+ All Categories
Home > Documents > Drug Delivery Systems a Review

Drug Delivery Systems a Review

Date post: 16-Apr-2015
Category:
Upload: abhijitgothoskar6039
View: 345 times
Download: 14 times
Share this document with a friend
416
Drug Delivery Systems: A Review Editor A V Gothoskar
Transcript
  • Drug Delivery Systems:

    A Review

    Editor

    A V Gothoskar

  • Dedicated to pharmacy profession

  • Acknowledgement:

    SCES's Indira College of

    Pharmacy, Pune

  • PREFACE

    Men and medicine are inseparable from times immemorial. Although the physical forms of medication have not

    changed dramatically, the attitude of the public toward accepting medicines have changed with the passage of time.

    This fact is also reflected in the strategies adopted by the pharmaceutical companies in the field of research. The cost

    involved, both in terms of time and money, has made it mandatory for the companies to reconsider their research

    focus. In an attempt to reduce the cost of drug development process and advantageously reap the benefits of the

    patent regime, drug delivery systems have become an integral part of the said process.

    Drug delivery system is a dosage form, containing an element that exhibits temporal and/or spatial control over the

    drug release. The ultimate aim of such systems is tailoring of the drug formulation to individual requirements under

    the control of pathophysiological or in-vivo conditions rather than in-vitro characteristics.

    This field of drug delivery systems is dynamic and extensive. Probably it would need an encyclopedia to cover all

    the types of drug delivery systems. The aim of this book is to compile major drug delivery systems and offer a

    source of information for all those working in pharmaceutical academia as well as industry.

    The book is made available free of charge to all who are interested in the subject for dissemination of knowledge.

    Authors feel proud to be a part of first of its kind of experiment wherein a technical book is offered for free

    download through a blog.

    We welcome suggestions and criticisms for our readers.

    A V Gothoskar

    PhD, MBA

  • Contributors

    Bajaj Amruta Basrur Pooja Bhuruk Manisha

    B.Pharm B.Pharm B.pharm, D.Pharm

    Chavan Shankar Deshpande Tanvee Gothoskar Abhijit

    B.Pharm B.Pharm PhD, MBA

    Hastak Vishakha Kamble Pranay Katedeshmukh Ramesh

    M.Pharm (Pharmaceutics) B.Pharm M.Pharm (Pharmaceutics)

  • Khan Halimunnisa Kulkarni Akshada Maravaniya Pathik Kumar

    B.Pharm B.Pharm B.Pharm

    Mogal Rajendra Patel Ruchita Pawar Sandesh M.Pharm (Pharmaceutics) B.Pharm B.Pharm, D.Pharm

    Pawar Yogesh Satam Madhavi Sawant Sandip

    M.Pharm (Pharmaceutics) B.Pharm B.Pharm

  • Shaikh Amir Shinde Rohit Suryavanshi Kiran

    M.Pharm (Pharmaceutics) B.Pharm, D.Pharm B.Pharm, D.Pharm

    Wayal Abhijit Zarikar Nitin

    B.Pharm B.Pharm

  • Table of contents

    1. Fundamentals of Drug Delivery System - 10

    Suryavanshi Kiran, Mogal Rajendra, Pawar Yogesh, Shaikh Aamir

    2. Oral Controlled Drug Delivery System - 18

    Bajaj Amruta, Katedeshmukh Ramesh

    3. Gastroretentive Drug Delivery System - 43

    Basrur Pooja, Hastak Vishakha

    4. Colon Specific Drug Delivery System - 59

    Bhuruk Manisha, Pawar Yogesh

    5. Chronopharmaceutical Drug Delivery System - 83

    Chavan Shankar, Shaikh Aamir

    6. Self Dispersing Formulations-101

    Deshpande Tanvee, Mogal Rajendra

    7. Introduction To Bioadhesion/Mucoadhesion - 114

    Kamble Pranay, Katedeshmukh Ramesh

    8. Mucoadhesive Drug Delivery System - Nasal - 127

    Khan Halimunnisa, Hastak Vishakha

    9. Mucoadhesive Drug Delivery System - Rectal - 149

    Kulkarni Akshasa, Pawar Yogesh

    10. Mucoadhesive Drug Delivery System - Vaginal - 160

    Deshpande Tanvee, Shinde Rohit, Pawar Yogesh

    11. Parenteral Controlled Drug Delivery System 182

    Maravaniya Pathikkumar, Shaikh Aamir

  • 12. Parenteral Implants 194

    Patel Ruchita, Mogal Rajendra

    13. Transdermal Drug Delivery System - 206

    Pawar Sandesh, Katedeshmukh Ramesh

    14. Particulate Drug Delivery System-Liposomes - 224

    Satam Madhavi, Hastak Vishakha

    15. Particulate Drug Delivery System- Microcapsules 241

    Sawant Sandip, Pawar Yogesh

    16. Particulate Drug Delivery System- Microspheres -253

    Sawant Sandip, Pawar Yogesh

    17. Particulate Drug Delivery System-Resealed Erythrocytes-266

    Shinde Rohit, Shaikh Aamir

    18. Particulate Drug Delivery System-Monoclonal Antibodies -281

    Suryavanshi Kiran, Mogal Rajendra

    19. Intranasal Drug Delivery System - 291

    Wayal Abhijit, Katedeshmukh Ramesh

    20. Protein And Peptide Drug Delivery System - 302

    Zarikar Nitin, Hastak Vishakha

    21. Intraocular Drug Delivery System - 318

    Maravaniya Pathikkumar , Zarikar Nitin , Pawar Yogesh

    22. Pulmonary Drug Delivery System 326

    Kamble Pranay, Suryavanshi Kiran, Shaikh Aamir

    23. Nanopharmaceuticals 334

    Kulkarni Akshada, Patel Ruchita, Mogal Rajendra

  • 24. Medicated Chewing Gums 347

    Basrur Pooja, Katedeshmukh Ramesh

    25. Oral Thin Film 357

    Bhuruk Manisha, Satam Madhavi, Hastak Vishakha

    26. Nail Drug Delivery System 367

    Basrur Pooja, Suryavanshi Kiran, Katedeshmukh Ramesh

    27. Regulatory Aspects of Drug Delivery System- 377

    Chavan Shankar, Mogal Rajendra, Pawar Yogesh, Shaikh Aamir

  • Drug Delivery Systems - A Review

    10

    FUNDAMENTALS OF DRUG DELIVERY SYSTEMS Suryavanshi Kiran, Mogal Rajendra, Pawar Yogesh, Shaikh Aamir

    Need for Controlled Release Systems:

