SlideShare a Scribd company logo
FORMULATION AND EVALUATION OF GASTRO-
RETENTIVE MUCOADHESIVE MICROBALLONS OF
NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER
Dissertation Part-II report submitted to
RAJIV GANDHI PROUDYOGIKI VISHWAVIDYALAYA, BHOPAL (M.P.)
For the partial fulfillment of the degree of
MASTER OF PHARMACY
Pharmaceutics
Supervised by
Dr.SarangJain
Principal
Submitted by
PARMANAND DHAKAD
Enroll.no 0148PY16MP09
Co-GUIDE
Ms. Swati Saxena
Associate Professor
RAJEEV GANDHI COLLEGE OF PHARMACY,BHOPAL
SESSION2020-2021
RAJEEV GANDHI COLLEGE OF PHARMACY
Village: Salaiya, Via Danish Kunj, Kolar Road, Bhopal
Department of Pharmaceutics
CERTIFICATE
This is to certify that the dissertation entitled “FORMULATION AND EVALUATION
OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF
NIZATIDINE FOR MANAGEMENT OF
PEPTIC ULCER” submitted to Rajiv Gandhi Proudyogiki Vishwavidhyalaya, Bhopal
by Mr. Parmanand Dhakad is a partial fulfillment of the requirement for the award of
the degree of the M.Pharm. with specialization in Pharmaceutics. The matter embodied
is the actual work by Mr. Parmanand Dhakad and this work has not been submitted
earlier in part or full for the award of any otherdegree.
PARMANAND DHAKAD
Enroll. No. 0148PY16MP09
Dr. Sarang Jain
Principal
Ms. Swati Saxena
Associate Professor
RAJEEV GANDHI COLLEGE OF PHARMACY
BHOPAL
DECLARATION
I hereby declare that the work, which is being presented in the dissertation, entitled
“FORMULATION AND EVALUATION OF GASTRO- RETENTIVE
MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT
OF PEPTIC ULCER” submitted
by Mr. Parmanand Dhakad (0148PY16MP09) for the award of degree of “Master of
Pharmacy” degree with specialization in Pharmaceutics, comprises of bonafide research
work carried out by me in our laboratories, library and computer centre under the
guidance of Dr. Sarang Jain, Principal and Ms Swati Saxena, Associate Professor.,
Department of Pharmaceutics, Rajeev Gandhi College of Pharmacy, Bhopal.
I also declare that the present work embodies has not formed the basis for the award of
any other degree or fellowship previously. The particulars given in this thesis are to best
of myknowledge.
Date
Place: Bhopal
PARMANAND DHAKAD
Enroll. No. 0148PY16MP09
RAJEEV GANDHI COLLEGE OF PHARMACY
BHOPAL
DECLARATION
I hereby declare that the work, which is being presented in the dissertation, entitled
“Formulation And Evaluation of Gastro-retentive Mucoadhesive Microballons of
Nizatidine For Management of Peptic Ulcer” partial fulfillment of the requirements
for the award of degree of Master of Pharmacy in Pharmaceutics submitted in the
department of Pharmaceutics (Rajeev Gandhi College of Pharmacy, Bhopal M.P.) is an
authentic record of my own work carried under the guidance of Dr.Sarang Jain and Ms
Swati Saxena. I have not submitted the matter embodiedin this report for award of any
otherdegree.
I also declare that “A check for Plagiarism has been carried out on the thesis/ project
report/ dissertation and is found within the acceptable limit and report of which is
enclosed herewith.”
Name of Student
PARMANAND DHAKAD
Enroll. No. 0148PY16MP09
Name of Supervisor
Dr. Sarang Jain
Principal
Name of Co-Supervisor
Ms. Swati Saxena
Associate Professor
ACKNOWLEDGEMENT
Firstly, I offer my adoration to God who created me, gave me the strength
and courage to complete my dissertation and gave me the opportunity to thank all those
people through whom his Grace was delivered to me.
With a deep sense of gratitude, I express my indebtedness to my guide, Dr.
Sarang Jain, Principal Rajeev Gandhi college of Pharmacy, Bhopal, for his valuable
guidance, boundless enthusiasm and constant inspiration throughout the entire course of
the work. I shall forever remain indebted to his for having inculcated in me a quest for
excellence, a spirit of diligenceIt is my privilege to express my heartfelt thanks to my Co-
guide Ms Swati Saxena, Department of Pharmaceutics, for her guidance and
encouragement throughout the research work.
I sincerely thank all the teaching staff and non-teaching staff, Mr Navneet
Dube, Mr Anuj Singhai, Mr. Bajpei, Mr.Pradeep and librarian Mr. Dharmendra. It
gives me immense pleasure to record my sincere thanks to my colleagues friends& Non
collegeous friends Risabh mishra, Sumit Raikwar, Nidhi Malviya ,and for their help and
co- operation.
I sincerely acknowledge the authorities of Rajeev Gandhi college of
Pharmacy, Bhopal, for providing the necessary facilities to carry out my research
work. I would like to thank all those who have helped me directly or indirectly to
complete this work successfully.
From deepest depth of my heart, I express my love and gratitude to my
beloved parents Mr Raghuraj Singh Dhakad , Mrs. Shusila Bai and all family members
for their love, support and constant encouragement throughout my career.
Thank you one & all
Date
Place: Bhopal
PARMANAND DHAKAD
Enroll. No. 0148PY16MP09
[not detected]
[not detected]
GoogleBooks Detected!
Wiki Detected!
Anti-cheating:
Ghostwriting services:
Google Books:
Wikipedia:
Plagiarism Detector v. 1517 - Originality Report
Analyzed document: 30-09-2020 16:51:52
" FORMULATION AND EVALUATION OF GASTRO-
RETENTIVE MUCOADHESIVE MICROBALLONS OF
NIZATIDINE FOR
MR.
MANAGEMENT OF PEPTIC ULCER”
PARMANAND DHAKAD
Enroll. No. 0148PY16MP09 M PHARM IV.docx
Check Type: Internet - via Google and Bing
Licensed Version
Relation chart:
Important notes:
Plagiarism Detector
Your right to know the authenticity!
CONTENT
Chapter Title Page no.
1 INTRODUCTION 1-24
1.1 Peptic ulcer 1
1.2 Drug Delivery System 6
1.3 Gastro-retentive drug delivery system 9
1.4 Mechanism of Bioadhesion 15
1.5 Mucoadhesive Microspheres 23
2 REVIEW OF LITERATURE 25-48
2.1 Drug profile 25
2.2 Excipients review 26
2.3 Literature review 39
3 PLAN OF WORK & HYPOTHESIS 49-50
3.1 Objective 49
3.2 Hypothesis 49
3.3 Plan of present work 49
4 MATERIAL AND METHODS 51-59
4.1 Analytical and Validation studies 51
4.2. Preformulation Studies 52
4.2.1 Organoleptic properties 52
4.2.2 Microscopic examination 52
4.2.3 Physical Characteristics 52
4.3 Preparation of mucoadhesive Microballons 55
4.4 Evaluation of mucoadhesive microballons 57
5 RESULT AND DISCUSSION 60-60
5.1 Analytical and Preformulation studies of model drug 60
5.2 Preformulation Studies 61
5.3 Evaluation of mucoadhesive microspheres 65
6 Summary and conclusion 81-82
7 References 83-67
LIST OF TABLES
Table
No.
Particulars Page No.
1.1 Routes of drug administration and dosage form / drug delivery systems 6
1.2 Different theories explaining the mechanism of bioadhesion 15
1.3 Bioadhesive strength of some polymers 21
1.4 Application of Bioadhesive Systems 22
1.5 Patents related to bioadhesive system 23
4.1 Drug-excipient combinations for compatibility study 55
4.2 Preparation of of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 56
5.1 Organoleptic characteristics of Nizatidine HCl 62
5.2 Flow properties of drug (n = 3) 62
5.3 The solubility of Nizatidine HCl at different pH medium (n=3) 62
5.4 Drug-excipient combinations for compatibility study 63
5.5 Results of physical observation 63
5.6 Results of content determination 63
5.7
Percentage yield of mucoadhesive microballons of Nizatidine HCl (A1 –
B3)
67
5.8 Particle size of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 67
5.9
Drug entrapment efficiency of mucoadhesive microballons of Nizatidine
HCl (A1 – B3)
68
5.10
Degree of swelling of mucoadhesive microballons of Nizatidine HCl (A1 –
B3)
68
5.11
Percent mucoadhesion of mucoadhesive microballons of Nizatidine HCl
(A1 – B3)
69
5.12 Buoyancy test of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 70
5.13
LIST OF FIGURES
70
Dissolution data of mucoadhesive microballons of Nizatidine HCl (A1 –B3)
5.14 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(A1)
71
5.15
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(A2)
72
5.16
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(CH1)
73
5.17
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(CH2)
74
5.18
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(CA1)
75
5.19
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(CA2)
76
5.20
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(B1)
77
5.21
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(B2)
78
5.22
in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl
(B3)
79
Figure
No.
Particulars Page No.
1.1
(a) Microspheres floats on stomach contents. (b & c) microspheres adhere
to stomach wall
14
1.2 Three regions within a mucoadhesive joint 16
2.1 Structure of drug Nizatidine 25
2.2 Chemical structure of microcrystalline cellulose 27
2.3 Chemical structure of anhydrous lactose 29
2.4 Chemical structure of sodium starch glycolate 30
2.5 Chemical structure of starch 32
2.6 Chemical structure of guar gum 35
2.7 Chemical structure of cellulose hydroxypropyl methyl ether 38
5.1
Absorption maxima (λ-max) of Nizatidine HCl in 0.1N HCl solution (10
μg/ml)
60
5.2 Standard curve of Nizatidine HCl in 0.1N HCl solution (228 nm) 61
5.3 The I. R. Spectrum of sample of pure Nizatidine HCl (S1) 64
5.4 The I. R. Spectrum of sample of Nizatidine HCl and all excipients (S2) 64
5.5 Photograph of microspheres (100X) 66
5.6 SEM photomicrograph of microspheres (650X) 66
5.7
Zero-order plots for mucoadhesive microballons of Nizatidine HCl (A1 –
B3)
80
5.8
First-order plots for for mucoadhesive microballons of Nizatidine HCl (A1
– B3)
80
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 1
1: INTRODUCTION
Peptic Ulcer
Drug Delivery System
Gastro-retentive Drug Delivery System
Mechanism of Bioadhesion
PEPTIC ULCER
Peptic ulcer occurs in that part of the gastrointestinal tract (g.i.t.) which is exposed to
gastric acid and pepsin, i.e. the stomach and duodenum. The etiology of peptic ulcer is
not clearly known. It results probably due to an imbalance between the aggressive (acid,
pepsin, bile and H. pylori) and the defensive (gastric mucus and bicarbonate secretion,
prostaglandins, nitric oxide, high mucosal blood flow, innate resistance of the mucosal
cells) factors. A variety of psychosomatic, humoral and vascular derangements have been
implicated and the importance of Helicobacter pylori infection as a contributor to ulcer
formation and recurrence has been recognized. In gastric ulcer, generally acid secretion is
normal or low, while deficient mucosal defence (mostly impaired mucus and bicarbonate
secretion) plays a greater role. In duodenal ulcer, acid secretion is high in about half of
the patients but normal in the rest. Notwithstanding whether production of acid is normal
or high, it does contribute to ulceration as an aggressive factor, reduction of which is the
main approach to ulcer treatment. An understanding of the mechanism and control of
gastric acid secretion will elucidate the targets of antisecretory drugaction.
An ulcer is a round or oval shaped hole (also called parietal defect), 2 to 4 cm in
diameter with perpendicular borders and a smooth base. A Peptic Ulcer is an ulcer in the
gastrointestinal tract that is characteristically acidic and thus extremely painful. It is also
called ulcers pepticum or peptic ulcer disease (PUD). Contrary to general belief peptic
ulcers happen more often in the duodenum first part of the small intestine than in the
stomach. Duodenal ulcers are usually benign whereas about 4% of stomach ulcers are
caused by a malignant tumor. The borders of the Peptic ulcer are not well-known in the
acute form but elevated and inflammatory in the chronic form. In the ulcerative form of
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 2
Heartburn
gastric cancer the borders are uneven. Due to parietal scarring the surrounding mucosa
(inner mucus lining) may come into view.
Types of Peptic ulcer
Peptic Ulcers are categorized by their site in the body:
Gastric Ulcer – in the stomach
Duodenal Ulcer – in the duodenum (the first part of the small intestine)
Esophageal Ulcer – in the esophagus
Meckel’s Diverticulum Ulcer – a small pouch in the wall at the junction
of the small and large intestines
Prepyloric Ulcer – between inner and outer walls of the stomach
Proximal gastroesophageal Ulcer – in the mucosa, submucosa, and
muscular layer on the lower esophagus, stomach, or duodenum to have
radial folds.
Symptoms of Peptic Ulcer
A patient with Peptic Ulcer would have some of the next symptoms
Bloating of the abdomen
Waterbrash – this is the rush of saliva in the mouth after an incident of
regurgitation in order to dilute the acid in the esophagus
Abdominal pain – duodenal ulcers are characteristically relieved by foodwhile
gastric ulcers are exacerbated by it
Nausea and a lot of vomiting
Loss of appetite
Weight loss
Hematemesis – vomiting of blood due to gastric ulcer or injure to esophagus from
frequent vomiting
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 3
Melena – tarry foul-smelling feces due to oxidized iron from the hemoglobin
Gastric or duodenal perforation (in some severe cases).
The timing of the symptoms in relation to meals may distinguish between gastric
and duodenal ulcers. In the gastric ulcer, the ache would be during or after meals
because the alkaline duodenal contents reflux into the stomach. In duodenal
ulcers the ache would manifest before a meal when acid manufacturing increases
due to hunger and is passed into the duodenum. However, this is not used as a
dependable method of diagnosis.
Causes of Peptic Ulcer
In most cases, tobacco smoking, anorexia (malnutrition), blood group, spicy food
and other factors that were suspected to source ulcers until late in the 20th century, are
actually of minor importance in the development of peptic ulcers. But these are factors
that make worse the symptoms of peptic ulcer, once formed. Some of the causes are:
Bacteria: As much as 60% of gastric and up to 90% of duodenal ulcers are linked with
Helicobacter pylori, a spiral-shaped bacterium that lives in the acidic surroundings of the
stomach. This bacterium colonies the antral mucosa (inner mucus lining) and grows
rapidly. The antibodies are not capable to clear the infection. This causes localized
erosion and damage of the mucosa and results in ulcer formation.
Reaction to medications: Ulcers can also be sourced or worsened by drugs such as
aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs). Normally,
prostaglandins kindle the secretion of mucus in the gastrointestinal tract to defend its
inner lining from gastric acid. NSAIDs block this function and the risk of ulceration
increases. Hence, peptic ulcer due to reaction to medication is found additional in aged
people suffering from arthritis or those with pain syndromes. Ulcerogenesis can also be
caused by making use of glucocorticoids.
Tobacco: Smoking leads to atherosclerosis and vascular spasms. This guides to ischemia
(localized reduction of blood flow) and ulcers.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 4
Heredity: A family history is often there in duodenal ulcers, especially when blood
group O is also present. But studies show that this is not a significant factor in the case of
gastric ulcers.
Zollinger Ellison syndrome: Gastrinomas, rare gastrin-secreting tumors cause multiple
ulcers which are hard to heal.
Diagnosis of Peptic Ulcer
Endoscopy: Esophagogastroduodenoscopy (EGD), a form of endoscopy, also
recognized as a gastroscopy, is carried out on patients in whom a peptic ulcer is
supposed. By direct visual identification, the location and harshness of an ulcer
can be diagnosed. Moreover, if no ulcer is there, EGD can often give an
alternative diagnosis. In patients over 45 years with more than two weeks of the
above symptoms, the odds for peptic ulceration are high sufficient to warrant
rapid investigation by EGD.
Findings of endoscopy: The microscopical emergence of the ulcer shows 4
zones: inflammatory exudate (fluid); fibrinoid necrosis dead cells; granulation
tissue; and fibrous tissue. The fibrous base of the ulcer may hold vessels with
thickened wall or with thrombosis (formation of blood clot).
Urea breath test: This is a non-invasive test and does not need an endoscopy.
Blood test: This is just to ensure for the presence of antibodies but is not a
sufficient diagnostic method for Peptic ulcer.
Stool antigen test: This is a non-invasive immunoassay to identify active
infection with Helicobacter pylori bacteria in fecal samples.
Histological examination: The possibility of other causes of ulcers, notably
malignancy (gastric cancer) needs to be reserved in mind. This is especially true
in ulcers of the greater (large) curvature of the stomach.
X-ray: This is carried out to rule out perforated Peptic Ulcer. If the air leaks from
the gastrointestinal tract (which contains air) to the peritoneal cavity (which never
contains air), there would be free gas within the peritoneal cavity. The gas will
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 5
drift to a position underneath the diaphragm and a chest X-ray would illustrate an
erect chest and supine, lateral abdomen. This is a sure signal of perforated peptic
ulcer and should be treated forthwith.
Treatment of Peptic Ulcer
Antacids: Young, otherwise fit patients with ulcer-like symptoms are treated with
antacids or H2 antagonists before Endoscopy (EGD) is undertaken. Bismuth
compounds may in fact reduce or even clear organisms and no further treatment is
required.
Antibiotics: When Helicobacter pylori infection is there, the most effective
treatments are mixtures of any two antibiotics like Clarithromycin, Amoxicillin,
Tetracycline or Metronidazole along with a proton pump inhibitor (PPI) and a
bismuth compound.
Surgery: Perforated peptic ulcer becomes a surgical emergency and requires
surgical mend of the perforation. Most bleeding ulcers need endoscopy urgently
to stop the bleeding with cautery destroying abnormal tissue by burning with
electrosurgery or with injection
Prevention of Peptic Ulcer
There is no proven way to stop peptic ulcer disease (PUD) but some measure
can be taken to decrease the risk:
Reduce use of NSAIDs: Use of non-steroidal anti-inflammatory drugs (NSAIDs)
appears to grounds ulcers by inhibiting the body’s production of prostaglandins,
hormones that defend the stomach lining. Patients should also be prescribed a
prostaglandin analogue in order to help stop peptic ulcers.
Tested for Helicobacter pylori: If a person has a family history of ulcers being
tested for disease by Helicobacter pylori bacterium can be a preventivemeasure
Quit smoking: Smokers are about twice as likely to extend ulcers as non-smokers
as they are more vulnerable to Helicobacter pylori, and provide more favorable
conditions for the bacteria to thrive and diminish gastric mucosaldefenses.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 6
Reduce stress: There’s conflicting evidence that pressure is one of the causes of
peptic ulcer and more so in duodenal ulcers. But relaxation exercises like yoga;
meditation; and other stress-reducing strategies can assist decrease blood pressure
and slow heart rate more as a general wellness practice.
Stop alcohol: Alcohol aggravates formation and uneasiness from ulcers although
it does not reason it 1
.
Drug Delivery System
The drug-delivery system should deliver drug at a rate dictated by the needs of the
body over a specified period of time. The goal of any drug delivery system is to provide a
therapeutic amount of drug to a proper site in the body, so that the desired drug
concentration can be achieved promptly and then maintained. The idealized objective
points to the two aspects most important to drug delivery, namely, spatial placement and
temporal delivery. Spatial placement relates to targeting drugs to specific organs, tissues,
cells, or even subcellular compartments; whereas temporal delivery refers to controlling
the rate of drug delivery to the target site 2,3
.
The various routes of drug administration and dosage forms 4
are shown in Table 1.
Table 1.1: Routes of drug administration and dosage form / drug deliverysystems
S. NO. TERM SITE PRIMARY DOSAGE
FORM
1 Oral Mouth Tablets, Capsules, Solutions,
Syrups, Elixirs, Suspensions,
Magmas, Gels, Powders
Peroral
(per os: to be
swalled)
Gastrointestinal
tract via mouth
2 Sublingual Under the tongue Tablets, Troches or Lozenges
3 Parenteral Other than the
gastrointestinal
tract (by
Solutions, Suspensions
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 7
injection)
Interavenous Vein
Interaarterial Artery
Interacardiac Heart
Interaspinal or
Interathecal
Spine
Interaosseous Bone
Interaarticular Joint
Interasynovial Joint-fluid area
Interacutaneous
or Interadermal
Skin
Subcutaneous Beneath the skin
Interamuscular Muscle
4 Epicutaneous
(Topical)
Skin surface Ointments, Creams,
Infusion pumps, Pastes,
Plasters, Powders,Aerosols,
Lotions, Transdermal patches,
Transdermal discs,
Transdermal solutions
5 Transdermal Skin surface
6 Conjunctival Conjunctiva Contact lenses inserts,
Ointments
7 Intraocular Eye Solutions, Suspensions
8 Intranasal Nose Solutions, Sprays,
Inhalants, Ointments
9 Aural Ear Solutions
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 8
Minimize or eliminate local side effects
10 Intrarepiratory Lung Aerosols
11 Rectal Rectum Solutions,
Ointments, Suppositories
12 Vaginal Vagina Solutions, Ointments,
Emulsion foams, Tablets,
Inserts, Suppositories, Sponge
13 Urethral Urethra Solutions, Suppositories
: Several types of modified-release drug products are recognized:
1.2.1.1: Extended-release drug products:
A dosage form that allows at least a twofold reduction in dosage frequency as
compared to that drug presented as an immediate-release (conventional) dosage form.
Examples of extended-release dosage forms include controlled-release, sustained-release,
and long-acting drug products5, 6
.
: Delayed-release drug products:
A dosage form that releases a discrete portion or portions of drug at a time or at
times other than promptly after administration, although one portion may be released
promptly after administration. Enteric-coated dosage forms are the most common
delayed-release products.
: Targeted-release drug products:
A dosage form that releases drug at or near the intended physiologic site of action.
Targeted-release dosage forms may have either immediate- or extended-release
characteristics 6, 7
.
: Advantages of modified drug therapy: 6
1. Avoid patient compliance problems
2. Employ less total drug
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 9
Minimize or eliminate systematic side effects
Obtain less potentiation or reduction in drug activity with chronic use
Minimize drug accumulation with chronic dosing
3. Improve efficiency in treatment
Cure and control condition more promptly
Improve control of condition (i.e., reduce fluctuation in drug level)
Improve bioavailability of somedrugs
4. Economic savings
Gastro-retentive drug delivery system:
Even though various drug delivery systems are used for maximizing therapeutic index
and reduction in the side effects of the drug, oral route remains the preferred, promising
and effective route for the administration of therapeutic agents. Because, low cost of
therapy, ease of administration, flexibility in formulation and handling leads to higher
level of patient compliance. Approximately 50% of the drug delivery systems available in
the market are oral drug delivery system 8
.
The novel design of an oral controlled drug delivery system during last two decades, it
has limited success in case of drugs with a poor absorption window throughout the GIT
(Gastro Intestinal Tract). This approach has several physiological difficulties such as
inability to restrain and locate the controlled drug delivery system within the desired
region of the gastrointestinal tract (GIT) due to variable gastric emptying and motility.
Furthermore, the relatively brief gastric emptying time in humans which normally
averages 2-3 h through the major absorption zone, i.e., stomach and upper part of the
intestine can result in incomplete drug release from the drug delivery system leading to
reduced efficacy of the administered dose 9
Advantages of GRDDs
1. Floating dosage forms such as tablets or capsules will remains in the solutionfor
prolonged time even at the alkaline pH of the intestine
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 10
2. FDDS are advantageous for drugs meant for local action in the stomach eg:
antacids
3. FDDS dosage forms are advantageous in case of vigorous intestinal movement
and in diarrhea to keep the drug in floating condition in stomach to get a relatively
better response.
4. Acidic substance like aspirin causes irritation on the stomach wall when come in
contact with it hence; HBS/FDDS formulations may be useful for the
administration of aspirin and other similar drugs.
5. The FDDS are advantageous for drugs absorbed through the stomach eg: Ferrous
salts, Antacids 10-12
.
Disadvantages of GRDDs
1. Floating systems are not feasible for those drugs that have solubility or stability
problems in gastric fluids.
2. Drugs such as Nifedipine, which is well absorbed along the entire GI tract and
which undergo significant first pass metabolism, may not be suitable candidates
for FDDS.
3. One of the disadvantages of floating systems is that they require a sufficiently
high level of fluids in the stomach, so that the drug dosage form float therein and
work efficiently.
4. These systems also require the presence of food to delay their gastric emptying 13-
14
.
Potential drug candidates for gastro retentive drug delivery Systems
1. Drugs those are locally active in the stomach e.g misoprostol, antacids etc.
2. Drugs that have narrow absorption window in gastrointestinal tract (GIT) e.g. L-
DOPA, para-aminobenzoic acid, furosemide, riboflavin etc.
3. Drugs those are unstable in the intestinal or colonic environment e.g.captopril,
ranitidine HCl,
4. Drugs that disturb normal colonic microbes e.g.antibiotics against Helicobacter
pylori.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 11
5. Drugs that exhibit low solubility at high pH values e.g. diazepam,
chlordiazepoxide,
6. verapamil HCl.
Drugs those are unsuitable for gastroretentive drug delivery systems
1. Drugs that have very limited acid solubility e.g. phenytoin etc.
2. Drugs that suffer instability in the gastric environment e.g. erythromycin etc.
3. Drugs intended for selective release in the colon. eg. 5- amino salicylic acid and
corticosteroids etc.15
.
Polymers and other ingredients used for the preparations of Floating drugs
i) Polymers: The following polymers used to preparations of floating drugs: HPMC K4
M, Calcium alginate, Eudragit S100 Eudragit RL, Propylene foam, Eudragit RS, ethyl
cellulose, poly methyl methacrylate, Methocel K4M, Polyethylene oxide, %
Cyclodextrin, HPMC 4000, HPMC 100, CMC, Polyethylene glycol, polycarbonate, PVA,
Polycarbonate, Sodium alginate, HPC-L, HPC, Eudragit S, HPMC, Metolose S.M. 100,
PVP, HPCH, HPC-M, HPMC K15, Polyox, HPMC K4, Acrylic polymer E4 M and
Carbopol.
ii) Inert fatty materials (5 - 75%): Edible, inert fatty materials having a specific
gravity of less than one can be used to decrease the hydrophilic property of formulation
and hence increase buoyancy. E.g. Beeswax, fatty acids, long chain fatty alcohols,
Gelucires® 39/01 and 43/01.
iii) Effervescent agents: Sodium bicarbonate, citric acid, tartaric acid, Di-SGC (Di-
Sodium Glycine Carbonate, CG (Citroglycine).
iv) Release rate accelerants (5 - 60%): lactose, mannitol.
v) Release rate retardants (5 - 60%): Dicalcium phosphate, talc, magnesium stearate
vi) Buoyancy increasing agents (upto 80%): Ethyl cellulose
vii) Low density material: Polypropylene foam powder (Accurel MP 1000®) 11-15
.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 12
Approaches to GRDDS
To formulate a successful stomach specific or gastroretentive drug several techniques are
currently used such as
Hydrodynamically balanced systems 17
Bioadhesive or Mucoadhesive 18
Raft systems incorporating alginate gels 19-22
Modified shape systems 19
High density systems 20-23
Swelling system 24-26
Magnetic systems 26-27
Floating drug delivery 28-29
Colloidal gel barrier system
Microporous compartment system
Alginate beads 30
Hollow microspheres / Microballons 31
Effervescent systems
Drug release from effervescent (gas generating) systems
This system can also be further described as:
(i) Volatile liquid containing systems 37
(ii) Gas-generating Systems
Among these the following have been studied extensively.
Floating drug delivery systems
Non effervescent systems
Gas generating systems
High density systems
Bioadhesive system
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 13
Floating system
Drug delivery system that float immediately upon contact with gastric fluids
present promising approach for increasing the bioavailability of drugs with absorption
window in the upper small intestine. However, immediate floating can only be achieved
if the density of the device is low at the very beginning. Devices with an initially high
density (which decreases with time) first settle down in the stomach and thus undergo the
risk of premature emptying. Inherent low density can, for example, be provided by the
entrapment of air (e.g. hollow chambers) or by the (additional) incorporation of low
density materials e.g. fatty substances or oils or foampowder.
Non effervescent systems
This type of system, after swallowing, swells unrestrained via imbibition of gastric
fluid to an extent that it prevents their exit from the stomach .These systems may be
referred to as the ‘plug-type systems’ since they have a tendency to remain lodged near
the pyloric sphincter. One of the formulation methods of such dosage forms involves the
mixing of drug with a gel, which swells in contact with gastric fluid after oral
administration and maintains a relative integrity of shape and a bulk density of less than
one within the outer gelatinous barrier. The air trapped by the swollen polymer confers
buoyancy to these dosage forms.
Gas generating systems
These buoyant systems utilise matrices prepared with swellable polymers like
methocel, polysaccharides like chitosan and effervescent components like sodium
bicarbonate, citric acid and tartaric acid or chambers containing a liquid that gasifies at
body temperature.
The optimal stoichiometric ratio of citric acid and sodium bicarbonate for gas
generation is reported to be 0.76:1. The common approach for preparing these systems
involves resin beads loaded with bicarbonate and coated with ethylcellulose. The coating,
which is insoluble but permeable, allows permeation of water. Thus, carbon dioxide is
released, causing the beads to float in the stomach.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 14
High density systems
Sedimentation has been employed as a retention mechanism for pellets that are
small enough to be retained in the rugae or folds of the stomach body near the pyloric
region, which is the part of the organ with the lowest position in an upright posture.
Dense pellets (approximately 3g/cm3
) trapped in rugae also tend to withstand the
peristaltic movements of the stomach wall. With pellets, the GI transit time can be
extended from an average of 5.8 to 25 hours, depending more on density than on
diameter of the pellets, although many conflicting reports stating otherwise also found in
the literature. Commonly used excipients are barium sulphate, zinc oxide, titanium
dioxide and iron powder, etc. These materials increase density by up to 1.5–2.4g/cm3
.
However, no successful system has reached the market.
Figure 1.1. (a) microspheres floats on stomach contents. (b & c)
microspheres adhere to stomach wall
Bioadhesive drug delivery systems
Bioadhesive drug delivery systems (BDDS) are used to localize a delivery device
within the lumen to enhance the drug absorption in a site-specific manner. This approach
involves the use of bioadhesive polymers which can adhere to the epithelial surface in the
stomach. A microbalance-based system is reported for measuring the forces of interaction
between the GI mucosa and the individual polymers. The Cahn Dynamic Contact Angle
