You are on page 1of 9

ISSN: 2277- 7695

CODEN Code: PIHNBQ


ZDB-Number: 2663038-2
IC Journal No: 7725

Vol. 1 No. 10 2012


Online Available at: www.thepharmajournal.com

THE PHARMA INNOVATION

Controlled Release Drug Delivery Systems

Debjit Bhowmik 1*, Harish Gopinath 1, B. Pragati Kumar 1, S. Duraivel 1, K. P. Sampath Kumar 2

1. Nimra College of Pharmacy, Vijayawada, Andhra Pradesh, India.


2. Department of pharmaceutical sciences, Coimbatore medical college, Coimbatore

Controlled drug delivery is one which delivers the drug at a predetermined rate, for locally or
systemically, for a specified period of time. Continuous oral delivery of drugs at predictable and
reproducible kinetics for predetermined period throughout the course of GIT. Controlled release drug
delivery employs drug-encapsulating devices from which therapeutic agents may be released at controlled
rates for long periods of time, ranging from days to months. Such systems offer numerous advantages
over traditional methods of drug delivery, including tailoring of drug release rates, protection of fragile
drugs and increased patient comfort and compliance.
Keyword: Controlled Drug Delivery, High Blood Level, Extended Release, Drug Toxicity

INTRODUCTION: Controlled drug delivery higher cost of controlled-release systems


systems can include the maintenance of drug compared with traditional pharmaceutical
levels within a desired range, the need for fewer formulations. The ideal drug delivery system
administrations, optimal use of the drug in should be inert, biocompatible, mechanically
question, and increased patient compliance. strong, comfortable for the patient, capable of
While these advantages can be significant, the achieving high drug loading, safe from accidental
potential disadvantages cannot be ignored like the release, simple to administer and remove, and
possible toxicity or non-biocompatibility of the easy to fabricate and sterilize. The goal of many
materials used, undesirable by-products of of the original controlled-release systems was to
degradation, any surgery required to implant or achieve a delivery profile that would yield a high
remove the system, the chance of patient blood level of the drug over a long period of time.
discomfort from the delivery device, and the With traditional drug delivery systems, the drug
level in the blood follows the in which the level
rises after each administration of the drug and
then decreases until the next administration. The
Corresponding Author’s Contact information: key point with traditional drug administration is
Debjit Bhowmik * that the blood level of the agent should remain
Nimra College of Pharmacy, Andhra Pradesh, India between a maximum value, which may represent
E-mail: debjit_cr@yahoo.com

