devi VK, Asha AN, et al. Oral dispersible Fluconazole tablets - preparation and
14. Mahajan HS, Kuchekar BS, et al. Mouth dissolve tablets of Sumatriptan Succinate.
15. Kuchekar BS, Badhan AC, et al. Mouth dissolving tablets of Salbutamol Sulphate: a
16. Nayak SM, and Gopalkumar P. Design and optimization of fast dissolving tablets for
17. Patel DM, Patel NM, et al. Studies in formulation of ODTs of Rofecoxib. Ind. J.
18. Desai SA, Ferade SV. Orodissolving tablets of Promethazine Hydrochloride. Ind. J.
19. Patel MM, and Patel DM. Fast dissolving Valdecoxib tablets containing solid
20. Poornima D, Anita R, et al. Patient compliant dosage form for Roxithromycin. Ind. J.
21. Gilman AG. The Pharmacological basis of therapeutics. 10th ed. NewYork: McGraw-
22. Tripathi KD. Essential of Medicinal Pharmacology 6th ed. New Delhi: Jaypee
23. Wilson, Gisvold, Block JH. Textbook of organic medicinal and pharmaceutical
chemistry. 11th ed. Philadelphia: Lippincott Williams and Wilkins; 2004: p. 722-27.
24. Tortora GJ, Grabowski SR. Principles of anatomy and physiology. U.S.A : John
25. http://www.cims.com
26. Guyton AC, Hall JE. Textbook of Medical Physiology. Singapore: Harcourt Brace
27. Jungnickel PW. Pantoprazole: A new proton pump inhibitor clinical therapeutics
28. Bell W, Staar U and Sewing KF. Pantoprazole: a novel H+/K+-ATPase inhibitor with
29. Li XQ et al. Comparison of inhibitory effects of the proton pump inhibiting drugs
cytochrome p450 activities. The Amer. Soc. for Pharmacol. and Expt. Therap. 2004;
32 (8): 821-7.
31. Raghunath AS et al. a review of the clinical and economic impact of using
32. Numans ME et al. Short-term treatment with proton-pump inhibitors as a test for
705–711.
1133-42.
and Pantoprazole Sodium Sesquihydrate. J. Pharma. Biomed. Ana. 2000; 22: 45–58.
37. Ramakrishna NVS et al. High-performance liquid chromatography method for the
40. Choi HG, Kim CK, et al. Development of omeprazole buccal adhesive tablets with
stability enhancement in human saliva. Journal of Controlled Release 2000; 68: 397-
404.
41. Choi HG, Jung JH, et al. Formulation and in vivo evaluation of omeprazole buccal
42. Gohel MC, Mehta PR, Dave RK, Bariya NH. A more relevant dissolution method for
43. Garcia CV, et al. Development and validation of a dissolution test for rabeprazole
41: 833-7.
44. Bladh N, et al. A new esomeprazole packet (sachet) formulation for suspension: in
45. F. Baldi, et al. Lansoprazole fast disintegrating tablet: a new formulation for an
46. Silver DI, et al. Lansoprazole orally disintegrating tabets. US 2007/0202169 A1.
47. Kundu S and Sahoo PK. Recent trends in the developments of orally disintegrating
48. Bhowmik D, et al. Fast dissolving tablet: an overview journal of chemical and
49. Hirani JJ, et. al. Orally disintegrating tablets: a review. Tropical Journal of
masking and clinical studies. Therapeutic Drug Carrier Systems 2004; 21(6): 433–75.
51. Fini A, et al. Fast dispersible/slow releasing ibuprofen tablets. European Journal of
52. Schiermeier S. and Schmidt PC. Fast dispersible ibuprofen tablets. European Journal
A1.
54. Puttewar TY. et al. Formulation and evaluation of orodispersible tablet of taste
masked doxylamine succinate using ion exchange resin. Journal of King Saud
56. Shoukri RA, et al. In vitro and in vivo evaluation of nimesulide lyophilized orally
73: 162–71.
58. Goverment of India ministry of health and family welfare. The Indian Pharmacopoeia
59. http://www.rxlist.com/protonix-drug.htm
60.http://en.winkipedia.org/winki/pantoprazole_sodium
61. Rowe RC, Sheskey PJ and Owen SC. Handbook of Pharmaceutical Excipients. 5th
62. Niazi SK. Hand book of preformulation. New York; informa health care; 2007: p. 1-
7.
63. Patel DM. et al. Optimization of fast dissolving Etoricoxib tablets prepared by
64 Sinko PJ. Martin‘s Physical Pharmacy and Pharmaceutical Sciences. 5th ed.
65. Aulton ME. Pharmaceutics the science of dosage form design. 2nd ed. Philadelphia:
66. Gohel MC, Sarvaiya KG, Shah AR, Brahmbhatt BK. Mathematical approach for the
assessment of similarity factor using a new scheme for calculating weight. Indian J
10. ANNEXURES
ERRATA