    (Kathryn E. Uhrich 1999)Controlled drug

    delivery technology represents one of the most

    rapidly advancing areas of science in which

    chemists and chemical engineers are contributing to

    human health care. Such delivery systems offer

    numerous advantages compared to conventional

    dosage forms including improved efficacy, reduced

    toxicity, and improved patient compliance and

    convenience. Such systems often use synthetic

    polymers as carriers for the drugs. By so doing,

    treatments that would not otherwise be possible are

    now in conventional use. Although the introduction

    of the first clinical controlled release systems

    occurred less than 25 years ago, 1997 sales of

    advanced drug delivery systems in the United

    States alone were approximately $14 billion

    dollars. Synthetic polymers used in the controlled

    release of drugs. Before considering the variety and

    the evolution of these polymeric structures, it is

    necessary to examine the motivation for achieving

    controlled release. This field of pharmaceutical

    technology has grown and diversified rapidly in

    recent years. Understanding the derivation of the

    methods of controlled release and the range of new

    polymers can be a barrier to involvement from the

    nonspecialist. All controlled release systems aim to

    improve the effectiveness of drug therapy. This

    improvement can take the form of increasing

    therapeutic activity compared to the intensity of

    side effects, reducing the number of drug

    administrations required during treatment, or

    eliminating the need for specialized drug

    administration (e.g., repeated injections).

    B. Methods of Controlled Release

    In temporal control, drug delivery systems aim to

    deliver the drug over an extended duration or at as

    specific time during treatment.

    Controlled release over an extended duration is

    highly beneficial for drugs that are rapidly

    metabolized and eliminated from the body after

    administration. An example of this benefit is shown

    schematically in Figure 1 in which the

    concentration of drug at the site of activity within

    the body is compared after immediate release from

    4 injections administered at 6 hourly intervals and

    after extended release from a controlled release

    system. Drug concentrations may fluctuate widely

    during the 24 h period when the drug is

    administered via bolus injection, and for only a

    portion of the treatment period is the drug

    concentration in the therapeutic window (i.e., the

    drug concentration that produces beneficial effects

    without harmful side effects). With the controlled

    release system, the rate of drug release matches the

    rate of drug elimination and, therefore, the drug

    concentration is within the therapeutic window for

    the vast majority of the 24 h period. Clinically,

    temporal control can produce a significant

    improvement in drug therapy. For example, when

    an opioids pain killer is administered to a patient

    with terminal cancer, any time that the drug

    concentration is below therapeutic concentrations

    the patient experiences pain. A temporally

    controlled release system would ensure that the

    maximum possible benefit is derived from the drug.

    In distribution control, drug delivery systems aim

    to target the release of the drug to the precise site of

    activity within the body. The benefit of this type of

    control is shown schematically in Figure 2 in which

    Figure 1. Drug concentrations at site of

    therapeutic action after delivery as a conventional

    injection (thin line) and as a temporal controlled

    release system (bold line). (Kathryn E. Uhrich

    1999)

  • Drug Delivery Systems - A Review

    11

    Figure 2. Drug delivery from an ideal distribution

    controlled release system. Bold line: Drug

    concentrations at site of therapeutic action. Thin

    line: Systemic levels at which side effects occur.

    (Kathryn E. Uhrich 1999)

    Drug concentrations at the site of activity and side

    effect production are compared. There are two

    principle situations in which distribution control

    can be beneficial. The first is when the natural

    distribution causes drug molecules to encounter

    tissues and cause major side effects that prohibit

    further treatment. This situation is often the cause

    of chemotherapy failure when bone marrow cell

    death prevents the patient from undergoing a

    complete drug treatment. The second situation is

    when the natural distribution of the drug does not

    allow drug molecules to reach their molecular site

    of action. For example, a drug molecule that acts

    on a receptor in the brain will not be active if it is

    distributed by the patients blood system but cannot

    cross the blood-brain barrier. A large number of

    classes of drugs can benefit from temporal or

    distribution controlled release. These classes

    include chemotherapeutic drugs,

    immunosuppressants, anti-inflammatory agents,

    Antibiotics, opioid antagonists, steroids, hormones,

    anesthetics, and vaccines. Recently, the need to

    develop new controlled release strategies has been

    intensified by advances in the design of peptide

    drugs and emergence of gene therapy. These

    biotechnology derived agents may dominate the

    next generation of drug design. However, their

    clinical success may be dependent on the design of

    controlled release devices that ensure that the drugs

    reach their target cells precisely at the required

    time. A discussion of the pharmacological and

    clinical motivations for controlling the release of

    the specific drug classes referred to above is

    beyond the limit of this article; however, a number

    of excellent reviews are available. In addition, it

    should be noted that controlled release technology

    is not confined to pharmaceutical applications but

    has also proven beneficial in agricultural and

    cosmetic industries. (Kathryn E. Uhrich 1999)

    Scope of Polymer Systems:

    In this review, a number of polymer

    backbones that are potentially degradable are

    detailed in the text. This restriction certainly does

    not reduce the impact and significance of C-C

    backbones for controlled release applications but is

    simply a mechanism to focus on an important

    subset of materials. To illustrate the diverse range

    of functionalities available from nonbiodegradable

    systems based on C-C backbones to heteroatom-

    containing polymer backbones that may confer

    biodegradability. (Langer 1998)

    Mechanisms of Controlled Drug Release Using

    Polymers:

    A diverse range of mechanisms have

    been developed to achieve both temporal and

    distribution controlled release of drugs using

    polymers. This diversity is a necessary

    consequence of different drugs imposing various

    restrictions on the type of delivery system

    employed. For example, a drug that is to be

    released over an extended period in a patients

    stomach where the pH is acidic and environmental

    conditions fluctuate widely will require a controlled

    release system very different from that of a drug

    that is to be delivered in a pulsatile manner within

    the blood system. An important consideration in

    designing polymers for any controlled release

    mechanism is the fate of the polymer after drug

    release. Polymers that are naturally excreted from

    the body are desirable for many controlled release

    applications. These polymers may be excreted

    directly via the kidneys or may be biodegraded into

    smaller molecules that are then excreted.

    Nondegradable polymers are acceptable in

    applications in which the delivery system can be

    recovered after drug release (e.g., removal of patch

    or insert) or for oral applications in which the

    polymer passes through the gastrointestinal tract.

    From a polymer chemistry perspective, it is

    important to appreciate that different mechanisms

    of controlled release require polymers with a

    variety of physicochemical properties. This

    requirement has stimulated the evolution of the

    new polymers that will be discussed in section IV.