Analyzer has been used to study the adherence.
Gastric mucoadhesion does not tend to be strong enough to impart to dosage forms
the ability to resist the strong propulsion forces of the stomach wall. The continuous
production of mucous by the gastric mucosa to replace the mucous that is lost through
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 15
peristaltic contractions and the dilution of the stomach content also seem to limit the
potential of mucoadhesion as a gastroretentive force. Some of the most promising
excipients that have been used commonly in these systems include polycarbophil,
carbopol, lectins, chitosan, CMC and gliadin, etc. Some investigators have tried out a
synergistic approach between floating and bioadhesion systems.
The major challenge for bioadhesive drug delivery systems is the high turnover
rate of the gastric mucus and the resulting limited retention times. Furthermore, it is
difficult to target specifically the gastric mucus with bioadhesive polymers. Most of
the latter (e.g. polycarbophil, Carbopol and chitosan) will stick to various other
surfaces that they come into contact with. In addition; the possibility of oesophageal
binding might present a challenge regarding safety aspects.
Mechanism of Bioadhesion
The mechanism of adhesion of polymers to mucosal tissues has involved both
chemical and physical binding. Both weak and strong interactions (i.e. Vander walls
interaction, hydrogen bonding and ionic bonding) can develop between certain types of
chemical groups on the polymer (i.e. –OH, -COOH) and the glycoprotein network of the
mucus layer or the glycoprotein chains attached to the epithelial cells.
Mucous membranes
Mucous membranes (mucosae) are the moist surfaces lining the walls of various
body cavities such as the gastrointestinal and respiratory tracts. They consist of a
connective tissue layer (the lamina propria) above which is an epithelial layer, the surface
of which is made moist usually by the presence of a mucus layer. The epithelia may be
either single layered (e.g. the stomach, small and large intestine and bronchi) or
multilayered/stratified (e.g. in the oesophagus, vagina and cornea). The former contain
goblet cells which secrete mucus directly onto the epithelial surfaces, the latter contain,
or are adjacent to tissues containing, specialised glands such as salivary glands that
secrete mucus onto the epithelial surface. Mucus is present as either a gel layer adherent
to the mucosal surface or as a luminal soluble or suspended form. The major components
of all mucus gels are mucin glycoproteins, lipids, inorganic salts and water, the latter
accounting for more than 95% of its weight, making it a highly hydrated system. The
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 16
mucin glycoproteins are the most important structure-forming component of the mucus
gel resulting in its characteristic gel-like, cohesive and adhesive properties. The thickness
of this mucus layer varies on different mucosal surfaces, from 50 to 450 µm in the
stomach to less than 1 µm in the oral cavity. The major functions of mucus are that of
protection and lubrication (they could be said to act as anti-adherents).
Figure 1.2. Three regions within a mucoadhesive joint
Mucoadhesives
The most widely investigated group of mucoadhesives is hydrophilic
macromolecules containing numerous hydrogen bond forming groups, the so-called first
generation mucoadhesives. Their initial use as mucoadhesives was in denture fixative
powders or pastes. The presence of hydroxyl, carboxyl or amine groups on the molecules
favours adhesion. They are called the wet adhesives in that they are activated by
moistening and will adhere non-specifically to many surfaces. Once activated, they will
show stronger adhesion to dry inert surfaces than those covered with mucus. Unless water
uptake is restricted, they may over hydrate to form slippery mucilage. Like typical
hydrocolloid glues, if the formed adhesive joint is allowed to dry then, they can form
very strong adhesive bonds. Typical examples are carbomers, chitosan, sodium alginate
and the cellulose derivatives.
Mucoadhesive / Mucosa Interaction
For adhesion to occur, molecules must bond across the interface. These bonds can
arise in the following way
Ionic bonds—where two oppositely charged ions attract each other via
electrostatic interactions to form a strong bond (e.g. in a salt crystal).
Covalent bonds—where electrons are shared, in pairs, between the bonded
atoms in order to fill the orbital in both. These are also strong bonds.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 17
Hydrogen bonds—here a hydrogen atom, when covalently bonded to
electronegative atoms such as oxygen, fluorine or nitrogen, carries a slight
positively charge and is therefore is attracted to other electronegative atoms. The
hydrogen can therefore be thought of as being shared, and the bond formed is
generally weaker than ionic or covalent bonds.
Vander-Waals bonds—these are some of the weakest forms of interaction that
arise from dipole–dipole and dipole-induced dipole attractions in polar
molecules, and dispersion forces with non-polar substances.
Hydrophobic bonds—more accurately described as the hydrophobic effect,
these are indirect bonds (such groups only appear to be attracted to each other)
that occur when non-polar groups are present in an aqueous solution. Water
molecules adjacent to non-polar groups form hydrogen bonded structures, which
lowers the system entropy.
Table 1.2: Different theories explaining the mechanism of bioadhesion
S. No. Theory Mechanism of bioadhesion
1 Electronic theory Attractive electrostatic forces between glycoprotein
mucin network and the bioadhesive material.
2 Adsorption theory Surfaces forces resulting in chemical bonding.
3 Wetting theory Ability of bioadhesive polymers to spread and develop
intimate contact with the mucus membranes.
4 Diffusion theory Physical entanglement of mucin strands and the flexible
polymer chains. Interpretation of mucin strands into the
porous structure of the polymer substrate.
5 Fracture theory Analyses the maximum tensile stress developed during
detachment of the BDDS from the mucosal surfaces.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 18
Factors Affecting Mucoadhesion
Polymer related factors
Molecular weight Low molecular weight polymer favors the inter
penetration of polymer molecules, high molecular
weight polymer favours physical entanglement.
Flexibility of polymer chains Required for interpenetration and entanglement.
Highly cross-linked polymers: mobility of
individual polymer chains decreases which leads
to decreased bioadhesive strength.
Concentration of polymer Solid BDDS: more is the polymer concentration
higher is the bioadhesive strength, Liquid BDDS:
optimum concentration is required for best
bioadhesion, High concentration may result in
coiling of polymer molecules and hence reduced
flexibility of the polymeric chains.
Methods to Study Bioadhesion
In-vitro methods for testing bioadhesion for the design and development of
bioadhesive controlled release systems and to ensure compatibility, physical and
mechanical stability of these systems (Peppas et al., 1985). These methods include:
i. Ex vivo methods : Methods based on measurement of tensile strength
or shear strength.
Adhesion weight method
Fluorescent probe method
Mechanical spectroscopic method
Falling liquid film method
Flow channel method
Colloidal gold staining method
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 19
Viscometric method
Thumb test
Adhesion number
Electrical Conduction
ii. In vivo methods
Use of radioisotopes
Use of gamma scintiography
Advantages and Limitations of Mucoadhesive Drug Delivery System
Mucoadhesive dosage forms have following advantages because of their property to
immobilize drug-carrying particles at the mucosal surface.
A prolonged residence time at the site of action or absorption thus permit onceor
twice a day dosing.
Increase in drug concentration gradient due to the intimate contact of the particles with
the mucosal surface.
Adirect contact with mucus membrane, which is the step earlier to particle absorption.
Bioadhesive system can prevent the first pass metabolism to certain protein drugs by
the liver through the introduction of the drugs via route bypassing the digestive tract.
Drugs that are absorbed through mucosal lining of tissues can enter directly into the
blood stream and prevented from enzymatic degradation in the GIT.
These dosage forms facilitate intimate contact of the formulation with the
underlying absorption surface. This allows modification of tissue permeability for
absorption of macromolecules such as peptides and proteins, inclusion of
penetration enhancers such as Sodium glycolate, Lysophosphatidylcholine, Sodium
taurocholate and Protease inhibitors in the mucoadhesive dosage forms resulted in
the better absorption of peptides/ proteins.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 20
Limitations
Gastric mucoadhesion does not tend to be strong enough to impart to dosage forms
the ability to resist the strong propulsion forces of the stomachwall.
The continuous production of mucous by the gastric mucosa to replace the mucous
that is lost through peristaltic contractions and the dilution of the stomach content
also seems to limit the potential of mucoadhesion as a gastro-retentiveforce.
Mucoadhesive Polymers
There are two broad classes of mucoadhesive polymers: water-soluble and/or
water insoluble polymers, having swellable networks, joined by cross-linking agents. In
large classes of hydrophillic polymers those containing carboxylic group exhibit the best
bioadhesive properties, Polyvinyl pyrrolidone, Methylcellulose, Sodiumcarboxy
methylcellulose, Hydroxypropyl cellulose and other cellulose derivatives.
Hydrogel are the class of polymeric biomaterial that exhibit the basic
characteristics of an hydrogel to swell by absorbing water interacting by means of
adhesion with the mucus that covers epithelia (Junginger et al., 1990) i.e.
Anionic group - Polyacrylates and their cross-linked modifications
Cationic group - Chitosan (Hango, 1998) and its derivatives
Neutral group - Eudragit-NE30D etc.
The polymer should possess optimal polarity to make sure that it is sufficiently
wetted by the mucus and optimal fluidity that permits the mutual adsorption.
An ideal mucoadhesive polymer should have following characteristics:
The polymer and its degradation products should be nontoxic, nonabsorbable
It should be non-irritant to the mucous membrane
Itshouldforma strongnoncovalent bondwiththe mucinepithelialcellsurfaces
It shouldadherequicklytomoisttissueandshouldpossesssomesitespecificity
The cost of the polymer should not be high
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 21
Thepolymermustnotdecomposeonstorageorduringshelflifeofthedosageform
It should allow easy incorporation of the drug and offers no hindrance to its
release.
Bioadhesive Strength of Some Polymers Table
1.3: Bioadhesive strength of some polymers
S. No. Polymer name Bioadhesive property
1 Polycarbophil +++
2 Tragacanth +++
3 Poly (acrylic acid/ divinyl benzene) +++
4 Sodium alginate +++
5 Carbopol 934 +++
6 Carboxymethyl cellulose
CMC (low viscosity) ++
CMC (high viscosity) +
CMC (medium viscosity) +
7 Hydroxyethyl cellulose +++
8 Gum karaya ++
9 Guar gum ++
10 Chitosan ++
11 Pectin +
12 Gelatin +
13 Thermally modified starch +
14 Hydroxyethyl methacrylate +
15 Amberlite–200 resin +
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 22
16 Hydroxypropyl cellulose +
17 Polyethylene glycol +
18 Polyvinyl pyrrolidone +
19 Chitosan (Dr. Knapczyk) +
20 Daichitosan H ++
+++ Excellent (8.0 to 12.0 mN/cm2
); ++ fair (1.5 to 8.0 mN/cm2
);+ poor (<1.5mN/cm2
).
Application of Bioadhesive Systems
Table 1.4: Application of Bioadhesive Systems
Drug Route of
administration
Polymer used Comment
Acyclovir Ocular Chitosan Slow release rate,
increased AUC
Gentamycin Nasal DSM+LPC Slow release rate
Insulin Nasal DSM+LPC Efficient delivery into
systemic circulation
via nasal route
Furosemide GI AD-MMS[PGEFs] Increased bioavailability
Amoxicillin GI AD-MMS[PGEFs] Greater H-pylori activity
Vancomycin Colonic PGEF coated with
Eudragit S 100
Well absorb even with
enhancer
Insulin Colonic PGEF coated with
Eudragit S 100
Absorbtion only in
presence of enhancer
Methyl
perdisolone
Ocular Hyaluronic acid Slow release rate
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 23
1.4.10 Patents Related to Bioadhesive Systems
Table 1.5: Patents related to bioadhesive system 32
Patent
number
Assignee/Inventor Year Title
US
6368586
Brown University Research
foundation
April
2002
Methods and compositions
for enhancing bioadhesive
properties of polymers
WO
0203955
Roversi Francesco, Cilurzo
Francesco
Jan.
2002
Fast release bioadhesive
microspheres for the
sublingual administration of
Proximate
US
627415
Immunex Corporation Aug
2001
Prolonged release of GM-CSF
US
6207197
West Pharmaceutical
Services Drug Delivery and
Clinical Research Centre
March
2001
Gastroretentive controlled
release microspheres
Mucoadhesive Microballons:
Microballons are small spherical particles, with diameters in the micrometer range
(typically 1µm to 1000µm or 1mm). Microballons are sometimes referred to as
microparticles. Microballons are defined as “the monolithic spheres or therapeutic agents
distributed throughout the matrix either as a molecular dispersion of particles”.
Microballons are small spherical particles with diameter in the micrometer range
and sometimes referred as microparticles. When adhesion is restricted to the
mucous layer lining of the mucosal surface it is termed as mucoadhesion.
Mucoadhesion offers prolonged residence time at the site of absorption,
localization of the drug delivery system at a given target site, increase in drug
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 24
concentration gradient due to the intestine contact of the particle with the
mucosal surface. Development of adhesive bond between polymer and biological
membrane or its coating can be achieved by two ways: initial contact between the
surfaces or formation of secondary bonds due to non covalent interaction.
Mucoadhesives must interact with mucin layer during the process of attachment.
Mucins are synthesized by globet cells and special exocrine glands with mucin
cells acnini. There are atleast two main targets which could be used for anchoring
of delivery system through mucoadhesive in the GIT, the mucosal tissue and
mucosal gel layer. The mucos layer is the first surface encountered by particulate
system and its complex structure offers many opportunities for the
development of adhesive interaction with small polymeric particles either through
non specific or specific interaction between complimentary structures. Due to all
above advantages Microsphere delivery is an better choice for drug delivery in
colon 33
.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 25
2: REVIEW OF LITERATURE
: DRUG PROFILE:
NIZATIDINE
Structure:
Figure 2.1: Structure of drug Nizatidine
Chemical name: N-[2-[[2-(dimethylaminomethyl)-1,3-thiazol-4-
yl]methylsulfanyl]ethyl]-N'-methyl-2-nitroethene-1,1-diamine
Molecular formula:
Molecular weight :
C12H21N5O2S2
331.5
Physical characteristics: A white to off–white crystalline solid.
Melting range:
Storage:
130-132ºC
Store in air tight, temper proof container, protected
from light.
Mechanism of action: Nizatidine is a competitive, reversible inhibitor of histamine at
the histamine H2 receptors, particularly those in the gastric parietal cells. By inhibiting
the action of histamine on stomach cells, nizatidine reduces stomach acid production.
Nizatidine competes with histamine for binding at the H2-receptors on the gastric
basolateral membrane of parietal cells. Competitive inhibitionresults
basal and nocturnal gastric acid secretions. The drug also decreases
in reduction of
the gastric acid
response to stimuli such as food, caffeine, insulin, betazole, or pentagastrin.
Plasma half life: 1.5 hrs
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 26
Disposition in the body: Nizatidine is readily and almost completely absorbed after oral
administration and peak plasma concentrations are reached within 1 to 3 h. Absorption is
increased by the presence of food and decreased by 10% in the presence of antacids such
as aluminium hydroxide gel and magnesium silicate. It is partially metabolised by the
liver, but does not inhibit the hepatic mixed function oxidase system. Three metabolites
have been identified, nizatidine N-2–oxide, nizatidine S-oxide and N-2–mono des methyl
nizatidine (60% activity of nizatidine). Nizatidine is widely distributed throughout the
body and has been detected in breast milk (0.1% of the administered dose). 90% of an
administered dose is excreted in urine, partly by active tubular secretion, with 60% as the
unchanged drug. Less than 6% of a dose is excreted in faeces.
Protein binding: 35%.
Dose: Orally the usual daily dose is 150 to 600 mg. The total intravenous daily dose
should not exceed 480 mg.
Pharmacokinetics parameter: Volume of distribution: Approx. 1.2 to 1.6 L/kg (single
150 mg dose), 1.1 to 1.9 L/kg (multiple doses). Clearance: Serum 37.5 to 41.4 L/h (single
150 mg dose), 39.1 to 44.6 L/h (multiple doses). Plasma 40 to 51 L/h.
Side effects: It include an allergic reaction (difficulty in breathing, closing of throat)
bleeding gums, irregular heartbeat, fever or yellowing of the skin or eyes, headache,
dizziness or diarrhea may occur 34
.
: EXCIPIENTS REVIEW
Cellulose, microcrystalline
Nonproprietary Names: BP: Microcrystalline cellulose, JP: Microcrystalline cellulose,
PhEur: Cellulosum microcristallinum, USPNF: Microcrystalline cellulose
Synonyms: Avicel PH, Celex, cellulose gel, Celphere, Ceolus KG, crystalline cellulose,
E460, Emcocel, Ethispheres, Fibrocel, Pharmacel, Tabulose, Vivapur.
Chemical Name: Cellulose
Empirical Formula and Molecular Weight: (C6H10O5)n ≈36 000 where n ≈ 220.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 27
Structural Formula:
Figure 2.2: Chemical structure of microcrystalline cellulose
Functional Category: Adsorbent, suspending agent, tablet and capsule diluents, tablet
disintegrant.
Applications in Pharmaceutical Formulation or Technology: Microcrystalline
cellulose is widely used in pharmaceuticals, primarily as a binder/diluent in oral tablet
and capsule formulations where it is used in both wet-granulation and direct compression
processes
Description: Microcrystalline cellulose is purified, partially depolymerized cellulose that
occurs as a white, odorless, tasteless, crystalline powder composed of porous particles.
Typical Properties:
Angle of repose: 49° for Ceolus KG; 34.4° for Emcocel 90M.9
Density (true): 1.512–1.668 gm / cm3
Flowability: 1.41 g / s for Emcocel 90M.9
Particle size distribution: Typical mean particle size is 20–200 μm. Different grades may
have a different nominal mean particle size; Avicel PH- 102a 100-60
Solubility: Slightly soluble in 5 % w/v sodium hydroxide solution; practically insoluble
in water, dilute acids, and most organic solvents.
Calcium phosphate, dibasic dihydrate
Nonproprietary Names: BP: Calcium hydrogen phosphate, JP: Dibasic calcium
phosphate, PhEur: Calcii hydrogenophosphas dihydricus, USP: Dibasic calcium
phosphate
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 28
Synonyms: Calcium hydrogen orthophosphate dehydrate, calcium monohydrogen
phosphate dehydrate, Di-Cafos; dicalcium orthophosphate, DI-TAB, E341, Emcompress,
phosphoric acid calcium salt (1:1) dihydrate; secondary calcium phosphate.
Chemical Name: Dibasic calcium phosphate dehydrate
Empirical Formula and Molecular Weight: CaHPO4·2H2O; 172.09
Structural Formula: CaHPO4·2H2O
Functional Category: Tablet and capsule diluent.
Applications in Pharmaceutical Formulation or Technology: Dibasic calcium
phosphate dihydrate is widely used in tablet formulations both as an excipient and as a
source of calcium and phosphorus in nutritional supplements.
Description: Dibasic calcium phosphate dihydrate is a white, odorless, tasteless powder
or crystalline solid. It occurs as monoclinic crystals.
Typical Properties:
Angle of repose: 28.3° for Emcompress.
Density (bulk): 0.915 gm / cm3
Density (tapped): 1.17 gm / cm3
Density (true): 2.389 gm / cm3
Particle size distribution: DI-TAB: average particle diameter 180 μm, Fine powder:
average particle diameter 9 μm
Solubility: Practically insoluble in ethanol, ether, and water; soluble in dilute acids.
Lactose, Anhydrous
Nonproprietary Names: BP: Anhydrous lactose, JP: Anhydrous lactose, PhEur:
Lactosum anhydricum, USPNF: Anhydrous lactose
Synonyms: Anhydrous Lactose NF 60M, Anhydrous Lactose NF Direct Tableting,
Lactopress Anhydrous, lactosum, lattioso, milk sugar, Pharmatose DCL 21, Pharmatose
DCL 22, saccharum lactis, Super-Tab Anhydrous.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 29
Chemical Name: O-β-D-galactopyranosyl-(1→4)-β-D-glucopyranose
Empirical Formula and Molecular Weight: C12H22O11 and 342.30
Structural Formula: Anhydrous lactose as O-β-D-galactopyranosyl-(1→4)-β-
Dglucopyranose; or a mixture of O-β-D-galactopyranosyl-(1→4)-α-D-glucopyranose and
O-β- D-galactopyranosyl-(1→4)-β-D-glucopyranose (Figure 2.3).
Figure 2.3: Chemical structure of anhydrous lactose
Functional Category: Binding agent, directly compressible tableting excipients,
lyophilization aid, tablet and capsule filler.
Applications in Pharmaceutical Formulation or Technology: Anhydrous lactose is
widely used in direct compression tableting applications and as a tablet and capsule filler
and binder. Anhydrous lactose can be used with moisture-sensitive drugs due to its low
moisture content.
Description: Lactose occurs as white to off-white crystalline particles or powder. Several
different brands of anhydrous lactose are commercially available which contain
anhydrous β-lactose and anhydrous α-lactose.
Typical Properties:
Angle of repose: 39° for Pharmatose DCL 21 and 38° for Super-Tab Anhydrous.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 30
Density (true): 1.589 gm / cm3
for anhydrous β-lactose; 1.567 gm / cm3
for Super-Tab
Anhydrous.
Density (bulk): 0.68 gm / cm3
for Pharmatose DCL 21; 0.67 gm / cm3
for Pharmatose
DCL 22; 0.65 gm / cm3
for Super-TabAnhydrous.
Density (tapped): 0.88 gm / cm3
for Pharmatose DCL 21; 0.79 gm / cm3
for Pharmatose
DCL 22; 0.87 gm / cm3
for Super-TabAnhydrous.
Solubility: soluble in water; sparingly soluble in ethanol (95 %) and ether.
Sodium starch glycolate
Nonproprietary Names: BP: Sodium starch glycollate, PhEur: Carboxymethylamylum
natricum, USPNF: Sodium starch glycolate
Synonyms: Carboxymethyl starch, sodium salt, Explosol, Explotab, Glycolys, Primojel,
starch carboxymethyl ether, sodium salt, Tablo, Vivastar P.
Chemical Name: Sodium carboxymethyl starch
Empirical Formula and Molecular Weight: Sodium starch glycolate is the sodium salt
of a carboxymethyl ether of starch, containing 2.8–4.2 % sodium. The molecular weight
is typically 5 × 105–1 × 106.
Structural Formula:
Figure 2.4: Chemical structure of sodium starch glycolate
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 31
Functional Category: Tablet and capsule disintegrant.
Applications in Pharmaceutical Formulation or Technology: Sodium starch glycolate
is widely used in oral pharmaceuticals as a disintegrant in capsule and tablet
formulations. It is commonly used in tablets prepared by either directcompression or wet-
granulation processes. The usual concentration employed in a formulation is between 2 %
and 8 %, with the optimum concentration about 4 %, although in many cases 2 % is
sufficient. Disintegration occurs by rapid uptake of water followed by rapid and
enormous swelling. Although the effectiveness of many disintegrants is affected by the
presence of hydrophobic excipients such as lubricants, the disintegrant efficiency of
sodium starch glycolate is unimpaired. Increasing the tablet compression pressure also
appears to have no effect on disintegration time.10 – 14 sodium starch glycolate has also
been investigated for use as a suspending vehicle.
Description: Sodium starch glycolate is a white to off-white, odorless, tasteless, free-
flowing powder. It consists of oval or spherical granules, 30 – 100 μm in diameter, with
some less-spherical granules ranging from 10 – 35 μm in diameter.
Typical Properties:
Particle size distribution: 100 % of particles less than 106 μm in size. Average particle
size is 35 – 55 μm for Explotab.
Solubility: Sparingly soluble in ethanol (95 %), practically insoluble in water. At a
concentration of 2 % w/v sodium starch glycolate disperses in cold water and settles in
the form of a highly hydrated layer.
Swelling capacity: In water, sodium starch glycolate swells to up to 300 times its
volume.
Viscosity (dynamic): ≤ 200 mPa s (200 cP) for a 4 % w/v aqueous dispersion. Viscosity
is 4.26 mPa s for a 2% w/v aqueous dispersion
Starch
Nonproprietary Names: BP: Potato starch, JP: Potato starch, PhEur: Solani amylum
(potato starch), USPNF: Potato starch
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 32
Synonyms: Amido, amidon, amilo, amylum, Aytex P, C*PharmGel, Fluftex W, Instant
Pure-Cote, Melojel, Meritena, Paygel 55, Perfectamyl D6PH, Pure-Bind, Pure-Cote,
Pure-Dent, Pure- Gel, Pure-Set, Purity 21, Purity 826, Tablet White.
Chemical Name: Starch
Empirical Formula and Molecular Weight: (C6H10O5) n 50 000–160 000 where n =
300–1000. Starch consists of amylose and amylopectin, two polysaccharides based on α-
glucose.
Functional Category: Glidant, diluents, disintegrant, tablet binder.
Structural Formula:
Figure 2.5: Chemical structure of starch
Applications in Pharmaceutical Formulation or Technology: Starch is used as an
excipient primarily in oral solid-dosage formulations where it is utilized as a binder,
diluent, and disintegrant. As a diluent, starch is used for the preparation of standardized
triturates of colorants or potent drugs to facilitate subsequent mixing or blending
processes in manufacturing operations. Starch is also used in dry-filled capsule
formulations for volume adjustment of the fill matrix.1 In tablet formulations, freshly
prepared starch paste is used at a concentration of 5–25 % w/w in tablet granulations as a
binder.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 33
Description: Starch occurs as an odourless and tasteless, fine, white-colored powder
comprising very small spherical or ovoid granules whose size and shape are characteristic
for each botanical variety.
Typical Properties:
Density (bulk): 0.462 gm / cm3
for corn starch.
Density (tapped): 0.658 gm / cm3
for corn starch.
Density (true): 1.478 gm / cm3
for corn starch.
Flowability: Corn starch is cohesive and has poor flow characteristics.
Gelatinization temperature: 72 °C for potato starch
Particle size distribution: Potato starch: 10–100 μm;
Solubility: Practically insoluble in cold ethanol (95 %) and in cold water. Starch swells
instantaneously in water by about 5–10 % at 37 °C.2, 22 polyvalent cations produce more
swelling than monovalent ions, but pH has little effect.
Swelling temperature: 64°C for potato starch
Talc
Nonproprietary Names: BP: Purified talc, JP: Talc, PhEur: Talcum, USP: Talc
Synonyms: Altalc, E553b, hydrous magnesium calcium silicate, hydrous magnesium
silicate, Luzenac Pharma; magnesium hydrogen metasilicate, Magsil Osmanthus, Magsil
Star, powdered talc, purified French chalk, Purtalc, soapstone, steatite, Superiore.
Chemical Name: Talc
Empirical Formula and Molecular Weight: Talc is a purified, hydrated, magnesium
silicate, approximating to the formula Mg6(Si2O5)4(OH)4. It may contain small, variable
amounts of aluminum silicate and iron.
Functional Category: Anticaking agent, glidant, diluents, lubricant.
Applications in Pharmaceutical Formulation or Technology: Talc was once widely
used in oral solid dosage formulations as a lubricant and diluents.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 34
Typical Properties:
Hardness: 1.0 – 1.5
Moisture content: Talc absorbs insignificant amounts of water at 25 °C and relative
humidities up to about 90 %.
Solubility: It is practically insoluble in dilute acids and alkalis, organic solvents, and
water
Magnesium stearate
Nonproprietary Names: BP: Magnesium stearate, JP: Magnesium stearate, PhEur:
Magnesii stearas, USPNF: Magnesium stearate
Synonyms: Magnesium octadecanoate, octadecanoic acid, magnesium salt, stearic acid,
magnesium salt.
Chemical Name: Octadecanoic acid magnesium salt
Empirical Formula and Molecular Weight: C36H70MgO4 and 591.34
Structural Formula: [CH3 (CH2)16COO]2 Mg
Functional Category: Tablet and capsule lubricant.
Applications in Pharmaceutical Formulation or Technology: Magnesium stearate is
widely used in cosmetics, foods, and pharmaceutical formulations. It is primarily used as
a lubricant in capsule and tablet manufacture at concentrations between 0.25 % and 5.0 %
w/w.
Description: Magnesium stearate is a very fine, light white, precipitated or milled,
impalpable powder of low bulk density, having a faint odor of stearic acid and a
characteristic taste. The powder is greasy to the touch and readily adheres to the skin.
Typical Properties:
Density (bulk): 0.159 gm / cm3
Density (tapped): 0.286 gm / cm3
Density (true): 1.092 gm / cm3
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 35
Flowability: Poorly flowing, cohesive powder.
Solubility: Practically insoluble in ethanol, ethanol (95 %), ether and water; slightly
soluble in warm benzene and warm ethanol (95 %).
Guar Gum
Nonproprietary Names: BP: Guar galactomannan, PhEur: Guar galactomannanum,
USPNF: Guar gum
Synonyms: E412; Galactosol; guar flour; jaguar gum; Meyprogat; Meyprodor;
Meyprofin.
Chemical Name: Galactomannan polysaccharide
Empirical Formula and Molecular Weight: (C6H12O6) n ≈220 000
Structural Formula: Guar gum consists of linear chains of (1→4)-β-D-mannopyranosyl
units with α-Dgalactopyranosyl units attached by (1→6) linkages. The ratio of D-
galactose to D-mannose is between 1: 1.4 and 1: 2 (Figure 2.6).
Figure 2.6: Chemical structure of guar gum
Functional Category: Suspending agent, tablet binder, tablet disintegrant, viscosity-
increasing agent.
Description: Guar gum is obtained from the ground endosperms of Cyamopsis
tetragonolobus (L.) Taub. (Family: Leguminosae). It consists chiefly of a highmolecular-
weight hydrocolloidal polysaccharide, composed of galactan and mannan units combined
through glycoside linkages, which may be described chemically as a galactomannan.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 36
Typical Properties:
Density: 1.492 gm / cm3
Solubility: Practically insoluble in organic solvents. In cold or hot water, guar gum
disperses and swells almost immediately to form a highly viscous, thixotropic sol. The
optimum rate of hydration occurs at pH 7.5 to 9.0. Finely milled powders swell more
rapidly and are more difficult to disperse. Two to four hours in water at room temperature
are required to develop maximum viscosity.
Viscosity: 4.86 Pa s (4860 cP) for 1 % w/v dispersion at 25 ºC temperature.
Stability and Storage Conditions: Guar gum powder should be stored in a well-closed
container in a cool, dry place. Aqueous guar gum dispersions have a buffering action and
are stable at pH 4.0 to 10.5.
Xanthan Gum
Nonproprietary Names: BP: Xanthan gum, PhEur: Xanthani gummi, USPNF: Xanthan
gum
Synonyms: Corn sugar gum; E415; Keltrol; polysaccharide B-1459; Rhodigel; Vanzan
NF; Xantural.
Chemical Name: Xanthan gum
Empirical Formula and Molecular Weight: (C35H49O29)n Approximately 2 × 106
Xanthan gum is a high molecular weight polysaccharide gum. It contains D-glucose and
D-mannose as the dominant hexose units, along with D-glucuronic acid, and is prepared
as the sodium, potassium, or calcium salt.
Functional Category: Stabilizing agent; suspending agent; viscosity-increasing agent.
Description: Xanthan gum occurs as a cream- or white-colored, odorless, free-flowing,
fine powder.
Typical Properties:
