Uploaded by Ashutosh Shukla

CHOLIC ACID

advertisement
Introduction of dissertation
1. Introduction [1-12]:1.1.
Introduction of dosage form:-
A new era of science and technology has emerged in pharmaceutical
research with focus on developing novel drug delivery systems for oral
administration. Conventional dosage forms like tablets and capsules are
associated with a low bioavailability, frequent application, side effects and
hence patient noncompliance. By developing novel strategies for drug
delivery, researchers embraced an alternative to traditional drug delivery
systems. Out of those, fast dissolving drug delivery systems are very
eminent among pediatrics and geriatrics. Orally disintegrating films are
superior over fast dissolving tablets as the latter are assigned with the risk of
suffocation. Osmotic devices enable a controlled drug delivery independent
upon gastrointestinal conditions using osmosis as driving force. The
advances in nanotechnology and the variety of possible materials and
formulation factors enable a targeted delivery and triggered release.
Vesicular systems can be easily modified as required and provide a
controlled and sustained drug delivery to a specific site. This work provides
an insight of the novel approaches in drug delivery covering the critical
comparison between traditional and novel “advanced-designed” systems.
Tablets are preferred drug delivery system, precisely dosed, easily
manufactured, contribute to good patient compliance, and packaged on a
large scale. Tablets can be prepared by wet granulation, dry granulation, or
direct compression. Dry and wet granulation requires controlling more
processing variables than the direct compression. Over the years, significant
advances have taken place in the manufacturing processes of tablets,
including the evolution from wet granulation to direct compression method.
Direct compression method in tableting is widely used due to fewer
processing steps, simplified validation, elimination of heat and moisture,
economy and improved drug stability. Hence, the current trend in the
pharmaceutical industry is to adopt direct compression technology.
All the pharmaceutical products formulated for systemic delivery via the
oral route of administration irrespective of the mode of delivery (immediate,
sustained or controlled release) and the design of dosage forms (either solid
Ashutosh Shukla
Page 1
Introduction of dissertation
dispersion or liquid), must be developed within the intrinsic characteristics
of GI physiology, pharmacokinetics, Pharmacodynamic and formulation
design is necessary to achieve a systemic approach to the successful
development of an oral pharmaceutical dosage form compensation of
administering a single dose of a drug that is released over an extensive period
of time, instead of numerous doses, have been obvious to the Pharmaceutical
industry for some time. Drug delivery dosage forms can be traced to the
1938.
This work concerned coated pallets for extended release of drug and was
most probably forerunner to the development of the coated particle approach
to sustained drug delivery that introduced in the early 1950s. The novel
system of drug delivery offers a means of improving the therapeutic
effectiveness of included drugs by providing sustained, controlled delivery
or targeting the drug to desired site. The goal of any drug delivery system is
to make available a therapeutic quantity of drug to the proper site in the body
to achieve rapidly and then maintain the desired drug concentration.
Sustained release systems include any drug delivery system that achieves
slow. Release of drug over a comprehensive period of time. If the system is
successful in maintaining constant drug levels in the blood or target tissue,
it is considered as a controlled-release system.
Oral drug delivery is the most preferred and expedient option as the oral
route provides greatest active surface area among all drug delivery system
for administration of various drugs. The attractiveness of these dosage forms
is due to consciousness to toxicity and ineffectiveness of drugs when
administered by oral predictable method in the form of tablets & capsules.
There are several advantages of sustained release drug delivery over
conventional dosage forms like improved patient compliance due to less
frequent drug administration, maximum consumption of the drug, increased
safety margin of potent drug, reduction of fluctuation in steady-state drug
levels, decrease in healthcare costs through enhanced therapy and shorter
treatment period. The principal goal of sustained release forms is the
improvement of drug therapy assessed by the relationship between
advantages and disadvantages of the use of sustained release system.
Ashutosh Shukla
Page 2
Introduction of dissertation
“To liberate the drug at the right time in a right amount of concentration at
a specified target site” is the major objective of a drug delivery system. The
requirements for a successful drug delivery are usually determined by the
physicochemical characteristics of the therapeutic agent and bio-barriers like
the skin and membrane of body organs. Depending on size, chemical
composition, hydrophilicity and ability to bind specific receptor, drug
properties may vary greatly even when used to treat the same symptoms.
Many drugs suffer from an insufficient bioavailability due to insolubility in
physiological fluids and low permeability of different body organs. Hence,
the therapeutic performance is not merely dependent on the activity of the
applied drug, but also, on the bioavailability at the target side according to
evidence.
In the past decades, the treatment of serious diseases or chronic illnesses has
mainly consisted of rapid acting and simple compound that are administered
conventionally in form of as tablets, pills, capsules, cremes, liquids,
aerosols, suppositories, injectables or ointments. These conventional drug
delivery systems represent the classical method for delivery of drugs orally.
These common dosage forms are often accompanied by systemic adverse
effects that are primarily attributable to their unspecified bio-distribution
and missing controllability of the drug release characteristics. Furthermore,
conventional drug delivery systems have been found to have severe
constraints including non-controlled release, higher doses and a frequent
application. Another major challenge in the formulation of drugs is the
improvement of bioavailability.
Sustained release tablet is a kind of preparation which can release medicine
in a relatively long time. Because of the fast distribution, metabolism and
excretion of some drugs, in order to maintain the effective concentration of
drugs, patients need to take drugs continuously in a very short time, that is,
two or three times a day. Some methods of administration are painful,
patients may have poor compliance, or forget to take medicine. Therefore,
the drug is made into a sustained-release preparation. Through some special
techniques and means, the release time of the drug in the body is prolonged,
so that the drug can maintain the effective blood concentration in the body
for a long time. For example, some drugs can last for several days or even
Ashutosh Shukla
Page 3
Introduction of dissertation
longer, which can increase the interval of administration and reduce the pain
of patients.
1.2.
Characteristic of sustained release drug delivery
system :-
Sustained-release medications should not be used alone to adjust or titrate a
patient's uncontrolled pain. Using them for titration unduly prolongs the
process to bring the pain under control. However, once the pain is controlled,
changing to a sustained-release product may enhance the patient's quality of
life and improve compliance and adherence due to the decreased frequency
of dosing.
Sustained release allows delivery of a specific drug at a programmed rate
that leads to drug delivery for a prolonged period of time. This approach of
drug release is especially useful for drugs that are metabolized too fast and
are eliminated from the body shortly after administration. Sustained release
by adjusting the speed of drug release can keep the concentration of the drug
at a constantlevel in the blood or target tissue. A constant dosage of drug
within the therapeutic window is beneficial, for example, to the cancer
treatment. When the drug is dissolved in the aqueous body fluid, it can be
easily transported with the fluid to the target receptors. Some studies have
shown that one method to achieve sustained drug release is by preventing
drug molecules from entering completely the aqueous environment for a
manageable period of time. As depicted, this inhibition can be recognized
by adjusting the degradation speed of a carrier, or by adjusting the diffusion
rate of drug molecules over an insoluble polymer matrix or shell.
A drug delivery system (DDS) is defined as a formulation or a device that
enables the introduction of a therapeutic substance in the body and improves
its efficacy and safety by controlling the rate, time, and place of release of
drugs in the body. This process includes the administration of the therapeutic
product, the release of the active ingredients by the product, and the
subsequent transport of the active ingredients across the biological
membranes to the site of action. The term therapeutic substance also applies
to an agent such as gene therapy that will induce in vivo production of the
active therapeutic agent.
Ashutosh Shukla
Page 4
Introduction of dissertation
Drug delivery system is an interface between the patient and the drug. It may
be a formulation of the drug to administer it for a therapeutic purpose or a
device used to deliver the drug. This distinction between the drug and the
device is important, as it is the criterion for regulatory control of the delivery
system by the drug or medicine control agency. If a device is introduced into
the human body for purposes other than drug administration, such as
therapeutic effect by a physical modality or a drug may be incorporated into
the device for preventing complications resulting from the device, it is
regulated strictly as a device.
There is a wide spectrum between drugs and devices, and the allocation to
one or the other category is decided on a case by case basis. Sustained release
(SR) preparations are not new but several new modifications are being
introduced. They are also referred to as “long acting” or “delayed release”
when compared to “rapid” or “conventional” release preparations. The term
sometimes overlaps with “controlled release,” which implies more
sophisticated control of release and not just confined to the time dimension.
1.3.
Classification of sustained release drug delivery
system:-
1.3.1. Continuous release system:
These systems release the drug continuously for prolonged period of time
along the entire length of GIT with normal transit time.
1.3.2. Diffusion controlled systems

Reservoir devices: A core of drug (reservoir) surrounded by a polymeric
membrane characterizes them. The nature of the membrane determines
the rate of drug release. The characteristics of reservoir diffusion systems
are
1. Zero order drug release is possible.
2. The release rate is dependent on the type of polymer.
3. High molecular weight compounds are difficult to deliver through the
device.