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 24


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

a toxic level, and a minimum value, below which performance when compared with that presented
the drug is no longer effective. as a conventional dosage form (a solution or a
prompt drug-releasing dosage form). The terms
POLYMER USED IN CONTROL DRUG “controlled release (CR)”, “prolonged release”,
DELIVERY SYSTEM “sustained or slow release (SR)” and “long-acting
Polymers are becoming increasingly important in (LA)” have been used synonymously with
the field of drug delivery. The pharmaceutical “extended release”.
applications of polymers range from their use as
binders in tablets to viscosity and flow Nearly all of the currently marketed monolithic
controlling agents in liquids, suspensions and oral ER dosage forms fall into one of the
emulsions. Polymers can be used as film coatings following two technologies:
to disguise the unpleasant taste of a drug, to 1. Hydrophilic, hydrophobic or inert matrix
enhance drug stability and to modify drug release systems: These consist of a rate controlling
characteristics. The review focuses on the polymer matrix through which the drug is
significance of pharmaceutical polymer for dissolved or dispersed.
controlled drug delivery applications.Sixty 2. Reservoir (coated) systems where drug-
million patients benefit from advanced drug containing core is enclosed within a polymer
delivery systems today, receiving safer and more coating. Depending on the polymer used, two
effective doses of the medicines they need to types of reservoir systems are considered.
fight a variety of human ailments, including (a) Simple diffusion/erosion systems where a
cancer. Controlled Drug Delivery (CDD) occurs drug-containing core is enclosed within
when a polymer, whether natural or synthetic, is hydrophilic and/or water-insoluble polymer
judiciously combined with a drug or other active coatings. Drug release is achieved by diffusion of
agent in such a way that the active agent is the drug through the coating or after the erosion
released from the material in a predesigned of the polymer coating.
manner. The release of the active agent may be (b) Osmotic systems where the drug core is
constant over a long period, it may be cyclic over contained within a semi-permeable polymer
a long period, or it may be triggered by the membrane with a mechanical/laser drilled hole
environment or other external events. In any case, for drug delivery. Drug release is achieved by
the purpose behind controlling the drug delivery osmotic pressure generated within the tablet core.
is to achieve more effective therapies while
eliminating the potential for both under and Advantages and Limitations of Control
overdosing. Release Dosage Forms
Clinical Advantages2, 6
 Reduction in frequency of drug
CONTROL RELEASE DOSAGE FORM administration
The United States Pharmacopoeia (USP) defines1  Improved patient compliance
the modified-release (MR) dosage form as “the  Reduction in drug level fluctuation in
one for which the drug release characteristics of blood
time course and/or location are chosen to  Reduction in total drug usage when
accomplish therapeutic or convenience objectives compared with conventional therapy
not offered by conventional dosage forms such as  Reduction in drug accumulation with
solutions, ointments, or promptly dissolving chronic therapy
dosage forms”. One class of MR dosage form is  Reduction in drug toxicity
an extended-release (ER) dosage form and is (local/systemic)
defined as the one that allows at least a 2-fold  Stabilization of medical condition
reduction in dosing frequency or significant
(because of more uniform drug levels)
increase in patient compliance or therapeutic

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 25


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

 Improvement in bioavailability of some  Encapsulation Dissolution control


drugs because of spatial control  Seed or granule coated
 Economical to the health care providers  Micro encapsulation
and the patient  Matrix Dissolution control
2. Diffusion controlled release7
Commercial / Industrial Advantages  Reservoir type devices
 Illustration of innovative/technological  Matrix type devices
leadership 3. Diffusion and Dissolution controlled
 Product life-cycle extension systems
 Product differentiation 4. Ion exchange resins
 Market expansion 5. Osmotically controlled release
 Patent extension

Potential Limitations
 Delay in onset of drug action BIOPHARMACEUTIC AND
 Possibility of dose dumping in the case of PHARMACOKINETIC ASPECTS IN THE
a poor formulation strategy DESIGN OF CONTROLLED RELEASE PER
 Increased potential for first pass ORAL DRUG DELIVERY SYSTEMS
metabolism
 Greater dependence on GI residence time Controlled release drug delivery systems9, 21 are
of dosage form dosage forms from which the drug is released by
 Possibility of less accurate dose a predetermined rate which is based on a desired
adjustment in some cases therapeutic concentration and the drug’s
 Cost per unit dose is higher when pharmacokinetic characteristics
compared with conventional doses
Biological half-life (t ½ )
 Not all drugs are suitable for formulating
The shorter the t ½ of a drug the larger will be the
into ER dosage form
fluctuations between the maximum steady state
concentration and maximum steady state
Selection of drug for formulation into extended
concentration upon repetitive dosing. Thus drug
release dosage form is the key step. Following
product needs to be administered more
candidates are generally not suitable for ER
frequently.
dosage forms
Minimum effective concentration (MEC)
Characteristics That May Make A Drug
If a minimum effective concentration, MEC is
Unsuitable For Control release Dosage Form
required either frequent dosing of a conventional
 Short elimination half-life3, 4, 5. drug product is necessary or a controlled release
 Long elimination half-life preparation may be chosen.
 Narrow therapeutic index
 Poor absorption Dose size and Extent of duration
 Active absorption The longer the extent of duration the larger the
 Low or slow absorption total dose per unit delivery system needs to be.
 Extensive first pass effect Hence there is a limitation to the amount of drug
that can be practically incorporated into such a
Control release dosage form Release system.
Formulation Designs
1. Dissolution controlled release8