Acidity / alkalinity: pH = 6.0 – 8.0 for a 1 % w/v aqueous solution.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 37
Particle size distribution: Various grades with different particle sizes are available; for
example, 100 % less than 180 μm in size for Keltrol CG; 100 % less than 75 μm in size
for Keltrol CGF; 100 % less than 250 μm, 95 % less than 177 μm in size for Rhodigel;
100 % less than 177 μm, 92 % less than 74 μm in size for Rhodigel 200.
Solubility: Practically insoluble in ethanol and ether; soluble in cold or warm water.
Viscosity: 1200 to 1600 mPa s (1200–1600 cP) for a 1 % w/v aqueous solution at 25 °C.
Stability and Storage Conditions: Xanthan gum is a stable material. Aqueous solutions
are stable over a wide pH range (pH 3– 12), although they demonstrate maximum
stability at pH 4 – 10 and temperatures of 10 – 60°C. Xanthan gum solutions of less than
1 % w/v concentration may be adversely affected by higher than ambienttemperatures.
Hypromellose
Nonproprietary Names: • BP: Hypromellose,
• JP: Hydroxypropylmethylcellulose, • PhEur: Hypromellosum, • USP: Hypromellose
Synonyms: Benecel MHPC; E464; hydroxypropyl methylcellulose; HPMC; Methocel;
methylcellulose propylene glycol ether; methyl hydroxypropylcellulose; Metolose;
Tylopur.
Chemical Name: Cellulose hydroxypropyl methyl ether
Empirical Formula and Molecular Weight: The PhEur 2005 describes hypromellose as
a partly O-methylated and O-(2-hydroxypropylated) cellulose. It is available in several
grades that vary in viscosity and extent of substitution. Grades may be distinguished by
appending a number indicative of the apparent viscosity, in mPa s, of a 2 % w/w aqueous
solution at 20 °C. Hypromellose defined in the USP 28 specifies the substitution type by
appending a four-digit number to the nonproprietary name: e.g., hypromellose 1828. The
first two digits refer to the approximate percentage content of the methoxy group
(OCH3). The second two digits refer to the approximate percentage content of the
hydroxypropoxy group (OCH2CH (OH) CH3), calculated on a dried basis. It contains
methoxy and hydroxypropoxy groups conforming to the limits for the types of
hypromellose. Molecular weight is approximately 10 000 – 1 500 000.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 38
Structural Formula:
Figure 2.7: Chemical structure of cellulose hydroxypropyl methyl ether
Where R is H, CH3, or CH3CH (OH) CH2 (Figure 2.7)
Description: Hypromellose is an odorless and tasteless, white or creamy-white fibrous or
granular powder.
Typical Properties:
Density (bulk): 0.341 gm / cm3
Density (tapped): 0.557 gm / cm3
Density (true): 1.326 gm / cm3
Melting point: Browns at 190 – 200 °C; chars at 225 – 230 °C. Glass transition
temperature is 170 – 180 °C.
Moisture content: Hypromellose absorbs moisture from the atmosphere; the
amount of water absorbed depends upon the initial moisture content and the
temperature and relative humidity of the surrounding air.
Solubility: Soluble in cold water, forming a viscous colloidal solution; practically
insoluble in chloroform, ethanol (95 %), and ether, but soluble in mixtures of
ethanol and dichloromethane, mixtures of methanol and dichloromethane, and
mixtures of water and alcohol. Certain grades of hypromellose are soluble in
aqueous acetone solutions, mixtures of dichloromethane and propan-2-ol, and
other organic solvents 6, 15, 35
.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 39
2.3. Literature review
Rahamathulla et al., 2019 was develop valsartan floating tablets (VFT) via non-
effervescent technique using low density polypropylene foam powder, carbopol, and
xanthan gum by direct compression. Before compression, the particulate powdered
mixture was evaluated for pre-compression parameters. The prepared valsartan tablets
were evaluated for post-compression parameters, swelling index, floating lag time, in
vitro buoyancy studies, and in vitro and in vivo X-ray imaging studies in albino rabbits.
The result of all formulations for pre- and post-compression parameters were within the
limits of USP. FTIR and DSC studies revealed no interaction between the drug and
polymers used. The prepared floating tablets had good swelling and floating capabilities
for more than 12 h with zero floating lag time. The release of valsartan from optimized
formulation NF-2 showed sustained release up to 12 h; which was found to be non-
Fickian release. Moreover, the X-ray imaging of optimized formulation (NF-2) revealed
that tablet was constantly floating in the stomach region of the rabbit, thereby indicating
improved gastric retention time for more than 12 h. Consequently, all the findings and
outcomes have showed that developed valsartan matrix tablets could be effectively used
for floating drug delivery system 36
.
Jain 2018, developed a simple Gastroretentive Mucoadhesive tablets of Riboflavin.
Mucoadhesion is one of the approach to prolong gastric retention. In these systems,
Mucoadhesion is achieved by using Mucoadhesive polymers which adhere to the
epithelial surface of gastrointestinal tract. Narrow absorption window mainly from
upper part of GIT and short half -life makes riboflavin an ideal candidate for its
formulation as a Mucoadhesive tablets. Hence we intend to develop a simple
Gastroretentive Mucoadhesive tablets of Riboflavin and prepare sustained release
once daily 37
.
Wagh et al., 2018 Gastroretentive drug delivery system (GRDDS) is one of the novel
approaches in the area of oral sustained release dosage forms. Gastro retentive dosage
forms has received significant interest in the past few decades as they can improve
the limitation of conventional and oral controlled release drug delivery system
related to fast gastric emptying time. Drugs that are easily absorbed from
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 40
gastrointestinal tract (GIT) and have short half - lives are eliminated quickly
from the systemic circulation require frequent dosing to achieve suitable therapeutic
activity. To avoid these limitations, the development of oral sustain release GRDDS
is an attempt to release the drug slowly into the GIT and maintain an effective drug
concentration in the systemic circulation for a long time. After oral administration,
such a drug delivery would be retained in the stomach and release the drug in controlled
manner so, that the drug could be supplied continuously to its absorption sited in
the GIT 38
.
More et al., 2018 compiled the various gastroretentive approaches. In order to
understand various physiological difficulties to achieve gastric retention, we have
summarized important factors controlling gastric retention. Oral route of drug
administration is the most preferable route because of its flexibility in formulation, ease
of administration, and patient compliance. But this route has certain limitations like
limited gastric residence time (GRT) for sustained drug delivery system and for the drugs
which are absorb from specific region of gastrointestinal tract (GIT). To overcome these
limitations, various approaches have been proposed to increase the gastric retention time
of the delivery system in the upper part of gastrointestinal tract. Gastroretentive dosage
form (GRDF) prolongs the GRT by targeting site-specific drug release in upper part of
GIT. GRDFs enable continuous and the extended duration of drug release and improve
bioavailability of drugs that have narrow therapeutic window, by this way they prolong
dosing interval and increase compliance of the patient. The evaluation parameters of
gastroretentive drug delivery systems are covered. The present review addresses briefly
about the current status of various leading gastroretentive drug delivery technologies,
developed until now, i.e. high density (sinking), floating, bio- or mucoadhesive,
expandable, unfoldable, super porous hydrogel, magnetic systems etc. In addition,
important factors controlling gastroretention, advantages and finally, future potential are
discussed 39
.
El Nabarawi et al., 2017 developed a controlled-release floating matrix tablet and
floating raft system of Mebeverine HCl (MbH) and evaluate different excipients for their
floating behavior and in vitro controlled-release profiles. Oral pharmacokinetics of the
optimum matrix tablet, raft system formula, and marketed Duspatalin® 200 mg retard as
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 41
reference were studied in beagle dogs. The optimized tablet formula (FT-10) and raft
system formula (FRS-11) were found to float within 34±5 sec and 15±7 sec, respectively,
and both remain buoyant over a period of 12 h in simulated gastric fluid. FT-10
(Compritol/HPMC K100M 1:1) showed the slowest drug release among all prepared
tablet formulations, releasing about 80.2% of MbH over 8 h. In contrast, FRS-11
(Sodium alginate 3%/HPMC K100M 1%/Precirol 2%) had the greatest retardation,
providing sustained release of 82.1% within 8 h. Compared with the marketed MbH
product, the Cmax of FT-10 was almost the same, while FRS-11 maximum concentration
was higher. The tmax was 3.33, 2.167, and 3.0 h for marketed MbH product, FT-10, and
FRS-11, respectively. In addition, the oral bioavailability experiment showed that the
relative bioavailability of the MbH was 104.76 and 116.01% after oral administration of
FT-10 and FRS-11, respectively, compared to marketed product. These results
demonstrated that both controlled-released floating matrix tablet and raft system would
be promising gastroretentive delivery systems for prolonging drug action 40
.
Kumari et al, 2016 developed sustained release formulation of Verapamil Hydrochloride
to maintain constant therapeutic levels of the drug for over 12 hrs. Various grades of
HPMC polymers, Guar gum, and Xanthum gum were employed as polymers. Verapamil
Hydrochloride dose was fixed as 120 mg. Total weight of the tablet was considered as
400 mg. Polymers were used in the concentration of 60, 120 and 180 mg concentration.
All the formulations were passed various physicochemical evaluation parameters and
they were found to be within limits. Whereas from the dissolution studies it was evident
that the formulation (F6) showed better and desired drug release pattern i.e.,96.10 % in
12 hours containing Guar gum polymer in the concentration of 180mg. It followed zero
order release kinetics. For the optimized formulation alcohol effect has been studied by
using various concentrations of alcohol in dissolution medium. As the concentration of
alcohol increased the sustained action of polymer was decreased. Hence it was concluded
that alcohol has significant effect on drug release pattern 41
.
Dawang et al., 2015 developed a prolonged release gastro retentive (GT) formulation of
Verapamil hydrochloride. Drug was evaluated by UV and DSC. A variety of polymers
and effervescent properties were utilized to optimize the desired disposition profile.
Tablets were prepared by the direct compression technique and evaluated for physical
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 42
properties, swelling, floating, and drug release. The purpose of this research was to
formulate and evaluation of a floating tablet of Verapamil hydrochloride by using Gastro
retentive technology using 32 factorial design. Floating tablets were prepared by
incorporating HPMC K15M, sodium alginate, sodium bicarbonate and citric acid. A 32
Factorial design was applied systemically; the amount of HPMC K15M (X1) and sodium
alginate (X2) were selected as independent variables. The time required for 100% drug
release and floating lag time (FLT) were selected as dependent variables. It was found
that HPMC K4M, sodium alginate and their interaction had significant influence on the
% drug release and floating lag time of the delivery system 42
.
Bharat et al, 2014, optimized bilayer gastric floating drug delivery system of Verapamil
hydrochloride to study the effect of formulation variables especially, combination of
polymers on drug release showing prolonged gastric residence time and optimized by
using mathematical and statistical techniques. Three ratios of drug to total polymer
content and three ratios of HPMC K4M to CP934 were chosen for an optimal design. In
the preliminary trials the effect of sodium bicarbonate loading was studied on floating
properties and 12% concentration was found to be optimum for floating buoyancy.
Hardness of about 5 kg/cm2 was found to be optimum for floating buoyancy and to keep
two layers intact. Other physical parameters like weight variation, thickness and friability
were within pharmacopoeial limit. Percentage drug content in all BFT formulations was
found to be 98.47% - 99.96% which were within pharmacopoeial limit. Drug-polymer
ratio and the HPMC K4M-CP934 significantly affect the buoyancy and drug release. It
was concluded on the basis of buoyancy and in-vitro release kinetics that an optimized
formulation containing a ratio of Drug to polymer 1:1 and polymer to polymer 1:1 gave
the best in-vitro release of 99.42% in 12 hrs while for 3:1 and 1:3 in-vitro release was
91.05% and 93.71% respectively. A comparative study was done with the marketed
formulation of Verapamil hydrochloride (Calaptin – SR). FTIR studies show no evidence
of interaction between drug, polymers and other excipients. The In vitro data were fitted
to different kinetic models 43
.
Kondeti et al., 2014 Sustained release matrix tablets reduce the frequency of the dosing
and increase the effectiveness of the drug by localization at the site of action, providing
uniform drug delivery. Sustained release matrix tablets of verapamil hydrochloride were
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 43
prepared by using HPMC K15M, Xanthan and Guar Gum polymers with different
concentration in various batches of the formulations to facilitate the drug release which
cause patient compliance as the dosing frequency is reduced. Verapamil HCl was
considered as an ideal drug for designing sustained release formulation because of the
high frequency of administration and short biological half-life. The sustained release
matrices of Verapamil HCl were prepared by wet granulation technique. Drug release
was studied by using Dissolution testing apparatus 2 (paddle method) with 0.1 N HCl for
2 hours followed by pH 6.8 Phosphate buffer for 8 hours. The in vitro drug release of
various formulations was performed and compared. The results shows that the tablets
formulated with HPMC K15M polymer shows more sustained action when compared to
that of Xanthan and Guar gum. The high viscosity of the polymers binds the formulation
of matrix thus sustains the release of drug. It was also observed that the increase in
concentration of the polymer decreased the drug release from the polymer matrix 44
.
Vidyadhara et al., 2014, developed osmotic controlled extended release formulations of
verapamil hydrochloride an angiotensin II receptor antagonist with anti‑hypertensive
activity. Verapamil hydrochloride matrix tablets were prepared by direct compression
process using hydroxypropyl methylcellulose (HPMC) K 15M as polymeric material and
mannitol as osmogen at varied concentrations. The matrix tablets were further coated
with different compositions of ethylcellulose7cps and polyethylene glycol (PEG)‑4000
by pan coating method. Physical parameters such as weight uniformity, drug content,
hardness and friability were evaluated for uncoated tablets and were found to be within I.
P limits. The coating thickness and percentage of coating applied for various tablets were
also evaluated. The optimized coated tablets were further subjected to micro drilling on
the upper face to get 0.5 µm orifice diameter. All the tablets were further subjected to
dissolution studies by using USP apparatus II with 6.8 pH phosphate buffer as medium.
These studies indicated that all the tablets were found to release the drug up to 12 hours,
while coated tablets with orifice found to release the drug at zero order rate, which was in
good agreement with peppas n > 0.9 45
.
Tola and Li 2014 developed a hydrophobic polymer to overcome the issue of pH-
dependent release of weakly basic model drug verapamil hydrochloride from matrix
tablets without the use of organic buffers in the matrix formulations. Providing pH-
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 44
independent oral release of weakly basic drugs with conventional matrix tablets can be
challenging because of the pH-dependent solubility characteristics of the drugs and the
changing pH environment along the gastrointestinal tract. Silicone pressure-sensitive
adhesive (PSA) polymer was evaluated because of its unique properties of low surface
energy, hydrophobicity, low glass transition temperature, high electrical resistance, and
barrier to hydrogen ion diffusion. Drug release, hydrogen ion diffusion, tablet contact
angle, and internal tablet microenvironment pH with matrix tablets prepared using PSA
were compared with those using water-insoluble ethyl cellulose (EC). Silicone PSA films
showed higher resistance to hydrogen ion diffusion compared with EC films. Verapamil
hydrochloride tablets prepared using silicone PSA showed higher hydrophobicity and
lower water uptake than EC tablets. Silicone PSA tablets also showed pH-independent
release of verapamil and decreased in dimensions during drug dissolution. By contrast,
verapamil hydrochloride tablets prepared using EC did not achieve pH-independent
release 46
.
Syeda et al., 2013 formulated Gastro retentive controlled release drug delivery system of
Verapamil HCl to increase the gastric retention time of the dosage form with controlling
the drug release pattern. Different grades of hydroxy propyl methyl cellulose derivatives;
Methocel K4M and Methocel K15MCR were incorporated for their gel forming
properties. Tablet buoyancy was achieved by incorporating a mixture of gas generating
agents; sodium bicarbonate and anhydrous citric acid. In vitro dissolution studies were
carried out for eight hours using USP XXII paddle type apparatus using 0.1N HCl as the
dissolution medium. All the gastro retentive tablets showed good in vitro buoyancy.
Tablets swelled radially and axially during buoyancy study. The release kinetics were
explored and explained with zero order, first order, Higuchi and Korsmeyer equations.
The release rate, extent and mechanisms were found to be governed by polymer type and
content. Formulations were characterized by physical characterization, drug loading
content and Fourier Transform Infrared spectroscopy (FT-IR). Good results were
obtained in the tests and the FT-IR spectroscopic studies indicating no interaction and the
stability of Verapamil HCl in the used excipients. Based up on the results, it was proved
that a proper balance between rate retarding polymer and gas forming agents is obligatory
for efficient buoyancy and controlled drug release 47
.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 45
Sahi al., 2013, developed the Verapamil hydrochloride sustained-release floating matrix
tablets using gas-generation approach to prolong the gastric residence time. Floating
tablets were prepared using hydroxypropyl methylcellulose K4M (HPMC) as hydrophilic
gel material, sodium bicarbonate as gas-generating agent and Citric Acid as floating
assistant agent. A 32 factorial design was used to select the optimized formulation
wherein HPMC K4M (X1) and Citric Acid (X2) were taken as independent variables and
Floating lag time (FLT), amount of drug release after 24hrs. (Q24) were taken as
dependent variables. The release data were evaluated by the model-dependent (curve
fitting) method using PCP Disso v2.08 software. Optimisation studies were carried out by
using the Design Expert software (version 8.0.1). The floating tablets were evaluated for
uniformity of weight, hardness, thickness, swelling index, friability, drug content, FLT,
and in vitro release. The in vitro drug release followed Hixson-Crowell model and
mechanism of drug release was found to be anomalous or non-fickian type. The
optimized formulation was F3 containing HPMC K4M 15%, and Citric acid 3% having
minimum FLT and maximum drug release after 24 hrs 48
.
Ray and Gupta, 2013 formulated Matrix tablets of Verapamil Hydrochloride as
sustained release tablet employing sodium alginate, hydroxyl propyl methyl cellulose
polymer, Ethyl cellulose and the sustainedrelease tablets was investigated. Sustained
release matrix tablets contain 240 mg Verapamil Hydrochloride were developed using
different drug polymer concentration of HPMC, Sodium Alginate and Ethyl Cellulose.
Tablets were prepared by wet granulation using HPMCand water solution. Formulation
was optimized on the basis of acceptable tablet properties and in-vitro drug release. The
resulting formulation produced robust tablets with optimum hardness, thickness
consistent weight uniformity and low friability. All tablets but one exhibited gradual and
near completion sustained release for Verapamil Hydrochloride, and 99% to 101%
released at the end of 24 hrs. The results of dissolution studies indicated that formulation
F8, the most successful of the study. An increase in release kinetics of the drug was
observed on decreasing polymer concentration 49
.
Mathur et al., 2013 developed a sustained release matrix tablet of verapamil
hydrochloride (VH) using ethyl cellulose, methyl cellulose, Eudragit RS 100,
hydroxypropyl methylcellulose and carboxymethyl cellulose and to evaluate the drug
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 46
release kinetics.Verapamil hydrochloride (VH) is a calcium channel blocking agent used
in the treatment of hypertension, cardiac arrhythmia and angina pectoris. The short
half‑life and high frequency of administration of VH makes it a suitable candidate for
designing sustained drug delivery system. In order to achieve the required sustained
release profile, the tablets were prepared by a wet granulation method using avicel PH
101 and magnesium stearate as binder and lubricant, respectively. The formulated tablets
were characterized for pre‑compression and post‑compression parameters and they were
in the acceptable limits. The drug release data obtained after an in vitro dissolution study
was fitted to various release kinetic models in order to evaluate the release mechanism
and kinetics. The criterion for selecting the best fit model was linearity (coefficient of
correlation). Drug release mechanism was found to follow a complex mixture of
diffusion, swelling and erosion. Furthermore, to minimize the initial burst drug release,
batches were coated by using Eudragit RS100 polymer. After coating the tablets, a better
release profile of the formulated tablets was expected and the release rate of the drug was
compared with the marketed SR tablet of VH 50
.
Vidyadhara et al., 2013 developed controlled release matrix tablets of verapamil
hydrochloride to increase therapeutic efficacy, reduced frequency of administration and
improved patient compliance. Verapamil hydrochloride was formulated as oral controlled
release matrix tablets by using the polyethylene oxides (Polyox WSR 303). They
investigate the influence of polymer level and type of fillers namely lactose (soluble
filler), swellable filler (starch 1500), microcrystalline cellulose and dibasic calcium
phosphate (insoluble fillers) on the release rate and mechanism of release for verapamil
hydrochloride from matrix tablets prepared by direct compression process. Higher
polymeric content in the matrix decreased the release rate of drug. On the other hand,
replacement of lactose with anhydrous dibasic calcium phosphate and microcrystalline
cellulose has significantly retarded the release rate of verapamil hydrochloride.
Biopharmaceutical evaluation of satisfactory formulations were also carried out on New
Zealand rabbits and parameters such as maximum plasma concentration, time to reach
peak plasma concentration, area under the plasma concentration time curve(0‑t) and area
under first moment curve(0‑t) were determined. In vivo pharmacokinetic study proves
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 47
that the verapamil hydrochloride from matrix tablets showed prolonged release and were
be able to sustain the therapeutic effect up to 24 h 51
.
Patel et al., 2013 formulated novel gastro retentive controlled release drug delivery
system of verapamil HCl to increase the gastric retention time of the dosage form and to
control drug release. Hydroxypropylmethylcellulose (HPMC), carbopol, and xanthan
gum were incorporated for gel forming properties. Buoyancy was achieved by adding an
effervescent mixture of sodium bicarbonate and anhydrous citric acid. In vitro drug
release studies were performed, and drug release kinetics was evaluated using the linear
regression method. The optimized intragastric floating tablet composed of 3:2 of HPMC
K4M to xanthan gum exhibited 95.39% drug release in 24 h in vitro, while the buoyancy
lag time was 36.2 s, and the intragastric floating tablet remained buoyant for >24 h. Zero-
order and non-Fickian release transport was confirmed as the drug release mechanism
from the optimized formulation (F7). X-ray studies showed that total buoyancy time was
able to delay the gastric emptying of verapamil HCl intragastric floating tablet in mongrel
dogs for more than 4 h. Optimized intragastric floating tablet showed no significant
change in physical appearance, drug content, total buoyancy time, or in vitro dissolution
pattern after storage at 40°C/75% relative humidity for 3 months 52
.
Nikam et al., 2011 formulated A novel gastro retentive controlled release drug delivery
system of verapamil HCl to increase the gastric retention time of the dosage form and to
control drug release. Gastro retentive systems can remain in the gastric region for several
hours and hence significantly prolong the gastric residence time of drugs. Verapamil
HCL belongs to the class of calcium channel blockers. These medication block the
movement of the calcium into the muscle cells of the coronary arteries. They design and
evaluate verapamil HCL floating controlled release gastroretentive tablets using different
hydrocolloid polymers including Carbopol, Hydroxy propyl methyl cellulose, and
Xanthan gum incorporated for gel forming agent by direct compression technology. The
tablets were evaluated for the physicochemical parameters such as weight variation,
thickness, friability, hardness, drug content, in vitro buoyancy studies, in vitro dissolution
studies. The prepared tablets exhibited satisfactory physico-chemical characteristics.
Tablet buoyancy was achieved by adding an effervescent mixture of sodium bicarbonate
and anhydrous citric acid. The in vitro dissolution studies were carried out in a USP XXII
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 48
apparatus II in 0.1N HCl. All the gastroretentive tablets showed good in-vitro buoyancy.
The selected tablets (F3) containing Xanthan gum released approximately 94.43% drug in
24 h in vitro dissolution study, while the buoyancy lag time was 25.8 ± 4.2 second and
the tablet remained buoyancy for > 24 h. Zero order and non-Fickian release transport
was confirmed as the drug release mechanism for the selected tablets (F3) 53
.
Gangadharappa et al., 2010 developed a controlled release floating drug delivery
system (tablet) of verapamil hydrochloride. Floating tablets of verapamil hydrochloride
were engineering to extend gastric residence time and hence to enhance its
bioavailability. The floating matrix tablets were prepared by direct compression
technique using a combination of hydroxyl propyl methyl cellulose (HPMC) and karaya
gum as polymers and sodium bicarbonate as generating agent. The prepared floating
tablets were evaluated for weight variation test, hardness, thickness, swelling index, in
vitro floating capabilities, floating lag time, compatibility studies, and in vitro drug
release. This swellable hydrophilic natural karaya gum was used to control the release of
drug. The results showed that the optimized formulation F8 containing 23.3% of karaya
gum (70 mg) and 13.3% of HPMC (40 mg) had good floating capability, shorter floating
lag time, and sustained drug release for the period of 8 h 54
.
Molke et al., 2010, prepared a gastroretentive drug delivery system of Verapamil HCL.
Floating tablets of Verapamil HCL were prepared employing solid dispersion technique
with compritol-888ATO. By using different release enhancer like Lactose,
Microcrystaline cellulose & HPMC K 100 LV. Cetyl alcohol was incorporated as a lower
density agent. The floating tablets were evaluated for uniformity of weight, hardness,
friability, drug content, in vitro buoyancy and dissolution studies. The effect of release
enhancer on drug release profile and floating properties was investigated. The prepared
tablets exhibited satisfactory physico-chemical characteristics. The drug release from the
tablets was sufficiently sustained and Fickian transport of the drug from tablets was
confirmed. The fabricated floating tablet formulations were subjected for stability study
at 400C and relative humidity at 75 % for three months. The product was evaluated for,
buoyancy, drug content and in vitro dissolution test. After stability study drug release
increased slightly but there is no change in physical appearance 55
.
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 49
CHAPTER 3: PLAN OF WORK & HYPOTHESIS
3.1: OBJECTIVE:
The objective of the present investigation was to develop a formulation
gastroretentive nizatidine mucoadhesive microballons for treatment of peptic ulcer
mainly at gastric part of GIT, to improve gastric residence time and increase
bioavailability.
: HYPOTHESIS:
A major problem for gastric delivery is the attainment of an optimal concentration
at site of action with maximum bioavailability of drugs. The problem is associated with
the conventional dosage form for peptic ulcer diseases is frequent dosing due to the low
half life. The bioavailability of an instilled compound is generally low from 1.5 – 3.0 h
and low solubility, with only a small fraction reaching the target site.
In the present study an attempt was made to develop a mucoadhesive
Microballons of Nizatidine with variation in polysaccharide polymeric combination with
different ratios to increase mucoadhesion at gastric mucosa, which increase the gastric
residence time, thus increase the bioavailability.
: PLAN OF PRESENT WORK: The study included the following.
: Preformulation Study:
i. Exhaustive literature survey: The exhaustive literature survey about drugs and
various polymers used in present study was done.
ii. Analytical methods: The drug samples (Nizatidine) was studied for determination of
absorption maxima (λmax) in solvents i.e. gastric pH 0.1 N HCl. The analytical method
was validated in terms of preparation of calibration curve, specificity, repeatability
precision, intermediate precision and accuracy.
iii. Preformulation studies of drug sample: The drug samples were studied for
organoleptic properties, microscopic examination by using phase contrast microscope.
The physical characteristics of drug samples i.e. density, particle size, flow properties,
FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE
FOR MANAGEMENT OF PEPTIC ULCER
Page 50
compatibility, solubility in various dissolution medias, partition coefficient and drug-
excipients compatibility study were characterized.
: Formulation and characterization of mucoadhesive Microballons: These prepared
systems were evaluated with various parameters such as the physical properties i.e., Flow
properties determination, particle size measurement, shape and surface morphology,
mucoadhesive properties, swelling study, percentage yield, drug entrapment efficiency,
in-vitro drug release studies and Stability Studies etc.
: Result and discussion: This section was included all the results of present research
work.
: Summary and conclusion: This section was included summary and conclusion of
present research work.
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis
M.pharm thesis