Matrix devices: It consists of drug dispersed homogenously in a matrix.
The characteristics of matrix diffusion systems are
1. Zero order release cannot be obtained.
Ashutosh Shukla
Page 5
Introduction of dissertation
2. Easy to produce than reservoir devices.
3. High molecular weight compounds are delivered through the device.
1.3.3. Dissolution controlled systems

Matrix
dissolution
controlled
systems:
Aqueous
dispersions,
congealing, spherical agglomeration, etc. can be used.

Encapsulation dissolution controlled systems: Particles, seeds,
granules can be coated by techniques such as microencapsulation.

Diffusion and dissolution controlled systems: In a bioerodible matrix,
the drug is homogenously dispersed in a matrix and it is released either by
swelling controlled mechanism or by hydrolysis or by enzymatic attack.

Sustained release matrix tablets: One of the least complicated
approaches to the manufacture of sustained release dosage forms is the
direct compression of drug, release retardant, and additives to form a
tablet in which drug is embedded in a matrix core of retardant.
Alternatively drug retardant blend may be granulated prior to
compression. Such tablets are called as matrix tablets. Three classes of
release retarding materials are used for the formulation of matrix tablets.
They include,
1. Insoluble or ‘skeleton’ matrices
2. Water insoluble, erodable matrices
3. Hydrophilicmatrices.
1.3.4. Delayed–transit and continuous release system:
These systems are designed to prolong release of drug with increased
residence time of GIT. Such dosage forms are designed to remain in the
stomach. Therefore the drug presented in such system should be stable at
gastric pH.
1.3.5. Altered density system
If the residence time of drug in the stomach or intestine is prolonged in some
way, the frequency of dosing can be further reduced.
Altering the density of the drug particle,use of mucoadhesive polymers and
altering the size of the dosage form.
 High Density Pellets
 Low Density Pellets
Ashutosh Shukla
Page 6
Introduction of dissertation
1.3.6. Mucoadhesive system
A mucoadhesive or bioadhesivepolymer s.a. Cross linked polyacrylic acid,
when incorporated in a tablet, allow it to adhere to the gastric mucosa or
epithelium.
1.3.7. Size-based system
The diameter of tablet always greater than 2cm which cannot pass through
pylorus and can’t go in to intestine. Using high grade polymer like HPMC
K200 having high swelling property.
1.3.8. Delayed-release system:
These systems are fabricated to release the drug only at the specific site in
the GIT.
The drugs those are, 1. Destroyed in stomach or by intestinal enzymes.
2. Known to cause gastric irritation.
3. Absorbed from specific site in intestine are formulate
in such system.
The two types of delayed release systems are–

Intestinal release system
A drug may be enteric coated for intestinal release for several known reason
s.a. to prevent gastric irritation, destabilization in gastric pH.

Colonic release system
Drug are poorly absorbed through colon but may be delivered to such a site
for two reason –
(i) Local action as in the treatment of ulcerative colitis.
(ii) Systemic absorption of protein and peptide drug like insulin and
vasopressin.
1.4. Advantage and disadvantage:1.4.1. Advantage:

Following are the potential advantages of sustained release products.

Decreased local and systemic side effects reduced gastrointestinal
irritation.

Better drug utilization reduction in total amount of drug used.
Ashutosh Shukla
Page 7
Introduction of dissertation

Improved efficiency in treatment, optimized therapy, more uniform
blood concentration.

Reduction in fluctuation in drug level and hence more uniform
pharmacological response, cure of control of condition more promptly,
less reduction in drug activity with chronic use.

Method by which sustained release is achieved can improve the
bioavailability of some drugs e.g. drugs susceptible to enzymatic
inactivation can be protected by encapsulation in polymer systems
suitable for sustained release.

Improved patient compliance, less frequent dosing, reduced night-time
dosing, reduced patient care time. The importance of patient compliance
in successful drug therapy is well recognized. It has been found that there
is an inverse relationship between the number of dosages per day and the
compliance rate.

Although the initial unit cost of sustained release products is usually
greater than that of conventional dosage forms because of the special
nature of these products, the average cost of treatment over an extended
time period may be less. Economy may also result from a decrease in
nursing time and hospitalization time.

Improve absorption, utilization and thereby enhancing bioavailability.

Decreased local and systemic side effects reduced gastrointestinal
irritation.

Reduction in dosing frequency.

Better patient acceptance and compliance.

Reduced fluctuations in circulating drug levels.

Reduction in the health care cost.

Bioavailability of certain drugs can be increased.
1.4.2. Disadvantage :

The disadvantages of sustained release drug delivery system are

Decreased systemic availability in comparison to immediate release
conventional dosage forms, which may be due to incomplete release,
Ashutosh Shukla
Page 8
Introduction of dissertation
increased first-pass metabolism, increased instability, insufficient residence
time complete release, site specific absorption, pH dependent stability, etc.

Poor in vitro – in vivo correlation.

Retrieval of drug is difficult in case of toxicity, poisoning or hypersensitivity
reactions.

Reduced potential for dose adjustment of drugs normally administered in
varying strengths.

Dose dumping.

Dose adjustment is difficult.

Patient education is required for successful therapy.

Patient need to substantial additional information as to the proper used
sustained release product.

Higher cost of single unit as compared to cost of single conventional unit.

Stability problems.
1.5. Evaluation parameters:
All the prepared tablets were evaluated for following official and unofficial
parameters.
1.5.1. Pre-compression studies:
1.5.1.1. Angle of repose
Flow property was determined by measuring the angle of repose.
Tan (θ) = h / r
Where, θ = Angle of repose, h = Height of heap, r = Radius of pile.
1.5.1.2. Bulk density
Bulk density is a ratio of given mass of powder and its bulk volume.
Bulk density = M / V0
M = Mass of the powder, V0 = Bulk volume of powder.
1.5.1.3. Tapped density
It is generally given by the equation
Tapped density = M / Vr
M= Mass of powder, Vr = final tapping volume of powder.
Ashutosh Shukla
Page 9
Introduction of dissertation
1.5.1.4. Compressibility index and Hausner’s ratio
To measure the unsettled apparent volume, ( V0) and the final tapped
volume, (Vf) of the powder after tapping the material until no further volume
changes occur .given by the expression as follows.
𝟏−𝑩𝒖𝒍𝒌 𝑫𝒆𝒏𝒔𝒊𝒕𝒚
Compressibility index = 𝑻𝒂𝒑𝒑𝒆𝒅 𝑫𝒆𝒏𝒔𝒊𝒕𝒚 × 𝟏𝟎𝟎
Hausner ratio =
𝑻𝒂𝒑𝒑𝒆𝒅 𝑫𝒆𝒏𝒔𝒊𝒕𝒚
𝑩𝒖𝒍𝒌 𝑫𝒆𝒏𝒔𝒊𝒕𝒚
1.5.2. Post compression:
1.5.2.1. Hardness
Hardness is force required to break tablet across the diameter. The Hardness
of a tablet is an indication of its strength. The tablets should to stable to
mechanical stress during handling and transportation. The tablet was placed
horizontally in content with the lower plunger of the Monsanto hardness
tester and zero reading was adjusted. The tablet was than compressed by
forcing upper plunger until the tablets breaks. Thus force was noted. The
hardness of ten tablets was measured using Monsanto Hardness tester. It is
expressed in kg/cm2.
1.5.2.2. Friability
Friability is the loss of weight of tablets in the container/package, due to
removal of fine particle from the surface. The friability of the tablets was
determined using Roche friabilator. It is expressed in percentage (%). 10
tablets were initially weighed and transferred into the friabilator. The
friabilator was operated at 25 rpm for four minutes. After four minutes the
tablets were weighed again. The % friability was determined following the
formula
% Friability =
𝑰𝒏𝒊𝒕𝒊𝒂𝒍 𝑾𝒆𝒊𝒈𝒉𝒕− 𝑭𝒊𝒏𝒂𝒍 𝑾𝒆𝒊𝒈𝒉𝒕
𝑰𝒏𝒊𝒕𝒊𝒂𝒍 𝑾𝒆𝒊𝒈𝒉𝒕
× 𝟏𝟎𝟎
1.5.2.3. Weight variation
Twenty tablets were randomly selected from each batch and individually
weighed. The average weight and standard deviation of 20 tablets was
Ashutosh Shukla
Page 10
Introduction of dissertation
calculated. The batch passes the test for weight variation test. The %
deviation was calculated by using the following formula
% Deviation =
𝑰𝒏𝒅𝒊𝒗𝒊𝒅𝒖𝒂𝒍 𝑾𝒆𝒊𝒈𝒉𝒕− 𝑨𝒗𝒆𝒓𝒂𝒈𝒆 𝑾𝒆𝒊𝒈𝒉𝒕
𝑨𝒗𝒆𝒓𝒂𝒈𝒆 𝑾𝒆𝒊𝒈𝒉𝒕
× 𝟏𝟎𝟎
1.5.2.4. Estimation of drug content
Ensure the consistency of dosage units, each unit in a batch should have
active substance content within narrow range around the label claim. Dosage
units are defined as dosage forms containing a single dose or a part of a dose
of an active substance in each dosage unit.
Ten tablets were weighed and average weight is calculated. All the ten
tablets were crushed in mortar. Powder equivalent to 50mg of ramipril was
dissolved in 250ml distilled water and shaken for 20 mins. Solution was
filtered and 5 ml filtrate was diluted to the 100ml using distilled water.
Absorbance of resultant solution was measured at 235 nm using distilled
water as blank. Amount of drug present in one tablet was calculated.
1.5.2.5. Dissolution study
The vitro drug release sample were carried out using type-II (paddle type).
The dissolution medium 900ml 0.1N Hcl was placed in to dissolution flask
maintains temperature of 37 ±0.5oc and rpm of 50. One Ramipril tablet was
placed in each basket dissolution apparatus. The apparatus run for 24 hours
sample measuring 5ml.Where withdrawn after every 4 hours upto 24 hours
using 5 ml pipette. The fresh dissolution was replaced every time with the
same quantity of the sample. Collected sample with suitability diluted 0.1N
Hcl and analyzed at 235nm using 0.1N Hcl as a blank. The percentage drug
release was calculated.
1.5.2.6. Kinetic analysis of dissolution data
The results of in vitro release profile obtained for all the formulations were
fitted modes of data treatment as follows