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 26


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

Relatively long t1/2 or fluctuation desired at at various pH values. If the solubility is less than
steady state 0.1 μg/ml (in acidic medium) one may expect
It is the belief of some that neither a SR nor a variable and reduced bioavailability. If the
CRDDS is needed or useful for drugs having a t solubility is less than 0.01 μg/ml absorption and
½ of 12 hours or more. This is not so because availability most likely become dissolution
there are two cases for which a 12 or 24 CRDDS limited dissolution limited. Hence driving force
seems to be indicated: for diffusion may be inadequate.

1. A drug having a t ½ between 12 and 72 It seems that drugs are well absorbed by passive
hours may be designed for a CRDDS diffusion from the small intestine upon per oral
permitting application for every two to administration if at least 0.1 to 1% is non ionised
three days. The decline of the blood level form.
time curve after release of the drug from
the system will depend on the drug’s t ½. Apparent partition coefficient (APC)
Naturally, fluctuation between Css max Drugs being absorbed by passive diffusion must
and Css min may accordingly be have a certain minimal APC. The higher the APC
relatively large in other words on adds in an n-octanol/buffer system the higher is the
slow release to the slow elimination flux across a membrane for many drugs. The
process. APC should be determined for the entire pH
range in the GI tract. The APC must also be
For some drugs having a t1/2 between 20 and 100 applied for partition of the drug between CRDDS
hrs ,and which are intended for long term use and the biological fluid.
,one may desire small fluctuations between peaks
and troughs at steady General absorption mechanism
For a drug to be a variable candidate for per oral
2. states either to achieve a certain CRDDS, its absorption mechanism must be by
therapeutic effect or because the diffusion throughout the entire GI tract. The term
therapeutic range is narrow. diffusion here refers to the dual pathway of
absorption either by partitioning into the lipid
DESIRED BIOPHARMACEUTIC membrane (across the cells) or by passing
CHARACTERISTICS OF DRUG TO through water filled channels (between the cells).
QUALIFY FOR CDDS It is also important that absorption occurs from all
segments of the GI tract which may depend on
Molecular weight or size the drug’s pKa, the pH in the segment, binding of
Small molecules may pass through pores of a drug to mucus, blood flow rate, etc. The
membrane by convective transport. This applies absorption process seems to be highly dependent
to both, the drug release from the dosage form on the hydrodynamics in the GI lumen.
and the transport across a biologic membrane. For
biologic membranes the limit may be a molecular Even though that first order and square root of
weight of 150 and 400 respectively for spherical time release can result in highly effective drug
molecules and chain like compounds delivery systems it is widely believed that the
respectively. ultimate goal is zero order release profile.

Solubility Zero order release invitro release will produce


For all mechanisms of absorption the drug must zero order in vivo release and zero order in vivo
be present at the site of absorption in the form of absorption only if; (1) the entire GI tract behaves
solution. During the Preformulation study it is as a one compartment model, i.e. the various
necessary to determine the solubility of the drug segments throughout the GI tract are