More Related Content

What's hot

Scale up and post approval changes(supac)
Scale up and post approval changes(supac)Scale up and post approval changes(supac)
Scale up and post approval changes(supac)
bdvfgbdhg
 

What's hot (20)

EVALUATION OF TRANSDERMAL DRUG DELIVERY SYSTEMS
EVALUATION OF TRANSDERMAL DRUG DELIVERY SYSTEMSEVALUATION OF TRANSDERMAL DRUG DELIVERY SYSTEMS
EVALUATION OF TRANSDERMAL DRUG DELIVERY SYSTEMS
 
Mucoadhesive drug delivery system
Mucoadhesive drug delivery systemMucoadhesive drug delivery system
Mucoadhesive drug delivery system
 
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
ICH Q8 GUIDELINES OF QUALITY BY DESIGN(PRODUCT DEVELOPEMENT)
 
Evaluation of transdermal drug delivery system
Evaluation of transdermal drug delivery systemEvaluation of transdermal drug delivery system
Evaluation of transdermal drug delivery system
 
Alzet osmotic pump
Alzet osmotic pumpAlzet osmotic pump
Alzet osmotic pump
 
Gastro retentive drug delivery system (GRDDS)
Gastro retentive drug delivery system (GRDDS)Gastro retentive drug delivery system (GRDDS)
Gastro retentive drug delivery system (GRDDS)
 
Mucosal drug delivery system
Mucosal drug delivery systemMucosal drug delivery system
Mucosal drug delivery system
 
NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)NEW DRUG APPLICATION ( NDA)
NEW DRUG APPLICATION ( NDA)
 
polymer in pharmacy and application of polymers
polymer in pharmacy and application of polymerspolymer in pharmacy and application of polymers
polymer in pharmacy and application of polymers
 
Supac
Supac Supac
Supac
 
Nasopulmonary Drug Delivery System
Nasopulmonary Drug Delivery SystemNasopulmonary Drug Delivery System
Nasopulmonary Drug Delivery System
 
Rate-Controlled Drug Delivery System
Rate-Controlled Drug Delivery SystemRate-Controlled Drug Delivery System
Rate-Controlled Drug Delivery System
 
Penetration enhancer with their examples
Penetration enhancer with their examplesPenetration enhancer with their examples
Penetration enhancer with their examples
 
Investigational New Drug Application
Investigational New Drug ApplicationInvestigational New Drug Application
Investigational New Drug Application
 
Non clinical drug development. ppt
Non clinical drug development. pptNon clinical drug development. ppt
Non clinical drug development. ppt
 
Scale up and post approval changes(supac)
Scale up and post approval changes(supac)Scale up and post approval changes(supac)
Scale up and post approval changes(supac)
 
Quality by Design ( QbD )
Quality by Design ( QbD )Quality by Design ( QbD )
Quality by Design ( QbD )
 
Supac
SupacSupac
Supac
 
Pharmaceutical validation & it's types
 Pharmaceutical validation & it's types Pharmaceutical validation & it's types
Pharmaceutical validation & it's types
 
ICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validationICH & WHO GUIDELINES ON validation
ICH & WHO GUIDELINES ON validation
 

Similar to M.pharm thesis

Bioinformatics resources and search tools - report on summer training proj...
Bioinformatics   resources and search tools -  report on summer training proj...Bioinformatics   resources and search tools -  report on summer training proj...
Bioinformatics resources and search tools - report on summer training proj...
Sapan Anand
 
WHO and ICH Guidelines for the Assessment of Herbal Drug
WHO and ICH Guidelines for the Assessment of Herbal DrugWHO and ICH Guidelines for the Assessment of Herbal Drug
WHO and ICH Guidelines for the Assessment of Herbal Drug
ijtsrd
 

Similar to M.pharm thesis (20)

HPLC Drugs Analysis Report
HPLC Drugs Analysis ReportHPLC Drugs Analysis Report
HPLC Drugs Analysis Report
 
QUALITY EVALUATION ON PROMETHAZINE TABLETS MARKETED IN NEPAL
QUALITY EVALUATION ON PROMETHAZINE TABLETS MARKETED IN NEPALQUALITY EVALUATION ON PROMETHAZINE TABLETS MARKETED IN NEPAL
QUALITY EVALUATION ON PROMETHAZINE TABLETS MARKETED IN NEPAL
 