Ashutosh Shukla
Log cumulative percent drug remaining versus time(First order)
Page 11
Introduction of dissertation

Cumulative percent drug release versus square root of time (Higuchi
model)

Log cumulative percent drug release versus time (Zero order)

cumulative percent drug released versus log time (Koysmeyers
model)
Ashutosh Shukla
Page 12
Introduction of dissertation
2. INTODUCTION OF DRUG[13-23]:
CHOLIC ACID :
Table 1. :- Drug Information
General Properties :Name
Cholic Acid
(4R)-4-[(3R,5S,7R,8R,9S,10S,12S,13R,14S,17R)-3,7,12-
IUPAC Name
trihydroxy-10,13-dimethyl2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1Hcyclopenta[a]phenanthren-17-yl]pentanoic acid
Chemical
Formula
Synonym
C24H40O5

NSC 6135,

Cholalin,

Cholalic Acid,

Kolbam
Cholic acid is a bile acid that is 5beta-cholan-24-oic
acid bearing three alpha-hydroxyl substituent’s at
position 3, 7 and 12. It has a role as a human metabolite
Description
and a mouse metabolite. It is a bile acid, a C24-steroid,
a 3alpha-hydroxy steroid, a 7alpha-hydroxy steroid, a
12alpha-hydroxy steroid and a trihydroxy-5betacholanic acid. It is a conjugate acid of a cholate.
Appearance
Odour
CAS Number
Ashutosh Shukla
Solid white powder
Odourless
81-25-4
Page 13
Introduction of dissertation
Structure
Category
Molecular
Bile acid and Derivatives
408.6
Weight
Water
Solubility
175 mg / 1L (0.175 mg / ml)
Log P
2.02
Pka
4.98 at 20°C
Melting
Point
Hygroscop
ic
Identificati
on
197 – 201 °C
Hygroscopic
Potentiometric Titration
BCS Class
I
Dose
50, 250 mg
Absorbed by passive diffusion in GI tract.
Same metabolic pathway as endogenous cholic acid;
Absorption
undergoes conjugation with glycine or taurine in liver
to form bile salts.
Ashutosh Shukla
Page 14
Introduction of dissertation
40-80%
Bioavailability
Metabolism
Half life
Forms conjugation with glycine or taurine in liver
3 Hours (in adult)
Mechanism of
Cholic acid is a primary bile acid synthesized from
Action
cholesterol in the liver. Endogenous primary bile
acids (i.e., cholic acid, chenodeoxycholic acid)
improve bile flow and provide physiologic feedback
inhibition of bile acid synthesis.
Cholic acid and its conjugates are endogenous
ligands of the nuclear farnesoid X receptor (FXR),
which regulates enzymes and transporters involved in
bile acid synthesis and enterohepatic circulation to
maintain bile acid homeostasis under normal
physiologic conditions.
 Pharmacokinetics
Cholic acid, a primary bile acid, is partially absorbed in the ileum. The
remaining part is transformed by reduction of the 7α-hydroxy group to
deoxycholic acid (3α, 12α-dihydroxy) by intestinal bacteria. Deoxycholic
acid is a secondary bile acid. More than 90% of the primary and secondary
bile acids are reabsorbed in the ileum by a specific active transporter and are
recycled to the liver by the portal vein; the remainder is excreted in the
faeces. A small fraction of bile acids is excreted in urine
Baseline pharmacokinetic profiles were similar among dose groups,
although basal cholic acid exhibited high inter-individual variability, with a
range of 36-fold in Cmax and 9-fold in AUC. When MT921 was administered
to the submental area subcutaneously, systemic cholic acid level reached
Tmax before 0.5 h post-dose and returned to baseline at approximately 6 to 8
h post-dose. Systemic exposure to cholic acid increased as the dose of
MT921 increased, while that to deoxycholic acid was similar without
showing significant differences among dose groups. The power model for
Ashutosh Shukla
Page 15
Introduction of dissertation
baseline adjusted cholic acid revealed less than proportional characteristics
for Cmax, while the dose-proportionality of AUC0-24 could not be concluded.
Rdnm (90% CI) for Cmax and AUC0-24 were 0.7773 (0.6082–0.9935) and
0.9755 (0.5031–1.8912), respectively. There was no relationship between
PK parameters of cholic acid at baseline or after MT921 injection.
MT921 is a pharmaceutical product that is under development. MT921 has
been developed as a liquid injection with a pH of 7.4 containing a buffer and
salts with 1.5% cholic acid as the main ingredient. Cholic acid has both
hydrophilic and hydrophobic properties by adding a hydrophilic group to
cholesterol, which enables cholic acidact as a surfactant.
Figure.no.1 Mean Plasma concentration-time profile of cholic acid
before and after administration of MT921.
Ashutosh Shukla
Page 16
Introduction of dissertation
Figure1.2. Pharmacokinetic dose-proportionality assessment of cholic
acid after administration of MT921.
Table 1. Pharmacokinetic parameters of cholic acid and deoxycholic
acid by dose group.
Analyte
Parameter
Tmax (h)
Cholic
Cmax (ng/mL)
Placebo
60 mg
120 mg
150 mg
(N = 6)
(N =6)
(N = 6)
(N = 6)
0.25
0.25
0.08
(0.08–
(0.08–
(0.08–
0.5)
0.25)
0.25)
1568
2488 ±
3193
± 342
370
703
1413
2400 ±
2769
± 291
342
663
0.04
(0–15)
319 ± 451
Acid
Baseline
adjusted Cmax
(ng/mL)
Ashutosh Shukla
70 ± 156
Page 17
±
±
Introduction of dissertation
AUC0-24
1618
(ng∙h/mL)
1501
±
±
1185
2887
Baseline
adjusted C0-24
4262
−67 ± 2141
(ng∙h/mL)
±
1108
7071 ±
8344
2505
6851
6582 ±
6319
2344
4498
Data are presented as mean ± standard deviation, except for T max which is presented
as median (min–max).

Absorption
 Bioavailability
Absorbed by passive diffusion in GI tract.

Distribution
Endogenous cholic acid distributed into human milk; no data available in
nursing women or lactating animals to determine distribution of
exogenously administered cholic acid in milk.

Elimination
 Metabolism
Same metabolic pathway as endogenous cholic acid; undergoes conjugation
with glycine or taurine in liver to form bile salts.