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 27


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

homogeneous with respect to absorption, and (2) concentration as that found in blood. It is the
drug release rate is the rate limiting step in the proportionality constant relating the amount of
absorption process. drug in the body to the measured concentration in
the blood.
With first order release on the other hand, smaller
and smaller amounts are released per unit of time Among the trio CL, Vz, and t ½, the former two
with increasing time. Assuming that rate of parameters are the independent variables and the
absorption gets slower past the small intestine last one is the dependent variable.
due to increased viscosity, decreased mixing, and The Vz or CL is required to predict the
decreased intestinal surface area, less drug is concentration time profile.
absorbed.
Absolute bioavailability (F)
In any case, the drug release from the CRDDS The absolute bioavailability is the percentage of
should not be influenced by pH changes within drug taken up into systemic circulation upon
the GI tract, by enzymes present in the lumen, extravascular administration. For drugs to be
peristalsis, etc suitable for CRDDS one wants an F value to be
close to 100%.
For all practicality, the one compartment open
model is quite suitable to design CRDDS for Intrinsic absorption rate constant (Ka)
most drugs. The intrinsic absorption rate constant of the drug
administered peroral in the form of a solution
Pharmacokinetic parameters should be high, generally by an order of
Elimination half life (t ½) magnitude higher than the desired release rate
Drugs having a t ½ and 8 hours are ideally suited constant of the drug from the dosage form, in
for CRDDS. If the t ½ is less than 1 hour the dose order to insure that release process is the rate
size required to be incorporated for a 12 hour or controlling step.
24 hour duration dosage form may be too large. If
the t ½ is very long there is usually no need for a Therapeutic concentration (Css)
CRDDS, unless it is simply intended for a The therapeutic concentrations are the desired or
reduction in fluctuation of steady state blood target steady state peak concentrations (Css max),
levels. the desired or target steady state minimum
concentrations (Css min), and the mean steady
Total clearance (CL) state concentration (Css avg). The difference
CL is a measure of the volume of distribution between Css max and Css min is the fluctuation.
cleared of drug per unit of time. It is the key The smaller the desired fluctuation the greater
parameter in estimating the required dose rate for must be the precision of the dosage form
CRDDS, and predicting the steady state performance.
concentration.
The lower Css, the smaller Vz, the longer t ½, the
Terminal disposition rate constant (Ke or λz) higher F and The less amount of drug is required
The terminal disposition rate constant or to be incorporated into a CRDDS.
elimination rate constant can be obtained from
the t ½ and is required to predict a blood level
time profile. POLYMERS AS BIOMATERIALS FOR
DELIVERY-SYSTEMS
Apparent volume of distribution (Vz) A range of materials have been employed to
The Vz is the hypothetical volume of a drug control the release of drugs and other active
would occupy if it were dissolved at the same agents. The earliest of these polymers were

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 28


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

originally intended for other, nonbiological uses, Originally, polylactides and polyglycolides were
and were selected because of their desirable used as absorbable suture material, and it was a
physical properties, for example: natural step to work with these polymers in
controlled drug delivery systems. The greatest
 Poly(urethanes) for elasticity. advantage of these degradable polymers is that
 Poly(siloxanes) or silicones for insulating they are broken down into biologically acceptable
ability. molecules that are metabolized and removed from
 Poly(methyl methacrylate) for physical the body via normal metabolic pathways.
strength and transparency. However, biodegradable materials do produce
 Poly(vinyl alcohol) for hydrophilicity and degradation by-products that must be tolerated
strength. with little or no adverse reactions within the
 Poly(ethylene) for toughness and lack of biological environment.
swelling.
 Poly(vinyl pyrrolidone) for suspension These degradation products both desirable and
capabilities. potentially nondesirable must be tested
thoroughly, since there are a number of factors
To be successfully used in controlled drug that will affect the biodegradation of the original
delivery formulations, a material must be materials. The various important factors
chemically inert and free of leachable impurities. indicating the breadth of structural, chemical, and
It must also have an appropriate physical processing properties that can affect
structure, with minimal undesired aging, and be biodegradable drug delivery systems are listed
readily processable. Some of the materials that below:
are currently being used for controlled drug
delivery include  Chemical structure
 Chemical composition
 Poly(2-hydroxy ethyl methacrylate)  Distribution of repeat units in multimers
 Poly(N-vinyl pyrrolidone).  Presence of ionic groups
 Poly(methyl methacrylate).  Presence of unexpected units or chain
 Poly(vinyl alcohol). defects.
 Poly(acrylic acid).  Configuration structure.
 Polyacrylamide.  Molecular weight.
 Poly(ethylene-co-vinyl acetate).  Molecular-weight distribution.
 Poly(ethylene glycol).  Morphology (amorphous/semi crystalline,
 Poly(methacrylic acid). microstructures, residual stresses).
 Presence of low-molecular-weight
However, in recent years additional polymers compounds.
designed primarily for medical applications have  Processing conditions.
entered the arena of controlled release. Many of  Annealing.
these materials are designed to degrade within the  Sterilization process.
body, few of them among these include:  Storage history.
 Shape.
 Polylactides (PLA).  Site of implantation.
 Polyglycolides (PGA).  Adsorbed and absorbed compounds
 Poly(lactide-co-glycolides) (PLGA). (water, lipids, ions, etc.).
 Polyanhydrides.  Physicochemical factors (ion exchange,
 Polyorthoesters. ionic strength, pH).
 Physical factors (shape and size changes,
variations of diffusion coefficients,