Mynul_Islam_Mufti__pdf
Mynul_Islam_Mufti__pdfMynul_Islam_Mufti__pdf
Mynul_Islam_Mufti__pdf
 
shuna 22.pdf
shuna 22.pdfshuna 22.pdf
shuna 22.pdf
 
NOVEL SIMULTANEOUS SEPARATION AND QUANTITATIVE DETERMINATION OF FOUR SARTANS...
NOVEL SIMULTANEOUS SEPARATION AND QUANTITATIVE DETERMINATION OF  FOUR SARTANS...NOVEL SIMULTANEOUS SEPARATION AND QUANTITATIVE DETERMINATION OF  FOUR SARTANS...
NOVEL SIMULTANEOUS SEPARATION AND QUANTITATIVE DETERMINATION OF FOUR SARTANS...
 
charmee final binder
charmee final bindercharmee final binder
charmee final binder
 
Bioinformatics resources and search tools - report on summer training proj...
Bioinformatics   resources and search tools -  report on summer training proj...Bioinformatics   resources and search tools -  report on summer training proj...
Bioinformatics resources and search tools - report on summer training proj...
 
report
reportreport
report
 
INDUSTRIAL TRAINING REPORT
INDUSTRIAL TRAINING REPORTINDUSTRIAL TRAINING REPORT
INDUSTRIAL TRAINING REPORT
 
Full Thesis
Full ThesisFull Thesis
Full Thesis
 
srikanth final presentation
srikanth final presentationsrikanth final presentation
srikanth final presentation
 
Tretinoin Emulgel
Tretinoin EmulgelTretinoin Emulgel
Tretinoin Emulgel
 
Thesis Final
Thesis FinalThesis Final
Thesis Final
 
WHO and ICH Guidelines for the Assessment of Herbal Drug
WHO and ICH Guidelines for the Assessment of Herbal DrugWHO and ICH Guidelines for the Assessment of Herbal Drug
WHO and ICH Guidelines for the Assessment of Herbal Drug
 
Design and development of insitu gel of opthalmic drug delivery system
Design and development of insitu gel of opthalmic drug delivery systemDesign and development of insitu gel of opthalmic drug delivery system
Design and development of insitu gel of opthalmic drug delivery system
 
Curriculum Vitae
Curriculum VitaeCurriculum Vitae
Curriculum Vitae
 
srini resume
srini resumesrini resume
srini resume
 
Synopsis copy
Synopsis   copySynopsis   copy
Synopsis copy
 
new cv
new cvnew cv
new cv
 
Development and Dose Modification of Ayurvedic Formulations
Development and Dose Modification of Ayurvedic FormulationsDevelopment and Dose Modification of Ayurvedic Formulations
Development and Dose Modification of Ayurvedic Formulations
 

Recently uploaded

Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
muralinath2
 
Aerodynamics. flippatterncn5tm5ttnj6nmnynyppt
Aerodynamics. flippatterncn5tm5ttnj6nmnynypptAerodynamics. flippatterncn5tm5ttnj6nmnynyppt
Aerodynamics. flippatterncn5tm5ttnj6nmnynyppt
sreddyrahul
 
Anemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditionsAnemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditions
muralinath2
 
Mammalian Pineal Body Structure and Also Functions
Mammalian Pineal Body Structure and Also FunctionsMammalian Pineal Body Structure and Also Functions
Mammalian Pineal Body Structure and Also Functions
YOGESH DOGRA
 
THYROID-PARATHYROID medical surgical nursing
THYROID-PARATHYROID medical surgical nursingTHYROID-PARATHYROID medical surgical nursing
THYROID-PARATHYROID medical surgical nursing
Jocelyn Atis
 

Recently uploaded (20)

NuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final versionNuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final version
 
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdfSCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
 
Astronomy Update- Curiosity’s exploration of Mars _ Local Briefs _ leadertele...
Astronomy Update- Curiosity’s exploration of Mars _ Local Briefs _ leadertele...Astronomy Update- Curiosity’s exploration of Mars _ Local Briefs _ leadertele...
Astronomy Update- Curiosity’s exploration of Mars _ Local Briefs _ leadertele...
 
In silico drugs analogue design: novobiocin analogues.pptx
In silico drugs analogue design: novobiocin analogues.pptxIn silico drugs analogue design: novobiocin analogues.pptx
In silico drugs analogue design: novobiocin analogues.pptx
 
Transport in plants G1.pptx Cambridge IGCSE
Transport in plants G1.pptx Cambridge IGCSETransport in plants G1.pptx Cambridge IGCSE
Transport in plants G1.pptx Cambridge IGCSE
 
Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
Circulatory system_ Laplace law. Ohms law.reynaults law,baro-chemo-receptors-...
 
Aerodynamics. flippatterncn5tm5ttnj6nmnynyppt
Aerodynamics. flippatterncn5tm5ttnj6nmnynypptAerodynamics. flippatterncn5tm5ttnj6nmnynyppt
Aerodynamics. flippatterncn5tm5ttnj6nmnynyppt
 
INSIGHT Partner Profile: Tampere University
INSIGHT Partner Profile: Tampere UniversityINSIGHT Partner Profile: Tampere University
INSIGHT Partner Profile: Tampere University
 
biotech-regenration of plants, pharmaceutical applications.pptx
biotech-regenration of plants, pharmaceutical applications.pptxbiotech-regenration of plants, pharmaceutical applications.pptx
biotech-regenration of plants, pharmaceutical applications.pptx
 
Gliese 12 b, a temperate Earth-sized planet at 12 parsecs discovered with TES...
Gliese 12 b, a temperate Earth-sized planet at 12 parsecs discovered with TES...Gliese 12 b, a temperate Earth-sized planet at 12 parsecs discovered with TES...
Gliese 12 b, a temperate Earth-sized planet at 12 parsecs discovered with TES...
 
Richard's entangled aventures in wonderland
Richard's entangled aventures in wonderlandRichard's entangled aventures in wonderland
Richard's entangled aventures in wonderland
 
EY - Supply Chain Services 2018_template.pptx
EY - Supply Chain Services 2018_template.pptxEY - Supply Chain Services 2018_template.pptx
EY - Supply Chain Services 2018_template.pptx
 
GBSN - Biochemistry (Unit 5) Chemistry of Lipids
GBSN - Biochemistry (Unit 5) Chemistry of LipidsGBSN - Biochemistry (Unit 5) Chemistry of Lipids
GBSN - Biochemistry (Unit 5) Chemistry of Lipids
 
SAMPLING.pptx for analystical chemistry sample techniques
SAMPLING.pptx for analystical chemistry sample techniquesSAMPLING.pptx for analystical chemistry sample techniques
SAMPLING.pptx for analystical chemistry sample techniques
 
Anemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditionsAnemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditions
 
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
 
A Giant Impact Origin for the First Subduction on Earth
A Giant Impact Origin for the First Subduction on EarthA Giant Impact Origin for the First Subduction on Earth
A Giant Impact Origin for the First Subduction on Earth
 
National Biodiversity protection initiatives and Convention on Biological Di...
National Biodiversity protection initiatives and  Convention on Biological Di...National Biodiversity protection initiatives and  Convention on Biological Di...
National Biodiversity protection initiatives and Convention on Biological Di...
 
Mammalian Pineal Body Structure and Also Functions
Mammalian Pineal Body Structure and Also FunctionsMammalian Pineal Body Structure and Also Functions
Mammalian Pineal Body Structure and Also Functions
 
THYROID-PARATHYROID medical surgical nursing
THYROID-PARATHYROID medical surgical nursingTHYROID-PARATHYROID medical surgical nursing
THYROID-PARATHYROID medical surgical nursing
 