2.2. Elimination Route
Undergoes enterohepatic circulation. Conjugated cholic acid not absorbed
in colon is deconjugated and dehydroxylated to form cholic acid and
deoxycholic acid, which may be reabsorbed in colon or excreted in feces.
 Pharmacodynamic
Cholic acid is the predominant primary bile acid in man. In patients with
inborn deficiency of 3β-Hydroxy-Δ5 -C27-steroid oxidoreductase and Δ4 3-Oxosteroid-5β-reductase, the biosynthesis of primary bile acids is reduced
or absent. Both inborn diseases are extremely rare, with a prevalence in
Europe of about 3 to 5 patients with 3β-Hydroxy-Δ5 -C27-steroid
oxidoreductase deficiency per 10 million inhabitants, and an estimated ten-
Ashutosh Shukla
Page 18
±
±
Introduction of dissertation
fold lower prevalence for Δ4 -3-Oxosteroid-5β-reductase deficiency. In the
absence of treatment, un physiologiccholestatic and hepatotoxic bile acid
metabolites are predominant in the liver, serum and urine.
The rational basis for treatment consists of restoration of the bile acid
dependent component of bile flow enabling restoration of biliary secretion
and biliary elimination of toxic metabolites; inhibition of the production of
the toxic bile acid metabolites by negative feedback on cholesterol 7αhydroxylase, which is the rate-limiting enzyme in bile acid synthesis; and
improvement of the patient’s nutritional status by correcting intestinal
malabsorption of fats and fat-soluble vitamins.
Clinical experience has been reported in the literature from small cohorts of
patients and single case reports; absolute patient numbers are small due to
the rarity of the conditions. This rarity also made the conduct of controlled
clinical studies impossible. Overall, cholic acid treatment results for about
60 patients with 3β-Hydroxy-Δ5 -C27-steroid oxidoreductase deficiency are
reported in the literature. Detailed long-term data on treatment with cholic
acid monotherapy are available for 14 patients observed for up to 12.9 years.
Cholic acid treatment results for seven patients with Δ4 -3-Oxosteroid-5βreductase deficiency for up to 14 years are reported in the literature. Detailed
medium- to long-term data are available for 5 of these patients, of whom 1
has been treated with cholic acid monotherapy.
Oral cholic acid therapy has been shown to: postpone or obviate the need for
liver transplantation; restore normal laboratory parameters; improve
histological lesions of the liver, and significantly improve all of the patient’s
symptoms. Mass spectrometry analysis of urine during cholic acid therapy
shows the presence of cholic acid and a marked reduction, or even complete
elimination of the toxic bile acid metabolites. This reflects restoration of an
effective feedback control of bile acid synthesis and a metabolic
equilibrium. In addition, blood cholic acid concentration was normal and
fat-soluble vitamins were restored to their normal range.
Mean concentration–time profiles of free fatty acid, triglyceride, and total
cholesterol were comparable when measured at baseline and post-dose
regardless of the dose group. There were no statistically significant
differences in PD parameters among dose groups either.
Ashutosh Shukla
Page 19
Introduction of dissertation
 Mechanism of Action
Cholic acid is a primary bile acid synthesized from cholesterol in the
liver. Endogenous primary bile acids (i.e., cholic acid, chenodeoxycholic
acid) improve bile flow and provide physiologic feedback inhibition of bile
acid synthesis.
Cholic acid and its conjugates are endogenous ligands of the nuclear
farnesoid X receptor (FXR), which regulates enzymes and transporters
involved in bile acid synthesis and enterohepatic circulation to maintain bile
acid homeostasis under normal physiologic conditions. Activation of the
FXR through binding of cholic acid results in upregulation of transcription
of genes coding for hepatic conjugation enzymes, resulting in increased
metabolic conjugation of bile acids and bile acid-dependent bile flow.
Activated FXR also results in downregulation of cholesterol 7α-hydroxylase
encoded by the CYP7A1 gene, potentially resulting in reduction in de novo
synthesis of primary bile acids; clinical importance of such downregulation
is not fully known.
 Drug Administration

Usual Adult Dose for Bile Acid Synthesis Disorders

Initial dose: 10 to 15 mg/kg orally once a day or in 2 divided doses

Patients
with
Concomitant
Familial
Hypertriglyceridemia:
Initial dose: 11 to 17 mg/kg orally once a day or in 2 divided doses

Maintenance dose: The lowest dose that effectively maintains liver
function
 Comments:
Adequacy of dosing should be determined by patient monitoring of clinical
response and laboratory values; monitor more frequently during periods of
rapid growth, concomitant disease, and pregnancy
Concurrent elevations of serum gamma glutamyl transferase (GGT) and
ALT may indicate cholic acid overdose.
 Uses:

For the treatment of bile acid synthesis disorders due to single enzyme
defects
Ashutosh Shukla
Page 20
Introduction of dissertation

As adjunctive treatment of peroxisomal disorders including Zellweger
spectrum disorders in patients who exhibit manifestations of liver
disease, steatorrhea, or complications from decreased fat soluble
vitamin absorption.

Usual Pediatric Dose for Bile Acid Synthesis Disorders

Initial dose: 10 to 15 mg/kg orally once a day or in 2 divided doses

Patients with Concomitant Familial Hypertriglyceridemia:
Initial dose: 11 to 17 mg/kg orally once a day or in 2 divided doses

Maintenance dose: The lowest dose that effectively maintains liver
function
 Comments:

Adequacy of dosing should be determined by patient monitoring of
clinical response and laboratory values; monitor more frequently
during periods of rapid growth, concomitant disease, and pregnancy

Concurrent elevations of serum gamma glutamyl transferase (GGT)
and ALT may indicate cholic acid overdose.
 Uses:

For the treatment of bile acid synthesis disorders due to single enzyme
defects

As adjunctive treatment of peroxisomal disorders including Zellweger
spectrum disorders in patients who exhibit manifestations of liver
disease, steatorrhea, or complications from decreased fat soluble
vitamin absorption.

Liver Dose Adjustments

Use caution; patients are expected to present with some degree of
hepatic impairment which should improve with treatment

Discontinue treatment if liver function does not improve within 3
months of the start of treatment.

Discontinue treatment if complete biliary obstruction develops.

Interrupt treatment if at any time there are clinical or laboratory
indicators of worsening liver function or cholestasis; may consider
restarting treatment at a lower dose if liver function parameters return
to baseline.
Ashutosh Shukla
Page 21
Introduction of dissertation

Dose Adjustments

Patients with Concomitant Familial Hypertriglyceridemia: Initial
dose: 11 to 17 mg/kg orally once a day or in 2 divided doses

Patients with newly diagnosed or a family history of, familial
hypertriglyceridemia may have poor absorption of this drug from the
intestine and require a 10% increase in the recommended dosage to
account for losses due to malabsorption.

See manufacturer product information for weight-based dosing tables.

Precautions

Safety and efficacy have not been established in patients younger than
3
weeks
of
age.
Consult WARNINGS section for additional precautions.

Other Comments
 Other comments:
Administration advice:
 Take with food
 Take at least 1 hour before or 4 to 6 hours (or at as great an interval as
possible) after a bile acid resin or aluminum-based antacid
 Capsules should not be crushed or chewed
 For patients unable to swallow capsules whole, capsules may be opened
and mixed with 15 to 30 mL of infant formula, breast milk, soft food such
a mashed potatoes or apple puree
 To prepare, hold capsule over liquid/food and gently twist open capsule and
allow contents to fall; stir for 30 seconds; the capsule contents will not
dissolve but remain as fine granules
 Administer mixture immediately
General:

Treatment should be initiated and monitored by an experienced hepatologist
or pediatric gastroenterologist.

The safety and effectiveness of this drug on extrahepatic manifestations of
bile acid synthesis disorders due to single enzyme defects or peroxisomal
disorders including Zellweger spectrum disorders have not been established.
Ashutosh Shukla
Page 22
Introduction of dissertation

The utility of bile acid measurements in monitoring the clinical course of
patients and in decisions regarding dose adjustment has not been
demonstrated.

Hepatic: Monitor AST, ALT serum gamma glutamyltransferase (GGT),
alkaline phosphatase (ALP), bilirubin, and INR every month for the first 3
months, every 3 months for the next 9 months, every 6 months during the
subsequent 3 years, and annually thereafter

Monitor more frequently during periods of rapid growth, concomitant disease,
and pregnancy.
Patient advice:
 Instruct patients to report any signs or symptoms of worsening liver
impairment.
 Patients should speak to their physician or health care provider if they become
pregnant, intend to become pregnant, or are breastfeeding.
Ashutosh Shukla
Page 23
Introduction of dissertation
3. Aim of research work:
Enhancing the permeability of cholic acid by using different excipients.

The aim of research work is improve the drug half life.

Sustained release tablet is used to formulate delayed release drugs by using direct
compression or granulation prior compression.

Lack of bile acid synthesis can be treated by cholic acid and its derivatives.
3.1. Rationale:

Cholic acid to treat the bile acid synthesis disorder and belongs to the Categor Bile
acid and Derivatives and class I.

Its recommended 50, 250 mg dose is available as tablet dosage form.

Cholic acid half-life is 3 hrs in adults.

Its water solubility is 175mg/1 L because of Cholic acid is BCS class I drug.

Bioavailability is approximately 40-80%.
3.2. Objectives:

Enhance permeability by using direct compression or granulation prior
compression.

To perform identification and estimation by HPLC.

To formulate Sustained release tablet of Cholic acid.

To evaluate Sustained release tablet of Cholic acid.