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 29


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

mechanical stresses, stress- and solvent- Zero order


induced cracking, etc.). Drug dissolution from pharmaceutical dosage
 Mechanism of hydrolysis (enzymes versus form that doesn’t disaggregates and release the
water). drug slowly can be represented by the following
equation
Controlled drug delivery: Qt  Q0  K 0 t
The most exciting opportunities in controlled
drug delivery lie in the arena of responsive Where
delivery systems, with which it will be possible to Qt =amount of drug released in time t
deliver drugs through implantable devices in Q0= initial amount of drug in solution
K0 =zero order release constant
response to a measured blood level or to deliver a
drug precisely to a targeted site. Much of the
Application:
development of novel materials in controlled drug
This relation can be used to describe the drug
delivery is focusing on the preparation and use of
dissolution of several types of modified release
these responsive polymers with specifically
dosage forms as in the case of transdermal
designed macroscopic and microscopic structural
systems and matrix tablets with low solubility of
and chemical features. Such systems include:
drugs, coated forms, osmotic systems etc
Pharmaceutical dosage forms following this
 Copolymers with desirable
profile release the same amount of drug by unit of
hydrophilic/hydrophobic interactions.
time and it is ideal method of drug release in
 Block or graft copolymers.
order to achieve a pharmacological prolonged
 Complexation networks responding via
action
hydrogen or ionic bonding.
 Dendrimers or star polymers as
nanoparticles for immobilization of First order model
Application of this model to drug dissolution
enzymes, drugs, peptides, or other
study was first proposed by Gibaldi and Feldman
biological agents.
(1967) later by Wagner (1969)
 New biodegradable polymers.
In this model the decimal logarithm of amount
 New blends of hydrocolloids and
remained VS time will be linear. It indicates first
carbohydrate-based polymers.
order release and expressed by following
equation
MODELLING AND COMPARISON OF
log Qt=logQe+(Ki.t/2.303)
DISSOLUTION PROFILE
Several theories and kinetic 10, 11, 12, 13models Qt = amount of drug released in time t
were described the drug release characteristics of Qe = initial amount f drug in solution
immediate release and modified release dosage Ki = first order release constant
forms, by using dissolution data and quantitative
interpretation of values obtained in dissolution Higuchi model
assay if facilitated by the usage of the generic Higuchi in (1961, 1963) developed several
equation dosage form that mathematically theoretical models to study the release of water
translates the dissolution curve in function of soluble and low soluble drugs incorporated in
some parameters related with pharmaceutical solid matrices; mathematical expressions were
dosage form. obtained for drug particles dispersed in a uniform
In the present work, some analytical models were matrix behaving as diffusion media and this
used to study the mechanism of drug release of model describes the drug release characteristics
extended release by following models as diffusion process based on fick’s law related

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 30


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

with square root of time dependent and it


expressed by using the formula W01 / 3  Wt1 / 3  K s t
Qt=KH√t Where
W0 is the initial amount of drug in the pharmaceutical
Where dosage form
Qt = amount of drug released in time t Wt is the remaining amount of drug pharmaceutical dosage
KH=Higuchi Constant form at time t
√t =dependent square root of time Ks is the constant incorporating the surface volume
relation.
Application: Higuchi model can be used to
describe the drug dissolution of several types of
modified release dosage forms as in the case of
transdermal systems and matrix tablets with low
solubility of drugs