    Before consideration of these polymers, the major

    mechanisms of controlled release and polymeric

    characteristics that are required to carry out these

    mechanisms will be briefly. (Kathryn E. Uhrich

    1999)

  • Drug Delivery Systems - A Review

    12

    CLASSIFICATION OF DRUG

    DELIVERY SYSTEM:

    Classification of NDDS based on Physical means

    1) Osmotic Pressure Activated

    2) Hydrodynamic pressure activated

    3) Vapor pressure activated

    4) Mechanically activated

    5) Magnetically activated

    6) Sonophoresis

    7) Iontophoresis

    8) Hydration activated

    Classification of NDDS based on Chemical means

    1) Hydrolysis activated

    2) Ion activated

    3) pH activated

    Polymers Generally Used for Controlled Drug

    Delivery System:

    1) Poly(esters):

    Poly (esters) is the best characterized and

    most widely studied biodegradable system. The

    synthesis of poly (esters) has received as much

    attention as the degradation of these materials. A

    patent for the use of poly (lactic acid) (PLA) as a

    resorbable suture material was first filed in 1967.34

    The mechanism of degradation in poly (ester)

    materials is classified as bulk degradation with

    random hydrolytic scission of the polymer

    backbone

    Polymerization of the cyclic lactone

    alone is usually too slow to produce high molecular

    weight material (>20 000 amu). The rate of ring

    opening for the cyclic lactone can be increased by

    activation of a Zn- or Snbased catalyst with the

    carbonyl ester. However, the introduction of a

    catalyst invites concerns over traces of potentially

    cytotoxic material. Thus, stannous octoate SnII

    (CO2CH(nBu)(Et))2 is commonly used because

    has FDA approval as a food stabilizer.

    Fig:3 Ring- opening polymerization of selected

    cyclic lactones to give the following

    A) Poly(e-caprolactone)PCL

    B) Poly(glycolic acid) PGA

    C) Poly(L-lactic acid)PLA (Kathryn E.

    Uhrich 1999)

    1. Poly(lactic acid), Poly(glycolic acid), and Their

    Copolymers Poly(esters) based on poly(lactic acid)

    (PLA), poly- (glycolic acid) (PGA), and their

    copolymers, poly(lactic acid-co-glycolic acid)

    (PLGA), are some of the best defined biomaterials

    with regard to design and performance. Lactic acid

    contains an asymmetric R-carbon which is typically

    described as the D or L form in classical stereo

    chemical terms and sometimes as the R and S form,

    respectively. For homopolymers, the enantiomer

    forms are poly (D-lactic acid) (PDLA) and poly (L-

    lactic acid) (PLLA). The physicochemical

    properties of optically active PDLA and PLLA are

    nearly the same, whereas the racemic PLA has very

    different characteristics.41 For example, racemic

    PLA and PLLA have Tgs of 57 and 56 C,

    respectively, but PLLA is highly crystalline with a

    Tm of 170 C and racemic PLA is completely

    amorphous.

    Because the naturally occurring lactic acid

    is L (or S), PLLA is considered more

    biocompatible. The polymers are derived from

    monomers that are natural metabolites of the body;

    thus degradation of these materials yields the

    corresponding hydroxy acid, making them safe for

    in vivo use. Biocompatibility of the monomer is the

    foundation for biocompatibility of degradable

    polymer systems. To this end, the degradation

    products often define the biocompatibility of a

    polymers not necessarily the polymer itself. Even

    though PLGA is extensively used and represents

    the gold standard of degradable polymers,

    increased local acidity due to the degradation can

    lead to irritation at the site of the polymer

  • Drug Delivery Systems - A Review

    13

    employment. Introduction of basic salts has been

    investigated as a technique to control the pH in

    local environment of PLGA implants

    From a physical level of understanding,

    poly (esters) undergo bulk degradation. PLA

    homopolymers degrade slower than PGA

    homopolymers on the basis of crystallinity as well

    as stearic inhibition by the pendent methyl group of

    PLA to hydrolytic attack. However, the complexity

    of PLA, PGA, and PLGA degradation has been

    demonstrated by Vert45 and does not conform to a

    simple model. Vert and coworkers have

    demonstrated that size dependence for hydrolytic

    degradation exists for PLA systems. Other research

    efforts suggest that PLA-derived micro particles

    will degrade faster than nanoparticles derived from

    PLA. This is modeled on diffusion reaction

    phenomena. An autocatalytic effect at the interior

    of larger devices is thought to contribute to the

    initial heterogeneous degradation of larger devices

    as acidic byproducts cannot readily diffuse out

    from the interior as is the case for smaller

    constructs. Extensive degradation studies have also

    been reported for PLA, poly (caprolactone) (PCL),

    and their copolymers both in vitro and in vivo.

    Studies in hydrolytic degradation for poly (esters)

    have focused on understanding the effects of

    changes in polymer chain composition. A

    distinguishable effect based on end group

    composition for poly (ester) degradation

    demonstrated that terminal carboxyl groups have a

    catalytic effect on hydrolysis for PGA. The ability

    to tailor rates of protein release from PLGA

    microspheres was derived from the understanding

    of end-group effects. The commercial

    developmental process for formulating poly (esters)

    with selected drug candidates has been reviewed.

    The aforementioned review highlights the

    development of poly (ester) matrices containing

    human growth hormone that sustained levels of a

    therapeutic protein in humans for 1 month from a

    single dose. (Kathryn E. Uhrich 1999)

    2. Poly (ethylene glycol) Block Copolymers:

    Poly (ethylene glycol) (PEG) is also

    referred to as poly (ethylene oxide) (PEO) at high

    molecular weights. Biocompatibility is one of the

    most noted advantages of this material. Typically,

    PEG with molecular weights of 4000 amu is 98%

    excreted in man. One of the emerging uses for

    inclusion of PEG in a controlled release system

    arises from its protein resistivity. The hydrophilic

    nature of PEG is such that water hydrogen bonds

    tightly with the polymer chain and thus excludes,

    or inhibits, protein adsorption. Many research

    groups are investigating attachment of PEG chains

    to therapeutic proteins; PEG chains at the surface

    allow for longer circulation of the protein in the

    body by prolonging biological events such as

    endocytosis, phagocytosis, liver uptake and

    clearance, and other adsorptive processes.

    Fig.4 Synthesis of PLA-PEG Copolymer (Kathryn

    E. Uhrich 1999)

    PEG can be made with a range of terminal

    functionalities which lends to its easy incorporation

    into copolymer systems. PEG is commonly

    terminated with chain-end hydroxyl groups which

    provide a ready handle for synthetic modification.