To perform stability study of optimize formulation.
Ashutosh Shukla
Page 24
Introduction of dissertation
4. Literature review:4.1. Literature review on dosage form[24-37]:
Lakade SH et al., have studied to develop hydrophilic polymer (HPMC) and
hydrophobic polymer (Ethyl cellulose) based Nicorandil matrix sustained
release tablet for treating the anginal disorder which can release the drug up
to 24 hours in predetermined rate. The in-vitro release rate profile of
formulation F2 (Gaur gum) showed higher drug release rate than other
formulation.
Shanmugam S et al., has formulated and evaluated the sustained release
matrix tablets of Losartan potassium. The studies showed drug release from
the tablets was sufficiently sustained and non-fickian transport of the drug
from tablets was confirmed. The Losartan potassium sustained release tablets
were stable at 40°C/75% RH up to 3 months period of study.
Krishnaiah YSR et al., have designed oral controlled drug delivery system
for highly water soluble drugs using guar gum as a carrier in the form of three
layered matrix tablet and concluded that guar gum is potential carrier in this
system for a highly water soluble drugs. Muhammad A et al., has done the
formulation and in-vitro evaluation of Flurbiprofen controlled release matrix
tablets using cellulose derivative polymers. The studies showed ethyl
cellulose ether derivative polymer was effective release controlling polymer
for Flurbiprofen matrix tablet. HPMC also retarded the release rate of drug
when combined with ethyl cellulose. Tabandeh H et al., have prepared
sustained-release matrix tablets of Aspirin using ethylcellulose, eudragit
RS100, eudragit S 100 by direct compression method and reported that ethyl
cellulose with an little amount as little as 10 % in the formulation could make
sustained-release Aspirin tablets.
Phani K et al., has prepared and evaluated the sustained release matrix tablets
of Lornoxicam using tamarind seed polysaccharide(TSP). The studies showed
that the tablets with highest binder concentration showed maximum hardness
and minimum friability. After 24 hours tablets with 20% tamarind seed
polysaccharide binder showed maximum drug release and tablets with 40%
tamarind seed polysaccharide binder showed minimum drug release. With
increasing the percentage of natural polymer, release rate decreased, though
Ashutosh Shukla
Page 25
Introduction of dissertation
drug release pattern was mainly dependent on the type of polymer. Among all
the formulations, the formulation which contains 20% TSP binder releases
the drug which follows zero order kinetics via swelling, diffusion and erosion.
Yassin EH et al., formulated the novel sustained-release double-layer tablets
of Lornoxicam by using cyclodextrin and xanthan gum combination. Each of
the proposed DLTs (Double layered tablets) is composed a fast-release layer
and a sustained-release layer, anticipating rapid drug release that start in the
stomach to rapidly elevate the symptoms and continues in the intestine to
maintain protected analgesic effect.
Nayak RK et al., has formulated and evaluated the sustained release matrix
tablets of Lornoxicam. The tablet with guar gum in the ratio of drug: polymer
(1:2) exhibited greater swelling index and better dissolution profile than those
with pectin, xanthan gum, sodium alginate. The drug release of optimized
formulation follows the Higuchi kinetic model, and the mechanism was found
to be non-fickian/anomalous according to Korsmeyer-Peppas equation.
Uddin M et al., formulated sustained release matrix tablet of Valsartan by
direct compression method using Methocel K4M CR and Methocel K100M
CR as polymer. They evaluated powder blend for its evaluation involves three
micromeritic properties, physical property studies of tablets and in-vitro
release kinetics studies. The weight variation was observed to be within the
prescribed limits for each formulation. In-vitro release studies were carried
out using USP apparatus type II and dissolution medium consisted of 0.1N
hydrochloric acid for the first 2 hours and phosphate buffer pH 6.8 from 3 to
24 hours, maintained at 37±0.5°C. Kinetic modeling of in-vitro release
profiles revealed that the drug release mechanism from all proposed
formulations followed anomalous type or non-fickian transport. In this study
formulation F8, F9 and F10 showed better drug release compared to other
formulations. Drug release from the matrix occurred by combination of two
mechanism, diffusion and erosion of tablet.
Vinit Sharma et al., have developed Pregabalin sustained release matrix
tablets prepared by using Hydroxy propyl methyl cellulose. The matrix tablets
were prepared by direct compression method. Formulation F2, F3 to F5 failed
to sustain release and among all the formulation, F4 shows 99.25% of drug
release at the end of 12 hours. These results showed that above a particular
Ashutosh Shukla
Page 26
Introduction of dissertation
concentration of MCC 101, HPMC K-100 and PVP K-30 are capable of
providing sustained drug release.
Madhavi N et al., developed sustained release matrix tablet of Phenytoin
sodium using eudragit- RL100, eudragit-RS100, HPMC-E15, ethyl cellulose
(N-14), chitosan and HPMC as release controlling factor. Different
dissolution models were applied to drug release data in order to evaluate
release mechanisms and kinetics. Formulation F6 showed 60% of drug release
for 12 hours. Criteria for selecting the most appropriate model were based on
linearity (coefficient of correlation). Based on “n” value (0.168) the drug
release was follows Fickian diffusion. Also the drug release mechanism was
best explained by Higuchi order (correlation value is 0.9063) by using this
polymer.
Varsha B. et al., formulated and evaluated sustain release matrix tablets of
Pregabalin by direct
compression method using hydroxyl propyl
methylcellulose (HPMC K-100), polyvinylpyrrolidone (PVP K-30) and
microcrystalline cellulose (MCC 101 and MCC 102) in varying ratios.
Powder blends and prepared tablets were subjected to various precompression
and post-compression evaluations respectively. Formulation F5 (which
composed of HPMC K100 and MCC 102 in the ratio 1:3) and F7 (which
composed of HPMC K100 and MCC 102 in the ratio 2:1) exhibited 93.03%
and 95.80% of drug release respectively at the end of 12 hours. These findings
revealed that by using MCC 102 and HPMC K-100, exhibited sustained
release of Pregabalin for 12 hours.
Sajid et al., developed sustained release matrix tablets of phenytoin sodium
by the wet granulation method using water as granulating agent along with
matrix materials like guar gum, sodium alginate, tragacanth and xanthan gum
with varying percentage. The granules showed satisfactory flow properties,
compressibility, and drug content. All the tablet formulations showed
acceptable pharmacotechnical properties. In the further formulation
development process, formulation F8 (55% guar gum with 10% acacia)
exhibited satisfactory drug release up to 12 hours. The mechanism of drug
release from all the formulations was diffusion coupled with erosion.
Ashutosh Shukla
Page 27
Introduction of dissertation
Subramaniam K et al., has formulated and evaluated the sustained release
tablets of Aceclofenac using hydrophilic matrix system. Powder blends and
prepared tablets were subjected to various pre-compression and postcompression evaluations respectively. The kinetic treatment of selected
formulation (F8) showed that the release of drug follows zero order models.
It is concluded that the formulation of sustained release tablet of Aceclofenac
containing HPMC K100, mannitol and lactose (formulation F8) which are
taken as ideal or optimized formulation of sustained release tablet for 24 hours
release as it fulfills all the requirement of sustained release tablets.
Noorana et al., designed twice daily mini-tablets formulation of pregabalin.
For achieving the sustain release profile, various viscosity grades of hydroxyl
propyl methylcellulose polymer (HPMC K4M, K15M, K100M) were used.
The mini-tablets were prepared by directcompression method. The in-vitro
formulation showed nearly 99.57 % of drug was sustained for a period of 12
hours. The stability study revealed that the formulations were found to be
stable. It was concluded that matrix mini-tablets of Pregabalin along with
HPMC can be used to improve its half-life and improve its bio-availability.
Emami J et al., in the present study sustained-release matrix tablets of
flutamide were prepared by direct compression method using different