M.pharm thesis

  • 1. FORMULATION AND EVALUATION OF GASTRO- RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Dissertation Part-II report submitted to RAJIV GANDHI PROUDYOGIKI VISHWAVIDYALAYA, BHOPAL (M.P.) For the partial fulfillment of the degree of MASTER OF PHARMACY Pharmaceutics Supervised by Dr.SarangJain Principal Submitted by PARMANAND DHAKAD Enroll.no 0148PY16MP09 Co-GUIDE Ms. Swati Saxena Associate Professor RAJEEV GANDHI COLLEGE OF PHARMACY,BHOPAL SESSION2020-2021
  • 2.
  • 3.
  • 4. RAJEEV GANDHI COLLEGE OF PHARMACY Village: Salaiya, Via Danish Kunj, Kolar Road, Bhopal Department of Pharmaceutics CERTIFICATE This is to certify that the dissertation entitled “FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER” submitted to Rajiv Gandhi Proudyogiki Vishwavidhyalaya, Bhopal by Mr. Parmanand Dhakad is a partial fulfillment of the requirement for the award of the degree of the M.Pharm. with specialization in Pharmaceutics. The matter embodied is the actual work by Mr. Parmanand Dhakad and this work has not been submitted earlier in part or full for the award of any otherdegree. PARMANAND DHAKAD Enroll. No. 0148PY16MP09 Dr. Sarang Jain Principal Ms. Swati Saxena Associate Professor
  • 5. RAJEEV GANDHI COLLEGE OF PHARMACY BHOPAL DECLARATION I hereby declare that the work, which is being presented in the dissertation, entitled “FORMULATION AND EVALUATION OF GASTRO- RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER” submitted by Mr. Parmanand Dhakad (0148PY16MP09) for the award of degree of “Master of Pharmacy” degree with specialization in Pharmaceutics, comprises of bonafide research work carried out by me in our laboratories, library and computer centre under the guidance of Dr. Sarang Jain, Principal and Ms Swati Saxena, Associate Professor., Department of Pharmaceutics, Rajeev Gandhi College of Pharmacy, Bhopal. I also declare that the present work embodies has not formed the basis for the award of any other degree or fellowship previously. The particulars given in this thesis are to best of myknowledge. Date Place: Bhopal PARMANAND DHAKAD Enroll. No. 0148PY16MP09
  • 6. RAJEEV GANDHI COLLEGE OF PHARMACY BHOPAL DECLARATION I hereby declare that the work, which is being presented in the dissertation, entitled “Formulation And Evaluation of Gastro-retentive Mucoadhesive Microballons of Nizatidine For Management of Peptic Ulcer” partial fulfillment of the requirements for the award of degree of Master of Pharmacy in Pharmaceutics submitted in the department of Pharmaceutics (Rajeev Gandhi College of Pharmacy, Bhopal M.P.) is an authentic record of my own work carried under the guidance of Dr.Sarang Jain and Ms Swati Saxena. I have not submitted the matter embodiedin this report for award of any otherdegree. I also declare that “A check for Plagiarism has been carried out on the thesis/ project report/ dissertation and is found within the acceptable limit and report of which is enclosed herewith.” Name of Student PARMANAND DHAKAD Enroll. No. 0148PY16MP09 Name of Supervisor Dr. Sarang Jain Principal Name of Co-Supervisor Ms. Swati Saxena Associate Professor
  • 7. ACKNOWLEDGEMENT Firstly, I offer my adoration to God who created me, gave me the strength and courage to complete my dissertation and gave me the opportunity to thank all those people through whom his Grace was delivered to me. With a deep sense of gratitude, I express my indebtedness to my guide, Dr. Sarang Jain, Principal Rajeev Gandhi college of Pharmacy, Bhopal, for his valuable guidance, boundless enthusiasm and constant inspiration throughout the entire course of the work. I shall forever remain indebted to his for having inculcated in me a quest for excellence, a spirit of diligenceIt is my privilege to express my heartfelt thanks to my Co- guide Ms Swati Saxena, Department of Pharmaceutics, for her guidance and encouragement throughout the research work. I sincerely thank all the teaching staff and non-teaching staff, Mr Navneet Dube, Mr Anuj Singhai, Mr. Bajpei, Mr.Pradeep and librarian Mr. Dharmendra. It gives me immense pleasure to record my sincere thanks to my colleagues friends& Non collegeous friends Risabh mishra, Sumit Raikwar, Nidhi Malviya ,and for their help and co- operation. I sincerely acknowledge the authorities of Rajeev Gandhi college of Pharmacy, Bhopal, for providing the necessary facilities to carry out my research work. I would like to thank all those who have helped me directly or indirectly to complete this work successfully. From deepest depth of my heart, I express my love and gratitude to my beloved parents Mr Raghuraj Singh Dhakad , Mrs. Shusila Bai and all family members for their love, support and constant encouragement throughout my career. Thank you one & all Date Place: Bhopal PARMANAND DHAKAD Enroll. No. 0148PY16MP09
  • 8. [not detected] [not detected] GoogleBooks Detected! Wiki Detected! Anti-cheating: Ghostwriting services: Google Books: Wikipedia: Plagiarism Detector v. 1517 - Originality Report Analyzed document: 30-09-2020 16:51:52 " FORMULATION AND EVALUATION OF GASTRO- RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MR. MANAGEMENT OF PEPTIC ULCER” PARMANAND DHAKAD Enroll. No. 0148PY16MP09 M PHARM IV.docx Check Type: Internet - via Google and Bing Licensed Version Relation chart: Important notes: Plagiarism Detector Your right to know the authenticity!
  • 9. CONTENT Chapter Title Page no. 1 INTRODUCTION 1-24 1.1 Peptic ulcer 1 1.2 Drug Delivery System 6 1.3 Gastro-retentive drug delivery system 9 1.4 Mechanism of Bioadhesion 15 1.5 Mucoadhesive Microspheres 23 2 REVIEW OF LITERATURE 25-48 2.1 Drug profile 25 2.2 Excipients review 26 2.3 Literature review 39 3 PLAN OF WORK & HYPOTHESIS 49-50 3.1 Objective 49 3.2 Hypothesis 49 3.3 Plan of present work 49 4 MATERIAL AND METHODS 51-59 4.1 Analytical and Validation studies 51 4.2. Preformulation Studies 52 4.2.1 Organoleptic properties 52 4.2.2 Microscopic examination 52 4.2.3 Physical Characteristics 52 4.3 Preparation of mucoadhesive Microballons 55 4.4 Evaluation of mucoadhesive microballons 57
  • 10. 5 RESULT AND DISCUSSION 60-60 5.1 Analytical and Preformulation studies of model drug 60 5.2 Preformulation Studies 61 5.3 Evaluation of mucoadhesive microspheres 65 6 Summary and conclusion 81-82 7 References 83-67
  • 11. LIST OF TABLES Table No. Particulars Page No. 1.1 Routes of drug administration and dosage form / drug delivery systems 6 1.2 Different theories explaining the mechanism of bioadhesion 15 1.3 Bioadhesive strength of some polymers 21 1.4 Application of Bioadhesive Systems 22 1.5 Patents related to bioadhesive system 23 4.1 Drug-excipient combinations for compatibility study 55 4.2 Preparation of of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 56 5.1 Organoleptic characteristics of Nizatidine HCl 62 5.2 Flow properties of drug (n = 3) 62 5.3 The solubility of Nizatidine HCl at different pH medium (n=3) 62 5.4 Drug-excipient combinations for compatibility study 63 5.5 Results of physical observation 63 5.6 Results of content determination 63 5.7 Percentage yield of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 67 5.8 Particle size of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 67 5.9 Drug entrapment efficiency of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 68 5.10 Degree of swelling of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 68 5.11 Percent mucoadhesion of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 69 5.12 Buoyancy test of mucoadhesive microballons of Nizatidine HCl (A1 – B3) 70
  • 12. 5.13 LIST OF FIGURES 70 Dissolution data of mucoadhesive microballons of Nizatidine HCl (A1 –B3) 5.14 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (A1) 71 5.15 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (A2) 72 5.16 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (CH1) 73 5.17 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (CH2) 74 5.18 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (CA1) 75 5.19 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (CA2) 76 5.20 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (B1) 77 5.21 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (B2) 78 5.22 in-vitro dissolution data of mucoadhesive microballons of Nizatidine HCl (B3) 79
  • 13. Figure No. Particulars Page No. 1.1 (a) Microspheres floats on stomach contents. (b & c) microspheres adhere to stomach wall 14 1.2 Three regions within a mucoadhesive joint 16 2.1 Structure of drug Nizatidine 25 2.2 Chemical structure of microcrystalline cellulose 27 2.3 Chemical structure of anhydrous lactose 29 2.4 Chemical structure of sodium starch glycolate 30 2.5 Chemical structure of starch 32 2.6 Chemical structure of guar gum 35 2.7 Chemical structure of cellulose hydroxypropyl methyl ether 38 5.1 Absorption maxima (λ-max) of Nizatidine HCl in 0.1N HCl solution (10 μg/ml) 60 5.2 Standard curve of Nizatidine HCl in 0.1N HCl solution (228 nm) 61 5.3 The I. R. Spectrum of sample of pure Nizatidine HCl (S1) 64 5.4 The I. R. Spectrum of sample of Nizatidine HCl and all excipients (S2) 64 5.5 Photograph of microspheres (100X) 66 5.6 SEM photomicrograph of microspheres (650X) 66 5.7 Zero-order plots for mucoadhesive microballons of Nizatidine HCl (A1 – B3) 80 5.8 First-order plots for for mucoadhesive microballons of Nizatidine HCl (A1 – B3) 80
  • 14. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 1 1: INTRODUCTION Peptic Ulcer Drug Delivery System Gastro-retentive Drug Delivery System Mechanism of Bioadhesion PEPTIC ULCER Peptic ulcer occurs in that part of the gastrointestinal tract (g.i.t.) which is exposed to gastric acid and pepsin, i.e. the stomach and duodenum. The etiology of peptic ulcer is not clearly known. It results probably due to an imbalance between the aggressive (acid, pepsin, bile and H. pylori) and the defensive (gastric mucus and bicarbonate secretion, prostaglandins, nitric oxide, high mucosal blood flow, innate resistance of the mucosal cells) factors. A variety of psychosomatic, humoral and vascular derangements have been implicated and the importance of Helicobacter pylori infection as a contributor to ulcer formation and recurrence has been recognized. In gastric ulcer, generally acid secretion is normal or low, while deficient mucosal defence (mostly impaired mucus and bicarbonate secretion) plays a greater role. In duodenal ulcer, acid secretion is high in about half of the patients but normal in the rest. Notwithstanding whether production of acid is normal or high, it does contribute to ulceration as an aggressive factor, reduction of which is the main approach to ulcer treatment. An understanding of the mechanism and control of gastric acid secretion will elucidate the targets of antisecretory drugaction. An ulcer is a round or oval shaped hole (also called parietal defect), 2 to 4 cm in diameter with perpendicular borders and a smooth base. A Peptic Ulcer is an ulcer in the gastrointestinal tract that is characteristically acidic and thus extremely painful. It is also called ulcers pepticum or peptic ulcer disease (PUD). Contrary to general belief peptic ulcers happen more often in the duodenum first part of the small intestine than in the stomach. Duodenal ulcers are usually benign whereas about 4% of stomach ulcers are caused by a malignant tumor. The borders of the Peptic ulcer are not well-known in the acute form but elevated and inflammatory in the chronic form. In the ulcerative form of
  • 15. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 2 Heartburn gastric cancer the borders are uneven. Due to parietal scarring the surrounding mucosa (inner mucus lining) may come into view. Types of Peptic ulcer Peptic Ulcers are categorized by their site in the body: Gastric Ulcer – in the stomach Duodenal Ulcer – in the duodenum (the first part of the small intestine) Esophageal Ulcer – in the esophagus Meckel’s Diverticulum Ulcer – a small pouch in the wall at the junction of the small and large intestines Prepyloric Ulcer – between inner and outer walls of the stomach Proximal gastroesophageal Ulcer – in the mucosa, submucosa, and muscular layer on the lower esophagus, stomach, or duodenum to have radial folds. Symptoms of Peptic Ulcer A patient with Peptic Ulcer would have some of the next symptoms Bloating of the abdomen Waterbrash – this is the rush of saliva in the mouth after an incident of regurgitation in order to dilute the acid in the esophagus Abdominal pain – duodenal ulcers are characteristically relieved by foodwhile gastric ulcers are exacerbated by it Nausea and a lot of vomiting Loss of appetite Weight loss Hematemesis – vomiting of blood due to gastric ulcer or injure to esophagus from frequent vomiting
  • 16. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 3 Melena – tarry foul-smelling feces due to oxidized iron from the hemoglobin Gastric or duodenal perforation (in some severe cases). The timing of the symptoms in relation to meals may distinguish between gastric and duodenal ulcers. In the gastric ulcer, the ache would be during or after meals because the alkaline duodenal contents reflux into the stomach. In duodenal ulcers the ache would manifest before a meal when acid manufacturing increases due to hunger and is passed into the duodenum. However, this is not used as a dependable method of diagnosis. Causes of Peptic Ulcer In most cases, tobacco smoking, anorexia (malnutrition), blood group, spicy food and other factors that were suspected to source ulcers until late in the 20th century, are actually of minor importance in the development of peptic ulcers. But these are factors that make worse the symptoms of peptic ulcer, once formed. Some of the causes are: Bacteria: As much as 60% of gastric and up to 90% of duodenal ulcers are linked with Helicobacter pylori, a spiral-shaped bacterium that lives in the acidic surroundings of the stomach. This bacterium colonies the antral mucosa (inner mucus lining) and grows rapidly. The antibodies are not capable to clear the infection. This causes localized erosion and damage of the mucosa and results in ulcer formation. Reaction to medications: Ulcers can also be sourced or worsened by drugs such as aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs). Normally, prostaglandins kindle the secretion of mucus in the gastrointestinal tract to defend its inner lining from gastric acid. NSAIDs block this function and the risk of ulceration increases. Hence, peptic ulcer due to reaction to medication is found additional in aged people suffering from arthritis or those with pain syndromes. Ulcerogenesis can also be caused by making use of glucocorticoids. Tobacco: Smoking leads to atherosclerosis and vascular spasms. This guides to ischemia (localized reduction of blood flow) and ulcers.
  • 17. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 4 Heredity: A family history is often there in duodenal ulcers, especially when blood group O is also present. But studies show that this is not a significant factor in the case of gastric ulcers. Zollinger Ellison syndrome: Gastrinomas, rare gastrin-secreting tumors cause multiple ulcers which are hard to heal. Diagnosis of Peptic Ulcer Endoscopy: Esophagogastroduodenoscopy (EGD), a form of endoscopy, also recognized as a gastroscopy, is carried out on patients in whom a peptic ulcer is supposed. By direct visual identification, the location and harshness of an ulcer can be diagnosed. Moreover, if no ulcer is there, EGD can often give an alternative diagnosis. In patients over 45 years with more than two weeks of the above symptoms, the odds for peptic ulceration are high sufficient to warrant rapid investigation by EGD. Findings of endoscopy: The microscopical emergence of the ulcer shows 4 zones: inflammatory exudate (fluid); fibrinoid necrosis dead cells; granulation tissue; and fibrous tissue. The fibrous base of the ulcer may hold vessels with thickened wall or with thrombosis (formation of blood clot). Urea breath test: This is a non-invasive test and does not need an endoscopy. Blood test: This is just to ensure for the presence of antibodies but is not a sufficient diagnostic method for Peptic ulcer. Stool antigen test: This is a non-invasive immunoassay to identify active infection with Helicobacter pylori bacteria in fecal samples. Histological examination: The possibility of other causes of ulcers, notably malignancy (gastric cancer) needs to be reserved in mind. This is especially true in ulcers of the greater (large) curvature of the stomach. X-ray: This is carried out to rule out perforated Peptic Ulcer. If the air leaks from the gastrointestinal tract (which contains air) to the peritoneal cavity (which never contains air), there would be free gas within the peritoneal cavity. The gas will
  • 18. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 5 drift to a position underneath the diaphragm and a chest X-ray would illustrate an erect chest and supine, lateral abdomen. This is a sure signal of perforated peptic ulcer and should be treated forthwith. Treatment of Peptic Ulcer Antacids: Young, otherwise fit patients with ulcer-like symptoms are treated with antacids or H2 antagonists before Endoscopy (EGD) is undertaken. Bismuth compounds may in fact reduce or even clear organisms and no further treatment is required. Antibiotics: When Helicobacter pylori infection is there, the most effective treatments are mixtures of any two antibiotics like Clarithromycin, Amoxicillin, Tetracycline or Metronidazole along with a proton pump inhibitor (PPI) and a bismuth compound. Surgery: Perforated peptic ulcer becomes a surgical emergency and requires surgical mend of the perforation. Most bleeding ulcers need endoscopy urgently to stop the bleeding with cautery destroying abnormal tissue by burning with electrosurgery or with injection Prevention of Peptic Ulcer There is no proven way to stop peptic ulcer disease (PUD) but some measure can be taken to decrease the risk: Reduce use of NSAIDs: Use of non-steroidal anti-inflammatory drugs (NSAIDs) appears to grounds ulcers by inhibiting the body’s production of prostaglandins, hormones that defend the stomach lining. Patients should also be prescribed a prostaglandin analogue in order to help stop peptic ulcers. Tested for Helicobacter pylori: If a person has a family history of ulcers being tested for disease by Helicobacter pylori bacterium can be a preventivemeasure Quit smoking: Smokers are about twice as likely to extend ulcers as non-smokers as they are more vulnerable to Helicobacter pylori, and provide more favorable conditions for the bacteria to thrive and diminish gastric mucosaldefenses.
  • 19. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 6 Reduce stress: There’s conflicting evidence that pressure is one of the causes of peptic ulcer and more so in duodenal ulcers. But relaxation exercises like yoga; meditation; and other stress-reducing strategies can assist decrease blood pressure and slow heart rate more as a general wellness practice. Stop alcohol: Alcohol aggravates formation and uneasiness from ulcers although it does not reason it 1 . Drug Delivery System The drug-delivery system should deliver drug at a rate dictated by the needs of the body over a specified period of time. The goal of any drug delivery system is to provide a therapeutic amount of drug to a proper site in the body, so that the desired drug concentration can be achieved promptly and then maintained. The idealized objective points to the two aspects most important to drug delivery, namely, spatial placement and temporal delivery. Spatial placement relates to targeting drugs to specific organs, tissues, cells, or even subcellular compartments; whereas temporal delivery refers to controlling the rate of drug delivery to the target site 2,3 . The various routes of drug administration and dosage forms 4 are shown in Table 1. Table 1.1: Routes of drug administration and dosage form / drug deliverysystems S. NO. TERM SITE PRIMARY DOSAGE FORM 1 Oral Mouth Tablets, Capsules, Solutions, Syrups, Elixirs, Suspensions, Magmas, Gels, Powders Peroral (per os: to be swalled) Gastrointestinal tract via mouth 2 Sublingual Under the tongue Tablets, Troches or Lozenges 3 Parenteral Other than the gastrointestinal tract (by Solutions, Suspensions
  • 20. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 7 injection) Interavenous Vein Interaarterial Artery Interacardiac Heart Interaspinal or Interathecal Spine Interaosseous Bone Interaarticular Joint Interasynovial Joint-fluid area Interacutaneous or Interadermal Skin Subcutaneous Beneath the skin Interamuscular Muscle 4 Epicutaneous (Topical) Skin surface Ointments, Creams, Infusion pumps, Pastes, Plasters, Powders,Aerosols, Lotions, Transdermal patches, Transdermal discs, Transdermal solutions 5 Transdermal Skin surface 6 Conjunctival Conjunctiva Contact lenses inserts, Ointments 7 Intraocular Eye Solutions, Suspensions 8 Intranasal Nose Solutions, Sprays, Inhalants, Ointments 9 Aural Ear Solutions
  • 21. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 8 Minimize or eliminate local side effects 10 Intrarepiratory Lung Aerosols 11 Rectal Rectum Solutions, Ointments, Suppositories 12 Vaginal Vagina Solutions, Ointments, Emulsion foams, Tablets, Inserts, Suppositories, Sponge 13 Urethral Urethra Solutions, Suppositories : Several types of modified-release drug products are recognized: 1.2.1.1: Extended-release drug products: A dosage form that allows at least a twofold reduction in dosage frequency as compared to that drug presented as an immediate-release (conventional) dosage form. Examples of extended-release dosage forms include controlled-release, sustained-release, and long-acting drug products5, 6 . : Delayed-release drug products: A dosage form that releases a discrete portion or portions of drug at a time or at times other than promptly after administration, although one portion may be released promptly after administration. Enteric-coated dosage forms are the most common delayed-release products. : Targeted-release drug products: A dosage form that releases drug at or near the intended physiologic site of action. Targeted-release dosage forms may have either immediate- or extended-release characteristics 6, 7 . : Advantages of modified drug therapy: 6 1. Avoid patient compliance problems 2. Employ less total drug
  • 22. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 9 Minimize or eliminate systematic side effects Obtain less potentiation or reduction in drug activity with chronic use Minimize drug accumulation with chronic dosing 3. Improve efficiency in treatment Cure and control condition more promptly Improve control of condition (i.e., reduce fluctuation in drug level) Improve bioavailability of somedrugs 4. Economic savings Gastro-retentive drug delivery system: Even though various drug delivery systems are used for maximizing therapeutic index and reduction in the side effects of the drug, oral route remains the preferred, promising and effective route for the administration of therapeutic agents. Because, low cost of therapy, ease of administration, flexibility in formulation and handling leads to higher level of patient compliance. Approximately 50% of the drug delivery systems available in the market are oral drug delivery system 8 . The novel design of an oral controlled drug delivery system during last two decades, it has limited success in case of drugs with a poor absorption window throughout the GIT (Gastro Intestinal Tract). This approach has several physiological difficulties such as inability to restrain and locate the controlled drug delivery system within the desired region of the gastrointestinal tract (GIT) due to variable gastric emptying and motility. Furthermore, the relatively brief gastric emptying time in humans which normally averages 2-3 h through the major absorption zone, i.e., stomach and upper part of the intestine can result in incomplete drug release from the drug delivery system leading to reduced efficacy of the administered dose 9 Advantages of GRDDs 1. Floating dosage forms such as tablets or capsules will remains in the solutionfor prolonged time even at the alkaline pH of the intestine
  • 23. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 10 2. FDDS are advantageous for drugs meant for local action in the stomach eg: antacids 3. FDDS dosage forms are advantageous in case of vigorous intestinal movement and in diarrhea to keep the drug in floating condition in stomach to get a relatively better response. 4. Acidic substance like aspirin causes irritation on the stomach wall when come in contact with it hence; HBS/FDDS formulations may be useful for the administration of aspirin and other similar drugs. 5. The FDDS are advantageous for drugs absorbed through the stomach eg: Ferrous salts, Antacids 10-12 . Disadvantages of GRDDs 1. Floating systems are not feasible for those drugs that have solubility or stability problems in gastric fluids. 2. Drugs such as Nifedipine, which is well absorbed along the entire GI tract and which undergo significant first pass metabolism, may not be suitable candidates for FDDS. 3. One of the disadvantages of floating systems is that they require a sufficiently high level of fluids in the stomach, so that the drug dosage form float therein and work efficiently. 4. These systems also require the presence of food to delay their gastric emptying 13- 14 . Potential drug candidates for gastro retentive drug delivery Systems 1. Drugs those are locally active in the stomach e.g misoprostol, antacids etc. 2. Drugs that have narrow absorption window in gastrointestinal tract (GIT) e.g. L- DOPA, para-aminobenzoic acid, furosemide, riboflavin etc. 3. Drugs those are unstable in the intestinal or colonic environment e.g.captopril, ranitidine HCl, 4. Drugs that disturb normal colonic microbes e.g.antibiotics against Helicobacter pylori.
  • 24. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 11 5. Drugs that exhibit low solubility at high pH values e.g. diazepam, chlordiazepoxide, 6. verapamil HCl. Drugs those are unsuitable for gastroretentive drug delivery systems 1. Drugs that have very limited acid solubility e.g. phenytoin etc. 2. Drugs that suffer instability in the gastric environment e.g. erythromycin etc. 3. Drugs intended for selective release in the colon. eg. 5- amino salicylic acid and corticosteroids etc.15 . Polymers and other ingredients used for the preparations of Floating drugs i) Polymers: The following polymers used to preparations of floating drugs: HPMC K4 M, Calcium alginate, Eudragit S100 Eudragit RL, Propylene foam, Eudragit RS, ethyl cellulose, poly methyl methacrylate, Methocel K4M, Polyethylene oxide, % Cyclodextrin, HPMC 4000, HPMC 100, CMC, Polyethylene glycol, polycarbonate, PVA, Polycarbonate, Sodium alginate, HPC-L, HPC, Eudragit S, HPMC, Metolose S.M. 100, PVP, HPCH, HPC-M, HPMC K15, Polyox, HPMC K4, Acrylic polymer E4 M and Carbopol. ii) Inert fatty materials (5 - 75%): Edible, inert fatty materials having a specific gravity of less than one can be used to decrease the hydrophilic property of formulation and hence increase buoyancy. E.g. Beeswax, fatty acids, long chain fatty alcohols, Gelucires® 39/01 and 43/01. iii) Effervescent agents: Sodium bicarbonate, citric acid, tartaric acid, Di-SGC (Di- Sodium Glycine Carbonate, CG (Citroglycine). iv) Release rate accelerants (5 - 60%): lactose, mannitol. v) Release rate retardants (5 - 60%): Dicalcium phosphate, talc, magnesium stearate vi) Buoyancy increasing agents (upto 80%): Ethyl cellulose vii) Low density material: Polypropylene foam powder (Accurel MP 1000®) 11-15 .
  • 25. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 12 Approaches to GRDDS To formulate a successful stomach specific or gastroretentive drug several techniques are currently used such as Hydrodynamically balanced systems 17 Bioadhesive or Mucoadhesive 18 Raft systems incorporating alginate gels 19-22 Modified shape systems 19 High density systems 20-23 Swelling system 24-26 Magnetic systems 26-27 Floating drug delivery 28-29 Colloidal gel barrier system Microporous compartment system Alginate beads 30 Hollow microspheres / Microballons 31 Effervescent systems Drug release from effervescent (gas generating) systems This system can also be further described as: (i) Volatile liquid containing systems 37 (ii) Gas-generating Systems Among these the following have been studied extensively. Floating drug delivery systems Non effervescent systems Gas generating systems High density systems Bioadhesive system
  • 26. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 13 Floating system Drug delivery system that float immediately upon contact with gastric fluids present promising approach for increasing the bioavailability of drugs with absorption window in the upper small intestine. However, immediate floating can only be achieved if the density of the device is low at the very beginning. Devices with an initially high density (which decreases with time) first settle down in the stomach and thus undergo the risk of premature emptying. Inherent low density can, for example, be provided by the entrapment of air (e.g. hollow chambers) or by the (additional) incorporation of low density materials e.g. fatty substances or oils or foampowder. Non effervescent systems This type of system, after swallowing, swells unrestrained via imbibition of gastric fluid to an extent that it prevents their exit from the stomach .These systems may be referred to as the ‘plug-type systems’ since they have a tendency to remain lodged near the pyloric sphincter. One of the formulation methods of such dosage forms involves the mixing of drug with a gel, which swells in contact with gastric fluid after oral administration and maintains a relative integrity of shape and a bulk density of less than one within the outer gelatinous barrier. The air trapped by the swollen polymer confers buoyancy to these dosage forms. Gas generating systems These buoyant systems utilise matrices prepared with swellable polymers like methocel, polysaccharides like chitosan and effervescent components like sodium bicarbonate, citric acid and tartaric acid or chambers containing a liquid that gasifies at body temperature. The optimal stoichiometric ratio of citric acid and sodium bicarbonate for gas generation is reported to be 0.76:1. The common approach for preparing these systems involves resin beads loaded with bicarbonate and coated with ethylcellulose. The coating, which is insoluble but permeable, allows permeation of water. Thus, carbon dioxide is released, causing the beads to float in the stomach.
  • 27. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 14 High density systems Sedimentation has been employed as a retention mechanism for pellets that are small enough to be retained in the rugae or folds of the stomach body near the pyloric region, which is the part of the organ with the lowest position in an upright posture. Dense pellets (approximately 3g/cm3 ) trapped in rugae also tend to withstand the peristaltic movements of the stomach wall. With pellets, the GI transit time can be extended from an average of 5.8 to 25 hours, depending more on density than on diameter of the pellets, although many conflicting reports stating otherwise also found in the literature. Commonly used excipients are barium sulphate, zinc oxide, titanium dioxide and iron powder, etc. These materials increase density by up to 1.5–2.4g/cm3 . However, no successful system has reached the market. Figure 1.1. (a) microspheres floats on stomach contents. (b & c) microspheres adhere to stomach wall Bioadhesive drug delivery systems Bioadhesive drug delivery systems (BDDS) are used to localize a delivery device within the lumen to enhance the drug absorption in a site-specific manner. This approach involves the use of bioadhesive polymers which can adhere to the epithelial surface in the stomach. A microbalance-based system is reported for measuring the forces of interaction between the GI mucosa and the individual polymers. The Cahn Dynamic Contact Angle Analyzer has been used to study the adherence. Gastric mucoadhesion does not tend to be strong enough to impart to dosage forms the ability to resist the strong propulsion forces of the stomach wall. The continuous production of mucous by the gastric mucosa to replace the mucous that is lost through