    Diblock PLA/ PEG and triblock PLA/PEG/PLA

    systems have been synthesized and characterized

    with various PLA contents. The free hydroxyl

    groups of PEG are ring-opening initiators for

    lactide in forming the diblock or triblock materials

    (Figure 5a, b). Recently, Chen et al. have

    synthesized PLA-PEG multiblock copolymers from

    L-lactide and ethylene oxide, the monomer

    precursors for PLA and PEG, respectively (Figure

    5c). This approach is different in two respects: (i)

    use of bimetallic catalysts which proceed by

    anionic mechanisms; (ii) multiblock polymers are

    generated. Han and Hubbell further demonstrated

    the synthetic utility for PLA-PEG systems by

    introducing acrylate moieties to form cross-linked

    systems. Similarly, Jeong et al. prepared thermo

    sensitive PLA-PEO hydrogels that exhibit

    temperature-dependent gel-sol transition for use as

    injectable drug delivery systems.

    Poly (ortho esters):

    The motivation for designing poly (ortho

    esters) for drug delivery was the need to develop

    biodegradable polymers that inhibited drug release

  • Drug Delivery Systems - A Review

    14

    by diffusion mechanisms and allowed drug release

    only after the hydrolysis of polymer chains at the

    surface of the device.70 Most research on poly

    (ortho esters) has focused on the synthesis of

    polymers by the addition of polyols to diketene

    acetals. For example, Heller et al. have described

    the synthesis and application of the 3, 9-

    diethylidene-2, 4, 8, 10-tetraoxaspiro [5.5]

    undecane (DETOSU)-based poly (ortho esters).71

    The basic structure is formed by the addition of the

    DETOSUmonomer to a diol to form the chemical

    structure. The DETOSU-based poly (ortho esters)

    contain acid labile ortho ester linkages in their

    backbone structure. Within aqueous environments,

    the ortho ester groups are hydrolyzed to form

    Pentaerythritol dipropionate and diol monomers as

    breakdown products. The Pentaerythritol

    dipropionate is further hydrolyzed to

    Pentaerythritol and acetic acid. Acid-catalyzed

    hydrolysis of these polymers can be controlled by

    introducing acidic or basic Excipients into

    matrixes. Rates of hydrolysis can be increased by

    the addition of acidic excipients, such as suberic

    acid, as demonstrated by the zero-order release of

    5-fluorouracil over a 15 day period.72

    Alternatively, basic excipients stabilize the bulk of

    the matrix but diffuse out of the surface region,

    thereby facilitating surface-only erosion. This

    approach has been employed in the temporal

    controlled release of tetracycline over a period of

    weeks in the treatment of periodontal disease.

    Fig.5: Degradation of 3, 9(bis ethylidene-2, 4,8,10

    Tetraoxaspiro undecane (DETOSU) based poly

    ortho ester (Kathryn E. Uhrich 1999)

    A useful feature of the DETOSU systems is

    the ability to control the mechanical properties by

    changing the diol monomer ratios within the final

    polymeric structure. For example, Heller et al. have

    shown that the glass transition temperature of

    polymers containing a rigid diol monomer

    (transcyclohexanedimethanol) and a flexible

    monomer (1, 6- hexanediol) could be varied

    between 20 and 105 by increasing the proportion

    of the rigid diol. This control can also be achieved

    with the glycolide containing polymers.

    A number of applications have been

    described for cross-linked poly (ortho esters)

    formed by the substitution of 1, 2, 6-hexanetriol for

    1, 2-hexanediol, for example. The triol monomer

    allows cross-linked materials to be formed that are

    semisolid materials. It has been envisaged that

    these materials could be injected into the patient as

    a viscous liquid at slightly elevated temperatures

    that form nondeformable depot implants upon

    cooling. (V. Balmurlidhara 2011)

    Poly (anhydrides)

    To obtain a device that erodes

    heterogeneously, the polymer should be

    hydrophobic yet contain water sensitive linkages.

    One type of polymer system that meets this

    requirement is the poly (anhydrides). Poly-

    (anhydrides) undergoes hydrolytic bond cleavage

    to form water-soluble degradation products that can

    dissolve in an aqueous environment, thus resulting

    in polymer erosion. Poly (anhydrides) are believed

    to undergo predominantly surface erosion due to

    the high water liability of the anhydride bonds on

    the surface and the hydrophobicity which prevents

    water penetration into the bulk. This process is

    similar to the slow disappearance of a bar of soap

    over time. The decrease in the device thickness

    throughout the erosion process, maintenance of the

    structural integrity, and the nearly zero-order

    degradation kinetics suggest that heterogeneous

    surface erosion predominates. The majority of poly

    (anhydrides) are prepared by melt-condensation

    polymerization. Starting with a dicarboxylic acid

    monomer, a prepolymer of a mixed anhydride is

    formed with acetic anhydride. The final polymer is

    obtained by heating the prepolymer under vacuum

    to remove the acetic anhydride byproduct. The

    most widely studied poly (anhydrides) are based on

    sebacic acid (SA), p-(carboxyphenoxy) propane

    (CPP), and p-(carboxyphenoxy) hexane (CPH)

    Degradation rates of these polymers can be

    controlled by variations in polymer composition.

    The more hydrophobic the monomer, the more

    stable the anhydride bond is to hydrolysis.

    Aliphatic poly- (anhydrides) (e.g., SA) degrade

    within days whereas aromatic poly (anhydrides)

    (e.g., CPH) degrade over several years.

  • Drug Delivery Systems - A Review

    15

    Fig: 6 Structure of widely used aromatic poly

    (anhydrides) based on monomer of p-carboxy

    phenoxy propane (Kathryn E. Uhrich 1999)

    The biocompatibility of copolymers of SA

    and CPP has been well established. Evaluation of

    the toxicity of poly (anhydrides) show that they

    possess excellent in vivo biocompatibility.81

    Recent clinical trials have demonstrated that an

    intracranial device of SA/CPP copolymers

    improves the therapeutic efficacy of an antitumor

    agent, bischloronitrosourea, for patients suffering

    from a lethal type of brain cancer.