polymers. Cellulose ethers (HPMC and NaCMC), natural gums (guar and
xanthan gums) and compressible eudragits (RSPO and RLPO) and their
combinations were used in different ratios to examine their influence on tablet
properties and drug release profile. Almost in all formulations, with
increasing the percentage of polymer, release rate decreased, though drug
release pattern was mainly dependent on the type of polymer. It was
concluded that the formulations H2F4 (containing 25% HPMC) and S3F4
(containing around 40% RSPO) met the desired requirements for a sustainedrelease dosage form. These two formulations released their drug content with
a first order kinetic. Islam M S et al., has studied effect of polymers on
sustained release Theophylline matrix tablets prepared by direct compression
method using different release retardant polymers like HPMC, HPMCP,
kollidon, Eudragit L 100 and Eudragit RL PO. Prepared matrix tablets showed
satisfactory tableting properties. Matrix systems composed of Eudragit L 100
Ashutosh Shukla
Page 28
Introduction of dissertation
and Eudragit RL PO released almost 100% Theopylline within 5 hrs and 6 hrs
of dissolution respectively.
Moin A et al.,formulated sustained release matrix tablets of Diltiazem by
using microcrystalline cellulose, Hydroxy propyl methyl cellulose (HPMC),
locust bean gum and karaya gum. Matrix tablets of Diltiazem were prepared
at different ratios of drug: gum (1:1, 1:2 and 1:4) and of the gum blends
(karaya gum, karaya gum/locust bean gum, karaya gum/Hydroxy propyl
methyl cellulose and karaya gum/locust bean gum/hydroxyl propyl methyl
cellulose) by direct compression. The matrix tablets were evaluated for
hardness, friability, in-vitro release and drug content. It was concluded that
locust bean gum alone cannot efficiently control drug release, a suitable
combination of the two natural gums (karaya and locust bean gum) may be
successfully employed for formulating sustained release matrix tablets of
Diltiazem.
Ulla SN et al., has formulated and evaluated sustained release matrix tablets
of Lornoxicam. Lornoxicam, a potent non-steroidal anti-inflammatory drug
which has short half-life, makes the development of sustained release (SR)
forms extremely advantageous. However, due to its weak acidic nature, its
release from SR delivery systems is limited to the lower gastrointestinal tract
which consequently leads to a delayed onset of its analgesic action. Therefore,
the present investigation of this study was to develop Lornoxicam SR matrix
tablets that provide complete drug release that starts in the stomach to rapidly
alleviate the painful symptoms and continues in the intestine to maintain
analgesic effect. Various formulations were developed by using release rate
controlling and gel forming polymers like HPMC (K4M, K15M, K100M) by
direct compression method.
Sharma VK et al., has formulated floating sustained release matrix tablets
using hydroxyl propyl methyl cellulose (HPMC) K15M as matrix forming
polymer and sodium bicarbonate as a gas generator. Meloxicam was used as
model drug. It was observed that the buoyancy lasted for up to 24 hrs and
supported by in-vitro dissolution studies. Floating drug delivery system can
be successfully formulated by direct compression technique and combination
of polymers.
Ashutosh Shukla
Page 29
Introduction of dissertation
Rao V et al., have formulated and evaluated the release profile of matrix
tablets of losartan potassium prepared by using different concentrations of
chitosan and trisodium citrate as cross-linking agent with combination of
HPMC K100M, carbopol 934P, and xanthan gum as polymers. Matrix tablets
were prepared by direct compression. It was found that among the 12
formulations F11 (99.72%) and F12 (98.70%) showed good dissolution
profile to control the drug release. The above results concluded that by
combining different classes of polymers an acceptable release profile can be
obtained in the fluctuating in-vivo environment.
Michael M C et al., has studied the physico-chemical properties and
mechanism of drug release from ethyl cellulose matrix tablets prepared by
direct compression and hot-melt extrusion. The results of this study
demonstrated that the Guaifenesin release rate was dependent upon the
particle size of ethyl cellulose and the processing conditions employed to
prepare the tablets. The Guaifenesin release rate was slower in tablets
prepared with the “fine” ethyl cellulose particle size fraction due to the
presence of fewer soluble drug clusters within the matrix. Tablets prepared
by hot-melt extrusion exhibited considerably slower drug release relative to
those prepared by direct compression method.
Rakesh PP et al., formulated and evaluated sustained release matrix tablet of
Tizanidine Hydrochloride by direct compression technique. Tizanidine
hydrochloride tablets were prepared by melt direct compression technique
using xanthum gum, guar gum, glyceryl behenate, glyceryl monostearate and
stearic acid in different proportion. Sustained release tablets of Tizanidine
prolong the time for absorption as well as bioavailability and thus better
patient compliance can be achieved.
Ahmad QJ et al., has prepared bi-layer tablet of Lornoxicam (LOR) for the
effective treatment of arthiritis. LOR was formulated as immediate release
layer
and
sustained
release
layer
using
hydrophilic
matrix
(hydroxypropylmethylcellulose [HPMC K15M]). The effect of concentration
of hydrophilic matrix (HPMC K15M), binder (polyvinyl- pyrrolidone [PVP
K30]) and dissolution study of sustained release layer showed that an
increasing amount of HPMC or PVP K30 results in reduced Lornoxicam
release. The hydrophilic matrix of HPMC could control the Lornoxicam
Ashutosh Shukla
Page 30
Introduction of dissertation
release effectively for 24 hours. It is evident from the result that a matrix
tablets prepared with HPMC and binding agent (PVP, 4% w/v) is a better
system for once-daily sustained release of a highly water-insoluble drug like
Lornoxicam.
Basavaraj et al., designed and characterized sustained release matrix tablets
of Aceclofenac containing tamarind seed polysaccharide seed kernel of
Tamrindus indica belonging to family leguminaceae. It is practically
insoluble in water so it is suitable to develop sustained release matrix tablet
using hydrophilic polymer. Aceclofenac is non-steroidal antiinflammatory
drug (NSAID) used extensively in the treatment of rheumatoid arthritis,
osteoarthritis and ankylosing spondylitis. It is newer derivative of Diclofenac
and having less GIT complication, with short biological half-life 4 hrs, so
developed formulation provides the advantages of sustained release
formulations. The tamarind seed polysaccharide (TSP) was extracted from
tamarind kernel powder and this polysaccharide was utilized in the
formulation of matrix tablets containing Aceclofenac by wet granulation
technique and evaluated for its drug release characteristics. TSP is a
hydrophilic and rate controlling polymer. The matrix tablets were found to be
effective in sustaining the drug release up to 12 hours so, that the controlled
released profile is maintained for an extended period.
4.2. Literature review on drug[38-47]:
Afonso et al. showed that necroptosis was induced in a study of bile acidrelated cholestasis. Primary biliary cholangitis (PBC) patients are chronic
cholestatic liver disease characterized by the destruction of small intrahepatic
bile ducts. GCDCA is a major component of human serum and bile during
cholestasis. These studies showed increased RIPK3 expression and MLKL
phosphorylation in liver samples from human PBC patients. Bile duct ligation
(BDL) is a surgical model for serious obstructive cholestasis, resulting in
significant jaundice and hepatocellular damage. They found that the mRNA
and protein expression of RIPK3 and MLKL, and MLKL phosphorylation
were strongly increased in the liver of BDL mice. RIP1 mRNA levels were
not altered, but RIPK1 protein levels were also increased in whole liver cell
lysates from BDL mice. These results suggest that targeting necroptosis may
represent a therapeutic strategy for acute cholestasis.
Ashutosh Shukla
Page 31
Introduction of dissertation
Zhou et al. reported that bile acid induces necroptosis in chronic pancreatitis.
Bile acid has also been shown to induce acinar cell death through decreased
mitochondrial membrane potential, increased reactive oxygen species and
energy depletion. All of these are known to promote acinar cell apoptosis and
necrosis in pancreatitis. The most abundant primary bile acids in humans are