  • 28. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 15 peristaltic contractions and the dilution of the stomach content also seem to limit the potential of mucoadhesion as a gastroretentive force. Some of the most promising excipients that have been used commonly in these systems include polycarbophil, carbopol, lectins, chitosan, CMC and gliadin, etc. Some investigators have tried out a synergistic approach between floating and bioadhesion systems. The major challenge for bioadhesive drug delivery systems is the high turnover rate of the gastric mucus and the resulting limited retention times. Furthermore, it is difficult to target specifically the gastric mucus with bioadhesive polymers. Most of the latter (e.g. polycarbophil, Carbopol and chitosan) will stick to various other surfaces that they come into contact with. In addition; the possibility of oesophageal binding might present a challenge regarding safety aspects. Mechanism of Bioadhesion The mechanism of adhesion of polymers to mucosal tissues has involved both chemical and physical binding. Both weak and strong interactions (i.e. Vander walls interaction, hydrogen bonding and ionic bonding) can develop between certain types of chemical groups on the polymer (i.e. –OH, -COOH) and the glycoprotein network of the mucus layer or the glycoprotein chains attached to the epithelial cells. Mucous membranes Mucous membranes (mucosae) are the moist surfaces lining the walls of various body cavities such as the gastrointestinal and respiratory tracts. They consist of a connective tissue layer (the lamina propria) above which is an epithelial layer, the surface of which is made moist usually by the presence of a mucus layer. The epithelia may be either single layered (e.g. the stomach, small and large intestine and bronchi) or multilayered/stratified (e.g. in the oesophagus, vagina and cornea). The former contain goblet cells which secrete mucus directly onto the epithelial surfaces, the latter contain, or are adjacent to tissues containing, specialised glands such as salivary glands that secrete mucus onto the epithelial surface. Mucus is present as either a gel layer adherent to the mucosal surface or as a luminal soluble or suspended form. The major components of all mucus gels are mucin glycoproteins, lipids, inorganic salts and water, the latter accounting for more than 95% of its weight, making it a highly hydrated system. The
  • 29. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 16 mucin glycoproteins are the most important structure-forming component of the mucus gel resulting in its characteristic gel-like, cohesive and adhesive properties. The thickness of this mucus layer varies on different mucosal surfaces, from 50 to 450 µm in the stomach to less than 1 µm in the oral cavity. The major functions of mucus are that of protection and lubrication (they could be said to act as anti-adherents). Figure 1.2. Three regions within a mucoadhesive joint Mucoadhesives The most widely investigated group of mucoadhesives is hydrophilic macromolecules containing numerous hydrogen bond forming groups, the so-called first generation mucoadhesives. Their initial use as mucoadhesives was in denture fixative powders or pastes. The presence of hydroxyl, carboxyl or amine groups on the molecules favours adhesion. They are called the wet adhesives in that they are activated by moistening and will adhere non-specifically to many surfaces. Once activated, they will show stronger adhesion to dry inert surfaces than those covered with mucus. Unless water uptake is restricted, they may over hydrate to form slippery mucilage. Like typical hydrocolloid glues, if the formed adhesive joint is allowed to dry then, they can form very strong adhesive bonds. Typical examples are carbomers, chitosan, sodium alginate and the cellulose derivatives. Mucoadhesive / Mucosa Interaction For adhesion to occur, molecules must bond across the interface. These bonds can arise in the following way Ionic bonds—where two oppositely charged ions attract each other via electrostatic interactions to form a strong bond (e.g. in a salt crystal). Covalent bonds—where electrons are shared, in pairs, between the bonded atoms in order to fill the orbital in both. These are also strong bonds.
  • 30. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 17 Hydrogen bonds—here a hydrogen atom, when covalently bonded to electronegative atoms such as oxygen, fluorine or nitrogen, carries a slight positively charge and is therefore is attracted to other electronegative atoms. The hydrogen can therefore be thought of as being shared, and the bond formed is generally weaker than ionic or covalent bonds. Vander-Waals bonds—these are some of the weakest forms of interaction that arise from dipole–dipole and dipole-induced dipole attractions in polar molecules, and dispersion forces with non-polar substances. Hydrophobic bonds—more accurately described as the hydrophobic effect, these are indirect bonds (such groups only appear to be attracted to each other) that occur when non-polar groups are present in an aqueous solution. Water molecules adjacent to non-polar groups form hydrogen bonded structures, which lowers the system entropy. Table 1.2: Different theories explaining the mechanism of bioadhesion S. No. Theory Mechanism of bioadhesion 1 Electronic theory Attractive electrostatic forces between glycoprotein mucin network and the bioadhesive material. 2 Adsorption theory Surfaces forces resulting in chemical bonding. 3 Wetting theory Ability of bioadhesive polymers to spread and develop intimate contact with the mucus membranes. 4 Diffusion theory Physical entanglement of mucin strands and the flexible polymer chains. Interpretation of mucin strands into the porous structure of the polymer substrate. 5 Fracture theory Analyses the maximum tensile stress developed during detachment of the BDDS from the mucosal surfaces.
  • 31. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 18 Factors Affecting Mucoadhesion Polymer related factors Molecular weight Low molecular weight polymer favors the inter penetration of polymer molecules, high molecular weight polymer favours physical entanglement. Flexibility of polymer chains Required for interpenetration and entanglement. Highly cross-linked polymers: mobility of individual polymer chains decreases which leads to decreased bioadhesive strength. Concentration of polymer Solid BDDS: more is the polymer concentration higher is the bioadhesive strength, Liquid BDDS: optimum concentration is required for best bioadhesion, High concentration may result in coiling of polymer molecules and hence reduced flexibility of the polymeric chains. Methods to Study Bioadhesion In-vitro methods for testing bioadhesion for the design and development of bioadhesive controlled release systems and to ensure compatibility, physical and mechanical stability of these systems (Peppas et al., 1985). These methods include: i. Ex vivo methods : Methods based on measurement of tensile strength or shear strength. Adhesion weight method Fluorescent probe method Mechanical spectroscopic method Falling liquid film method Flow channel method Colloidal gold staining method
  • 32. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 19 Viscometric method Thumb test Adhesion number Electrical Conduction ii. In vivo methods Use of radioisotopes Use of gamma scintiography Advantages and Limitations of Mucoadhesive Drug Delivery System Mucoadhesive dosage forms have following advantages because of their property to immobilize drug-carrying particles at the mucosal surface. A prolonged residence time at the site of action or absorption thus permit onceor twice a day dosing. Increase in drug concentration gradient due to the intimate contact of the particles with the mucosal surface. Adirect contact with mucus membrane, which is the step earlier to particle absorption. Bioadhesive system can prevent the first pass metabolism to certain protein drugs by the liver through the introduction of the drugs via route bypassing the digestive tract. Drugs that are absorbed through mucosal lining of tissues can enter directly into the blood stream and prevented from enzymatic degradation in the GIT. These dosage forms facilitate intimate contact of the formulation with the underlying absorption surface. This allows modification of tissue permeability for absorption of macromolecules such as peptides and proteins, inclusion of penetration enhancers such as Sodium glycolate, Lysophosphatidylcholine, Sodium taurocholate and Protease inhibitors in the mucoadhesive dosage forms resulted in the better absorption of peptides/ proteins.
  • 33. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 20 Limitations Gastric mucoadhesion does not tend to be strong enough to impart to dosage forms the ability to resist the strong propulsion forces of the stomachwall. The continuous production of mucous by the gastric mucosa to replace the mucous that is lost through peristaltic contractions and the dilution of the stomach content also seems to limit the potential of mucoadhesion as a gastro-retentiveforce. Mucoadhesive Polymers There are two broad classes of mucoadhesive polymers: water-soluble and/or water insoluble polymers, having swellable networks, joined by cross-linking agents. In large classes of hydrophillic polymers those containing carboxylic group exhibit the best bioadhesive properties, Polyvinyl pyrrolidone, Methylcellulose, Sodiumcarboxy methylcellulose, Hydroxypropyl cellulose and other cellulose derivatives. Hydrogel are the class of polymeric biomaterial that exhibit the basic characteristics of an hydrogel to swell by absorbing water interacting by means of adhesion with the mucus that covers epithelia (Junginger et al., 1990) i.e. Anionic group - Polyacrylates and their cross-linked modifications Cationic group - Chitosan (Hango, 1998) and its derivatives Neutral group - Eudragit-NE30D etc. The polymer should possess optimal polarity to make sure that it is sufficiently wetted by the mucus and optimal fluidity that permits the mutual adsorption. An ideal mucoadhesive polymer should have following characteristics: The polymer and its degradation products should be nontoxic, nonabsorbable It should be non-irritant to the mucous membrane Itshouldforma strongnoncovalent bondwiththe mucinepithelialcellsurfaces It shouldadherequicklytomoisttissueandshouldpossesssomesitespecificity The cost of the polymer should not be high
  • 34. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 21 Thepolymermustnotdecomposeonstorageorduringshelflifeofthedosageform It should allow easy incorporation of the drug and offers no hindrance to its release. Bioadhesive Strength of Some Polymers Table 1.3: Bioadhesive strength of some polymers S. No. Polymer name Bioadhesive property 1 Polycarbophil +++ 2 Tragacanth +++ 3 Poly (acrylic acid/ divinyl benzene) +++ 4 Sodium alginate +++ 5 Carbopol 934 +++ 6 Carboxymethyl cellulose CMC (low viscosity) ++ CMC (high viscosity) + CMC (medium viscosity) + 7 Hydroxyethyl cellulose +++ 8 Gum karaya ++ 9 Guar gum ++ 10 Chitosan ++ 11 Pectin + 12 Gelatin + 13 Thermally modified starch + 14 Hydroxyethyl methacrylate + 15 Amberlite–200 resin +
  • 35. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 22 16 Hydroxypropyl cellulose + 17 Polyethylene glycol + 18 Polyvinyl pyrrolidone + 19 Chitosan (Dr. Knapczyk) + 20 Daichitosan H ++ +++ Excellent (8.0 to 12.0 mN/cm2 ); ++ fair (1.5 to 8.0 mN/cm2 );+ poor (<1.5mN/cm2 ). Application of Bioadhesive Systems Table 1.4: Application of Bioadhesive Systems Drug Route of administration Polymer used Comment Acyclovir Ocular Chitosan Slow release rate, increased AUC Gentamycin Nasal DSM+LPC Slow release rate Insulin Nasal DSM+LPC Efficient delivery into systemic circulation via nasal route Furosemide GI AD-MMS[PGEFs] Increased bioavailability Amoxicillin GI AD-MMS[PGEFs] Greater H-pylori activity Vancomycin Colonic PGEF coated with Eudragit S 100 Well absorb even with enhancer Insulin Colonic PGEF coated with Eudragit S 100 Absorbtion only in presence of enhancer Methyl perdisolone Ocular Hyaluronic acid Slow release rate
  • 36. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 23 1.4.10 Patents Related to Bioadhesive Systems Table 1.5: Patents related to bioadhesive system 32 Patent number Assignee/Inventor Year Title US 6368586 Brown University Research foundation April 2002 Methods and compositions for enhancing bioadhesive properties of polymers WO 0203955 Roversi Francesco, Cilurzo Francesco Jan. 2002 Fast release bioadhesive microspheres for the sublingual administration of Proximate US 627415 Immunex Corporation Aug 2001 Prolonged release of GM-CSF US 6207197 West Pharmaceutical Services Drug Delivery and Clinical Research Centre March 2001 Gastroretentive controlled release microspheres Mucoadhesive Microballons: Microballons are small spherical particles, with diameters in the micrometer range (typically 1µm to 1000µm or 1mm). Microballons are sometimes referred to as microparticles. Microballons are defined as “the monolithic spheres or therapeutic agents distributed throughout the matrix either as a molecular dispersion of particles”. Microballons are small spherical particles with diameter in the micrometer range and sometimes referred as microparticles. When adhesion is restricted to the mucous layer lining of the mucosal surface it is termed as mucoadhesion. Mucoadhesion offers prolonged residence time at the site of absorption, localization of the drug delivery system at a given target site, increase in drug
  • 37. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 24 concentration gradient due to the intestine contact of the particle with the mucosal surface. Development of adhesive bond between polymer and biological membrane or its coating can be achieved by two ways: initial contact between the surfaces or formation of secondary bonds due to non covalent interaction. Mucoadhesives must interact with mucin layer during the process of attachment. Mucins are synthesized by globet cells and special exocrine glands with mucin cells acnini. There are atleast two main targets which could be used for anchoring of delivery system through mucoadhesive in the GIT, the mucosal tissue and mucosal gel layer. The mucos layer is the first surface encountered by particulate system and its complex structure offers many opportunities for the development of adhesive interaction with small polymeric particles either through non specific or specific interaction between complimentary structures. Due to all above advantages Microsphere delivery is an better choice for drug delivery in colon 33 .
  • 38. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 25 2: REVIEW OF LITERATURE : DRUG PROFILE: NIZATIDINE Structure: Figure 2.1: Structure of drug Nizatidine Chemical name: N-[2-[[2-(dimethylaminomethyl)-1,3-thiazol-4- yl]methylsulfanyl]ethyl]-N'-methyl-2-nitroethene-1,1-diamine Molecular formula: Molecular weight : C12H21N5O2S2 331.5 Physical characteristics: A white to off–white crystalline solid. Melting range: Storage: 130-132ºC Store in air tight, temper proof container, protected from light. Mechanism of action: Nizatidine is a competitive, reversible inhibitor of histamine at the histamine H2 receptors, particularly those in the gastric parietal cells. By inhibiting the action of histamine on stomach cells, nizatidine reduces stomach acid production. Nizatidine competes with histamine for binding at the H2-receptors on the gastric basolateral membrane of parietal cells. Competitive inhibitionresults basal and nocturnal gastric acid secretions. The drug also decreases in reduction of the gastric acid response to stimuli such as food, caffeine, insulin, betazole, or pentagastrin. Plasma half life: 1.5 hrs
  • 39. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 26 Disposition in the body: Nizatidine is readily and almost completely absorbed after oral administration and peak plasma concentrations are reached within 1 to 3 h. Absorption is increased by the presence of food and decreased by 10% in the presence of antacids such as aluminium hydroxide gel and magnesium silicate. It is partially metabolised by the liver, but does not inhibit the hepatic mixed function oxidase system. Three metabolites have been identified, nizatidine N-2–oxide, nizatidine S-oxide and N-2–mono des methyl nizatidine (60% activity of nizatidine). Nizatidine is widely distributed throughout the body and has been detected in breast milk (0.1% of the administered dose). 90% of an administered dose is excreted in urine, partly by active tubular secretion, with 60% as the unchanged drug. Less than 6% of a dose is excreted in faeces. Protein binding: 35%. Dose: Orally the usual daily dose is 150 to 600 mg. The total intravenous daily dose should not exceed 480 mg. Pharmacokinetics parameter: Volume of distribution: Approx. 1.2 to 1.6 L/kg (single 150 mg dose), 1.1 to 1.9 L/kg (multiple doses). Clearance: Serum 37.5 to 41.4 L/h (single 150 mg dose), 39.1 to 44.6 L/h (multiple doses). Plasma 40 to 51 L/h. Side effects: It include an allergic reaction (difficulty in breathing, closing of throat) bleeding gums, irregular heartbeat, fever or yellowing of the skin or eyes, headache, dizziness or diarrhea may occur 34 . : EXCIPIENTS REVIEW Cellulose, microcrystalline Nonproprietary Names: BP: Microcrystalline cellulose, JP: Microcrystalline cellulose, PhEur: Cellulosum microcristallinum, USPNF: Microcrystalline cellulose Synonyms: Avicel PH, Celex, cellulose gel, Celphere, Ceolus KG, crystalline cellulose, E460, Emcocel, Ethispheres, Fibrocel, Pharmacel, Tabulose, Vivapur. Chemical Name: Cellulose Empirical Formula and Molecular Weight: (C6H10O5)n ≈36 000 where n ≈ 220.
  • 40. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 27 Structural Formula: Figure 2.2: Chemical structure of microcrystalline cellulose Functional Category: Adsorbent, suspending agent, tablet and capsule diluents, tablet disintegrant. Applications in Pharmaceutical Formulation or Technology: Microcrystalline cellulose is widely used in pharmaceuticals, primarily as a binder/diluent in oral tablet and capsule formulations where it is used in both wet-granulation and direct compression processes Description: Microcrystalline cellulose is purified, partially depolymerized cellulose that occurs as a white, odorless, tasteless, crystalline powder composed of porous particles. Typical Properties: Angle of repose: 49° for Ceolus KG; 34.4° for Emcocel 90M.9 Density (true): 1.512–1.668 gm / cm3 Flowability: 1.41 g / s for Emcocel 90M.9 Particle size distribution: Typical mean particle size is 20–200 μm. Different grades may have a different nominal mean particle size; Avicel PH- 102a 100-60 Solubility: Slightly soluble in 5 % w/v sodium hydroxide solution; practically insoluble in water, dilute acids, and most organic solvents. Calcium phosphate, dibasic dihydrate Nonproprietary Names: BP: Calcium hydrogen phosphate, JP: Dibasic calcium phosphate, PhEur: Calcii hydrogenophosphas dihydricus, USP: Dibasic calcium phosphate
  • 41. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 28 Synonyms: Calcium hydrogen orthophosphate dehydrate, calcium monohydrogen phosphate dehydrate, Di-Cafos; dicalcium orthophosphate, DI-TAB, E341, Emcompress, phosphoric acid calcium salt (1:1) dihydrate; secondary calcium phosphate. Chemical Name: Dibasic calcium phosphate dehydrate Empirical Formula and Molecular Weight: CaHPO4·2H2O; 172.09 Structural Formula: CaHPO4·2H2O Functional Category: Tablet and capsule diluent. Applications in Pharmaceutical Formulation or Technology: Dibasic calcium phosphate dihydrate is widely used in tablet formulations both as an excipient and as a source of calcium and phosphorus in nutritional supplements. Description: Dibasic calcium phosphate dihydrate is a white, odorless, tasteless powder or crystalline solid. It occurs as monoclinic crystals. Typical Properties: Angle of repose: 28.3° for Emcompress. Density (bulk): 0.915 gm / cm3 Density (tapped): 1.17 gm / cm3 Density (true): 2.389 gm / cm3 Particle size distribution: DI-TAB: average particle diameter 180 μm, Fine powder: average particle diameter 9 μm Solubility: Practically insoluble in ethanol, ether, and water; soluble in dilute acids. Lactose, Anhydrous Nonproprietary Names: BP: Anhydrous lactose, JP: Anhydrous lactose, PhEur: Lactosum anhydricum, USPNF: Anhydrous lactose Synonyms: Anhydrous Lactose NF 60M, Anhydrous Lactose NF Direct Tableting, Lactopress Anhydrous, lactosum, lattioso, milk sugar, Pharmatose DCL 21, Pharmatose DCL 22, saccharum lactis, Super-Tab Anhydrous.
  • 42. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 29 Chemical Name: O-β-D-galactopyranosyl-(1→4)-β-D-glucopyranose Empirical Formula and Molecular Weight: C12H22O11 and 342.30 Structural Formula: Anhydrous lactose as O-β-D-galactopyranosyl-(1→4)-β- Dglucopyranose; or a mixture of O-β-D-galactopyranosyl-(1→4)-α-D-glucopyranose and O-β- D-galactopyranosyl-(1→4)-β-D-glucopyranose (Figure 2.3). Figure 2.3: Chemical structure of anhydrous lactose Functional Category: Binding agent, directly compressible tableting excipients, lyophilization aid, tablet and capsule filler. Applications in Pharmaceutical Formulation or Technology: Anhydrous lactose is widely used in direct compression tableting applications and as a tablet and capsule filler and binder. Anhydrous lactose can be used with moisture-sensitive drugs due to its low moisture content. Description: Lactose occurs as white to off-white crystalline particles or powder. Several different brands of anhydrous lactose are commercially available which contain anhydrous β-lactose and anhydrous α-lactose. Typical Properties: Angle of repose: 39° for Pharmatose DCL 21 and 38° for Super-Tab Anhydrous.
  • 43. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 30 Density (true): 1.589 gm / cm3 for anhydrous β-lactose; 1.567 gm / cm3 for Super-Tab Anhydrous. Density (bulk): 0.68 gm / cm3 for Pharmatose DCL 21; 0.67 gm / cm3 for Pharmatose DCL 22; 0.65 gm / cm3 for Super-TabAnhydrous. Density (tapped): 0.88 gm / cm3 for Pharmatose DCL 21; 0.79 gm / cm3 for Pharmatose DCL 22; 0.87 gm / cm3 for Super-TabAnhydrous. Solubility: soluble in water; sparingly soluble in ethanol (95 %) and ether. Sodium starch glycolate Nonproprietary Names: BP: Sodium starch glycollate, PhEur: Carboxymethylamylum natricum, USPNF: Sodium starch glycolate Synonyms: Carboxymethyl starch, sodium salt, Explosol, Explotab, Glycolys, Primojel, starch carboxymethyl ether, sodium salt, Tablo, Vivastar P. Chemical Name: Sodium carboxymethyl starch Empirical Formula and Molecular Weight: Sodium starch glycolate is the sodium salt of a carboxymethyl ether of starch, containing 2.8–4.2 % sodium. The molecular weight is typically 5 × 105–1 × 106. Structural Formula: Figure 2.4: Chemical structure of sodium starch glycolate
  • 44. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 31 Functional Category: Tablet and capsule disintegrant. Applications in Pharmaceutical Formulation or Technology: Sodium starch glycolate is widely used in oral pharmaceuticals as a disintegrant in capsule and tablet formulations. It is commonly used in tablets prepared by either directcompression or wet- granulation processes. The usual concentration employed in a formulation is between 2 % and 8 %, with the optimum concentration about 4 %, although in many cases 2 % is sufficient. Disintegration occurs by rapid uptake of water followed by rapid and enormous swelling. Although the effectiveness of many disintegrants is affected by the presence of hydrophobic excipients such as lubricants, the disintegrant efficiency of sodium starch glycolate is unimpaired. Increasing the tablet compression pressure also appears to have no effect on disintegration time.10 – 14 sodium starch glycolate has also been investigated for use as a suspending vehicle. Description: Sodium starch glycolate is a white to off-white, odorless, tasteless, free- flowing powder. It consists of oval or spherical granules, 30 – 100 μm in diameter, with some less-spherical granules ranging from 10 – 35 μm in diameter. Typical Properties: Particle size distribution: 100 % of particles less than 106 μm in size. Average particle size is 35 – 55 μm for Explotab. Solubility: Sparingly soluble in ethanol (95 %), practically insoluble in water. At a concentration of 2 % w/v sodium starch glycolate disperses in cold water and settles in the form of a highly hydrated layer. Swelling capacity: In water, sodium starch glycolate swells to up to 300 times its volume. Viscosity (dynamic): ≤ 200 mPa s (200 cP) for a 4 % w/v aqueous dispersion. Viscosity is 4.26 mPa s for a 2% w/v aqueous dispersion Starch Nonproprietary Names: BP: Potato starch, JP: Potato starch, PhEur: Solani amylum (potato starch), USPNF: Potato starch
  • 45. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 32 Synonyms: Amido, amidon, amilo, amylum, Aytex P, C*PharmGel, Fluftex W, Instant Pure-Cote, Melojel, Meritena, Paygel 55, Perfectamyl D6PH, Pure-Bind, Pure-Cote, Pure-Dent, Pure- Gel, Pure-Set, Purity 21, Purity 826, Tablet White. Chemical Name: Starch Empirical Formula and Molecular Weight: (C6H10O5) n 50 000–160 000 where n = 300–1000. Starch consists of amylose and amylopectin, two polysaccharides based on α- glucose. Functional Category: Glidant, diluents, disintegrant, tablet binder. Structural Formula: Figure 2.5: Chemical structure of starch Applications in Pharmaceutical Formulation or Technology: Starch is used as an excipient primarily in oral solid-dosage formulations where it is utilized as a binder, diluent, and disintegrant. As a diluent, starch is used for the preparation of standardized triturates of colorants or potent drugs to facilitate subsequent mixing or blending processes in manufacturing operations. Starch is also used in dry-filled capsule formulations for volume adjustment of the fill matrix.1 In tablet formulations, freshly prepared starch paste is used at a concentration of 5–25 % w/w in tablet granulations as a binder.
  • 46. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 33 Description: Starch occurs as an odourless and tasteless, fine, white-colored powder comprising very small spherical or ovoid granules whose size and shape are characteristic for each botanical variety. Typical Properties: Density (bulk): 0.462 gm / cm3 for corn starch. Density (tapped): 0.658 gm / cm3 for corn starch. Density (true): 1.478 gm / cm3 for corn starch. Flowability: Corn starch is cohesive and has poor flow characteristics. Gelatinization temperature: 72 °C for potato starch Particle size distribution: Potato starch: 10–100 μm; Solubility: Practically insoluble in cold ethanol (95 %) and in cold water. Starch swells instantaneously in water by about 5–10 % at 37 °C.2, 22 polyvalent cations produce more swelling than monovalent ions, but pH has little effect. Swelling temperature: 64°C for potato starch Talc Nonproprietary Names: BP: Purified talc, JP: Talc, PhEur: Talcum, USP: Talc Synonyms: Altalc, E553b, hydrous magnesium calcium silicate, hydrous magnesium silicate, Luzenac Pharma; magnesium hydrogen metasilicate, Magsil Osmanthus, Magsil Star, powdered talc, purified French chalk, Purtalc, soapstone, steatite, Superiore. Chemical Name: Talc Empirical Formula and Molecular Weight: Talc is a purified, hydrated, magnesium silicate, approximating to the formula Mg6(Si2O5)4(OH)4. It may contain small, variable amounts of aluminum silicate and iron. Functional Category: Anticaking agent, glidant, diluents, lubricant. Applications in Pharmaceutical Formulation or Technology: Talc was once widely used in oral solid dosage formulations as a lubricant and diluents.
  • 47. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 34 Typical Properties: Hardness: 1.0 – 1.5 Moisture content: Talc absorbs insignificant amounts of water at 25 °C and relative humidities up to about 90 %. Solubility: It is practically insoluble in dilute acids and alkalis, organic solvents, and water Magnesium stearate Nonproprietary Names: BP: Magnesium stearate, JP: Magnesium stearate, PhEur: Magnesii stearas, USPNF: Magnesium stearate Synonyms: Magnesium octadecanoate, octadecanoic acid, magnesium salt, stearic acid, magnesium salt. Chemical Name: Octadecanoic acid magnesium salt Empirical Formula and Molecular Weight: C36H70MgO4 and 591.34 Structural Formula: [CH3 (CH2)16COO]2 Mg Functional Category: Tablet and capsule lubricant. Applications in Pharmaceutical Formulation or Technology: Magnesium stearate is widely used in cosmetics, foods, and pharmaceutical formulations. It is primarily used as a lubricant in capsule and tablet manufacture at concentrations between 0.25 % and 5.0 % w/w. Description: Magnesium stearate is a very fine, light white, precipitated or milled, impalpable powder of low bulk density, having a faint odor of stearic acid and a characteristic taste. The powder is greasy to the touch and readily adheres to the skin. Typical Properties: Density (bulk): 0.159 gm / cm3 Density (tapped): 0.286 gm / cm3 Density (true): 1.092 gm / cm3
  • 48. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 35 Flowability: Poorly flowing, cohesive powder. Solubility: Practically insoluble in ethanol, ethanol (95 %), ether and water; slightly soluble in warm benzene and warm ethanol (95 %). Guar Gum Nonproprietary Names: BP: Guar galactomannan, PhEur: Guar galactomannanum, USPNF: Guar gum Synonyms: E412; Galactosol; guar flour; jaguar gum; Meyprogat; Meyprodor; Meyprofin. Chemical Name: Galactomannan polysaccharide Empirical Formula and Molecular Weight: (C6H12O6) n ≈220 000 Structural Formula: Guar gum consists of linear chains of (1→4)-β-D-mannopyranosyl units with α-Dgalactopyranosyl units attached by (1→6) linkages. The ratio of D- galactose to D-mannose is between 1: 1.4 and 1: 2 (Figure 2.6). Figure 2.6: Chemical structure of guar gum Functional Category: Suspending agent, tablet binder, tablet disintegrant, viscosity- increasing agent. Description: Guar gum is obtained from the ground endosperms of Cyamopsis tetragonolobus (L.) Taub. (Family: Leguminosae). It consists chiefly of a highmolecular- weight hydrocolloidal polysaccharide, composed of galactan and mannan units combined through glycoside linkages, which may be described chemically as a galactomannan.
  • 49. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 36 Typical Properties: Density: 1.492 gm / cm3 Solubility: Practically insoluble in organic solvents. In cold or hot water, guar gum disperses and swells almost immediately to form a highly viscous, thixotropic sol. The optimum rate of hydration occurs at pH 7.5 to 9.0. Finely milled powders swell more rapidly and are more difficult to disperse. Two to four hours in water at room temperature are required to develop maximum viscosity. Viscosity: 4.86 Pa s (4860 cP) for 1 % w/v dispersion at 25 ºC temperature. Stability and Storage Conditions: Guar gum powder should be stored in a well-closed container in a cool, dry place. Aqueous guar gum dispersions have a buffering action and are stable at pH 4.0 to 10.5. Xanthan Gum Nonproprietary Names: BP: Xanthan gum, PhEur: Xanthani gummi, USPNF: Xanthan gum Synonyms: Corn sugar gum; E415; Keltrol; polysaccharide B-1459; Rhodigel; Vanzan NF; Xantural. Chemical Name: Xanthan gum Empirical Formula and Molecular Weight: (C35H49O29)n Approximately 2 × 106 Xanthan gum is a high molecular weight polysaccharide gum. It contains D-glucose and D-mannose as the dominant hexose units, along with D-glucuronic acid, and is prepared as the sodium, potassium, or calcium salt. Functional Category: Stabilizing agent; suspending agent; viscosity-increasing agent. Description: Xanthan gum occurs as a cream- or white-colored, odorless, free-flowing, fine powder. Typical Properties: Acidity / alkalinity: pH = 6.0 – 8.0 for a 1 % w/v aqueous solution.