    Poly (anhydride-esters)

    Other modifications of poly (anhydrides)

    include poly (anhydride-esters), which include two

    different types of hydrolytically cleavable bonds in

    the polymer backbone. In one example, low

    molecular weight carboxylic acid-terminated

    prepolymer of poly (_- caprolactone) were coupled

    via anhydride linkages. The intent of this research

    was to design polymers that displayed two-stage

    degradation profiles: anhydride bonds rapidly

    hydrolyzed to the poly (ester) prepolymer which

    degraded much more slowly. In another example,

    carboxylic acid-terminated monomers that contain

    ester bonds are activated and then polymerized

    using the same chemistry described for the poly

    (anhydrides). A unique aspect of these poly

    (anhydride-esters) is that hydrolytic degradation of

    the polymer backbone yields a therapeutically

    useful compound, salicylic acid. Polymers

    degradation products are potentially beneficial

    Fig: 7 Poly (anhydride ester that undergo into

    salicylic acid, an anti inflammatory agent (Kathryn

    E. Uhrich 1999)

    Poly (amides):

    The most interesting class of poly

    (amides) for controlled release are the poly (amino

    acids). The synthetic ability to manipulate amino

    acid sequences has seen its maturity over the last

    two decades with new techniques and strategies

    continually being introduced. An excellent review

    of the histo4ry of amino acid-derived polymers is

    given by Nathan and Kohn.93 Poly(amino acids)

    have been used predominantly to deliver low

    molecular weight drugs, are usually tolerated when

    implanted in animals,94 and are metabolized to

    relatively nontoxic products. These results suggest

    good biocompatibility, but their mildly antigenic

    nature makes their widespread use uncertain.

    Another concern with poly (amino acids) is the

    intrinsic hydrolytic stability of the amide bond

    which must rely upon enzymes for bond cleavage.

    The dependence on enzymes generally results in

    poor controlled release in vivo. The expense and

    difficulty in production of elaborate polypeptide

    sequences has limited the composition to

    homopolymers, predominantly poly (glutamic acid)

    and poly (aspartic acid). Poly(amino acids) are

    generally hydrophilic with degradation rates

    dependent upon hydrophilicity of the amino

    acids.96,97 Amino acids are attractive due to the

    functionality they can provide a polymer. For

    example, poly (lactic acid-co lysine) (PLAL) was

    synthesized using a stannous octoate catalyst from

    lactide and a lysine-containing monomer analogous

    to lactide. Inclusion of the amino acid lysine

    provides an amino group that allows for further

    modification of the PLAL system. Recently,

    peptide sequences that promote cell adhesion have

    been attached to PLAL.

  • Drug Delivery Systems - A Review

    16

    Fig.8 Poly (lactic acid-co-amino acid) PLAL

    Polymer system (Kathryn E. Uhrich 1999)

    Currently marketed oral controlled-release

    systems:

    Advances in oral controlled-release

    technology are attributed to the development of

    novel biocompatible polymers and machineries that

    allow preparation of novel design dosage forms in a

    reproducible manner. The main oral drug-delivery

    approaches that have survived through the ages are

    as follows:

    Coating technology using various polymers for

    coating tablets, nonpareil sugar beads, and granules

    Matrix systems made of swell able or

    nonswellable polymers

    Slowly eroding devices

    Osmotically controlled devices.

    Conventional tablet formulations are still popular in

    the design of single-unit, matrix-type controlled

    release dosage forms. The advancement of

    granulation technology and the array of polymers

    available with various physicochemical properties

    (such as modified cellulose or starch derivatives)

    have made the development of novel oral

    controlled release systems possible. Matrix devices

    made with cellulose or acrylic acid derivatives,

    which release the homogeneously dispersed drug

    based on the penetration of water through the

    matrix, have gained steady popularity because of

    their simplicity in design. The drawback of matrix-

    type delivery systems is their first-order drug

    delivery mechanism caused by changing surface

    area and drug diffusional path length with time.

    This drawback has been addressed by osmotic

    delivery systems, which maintain a zero-order drug

    release irrespective of the pH and hydrodynamics

    of the GI tract. Multiparticulate systems are gaining

    favor over single-unit dosage forms because of

    their desirable distribution characteristics,

    reproducible transit time, and reduced chance of

    gastric irritation owing to the localization of drug

    delivery.

    Although several technologies for the production of

    microparticulate systems have been designed, thus

    far the mainstream technologies are still based on

    spray-drying, spheronization, and film-coating

    technology.

    FDA regulation of oral Controlled-

    release drugs:

    In the 1980s, FDA introduced rigorous

    regulations governing bioequivalence and in vitro

    in vivo correlations for controlled-release products.

    Required pharmacokinetic evaluations involve

    relative bioavailability following single dose

    relative bioavailability following multiple doses

    effect of food

    dose proportionality

    unit dosage strength proportionality

    single-dose bioequivalence study

    (Experimental versus marketed formulations at

    various strengths)

    In vivoin vitro correlation

    Pharmacokinetic/pharmacodynamic (PK/PD)

    relationship.

    In general, for drugs in which the exposure

    response relationship has not been established or is

    unknown, applications for changing the

    formulation from immediate release to controlled

    release requires demonstration of the safety and

    efficacy of the product in the target patient

    population. When an NME is developed as a

    controlled-release dosage form, additional studies

    to characterize its absorption, distribution,

    metabolism, and excretion (ADME) characteristics

    are recommended.

    The future of Drug Delivery System:

    The future of controlled-release products

    is promising, especially in the following areas that

    present high promise and acceptability:

    Particulate systems: The micro particle and

    nanoparticle approach that involves biodegradable

    polymers and is aimed at the uptake of intact drug-

    loaded particles via the Peyers patches in the small

    intestine could be useful for delivery of peptide

    drugs that cannot, in general, be given orally.

    Chronopharmacokinetic systems: Oral controlled

    drug delivery with a pulsatile release regimen could

    effectively deliver drugs where need exists to

    counter naturally occurring processes such as

    bacterial/parasitical growth patterns (e.g., the once-

    daily oral Pulsys system introduced by Advancis

    Pharmaceutical Corp., which could potentially

    inhibit the emergence of resistant strains of

    microorganisms).

    Targeted drug delivery: Oral controlled drug

    delivery that targets regions in the GI tract and

  • Drug Delivery Systems - A Review

    17

    releases drugs only upon reaching that site could

    offer effective treatment for certain disease states

    (e.g. colon-targeted delivery of antineoplastics in

    the treatment of colon cancer).

    Mucoadhesive delivery: This is a promising

    technique for buccal and sublingual drug delivery,

    which can offer rapid onset of action and superior

    bioavailability compared with simple oral delivery

    because it bypasses firstpass metabolism in the

    liver. (Das 2003)

    Advantages of controlled drug delivery

    systems:

    1. Improved patient convenience and

    compliance

    2. Reduction in fluctuation in steady

    state levels.

    3. Increased safety margin of high

    potency drugs.