GCDCA and TCA. Therefore, they confirmed that pancreatic cell lines
exposed to GCDCA and TCA increased the expression of the nuclear bile
acid receptor known as the farnesoid X receptor (FXR), and decreased the
expression of the essential autophagy-associated protein ATG7. Bile acid was
also increased in pancreatic tissue from patients with human chronic
pancreatitis, which correlated with increased FXR, and ATG7 expression was
associated with locally reduced autophagic activity. These results
demonstrated a cascade of events in which the local accumulation of bile acid
signals through FXR inhibits autophagy in pancreatic acinar cells, thereby
triggering acinar cell apoptosis and necroptosis.
Katona et al. found that the synthetic enantiomers of lithocholic acid
(entLCA), chenodeoxycholic acid (ent-CDCA), and deoxycholic acid (entDCA) induced toxicity and apoptosis in HT-29 and HCT-116 colorectal
cancer cells (Figure 3C). Native bile acids induced more apoptosis and
cleavage of capase-3 and -9 compared to enantiomeric bile acids. However,
natural and enantiomeric bile acids had similar effects on cell proliferation.
Among them, LCA- and ent-LCA-mediated apoptosis were prevented by both
the pancaspase and selective caspase-8 inhibitors, whereas selective caspase2 inhibitors provided no protection. In addition, LCA and ent-LCA increased
CD95 localization in the plasma membranes. Bile acid-mediated caspase-8
activation in hepatocytes is induced by CD95 oligomerization and
translocation to the cell membrane. This showed that LCA and entLCA
induced apoptosis selectively through CD95 activation, which induced procaspase-8 cleavage due to increased ROS production.
Agarwal et al. showed that the bile acid-added triazolyl aryl ketones 6af and
6cf induced apoptosis in MCF-7 breast carcinoma cells (Figure 3D). In
particular, compound 6cf induced apoptosis by 46.09% in MCF-7 cells, and
compound 6af induced apoptosis by 33.89%, showing that 6cf was more
effective in inducing apoptosis.
Ashutosh Shukla
Page 32
Introduction of dissertation
Melloni et al. showed that CDC-PTX and UDC-PTX combined with
paclitaxel (PTX), an anticancer drug, induced apoptosis in HL60 and NB4
acute promyelocytic leukemia cells through a high-yield condensation
reaction of CDCA and UDCA. It was also shown that CDC-PTX and UDCPTX RKO induced apoptosis in HCT-116 colon cancer cells. In particular, in
all four cell lines (HL60 and NB4 human leukemic cell lines, Int. J. Mol. Sci.
2022, 23, 7184 23 of 31 RKO, and HCT-116 colon cancer cells), CDC-PTX
induced more apoptosis than UDC-PTX. In addition, Pacific Blue (PB)conjugated derivatives of CDC-PTX and UDC-PTX (CDCPTX-PB and
UDC-PTX-PB) were prepared through multi-step synthesis to evaluate their
ability to enter tumor cells. CDC-PTX-PB showed a greater ability to cross
the plasma membrane than UDC-PTX-PB.
Brossard et al. showed that 7b, a new piperazinyl bile acid derivative,
induced apoptosis in KMS-11 multiple myeloma and HCT-116 colon cancer
cells (Figure 3F). Moreover, the apoptosis rate was higher in KMS-11
multiple myeloma cells than in HCT-116 colon cancer cells. Compound 7b
was also shown to induce DNA fragmentation, a characteristic of apoptosis,
in KMS-11 cells.
Singh et al. showed that four cationic bile acid-based facial amphiphilic
substances, LCA-TMA1, CDCA-TMA2, DCA-TMA2, and CA-TMA3
characterized by trimethyl ammonium head groups, induced apoptosis in
HCT-116 and DLD-1 colon cancer cells (Figure 3G). LCA-TMA1 induced
the highest level of apoptosis in both cells.
Kihel et al. synthesized six novel bile acid (LCA and CDCA)-substituted
piperazine conjugates lysocholic acids and chenodeoxycholic acid
piperazinylcarboxamides, and showed that compound IIIb caused apoptosis
in KMS-11 multiple myeloma cells (Figure 3H). Of the six compounds, IIIb
showed the best apoptotic activity in KMS-11 multiple myeloma cells. This
revealed that apoptosis is involved in Mcl-1 and PARP-1 cleavage, the
inhibition of NF-κB signaling, and DNA fragmentation.
Singh et al. synthesized cationic amphiphilic materials with different cationic
charge head group characteristics using LCA. Among them, it was confirmed
that the lithocholic acid-based amphiphilic substance carrying the piperidine
head group (LCA-PIP1) was 10 times more cytotoxic to colorectal cancer
Ashutosh Shukla
Page 33
Introduction of dissertation
cells than the precursor (Figure 3I). This confirmed that LCA-PIP1 induced
greater levels of apoptosis in HCT-116 colorectal cancer cells compared to
LCA. LCA-PIP1 induced sub-G0 arrest and the cleavage of caspase-3, -7, and
-8. These effects of LCA-PIP1 were also confirmed in a tumor xenograft
model of HCT-116 cells; tumor volume was reduced by up to 75%.
Sreekanth et al. synthesized a bile acid conjugate of tamoxifen using three
bile acids LCA, DCA, and CA. Among them, the free amine headgroup-based
CA-tamoxifen conjugate (CA-Tam3-Am) was shown to be the most potent
anticancer conjugate in breast cancer cells compared to tamoxifen, and it
induced apoptosis. CA-Tam3-Am induced more apoptosis than tamoxifen in
4T1, MCF-7, T47D, and MDA-MB 231 breast cancer cells, and showed cell
arrest at the G0 phase. In addition, the treatment of MCF7 cells, which are
estrogen receptor-positive, with CA-Tam3-Am induced apoptosis through the
intrinsic and extrinsic pathways, whereas treatment of MDA-MB-231 cells,
which are estrogen receptor-negative, resulted in apoptosis via the intrinsic
pathway.
Tang et al. showed that norUDCA, a side-shortened C23 homologue of
UDCA, induces autophagy in HTOZ cervical cancer cells (Figure 3K). Alpha1 antitrypsin (α1AT) deficiency is a genetic disorder that causes the
accumulation of the α1AT mutant Z (α1ATZ) protein in the ER of
hepatocytes, leading to chronic liver damage, liver fibrosis, and
hepatocellular carcinoma. NorUDCA inhibited the accumulation of α1ATZ
through the autophagy-mediated degradation of α1ATZ in HTOZ cells. They
showed that AMPK activation is required for norUDCA-induced autophagy
and α1ATZ degradation. Furthermore, they demonstrated that mTOR/ULK1
was involved in nor UDCA-induced AMPK activation and autophagy in
HTOZ cells.
Markov et al. showed that compound 9, among a series of novel DCA
derivatives containing an aliphatic diamine and amino alcohol or morpholine
moiety at the C3 position, induces apoptosis and autophagy in HuTu-80
duodenal carcinoma cells (Figure 3L). They showed that compound 9 causes
ROS-dependent cell death by activating the intrinsic caspase-dependent
pathway of apoptosis and cell-destructive autophagy in HuTu-80 duodenal
carcinoma cells.
Ashutosh Shukla
Page 34
Introduction of dissertation
5. Materials And Equipment:5.1. Material
Table no. 5.1. : List of Proposed materials to be used
Sr.
Material proposed to be used
Function
Cholic acid
Active
no.
1
Pharmaceutical
Ingredients
Sodium Starch Glycolate
2
Disintegrant
Croscarmellose Sodium
3
Gelatin, agarose, starch
Polymers
4
Corn starch, talc
Lubricant
Magnesium Stearate
5.2. Equipment
Table no. 5.2. : List of Proposed Equipments to be used
Sr.
Equipments
no.
Ashutosh Shukla
1
Digital weighing balance
2
Dissolution apparatus
3
Potentiometer
4
Roche Friabilator
5
Monsanto Hardness tester
6
Tablet compression machine
Page 35
Introduction of dissertation
6. Future plan of work:
Table no.6.1. : Future plan of work
Stages
Work
Phase I
Literature Review
Procurement of API and Excipients
Phase II
Identification and estimation of drug using potentiometer
Phase III
Preformulation Studies.
Phase IV
Preparation of Sustained Release tablet.
Phase V
Evaluation of Sustained Release tablet.
 Pre-compression studies:
 Angle of repose
 Bulk density
 Tapped density
 Carr’s index
 Hausner’s ratio
 Post-compression studies:
 General appearance
 Size and shape
 Weight variation
 Friability study
 Hardness test
 Solubility
 Disintegration Time
 Dissolution Study
Phase VI
Ashutosh Shukla
Stability Study
Page 36
Introduction of dissertation
7. References:
1. Rout S, Kar D, A brief Review on Modified Release Solid Dosage Form with
special reference to Design, International Journal of Research in Ayurveda and
Pharmacy, 2011, page no.-1701-1708.
2. Chien Y W Novel Drug delivery system,2nd edition Dekker, New York (1992), Del
cavillo, Mullol J, Barta J, Davila J, Montoro J, Sastre J,Valero AL, Comparative
pharmacology of the H 1 antihistamines 16, 2006, 3-12.
3. Mamidala R , Ramana V, Yamsani M, Factor influencing the design and
performance of oral sustained/controlled release dosage form, International journal