Korsmayer’s and Peppa’s model


Korsmayer’s and Peppa’s in 1983 developed a
simple empirical model relating exponentially the
drug release to the elapsed time by using ‘n’
values, in order to characterize several release SUSTAIN RELEASE DRUG DELIVERY
mechanisms . Under some experimental SYSTEM
conditions the release mechanism deviate from
the fick’s equations following an anomalous There are certain considerations for the formation
behaviour in this case it should be expressed by of sustained release formulation:
the following equation
 If the active compound has a long half-life
(over 6 hours), it is sustained on its own.
log(mt m f )  log K  n. log t  If the pharmacological activity of the
active compound is not related to its blood
levels, time releasing then has no purpose.
Where
mt=amount of drug released at time t
 If the absorption of the active compound
mf=amount of drug released at infinite time t involves an active transport, the
K=release rate constant development of a time-release product
n= diffusion expression (drug release mechanism) may be problematic.
 Finally, if the active compound has a short
half-life, it would require a large amount
to maintain a prolonged effective dose. In
Application This model generally used to this case, a broad therapeutic window is
analyze the release of pharmaceutical polymeric necessary to avoid toxicity; otherwise, the
dosage form, when the release mechanism is not risk is unwarranted and another mode of
well known or when more than one type release administration would be recommended.
mechanism could be involved.
The difference between controlled release and
Hixson-Crowell model sustained release is that controlled release is a
Hixson-Crowell (1931) recognising that particle perfectly zero order release; that is, the drug
regular area is proportional to the cubic root of its releases over time irrespective of concentration.
volume derived an equation that can be described Sustained release implies slow release of the drug
in the following manner:

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 31


“THE PHARMA INNOVATION” JOURNAL www.thepharmajournal.com

over a time period. It may or may not be


controlled release.

CONLUSION
The best new therapeutic entity in the world is of
little value without an appropriate delivery
system. Tablet delivery system can range from
simple immediate release formulations to
complex extended or modified release dosage
forms. The most important role of drug delivery
system is to get the drug delivered to the site of
action in sufficient amount & at the appropriate
rate. However it should meet other important
criteria such as physical & chemical stability,
ability to be mass-produced in a manner that
assures content uniformity.

REFERENCE
3. Howard C. Ansel, NIcholos G. Popvich,
lyold V. Allen , pharmaceutical dosage
forms and Drug Delivery system. 1st ed.;
1995.p.78.
4. Jain N.K and Sharma S.N. A text book of
professional pharmacy. 1st ed.; 1995.p.78.
5. Samuel Harder and GlennV. Buskirk.
Pilot Plant Scale-Up Techniques. In The
Theory and Practice of Industrial
Pharmacy. 3rd ed., 1991, p. 687-702
6. Remington, “The Science and Practice of
pharmacy”, 20 th Edn, vol.I, pg.no.903-
913.
7. Lachman et al Theory and Practice of
Industrial Pharmacy. 3rd ed Philadelphia,
1991, p. 303-314.
8. Lachman et al Theory and Practice of
Industrial Pharmacy. 3rd ed Philadelphia,
1991, p. 314-317.
9. Lachman, L.; Lieberman, H. A.; Kanig J.
L. Eds.; Theory and Practice of Industrial
Pharmacy. 3rd ed Philadelphia, 1991, p.
346-373.
10. Lutfi Genç, Hadi Bilaç and Erden Güler
Preparation of controlled release dosage
forms of diphenhydramine was prepared
with different polymers. International
Journal of Pharmaceutics, Volume 169,
Issue 2, 15 July 1998, Pages 232-235.

Vol. 1 No. 10 2012 www.thepharmajournal.com Page | 32

You might also like