    4. Reduction in dose.

    5. Reduction in health care cost.

    Disadvantages of controlled drug

    delivery systems:

    1. Decreased systemic availability

    2. Poor invitro-invivo correlations

    3. Chances of dose dumping

    4. Dose withdrawal is not possible.

    5. Higher cost of formulation

    Applications of controlled drug delivery

    system:

    1) Mucoadhesive drug delivery

    system

    2) Colon drug delivery system

    3) Pulmonary drug delivery system

    4) Ocular drug delivery system

    5) Oral thin films

    6) Nasal drug delivery system

    7) Gastro retentive drug delivery

    system

    8) Vaginal drug delivery system

    9) Resealed erythrocytes

    References:

    1. Www.Farmacist.Blogspot.Com.

    2. Blanco Md, Alonso Mj. "Development

    And Characterization Of ." Eur J. Pharma

    Biopharm, 1997: 387-422.

    3. Brouwers. "J. R. B. J." Pharm. World Sci,

    1998.

    4. Das, Nandita G. Das And Sudip K.

    "Controlled-Release Of Oral Dosage

    Forms." 2003: 10-16.

    5. Kathryn E. Uhrich, Scott M. Cannizzaro

    And Robert S. Langer,Kevin M.

    Shakesheff. "Polymeric Systems For

    Controlled Drug Release." 1999: 3181-

    3198.

    6. Katre, N. "Adv. Drug Delivery Review."

    1993.

    7. Langer, R. Nature 1998. 1998: 392.5.

    8. Mehreganym, Gabriel KJ, Trimmer WSN.

    1998: 35:719.

    9. Paolino, Donatella. "Drug Delivery

    System." Encyclopedia Of Medical

    Devices And Instrumentation, 2006: 437-

    485.

    10. Ranade VV, Hollinger MA. "Drug

    Delivery Systems." CRC Press, 1996.

    11. Shivkumar, Vishal Gupta N. And.

    "Development Of Drug Delivery System."

    Trop J. Pharma, 2009.

    12. Smith BR, Et Al. "A Biological

    Perspective Of Particulate Nanoporous."

    2004: 19-16.

    13. V. Balmurlidhara, T.M. Pramodkumar.

    "Ph Sensitive Drug Delivery System- A

    Review." American Journal Of Drug

    Delivery And Development, 2011: 24-48.

  • Drug Delivery Systems - A Review

    18

    ORAL CONTROLLED DRUG DELIVERY SYSTEM Bajaj Amruta, Katedeshmukh Ramesh

    Introduction:

    Oral drug delivery is the most widely utilized route

    of administration among all the routes [nasal,

    ophthalmic, rectal, transdermal and Parenteral

    routes] that have been explored for systemic

    delivery of drugs via pharmaceutical products of

    different dosage form. Oral route is considered

    most natural, uncomplicated, convenient and safe

    [in respect to Parenteral route] due to its ease of

    administration, patient acceptance, and cost-

    effective manufacturing process. Pharmaceutical

    products designed for oral delivery are mainly

    immediate release type or conventional drug

    delivery systems, which are designed for

    immediate release of drug for rapid absorption.

    These immediate release dosage forms have some

    limitations such as:

    1) Drugs with short half-life requires frequent

    administration, which increases chances of missing

    dose of drug leading to poor patient compliance.

    2) A typical peak-valley plasma concentration-time

    profile is obtained which makes attainment of

    steady state condition difficult.

    3) The unavoidable fluctuations in the drug

    concentration may lead to under medication or

    overmedication as the CSS values fall or rise

    beyond the therapeutic range.

    4) The fluctuating drug levels may lead to

    precipitation of adverse effects especially of a drug

    with small therapeutic index, whenever

    overmedication occurs.

    In order to overcome the drawbacks of

    conventional drug delivery systems, several

    technical advancements have led to the

    development of controlled drug delivery system

    that could revolutionize method of medication and

    provide a number of therapeutic benefits.

    (Hemnani M. 2011)

    TABLE 1:-Benefit Characteristics Of Oral Controlled-Release Drug Delivery System. (Das N. 2003)

    Benefit Reason

    Therapeutic advantage

    Reduction in drug plasma level fluctuations;

    maintenance of a steady plasma level of the drug over

    a prolonged time period, ideally simulating an

    intravenous infusion of a drug.

    Reduction in adverse side effects and improvement in

    tolerability

    Drug plasma levels are maintained within a narrow

    window with no sharp peaks and with AUC of plasma

    concentration versus time curve comparable with total

    AUC from multiple dosing with immediate release

    dosage forms. This greatly reduces the possibility of

    side effects, as the scale of side effects increase as we

    approach the MSC.

    Patient comfort and compliance

    Oral drug delivery is the most common and

    convenient for patients, and a reduction in dosing

    frequency enhances compliance.

    Reduction in healthcare cost

    The total cost of therapy of the controlled release

    product could be comparable or lower than the

    immediate-release product. With reduction in side

    effects, the overall expense in disease management

    also would be reduced.

    Controlled Drug Delivery Systems: (Hemnani M.

    2011)

    Controlled drug delivery systems have been

    developed which are capable of controlling the rate

    of drug delivery, sustaining the duration of

    therapeutic activity and/or targeting the delivery of

    drug to a tissue. Controlled drug delivery or

    modified drug delivery systems are conveniently

    divided into four categories.

    1) Delayed release

    2) Sustained release

  • Drug Delivery Systems - A Review

    19

    3) Site-specific targeting

    4) Receptor targeting

    More precisely, controlled delivery can be defined

    as:

    1) Sustained drug action at a predetermined rate by

    maintaining a relatively constant, effective drug

    level in the body with concomitant minimization of

    undesirable side effects.

    2) Localized drug action by spatial placement of a

    controlled release system adjacent to or in the

    diseased tissue.

    3) Targeted drug action by using carriers or

    chemical derivatives to deliver drug to a particular

    target cell type.

    4) Provide a physiologically/therapeutically based

    drug release system. In other words, the amount

    and the rate of drug release are determined by the

    physiological/ therapeutic needs of the body.

    A controlled drug delivery system is usually

    designed to deliver the drug at particular rate. Safe

    and effective blood levels are maintained for a

    period as long as the system continues to deliver

    the drug. Controlled drug delivery usually results in

    substantially constant blood levels of the active

    ingredient as compared to the uncontrolled

    fluctuations observed when multiple doses of quick

    releasing conventional dosage forms are

    administered to a patient.