of pharmaceutical sciences and Nanotechnology, volume 2, 2009.
4. Zameruddin M, Namdev H, Jadhav S B, Kadam V S, Bharkd V B, Recent Advances
of Sustained Release Oral Drug Delivery System: A Review, World Journal of
Pharmacy and Pharmaceutical Science, Volume 3, 2014.
5. Sampath k, Bhowmik D, Shrivastava S, Sutained Release Drug Delivery System
potential, The pharma Innovation, Volume 1, 2012, page no.-48-52.
6. Lechmen L, Liberman H, The Theory and Practice of Industrial Pharmacy,
Sustained release dosage form, Third edition, year-1991, Page no.-430-456.
7. Patnaik A, Nagarjuna T, Sustained Release Drug Delivery System: A moderate
formulation Approach, International journal of Research in Pharmaceutical and
Nano sciences, 2013.
8. Zalte H D, Saudagar R B, Review on sustained release matrix tablet, International
Journal of Pharmacy and Biological sciences, Volume 3, 2013.
9. Parashar T, Singh V, Singh G, Tyagi S, Patel C, Gupta A, Novel oral sustained
release technology: A concise review, International journal of Research and
Development in Pharmacy and Life sciences, Volume 2, 2013.
10. Gupta M, Ray B, A review on Sustained Release Technology, International Journal
of Therapeutic Application, Volume 8, 2012.
11. Remington AR, The Science and Practice of Pharmacy, 2002.
12. Madhukat, Doshi M, Milind, Joshi D,Mehta B P, Pharmaceutical Composition for
Controlled Drug Delivery System, Patent No. US 7, 157, 100 B2, Jan 2(2007).
13. Robinson JR, Vincent H, Lee L, Controlled Drug Delivery Fundamental and
application, 2002.
Ashutosh Shukla
Page 37
Introduction of dissertation
14. Jain NK, Controlled and Novel Drug Delivery, CBS publishers and Distribution
1997.
15. Venkataraman, Daar SN, Chester A, Kliener L. An overview of controlled release
systems. Handbook of pharmaceutical controlled release technology. Marcel
Dekker Inc., 2000.
16. Wagnaer JG, Biopharmaceutics and pharmacokinetics Org Intelligence publishers,
1971.
17. Ratilal D, Gaikwad Priti D, Banka Vidyadhar H, Pawalsunil P, A review on
sustained release technology, International journal of Research in Ayurveda and
Pharmacy, 2011.
18. Rudnic E. Schawartz JB. Oral solid dosage forms. Remington’s Pharmaceutical
sciences, Mark Publishing Company Easton 2000, 1965.
19. Staney DS,Formulation Strategies for absorption windows. Drug Discovery today
2005.
20. Garg S, SharmaS,Gastroretentive Drug delivery system. Businessbriefing,
Pharmatech 2003.
21. Marc MP, Julie HR, Burnies M Tasosartin, Enolatasosartan and Angiotensin 2
Receptor Blockade; The confounding Role of Protein Binding, The journal of
Pharmacology and environmental Therapeutics, 2005.
22. Cavillo D, MullolJ, BartaJ, Davila, Jauregui, MontoroJ, Sastre J Valero AL.
Comparative
pharmacology
of
the
HJ
anti
Histamine
J
Investig
AllergolClinImmunol, 2006.
23. Swarbrick J, Boylan J C Encyclopedia of Pharmaceutical Technology, 2007.
24. SH Lakade and MR Bhalekar. Formulation and Evaluation of Sustained Release
Matrix Tablet of Anti-Anginal Drug Influence of Combination of Hydrophobic and
Hydrophlic Matrix Former. Research J. Pharm.2008;1(4):410-13.
25. Shanmugam S, Ramya C, Sundaramoorthy K, Ayyappan T, Vetrichelvan T.
Formulation and evaluation of sustained release matrix tablets of Losartan
potassium. IJPRIF 2011;3(1):526-34.
26. Krishnaiah YSR, Karthikeyan RS, Satyanarayana V. A three-layer guar gum matrix
tablet for oral controlled delivery of highly soluble metoprolol tartrate. Int J Pharm
2002; (241): 353-66.
27. Akhlaq M, Majid GK, Abdul W, Abid H, Arshad K, Asif N, Kifayat US.
Formulation and in-vitro evaluation of Flurbiprofen controlled release matrix
Ashutosh Shukla
Page 38
Introduction of dissertation
tablets using cellulose derivative polymers. Pak. J. Pharm 2007-2010;2023(1&2):23-29.
28. Tabandeh H, Mortazavi SA, Guilani TB. Preparation of sustained-release matrix
tablet of asprin with ethyl cellulose, eudragit RS100 and studying the release
profiles and their sensitivity to tablet hardness. Iranian J Pharm Res 2003; 2: 20106.
29. Phani Kumar GK, Gangarao B, LovaRaju NSK. Preparation and evaluation of
sustained release matrix tablets of Lornoxicam using tamarind seed polysaccharide.
IJPRD 2011;2(12):89-98.
30. Yassin EI- Said Hamza, Mona Hassan Aburahma. Design and invitro evalution of
novel sustained- release Double- layer tablets of Lorinoxicam: Utility of
cyclodextrin and xantan gum combination.AAPS pharm sci Tech.2009;10(4):135767.
31. Ravi KN, Narayanaswamy VB, Senthil A, Mehul D, Tejas L, Mahalaxmi R.
Formulation and evaluation of sustained release matrix tablets of Lornoxicam. IndoGlobal Research Journal of Pharmaceutical Sciences 2011;1(3):92-99.
32. Uddin M. Development of sustained release tablet of Valsartan. World J Pharm
Pharma Sci. 2015;3(5):1196-05.
33. Sharma V, Sharma S, Khokra SL, Sahu RKR, Jangde R, Singh J. Formulation,
development and evaluation of Pregabalin Sustained release matrix tablets. Der
Pharmacia Lettre. 2011; 3(5):326-31.
34. Madhavi N, Sudhakar B, Ravikanth PV, Mohon K, Kolapalli RM. Formulation and
Evaluation of Phenytoin Sodium Sustained Release Matrix Tablet. Bioequivalence
and Bioavailability. J Bioequiv Availab. 2012; 4 (7):128-133.
35. Katare VB, Bhutkar MA, Kumbhar AP, Pol SK, Katare PB. Formulation and
Evaluation of Sustained Release Matrix Tablets of Pregabalin. Research Journal of
Pharmacy and Technology. 2013; 6: 1190-94.
36. Ali MS, Singh S, kumar A, singh S, Ansari MT, pattnaik G. Preparation and invitro
evaluation of sustained release matrix tablets of phenytoin sodium using natural
polymers. International Journal of Pharmacy and Pharmaceutical Sciences. 2010;
2(3):174-179.
37. Subramaniam K, Rangasamy M, Kugalur G, Parthiban KN, Senthil NK.
Formulation and evaluation of sustained release tablets of Aceclofenac using
hydrophilic matrix system. IJPRIF 2010;2(3):1775-78.
Ashutosh Shukla
Page 39
Introduction of dissertation
38. Katona, B.W.; Anant, S.; Covey, D.F.; Stenson, W.F. Characterization of
enantiomeric bile acid-induced apoptosis in colon cancer cell lines. J. Biol. Chem.
2009, 284, 3354–3364.
39. Agarwal, D.S.; Mazumdar, S.; Italiya, K.S.; Chitkara, D.; Sakhuja, R. Bile-acidappended triazolyl aryl ketones: Design, synthesis, in vitro anticancer activity and
pharmacokinetics in rats. Molecules 2021, 26, 5741.
40. Melloni, E.; Marchesi, E.; Preti, L.; Casciano, F.; Rimondi, E.; Romani, A.;
Secchiero, P.; Navacchia, M.L.; Perrone, D. Synthesis and biological investigation
of bile acid-paclitaxel hybrids. Molecules 2022, 27, 471.
41. Brossard, D.; El Kihel, L.; Clément, M.; Sebbahi, W.; Khalid, M.; Roussakis, C.;
Rault, S. Synthesis of bile acid derivatives and in vitro cytotoxic activity with proapoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme
(GBM), and colonic carcinoma (HCT-116) human cell lines. Eur. J. Med. Chem.
2010, 45, 2912–2918.
42. Singh, M.; Singh, A.; Kundu, S.; Bansal, S.; Bajaj, A. Deciphering the role of
charge, hydration, and hydrophobicity for cytotoxic activities and membrane
interactions of bile acid based facial amphiphiles. Biochim. Biophys. Acta 2013,
1828, 1926–1937.
43. El Kihel, L.; Clement, M.; Bazin, M.A.; Descamps, G.; Khalid, M.; Rault, S. New
lithocholic and chenodeoxycholic piperazinylcarboxamides with antiproliferative
and pro-apoptotic effects on human cancer cell lines. Bioorg. Med. Chem. 2008,
16, 8737–8744.
44. Singh, M.; Bansal, S.; Kundu, S.; Bhargava, P.; Singh, A.; Motiani, R.K.; Shyam,
R.; Sreekanth, V.; Sengupta, S.; Bajaj, A. Synthesis, structure-activity relationship,
and mechanistic investigation of lithocholic acid amphiphiles for colon cancer
therapy. Medchemcomm 2015, 6, 192–201.
45. Sreekanth, V.; Bansal, S.; Motiani, R.K.; Kundu, S.; Muppu, S.K.; Majumdar, T.D.;
Panjamurthy, K.; Sengupta, S.; Bajaj, A. Design, synthesis, and mechanistic
investigations of bile acid-tamoxifen conjugates for breast cancer therapy.
Bioconjug. Chem. 2013, 24, 1468–1484.
46. Tang, Y.; Blomenkamp, K.S.; Fickert, P.; Trauner, M.; Teckman, J.H. NorUDCA
promotes degradation of α1-antitrypsin mutant Z protein by inducing autophagy
through AMPK/ULK1 pathway. PLoS ONE 2018, 13, e0200897.
Ashutosh Shukla
Page 40
Introduction of dissertation
47. Markov, A.V.; Babich, V.O.; Popadyuk, I.I.; Salomatina, O.V.; Logashenko, E.B.;
Salakhutdinov, N.F.; Zenkova, M.A. Novel derivatives of deoxycholic acid bearing
linear aliphatic diamine and aminoalcohol moieties and their cyclic analogs at the
C3 position: Synthesis and evaluation of their in vitro antitumor potential.
Molecules 2019, 24, 2644.
Ashutosh Shukla
Page 41
Download