  • 50. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 37 Particle size distribution: Various grades with different particle sizes are available; for example, 100 % less than 180 μm in size for Keltrol CG; 100 % less than 75 μm in size for Keltrol CGF; 100 % less than 250 μm, 95 % less than 177 μm in size for Rhodigel; 100 % less than 177 μm, 92 % less than 74 μm in size for Rhodigel 200. Solubility: Practically insoluble in ethanol and ether; soluble in cold or warm water. Viscosity: 1200 to 1600 mPa s (1200–1600 cP) for a 1 % w/v aqueous solution at 25 °C. Stability and Storage Conditions: Xanthan gum is a stable material. Aqueous solutions are stable over a wide pH range (pH 3– 12), although they demonstrate maximum stability at pH 4 – 10 and temperatures of 10 – 60°C. Xanthan gum solutions of less than 1 % w/v concentration may be adversely affected by higher than ambienttemperatures. Hypromellose Nonproprietary Names: • BP: Hypromellose, • JP: Hydroxypropylmethylcellulose, • PhEur: Hypromellosum, • USP: Hypromellose Synonyms: Benecel MHPC; E464; hydroxypropyl methylcellulose; HPMC; Methocel; methylcellulose propylene glycol ether; methyl hydroxypropylcellulose; Metolose; Tylopur. Chemical Name: Cellulose hydroxypropyl methyl ether Empirical Formula and Molecular Weight: The PhEur 2005 describes hypromellose as a partly O-methylated and O-(2-hydroxypropylated) cellulose. It is available in several grades that vary in viscosity and extent of substitution. Grades may be distinguished by appending a number indicative of the apparent viscosity, in mPa s, of a 2 % w/w aqueous solution at 20 °C. Hypromellose defined in the USP 28 specifies the substitution type by appending a four-digit number to the nonproprietary name: e.g., hypromellose 1828. The first two digits refer to the approximate percentage content of the methoxy group (OCH3). The second two digits refer to the approximate percentage content of the hydroxypropoxy group (OCH2CH (OH) CH3), calculated on a dried basis. It contains methoxy and hydroxypropoxy groups conforming to the limits for the types of hypromellose. Molecular weight is approximately 10 000 – 1 500 000.
  • 51. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 38 Structural Formula: Figure 2.7: Chemical structure of cellulose hydroxypropyl methyl ether Where R is H, CH3, or CH3CH (OH) CH2 (Figure 2.7) Description: Hypromellose is an odorless and tasteless, white or creamy-white fibrous or granular powder. Typical Properties: Density (bulk): 0.341 gm / cm3 Density (tapped): 0.557 gm / cm3 Density (true): 1.326 gm / cm3 Melting point: Browns at 190 – 200 °C; chars at 225 – 230 °C. Glass transition temperature is 170 – 180 °C. Moisture content: Hypromellose absorbs moisture from the atmosphere; the amount of water absorbed depends upon the initial moisture content and the temperature and relative humidity of the surrounding air. Solubility: Soluble in cold water, forming a viscous colloidal solution; practically insoluble in chloroform, ethanol (95 %), and ether, but soluble in mixtures of ethanol and dichloromethane, mixtures of methanol and dichloromethane, and mixtures of water and alcohol. Certain grades of hypromellose are soluble in aqueous acetone solutions, mixtures of dichloromethane and propan-2-ol, and other organic solvents 6, 15, 35 .
  • 52. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 39 2.3. Literature review Rahamathulla et al., 2019 was develop valsartan floating tablets (VFT) via non- effervescent technique using low density polypropylene foam powder, carbopol, and xanthan gum by direct compression. Before compression, the particulate powdered mixture was evaluated for pre-compression parameters. The prepared valsartan tablets were evaluated for post-compression parameters, swelling index, floating lag time, in vitro buoyancy studies, and in vitro and in vivo X-ray imaging studies in albino rabbits. The result of all formulations for pre- and post-compression parameters were within the limits of USP. FTIR and DSC studies revealed no interaction between the drug and polymers used. The prepared floating tablets had good swelling and floating capabilities for more than 12 h with zero floating lag time. The release of valsartan from optimized formulation NF-2 showed sustained release up to 12 h; which was found to be non- Fickian release. Moreover, the X-ray imaging of optimized formulation (NF-2) revealed that tablet was constantly floating in the stomach region of the rabbit, thereby indicating improved gastric retention time for more than 12 h. Consequently, all the findings and outcomes have showed that developed valsartan matrix tablets could be effectively used for floating drug delivery system 36 . Jain 2018, developed a simple Gastroretentive Mucoadhesive tablets of Riboflavin. Mucoadhesion is one of the approach to prolong gastric retention. In these systems, Mucoadhesion is achieved by using Mucoadhesive polymers which adhere to the epithelial surface of gastrointestinal tract. Narrow absorption window mainly from upper part of GIT and short half -life makes riboflavin an ideal candidate for its formulation as a Mucoadhesive tablets. Hence we intend to develop a simple Gastroretentive Mucoadhesive tablets of Riboflavin and prepare sustained release once daily 37 . Wagh et al., 2018 Gastroretentive drug delivery system (GRDDS) is one of the novel approaches in the area of oral sustained release dosage forms. Gastro retentive dosage forms has received significant interest in the past few decades as they can improve the limitation of conventional and oral controlled release drug delivery system related to fast gastric emptying time. Drugs that are easily absorbed from
  • 53. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 40 gastrointestinal tract (GIT) and have short half - lives are eliminated quickly from the systemic circulation require frequent dosing to achieve suitable therapeutic activity. To avoid these limitations, the development of oral sustain release GRDDS is an attempt to release the drug slowly into the GIT and maintain an effective drug concentration in the systemic circulation for a long time. After oral administration, such a drug delivery would be retained in the stomach and release the drug in controlled manner so, that the drug could be supplied continuously to its absorption sited in the GIT 38 . More et al., 2018 compiled the various gastroretentive approaches. In order to understand various physiological difficulties to achieve gastric retention, we have summarized important factors controlling gastric retention. Oral route of drug administration is the most preferable route because of its flexibility in formulation, ease of administration, and patient compliance. But this route has certain limitations like limited gastric residence time (GRT) for sustained drug delivery system and for the drugs which are absorb from specific region of gastrointestinal tract (GIT). To overcome these limitations, various approaches have been proposed to increase the gastric retention time of the delivery system in the upper part of gastrointestinal tract. Gastroretentive dosage form (GRDF) prolongs the GRT by targeting site-specific drug release in upper part of GIT. GRDFs enable continuous and the extended duration of drug release and improve bioavailability of drugs that have narrow therapeutic window, by this way they prolong dosing interval and increase compliance of the patient. The evaluation parameters of gastroretentive drug delivery systems are covered. The present review addresses briefly about the current status of various leading gastroretentive drug delivery technologies, developed until now, i.e. high density (sinking), floating, bio- or mucoadhesive, expandable, unfoldable, super porous hydrogel, magnetic systems etc. In addition, important factors controlling gastroretention, advantages and finally, future potential are discussed 39 . El Nabarawi et al., 2017 developed a controlled-release floating matrix tablet and floating raft system of Mebeverine HCl (MbH) and evaluate different excipients for their floating behavior and in vitro controlled-release profiles. Oral pharmacokinetics of the optimum matrix tablet, raft system formula, and marketed Duspatalin® 200 mg retard as
  • 54. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 41 reference were studied in beagle dogs. The optimized tablet formula (FT-10) and raft system formula (FRS-11) were found to float within 34±5 sec and 15±7 sec, respectively, and both remain buoyant over a period of 12 h in simulated gastric fluid. FT-10 (Compritol/HPMC K100M 1:1) showed the slowest drug release among all prepared tablet formulations, releasing about 80.2% of MbH over 8 h. In contrast, FRS-11 (Sodium alginate 3%/HPMC K100M 1%/Precirol 2%) had the greatest retardation, providing sustained release of 82.1% within 8 h. Compared with the marketed MbH product, the Cmax of FT-10 was almost the same, while FRS-11 maximum concentration was higher. The tmax was 3.33, 2.167, and 3.0 h for marketed MbH product, FT-10, and FRS-11, respectively. In addition, the oral bioavailability experiment showed that the relative bioavailability of the MbH was 104.76 and 116.01% after oral administration of FT-10 and FRS-11, respectively, compared to marketed product. These results demonstrated that both controlled-released floating matrix tablet and raft system would be promising gastroretentive delivery systems for prolonging drug action 40 . Kumari et al, 2016 developed sustained release formulation of Verapamil Hydrochloride to maintain constant therapeutic levels of the drug for over 12 hrs. Various grades of HPMC polymers, Guar gum, and Xanthum gum were employed as polymers. Verapamil Hydrochloride dose was fixed as 120 mg. Total weight of the tablet was considered as 400 mg. Polymers were used in the concentration of 60, 120 and 180 mg concentration. All the formulations were passed various physicochemical evaluation parameters and they were found to be within limits. Whereas from the dissolution studies it was evident that the formulation (F6) showed better and desired drug release pattern i.e.,96.10 % in 12 hours containing Guar gum polymer in the concentration of 180mg. It followed zero order release kinetics. For the optimized formulation alcohol effect has been studied by using various concentrations of alcohol in dissolution medium. As the concentration of alcohol increased the sustained action of polymer was decreased. Hence it was concluded that alcohol has significant effect on drug release pattern 41 . Dawang et al., 2015 developed a prolonged release gastro retentive (GT) formulation of Verapamil hydrochloride. Drug was evaluated by UV and DSC. A variety of polymers and effervescent properties were utilized to optimize the desired disposition profile. Tablets were prepared by the direct compression technique and evaluated for physical
  • 55. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 42 properties, swelling, floating, and drug release. The purpose of this research was to formulate and evaluation of a floating tablet of Verapamil hydrochloride by using Gastro retentive technology using 32 factorial design. Floating tablets were prepared by incorporating HPMC K15M, sodium alginate, sodium bicarbonate and citric acid. A 32 Factorial design was applied systemically; the amount of HPMC K15M (X1) and sodium alginate (X2) were selected as independent variables. The time required for 100% drug release and floating lag time (FLT) were selected as dependent variables. It was found that HPMC K4M, sodium alginate and their interaction had significant influence on the % drug release and floating lag time of the delivery system 42 . Bharat et al, 2014, optimized bilayer gastric floating drug delivery system of Verapamil hydrochloride to study the effect of formulation variables especially, combination of polymers on drug release showing prolonged gastric residence time and optimized by using mathematical and statistical techniques. Three ratios of drug to total polymer content and three ratios of HPMC K4M to CP934 were chosen for an optimal design. In the preliminary trials the effect of sodium bicarbonate loading was studied on floating properties and 12% concentration was found to be optimum for floating buoyancy. Hardness of about 5 kg/cm2 was found to be optimum for floating buoyancy and to keep two layers intact. Other physical parameters like weight variation, thickness and friability were within pharmacopoeial limit. Percentage drug content in all BFT formulations was found to be 98.47% - 99.96% which were within pharmacopoeial limit. Drug-polymer ratio and the HPMC K4M-CP934 significantly affect the buoyancy and drug release. It was concluded on the basis of buoyancy and in-vitro release kinetics that an optimized formulation containing a ratio of Drug to polymer 1:1 and polymer to polymer 1:1 gave the best in-vitro release of 99.42% in 12 hrs while for 3:1 and 1:3 in-vitro release was 91.05% and 93.71% respectively. A comparative study was done with the marketed formulation of Verapamil hydrochloride (Calaptin – SR). FTIR studies show no evidence of interaction between drug, polymers and other excipients. The In vitro data were fitted to different kinetic models 43 . Kondeti et al., 2014 Sustained release matrix tablets reduce the frequency of the dosing and increase the effectiveness of the drug by localization at the site of action, providing uniform drug delivery. Sustained release matrix tablets of verapamil hydrochloride were
  • 56. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 43 prepared by using HPMC K15M, Xanthan and Guar Gum polymers with different concentration in various batches of the formulations to facilitate the drug release which cause patient compliance as the dosing frequency is reduced. Verapamil HCl was considered as an ideal drug for designing sustained release formulation because of the high frequency of administration and short biological half-life. The sustained release matrices of Verapamil HCl were prepared by wet granulation technique. Drug release was studied by using Dissolution testing apparatus 2 (paddle method) with 0.1 N HCl for 2 hours followed by pH 6.8 Phosphate buffer for 8 hours. The in vitro drug release of various formulations was performed and compared. The results shows that the tablets formulated with HPMC K15M polymer shows more sustained action when compared to that of Xanthan and Guar gum. The high viscosity of the polymers binds the formulation of matrix thus sustains the release of drug. It was also observed that the increase in concentration of the polymer decreased the drug release from the polymer matrix 44 . Vidyadhara et al., 2014, developed osmotic controlled extended release formulations of verapamil hydrochloride an angiotensin II receptor antagonist with anti‑hypertensive activity. Verapamil hydrochloride matrix tablets were prepared by direct compression process using hydroxypropyl methylcellulose (HPMC) K 15M as polymeric material and mannitol as osmogen at varied concentrations. The matrix tablets were further coated with different compositions of ethylcellulose7cps and polyethylene glycol (PEG)‑4000 by pan coating method. Physical parameters such as weight uniformity, drug content, hardness and friability were evaluated for uncoated tablets and were found to be within I. P limits. The coating thickness and percentage of coating applied for various tablets were also evaluated. The optimized coated tablets were further subjected to micro drilling on the upper face to get 0.5 µm orifice diameter. All the tablets were further subjected to dissolution studies by using USP apparatus II with 6.8 pH phosphate buffer as medium. These studies indicated that all the tablets were found to release the drug up to 12 hours, while coated tablets with orifice found to release the drug at zero order rate, which was in good agreement with peppas n > 0.9 45 . Tola and Li 2014 developed a hydrophobic polymer to overcome the issue of pH- dependent release of weakly basic model drug verapamil hydrochloride from matrix tablets without the use of organic buffers in the matrix formulations. Providing pH-
  • 57. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 44 independent oral release of weakly basic drugs with conventional matrix tablets can be challenging because of the pH-dependent solubility characteristics of the drugs and the changing pH environment along the gastrointestinal tract. Silicone pressure-sensitive adhesive (PSA) polymer was evaluated because of its unique properties of low surface energy, hydrophobicity, low glass transition temperature, high electrical resistance, and barrier to hydrogen ion diffusion. Drug release, hydrogen ion diffusion, tablet contact angle, and internal tablet microenvironment pH with matrix tablets prepared using PSA were compared with those using water-insoluble ethyl cellulose (EC). Silicone PSA films showed higher resistance to hydrogen ion diffusion compared with EC films. Verapamil hydrochloride tablets prepared using silicone PSA showed higher hydrophobicity and lower water uptake than EC tablets. Silicone PSA tablets also showed pH-independent release of verapamil and decreased in dimensions during drug dissolution. By contrast, verapamil hydrochloride tablets prepared using EC did not achieve pH-independent release 46 . Syeda et al., 2013 formulated Gastro retentive controlled release drug delivery system of Verapamil HCl to increase the gastric retention time of the dosage form with controlling the drug release pattern. Different grades of hydroxy propyl methyl cellulose derivatives; Methocel K4M and Methocel K15MCR were incorporated for their gel forming properties. Tablet buoyancy was achieved by incorporating a mixture of gas generating agents; sodium bicarbonate and anhydrous citric acid. In vitro dissolution studies were carried out for eight hours using USP XXII paddle type apparatus using 0.1N HCl as the dissolution medium. All the gastro retentive tablets showed good in vitro buoyancy. Tablets swelled radially and axially during buoyancy study. The release kinetics were explored and explained with zero order, first order, Higuchi and Korsmeyer equations. The release rate, extent and mechanisms were found to be governed by polymer type and content. Formulations were characterized by physical characterization, drug loading content and Fourier Transform Infrared spectroscopy (FT-IR). Good results were obtained in the tests and the FT-IR spectroscopic studies indicating no interaction and the stability of Verapamil HCl in the used excipients. Based up on the results, it was proved that a proper balance between rate retarding polymer and gas forming agents is obligatory for efficient buoyancy and controlled drug release 47 .
  • 58. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 45 Sahi al., 2013, developed the Verapamil hydrochloride sustained-release floating matrix tablets using gas-generation approach to prolong the gastric residence time. Floating tablets were prepared using hydroxypropyl methylcellulose K4M (HPMC) as hydrophilic gel material, sodium bicarbonate as gas-generating agent and Citric Acid as floating assistant agent. A 32 factorial design was used to select the optimized formulation wherein HPMC K4M (X1) and Citric Acid (X2) were taken as independent variables and Floating lag time (FLT), amount of drug release after 24hrs. (Q24) were taken as dependent variables. The release data were evaluated by the model-dependent (curve fitting) method using PCP Disso v2.08 software. Optimisation studies were carried out by using the Design Expert software (version 8.0.1). The floating tablets were evaluated for uniformity of weight, hardness, thickness, swelling index, friability, drug content, FLT, and in vitro release. The in vitro drug release followed Hixson-Crowell model and mechanism of drug release was found to be anomalous or non-fickian type. The optimized formulation was F3 containing HPMC K4M 15%, and Citric acid 3% having minimum FLT and maximum drug release after 24 hrs 48 . Ray and Gupta, 2013 formulated Matrix tablets of Verapamil Hydrochloride as sustained release tablet employing sodium alginate, hydroxyl propyl methyl cellulose polymer, Ethyl cellulose and the sustainedrelease tablets was investigated. Sustained release matrix tablets contain 240 mg Verapamil Hydrochloride were developed using different drug polymer concentration of HPMC, Sodium Alginate and Ethyl Cellulose. Tablets were prepared by wet granulation using HPMCand water solution. Formulation was optimized on the basis of acceptable tablet properties and in-vitro drug release. The resulting formulation produced robust tablets with optimum hardness, thickness consistent weight uniformity and low friability. All tablets but one exhibited gradual and near completion sustained release for Verapamil Hydrochloride, and 99% to 101% released at the end of 24 hrs. The results of dissolution studies indicated that formulation F8, the most successful of the study. An increase in release kinetics of the drug was observed on decreasing polymer concentration 49 . Mathur et al., 2013 developed a sustained release matrix tablet of verapamil hydrochloride (VH) using ethyl cellulose, methyl cellulose, Eudragit RS 100, hydroxypropyl methylcellulose and carboxymethyl cellulose and to evaluate the drug
  • 59. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 46 release kinetics.Verapamil hydrochloride (VH) is a calcium channel blocking agent used in the treatment of hypertension, cardiac arrhythmia and angina pectoris. The short half‑life and high frequency of administration of VH makes it a suitable candidate for designing sustained drug delivery system. In order to achieve the required sustained release profile, the tablets were prepared by a wet granulation method using avicel PH 101 and magnesium stearate as binder and lubricant, respectively. The formulated tablets were characterized for pre‑compression and post‑compression parameters and they were in the acceptable limits. The drug release data obtained after an in vitro dissolution study was fitted to various release kinetic models in order to evaluate the release mechanism and kinetics. The criterion for selecting the best fit model was linearity (coefficient of correlation). Drug release mechanism was found to follow a complex mixture of diffusion, swelling and erosion. Furthermore, to minimize the initial burst drug release, batches were coated by using Eudragit RS100 polymer. After coating the tablets, a better release profile of the formulated tablets was expected and the release rate of the drug was compared with the marketed SR tablet of VH 50 . Vidyadhara et al., 2013 developed controlled release matrix tablets of verapamil hydrochloride to increase therapeutic efficacy, reduced frequency of administration and improved patient compliance. Verapamil hydrochloride was formulated as oral controlled release matrix tablets by using the polyethylene oxides (Polyox WSR 303). They investigate the influence of polymer level and type of fillers namely lactose (soluble filler), swellable filler (starch 1500), microcrystalline cellulose and dibasic calcium phosphate (insoluble fillers) on the release rate and mechanism of release for verapamil hydrochloride from matrix tablets prepared by direct compression process. Higher polymeric content in the matrix decreased the release rate of drug. On the other hand, replacement of lactose with anhydrous dibasic calcium phosphate and microcrystalline cellulose has significantly retarded the release rate of verapamil hydrochloride. Biopharmaceutical evaluation of satisfactory formulations were also carried out on New Zealand rabbits and parameters such as maximum plasma concentration, time to reach peak plasma concentration, area under the plasma concentration time curve(0‑t) and area under first moment curve(0‑t) were determined. In vivo pharmacokinetic study proves
  • 60. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 47 that the verapamil hydrochloride from matrix tablets showed prolonged release and were be able to sustain the therapeutic effect up to 24 h 51 . Patel et al., 2013 formulated novel gastro retentive controlled release drug delivery system of verapamil HCl to increase the gastric retention time of the dosage form and to control drug release. Hydroxypropylmethylcellulose (HPMC), carbopol, and xanthan gum were incorporated for gel forming properties. Buoyancy was achieved by adding an effervescent mixture of sodium bicarbonate and anhydrous citric acid. In vitro drug release studies were performed, and drug release kinetics was evaluated using the linear regression method. The optimized intragastric floating tablet composed of 3:2 of HPMC K4M to xanthan gum exhibited 95.39% drug release in 24 h in vitro, while the buoyancy lag time was 36.2 s, and the intragastric floating tablet remained buoyant for >24 h. Zero- order and non-Fickian release transport was confirmed as the drug release mechanism from the optimized formulation (F7). X-ray studies showed that total buoyancy time was able to delay the gastric emptying of verapamil HCl intragastric floating tablet in mongrel dogs for more than 4 h. Optimized intragastric floating tablet showed no significant change in physical appearance, drug content, total buoyancy time, or in vitro dissolution pattern after storage at 40°C/75% relative humidity for 3 months 52 . Nikam et al., 2011 formulated A novel gastro retentive controlled release drug delivery system of verapamil HCl to increase the gastric retention time of the dosage form and to control drug release. Gastro retentive systems can remain in the gastric region for several hours and hence significantly prolong the gastric residence time of drugs. Verapamil HCL belongs to the class of calcium channel blockers. These medication block the movement of the calcium into the muscle cells of the coronary arteries. They design and evaluate verapamil HCL floating controlled release gastroretentive tablets using different hydrocolloid polymers including Carbopol, Hydroxy propyl methyl cellulose, and Xanthan gum incorporated for gel forming agent by direct compression technology. The tablets were evaluated for the physicochemical parameters such as weight variation, thickness, friability, hardness, drug content, in vitro buoyancy studies, in vitro dissolution studies. The prepared tablets exhibited satisfactory physico-chemical characteristics. Tablet buoyancy was achieved by adding an effervescent mixture of sodium bicarbonate and anhydrous citric acid. The in vitro dissolution studies were carried out in a USP XXII
  • 61. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 48 apparatus II in 0.1N HCl. All the gastroretentive tablets showed good in-vitro buoyancy. The selected tablets (F3) containing Xanthan gum released approximately 94.43% drug in 24 h in vitro dissolution study, while the buoyancy lag time was 25.8 ± 4.2 second and the tablet remained buoyancy for > 24 h. Zero order and non-Fickian release transport was confirmed as the drug release mechanism for the selected tablets (F3) 53 . Gangadharappa et al., 2010 developed a controlled release floating drug delivery system (tablet) of verapamil hydrochloride. Floating tablets of verapamil hydrochloride were engineering to extend gastric residence time and hence to enhance its bioavailability. The floating matrix tablets were prepared by direct compression technique using a combination of hydroxyl propyl methyl cellulose (HPMC) and karaya gum as polymers and sodium bicarbonate as generating agent. The prepared floating tablets were evaluated for weight variation test, hardness, thickness, swelling index, in vitro floating capabilities, floating lag time, compatibility studies, and in vitro drug release. This swellable hydrophilic natural karaya gum was used to control the release of drug. The results showed that the optimized formulation F8 containing 23.3% of karaya gum (70 mg) and 13.3% of HPMC (40 mg) had good floating capability, shorter floating lag time, and sustained drug release for the period of 8 h 54 . Molke et al., 2010, prepared a gastroretentive drug delivery system of Verapamil HCL. Floating tablets of Verapamil HCL were prepared employing solid dispersion technique with compritol-888ATO. By using different release enhancer like Lactose, Microcrystaline cellulose & HPMC K 100 LV. Cetyl alcohol was incorporated as a lower density agent. The floating tablets were evaluated for uniformity of weight, hardness, friability, drug content, in vitro buoyancy and dissolution studies. The effect of release enhancer on drug release profile and floating properties was investigated. The prepared tablets exhibited satisfactory physico-chemical characteristics. The drug release from the tablets was sufficiently sustained and Fickian transport of the drug from tablets was confirmed. The fabricated floating tablet formulations were subjected for stability study at 400C and relative humidity at 75 % for three months. The product was evaluated for, buoyancy, drug content and in vitro dissolution test. After stability study drug release increased slightly but there is no change in physical appearance 55 .
  • 62. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 49 CHAPTER 3: PLAN OF WORK & HYPOTHESIS 3.1: OBJECTIVE: The objective of the present investigation was to develop a formulation gastroretentive nizatidine mucoadhesive microballons for treatment of peptic ulcer mainly at gastric part of GIT, to improve gastric residence time and increase bioavailability. : HYPOTHESIS: A major problem for gastric delivery is the attainment of an optimal concentration at site of action with maximum bioavailability of drugs. The problem is associated with the conventional dosage form for peptic ulcer diseases is frequent dosing due to the low half life. The bioavailability of an instilled compound is generally low from 1.5 – 3.0 h and low solubility, with only a small fraction reaching the target site. In the present study an attempt was made to develop a mucoadhesive Microballons of Nizatidine with variation in polysaccharide polymeric combination with different ratios to increase mucoadhesion at gastric mucosa, which increase the gastric residence time, thus increase the bioavailability. : PLAN OF PRESENT WORK: The study included the following. : Preformulation Study: i. Exhaustive literature survey: The exhaustive literature survey about drugs and various polymers used in present study was done. ii. Analytical methods: The drug samples (Nizatidine) was studied for determination of absorption maxima (λmax) in solvents i.e. gastric pH 0.1 N HCl. The analytical method was validated in terms of preparation of calibration curve, specificity, repeatability precision, intermediate precision and accuracy. iii. Preformulation studies of drug sample: The drug samples were studied for organoleptic properties, microscopic examination by using phase contrast microscope. The physical characteristics of drug samples i.e. density, particle size, flow properties,
  • 63. FORMULATION AND EVALUATION OF GASTRO-RETENTIVE MUCOADHESIVE MICROBALLONS OF NIZATIDINE FOR MANAGEMENT OF PEPTIC ULCER Page 50 compatibility, solubility in various dissolution medias, partition coefficient and drug- excipients compatibility study were characterized. : Formulation and characterization of mucoadhesive Microballons: These prepared systems were evaluated with various parameters such as the physical properties i.e., Flow properties determination, particle size measurement, shape and surface morphology, mucoadhesive properties, swelling study, percentage yield, drug entrapment efficiency, in-vitro drug release studies and Stability Studies etc. : Result and discussion: This section was included all the results of present research work. : Summary and conclusion: This section was included summary and conclusion of present research work.