    Figure 1: A hypothetical plasma concentration-

    time profile from conventional multiple dosing

    and single doses of sustained and controlled

    delivery formulations. Rationale of Controlled

    Drug Delivery

    The basic rationale for controlled drug delivery is

    to alter the pharmacokinetics and

    pharmacodynamics of pharmacologically active

    moieties by using novel drug delivery system or by

    modifying the molecular structure and/or

    physiological parameters inherent in a selected

    route of administration. It is desirable that the

    duration of drug action become more a design

    property of a rate controlled dosage form, and less,

    or not at all, a property of the molecules inherent

    kinetic properties. Thus optimal design of

    controlled release systems necessitates a thorough

    understanding of the pharmacokinetics and

    pharmacodynamics of the drug. The primary

    objectives of controlled drug delivery are to ensure

    safety and to improve efficacy of drugs as well as

    patient compliance. This is achieved by better

    control of plasma drug levels and less frequent

    dosing. The dose and dosing interval can be

    modified in case of conventional dosage forms.

    However, therapeutic window of plasma

    concentration below which no therapeutic effect is

    exhibited and above which undesirable effects are

    manifested. Therapeutic index is the prime

    parameter for development of a controlled delivery

    system of a particular drug candidate.

    Factors Affecting the Design and

    Performance of Controlled Drug

    Delivery: (Hemnani M. 2011)

    1. Drug Properties:

    Partition coefficient

    Drug stability

    Protein binding

    Molecular size and diffusivity

    2. Biological Properties:

    Absorption

    Metabolism

    Elimination and biological half life

    Dose size

    Route of administration

    Target sites

    Acute or chronic therapy

    Disease condition

    Advantages of Controlled Drug Delivery

    System: (Patel H. Nov Dec 2011):

    1. Avoid patient compliance problems.

    2. Employ less total drug

    3. Minimize or eliminate local side effects

    4. Minimize or eliminate systemic side

    effects

    5. Obtain less potentiating or reduction in

    drug activity with chronic use.

    6. Minimize drug accumulation with

    chronic dosing.

  • Drug Delivery Systems - A Review

    20

    7. Improve efficiency in treatment

    8. Cures or controls condition more

    promptly.

    9. Improves control of condition i.e.,

    reduced fluctuation in drug level.

    10. Improves bioavailability of some drugs.

    11. Make use of special effects, E.g.

    Sustained-release aspirin for morning

    relief of arthritis by dosing before bed

    time.

    12. Economy i.e. reduction in health care

    costs. The average cost of treatment over

    an extended time period may be less, with

    less frequency of dosing, enhanced

    therapeutic benefits and reduced side

    effects.

    13. The time required for health care

    personnel to dispense and administer the

    drug and monitor patient is also reduced.

    Disadvantages: (Kamboj S 2009)

    1) Decreased systemic availability in

    comparison to immediate release

    conventional dosage forms, which may be

    due to incomplete release, increased first-

    pass metabolism, increased instability,

    insufficient residence time for complete

    release, site specific absorption, pH

    dependent stability etc.

    2) Poor in vitro in vivo correlation.

    3) Possibility of dose dumping due to food,

    physiologic or formulation variables or

    chewing or grinding of oral formulations

    by the patient and thus, increased risk of

    toxicity.

    4) Retrieval of drug is difficult in case of

    toxicity, poisoning or hypersensitivity

    reactions.

    5) Reduced potential for dosage adjustment

    of drugs normally administered in varying

    strengths.

    6) Stability problems.

    7) Increased cost.

    8) More rapid development of tolerance and

    counseling.

    9) Need for additional patient education and

    counseling

    Oral Controlled Drug Delivery Systems:

    (Hemnani M. 2011)

    Oral controlled release drug delivery is a drug

    delivery system that provides the continuous oral

    delivery of drugs at predictable and reproducible

    kinetics for a predetermined period throughout the

    course of GI transit and also the system that target

    the delivery of a drug to a specific region within the

    GI tract for either a local or systemic action.

    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 form (either solid dispersion or

    liquid), must be developed within the intrinsic

    characteristics of GI physiology. Therefore the

    scientific framework required for the successful

    development of oral drug delivery systems consists

    of basic understanding of

    (i) physicochemical, pharmacokinetic

    and pharmacodynamic characteristics

    of the drug

    (ii) the anatomic and physiologic

    characteristics of the gastrointestinal

    tract

    (iii) physicochemical characteristics and

    the drug delivery mode of the dosage

    form to be designed.

    The main areas of potential challenge in the

    development of oral controlled drug delivery

    systems are:-

    1) Development of a drug delivery system: To

    develop a viable oral controlled release drug

    delivery system capable of delivering a drug at a

    therapeutically effective rate to a desirable site for

    duration required for optimal treatment.

    2) Modulation of gastrointestinal transit time: To

    modulate the GI transit time so that the drug

    delivery system developed can be transported to a

    target site or to the vicinity of an absorption site

    and reside there for a prolonged period of time to

    maximize the delivery of a drug dose.

    3) Minimization of hepatic first pass elimination: If

    the drug to be delivered is subjected to extensive

    hepatic first-pass elimination, preventive measures

    should be devised to either bypass or minimize the

    extent of hepatic metabolic effect.

  • Drug Delivery Systems - A Review

    21

    Methods Used To Achieve Controlled Release

    Of Orally Administered Drugs:

    A. Diffusion Controlled System:

    Basically diffusion process shows the movement of

    drug molecules from a region of a higher

    concentration to one of lower concentration.

    This system is of two types:

    a) Reservoir type: A core of drug surrounded by

    polymer membrane, which controls the release rate,

    characterizes reservoir devices.

    b) Matrix type: Matrix system is characterized by

    a homogenous dispersion of solid drug in a

    polymer mixture.

    B. Dissolution Controlled Systems:

    a) Reservoir type: Drug is coated with a given

    thickness coating, which is slowly dissolved in the

    contents of gastrointestinal tract. By alternating

    layers of drug with the rate controlling coats as

    shown in figure no.2, a pulsed delivery can be

    achieved. If the outer layer is quickly releasing

    bolus dose of the drug, initial levels of the drug in

    the body can be quickly established with pulsed

    intervals

    Figure 2: Schematic representation of diffusion

    controlled drug release reservoir system.

    b) Matrix type: The more common type of

    dissolution controlled dosage form as shown in

    figure .3. It can be either a drug impregnated sphere

    or a drug impregnated tablet, which will be

    subjected to slow erosion.

    Figure 3: Schematic representation of diffusion

    controlled drug release matrix system.

    C. Bioerodable and Combination of Diffusion

    and Dissolution Systems:

    It is characterized by a homogeneous

    dispersion of drug in an erodible matrix. (Shown in

    figure.4)

    Figure 4: Drug delivery from (a) bulk-eroding

    and (b) surface-eroding Bio erodible systems.

    D. Methods using Ion Exchange: It is based on

    the drug resin complex formation when an ionic

    solution is kept in contact with ionic resins. The

    drug from these complexes gets exchanged in

    gastrointestinal tract and released with excess of

    Na+ and Cl- present in gastrointestinal tract.

    E. Methods using osmotic pressure: It is

    characterized by drug surrounded by semi

    permeable membrane and release governed by

    osmotic pressure.

    F. pH Independent formulations: A buffered

    controlled release formulation as shown in figure 5,

    is prepared by mixing a basic or acidic drug with

    one or more buffering agents, granulating with

    appropriate pharmaceutical excipients and coating

    with GI fluid permeable film forming polymer.

    When GI fluid permeates through the membrane

    the buffering agent adjusts the fluid inside to

    suitable constant pH thereby rendering a constant

    rate of drug release.

  • Drug Delivery Systems - A Review

    22

    Figure 5: Drug delivery from environmentally

    pH sensitive release systems.

    G. Altered density formulations: Several

    approaches have been developed to prolong the

    residence time of drug delivery system in the

    gastrointestinal tract.

    High-density approach

    Low-density approach

    Matrix Tablet: (Patel H. Nov Dec 2011)

    Advantages of matrix tablet:

    Easy to manufacture

    Versatile, effective and low cost

    Can be made to release high molecular

    weight compounds

    The sustained release formulations may

    maintain therapeutic concentrations over

    prolonged periods.

    The use of sustain release formulations

    avoids the high blood concentration.

    Sustain release formulations have the

    potential to improve the patient

    compliance.

    Reduce the toxicity by slowing drug

    absorption.

    Increase the stability by protecting the

    drug from hydrolysis or other derivative

    changes in gastrointestinal tract.

    Minimize the local and systemic side

    effects.

    Improvement in treatment efficacy.

    Minimize drug accumulation with chronic

    dosing.

    Usage of less total drug.

    Improvement the bioavailability of some

    drugs.

    Improvement of the ability to provide

    special effects. Ex: Morning relief of

    arthritis through bed time dosing.

    Disadvantages of matrix tablet: (Patel H. Nov

    Dec 2011)

    The remaining matrix must be removed after the

    drug has been released.

    High cost of preparation.

    The release rates are affected by various factors

    such as, food and the rate transit through the gut.

    The drug release rates vary with the square root of

    time. Release rate continuously diminishes due to

    an increase in diffusional resistance and/or a

    decrease in effective area at the diffusion front.

    However, a substantial sustained effect can be

    produced through the use of very slow release

    rates, which in many applications are

    indistinguishable from zero-order.

    Classification Of Matrix Tablets:

    On the Basis of Retardant Material Used: Matrix

    tablets can be divided in to 5 types.

    1. Hydrophobic Matrices (Plastic matrices):

    The concept of using hydrophobic or inert

    materials as matrix materials was first introduced in

    1959. In this method of obtaining sustained release

    from an oral dosage form, drug is mixed with an

    inert or hydrophobic polymer and then compressed

    in to a tablet. Sustained release is produced due to

    the fact that the dissolving drug has diffused

    through a network of channels that exist between

    compacted polymer particles. Examples of

    materials that have been used as inert or

    hydrophobic matrices include polyethylene,

    polyvinyl chloride, ethyl cellulose and acrylate

    polymers and their copolymers. The rate-

    controlling step in these formulations is liquid

    penetration into the matrix. The possible

    mechanism of release of drug in such type of

    tablets is diffusion. Such types of matrix tablets

    become inert in the presence of water and

    gastrointestinal fluid.

    2. Lipid Matrices:

    These matrices prepared by the lipid waxes and

    related materials. Drug release from such matrices

    occurs through both pore diffusion and erosion.

    Release characteristics are therefore more sensitive

    to digestive fluid composition than to totally

    insoluble polymer matrix. Carnauba wax in

    combination with stearyl alcohol or stearic acid has

    been utilized for retardant base for many sustained

    release formulation.

  • Drug Delivery Systems - A Review

    23

    3. Hydrophilic Matrices:

    Hydrophilic polymer matrix systems are widely

    used in oral controlled drug delivery because of

    their flexibility to obtain a desirable drug release

    profile, cost effectiveness, and broad regulatory

    acceptance. The formulation of the drugs in

    gelatinous capsules or more frequently, in tablets,

    using hydrophilic polymers with high gelling

    capacities as base excipients is of particular interest

    in the field of controlled release. Infect a matrix is

    defined as well mixed composite of one or more

    drugs with a gelling agent (hydrophilic polymer).

    These systems are called swellable controlled

    release systems. The polymers used in the

    preparation of hydrophilic matrices are divided in

    to three broad groups,

    A. Cellulose derivatives: Methylcellulose 400 and

    4000cPs, HEC; HPMC 25, 100, 4000 and

    15000cPs; and Sodium carboxymethylcellulose.

    B. Non cellulose natural or semi synthetic

    polymers: Agar-Agar; Carob gum; Alginates;

    Molasses; Polysaccharides of mannose and

    galactose, Chitosan and Modified starches.

    4. Biodegradable Matrices: These consist of the

    polymers which comprised of monomers linked to

    one another through functional groups and have

    unstable linkage in the backbone. They are

    biologically degraded or eroded by enzymes

    generated by surrounding living cells or by

    nonenzymetic process in to oligomers and

    monomers that can be metabolized or excreted.

    Examples are natural polymers such as proteins and

    polysaccharides; modified natural polymers;

    synthetic polymers such as aliphatic poly (esters)

    and poly anhydrides.

    5. Mineral Matrices: These consist of polymers

    which are obtained from various species of

    seaweeds. Example is Alginic acid which is a

    hydrophilic carbohydrate obtained from species of

    brown seaweeds (Phaephyceae) by the use of dilute

    alkali.

    On the Basis of Porosity of Matrix: Matrix

    system can also be classified according to their

    porosity and consequently, Macro porous; Micro

    porous and Non-porous systems can be identified:

    1. Macro porous Systems: In such systems the

    diffusion of drug occurs through pores of matrix,

    which are of size range 0.1 to 1 m. This pore size

    is larger than diffusant molecule size.

    2. Micro porous System: Diffusion in this type of

    system occurs essentially through pores. For micro

    porous systems, pore size ranges between 50 200

    A, which is slightly larger than diffusant

    molecules size.

    3. Non-porous System: Non-porous systems have

    no pores and the molecules diffuse through the

    network meshes. In this case, only the polymeric

    phase exists and no pore phase is present.

    Polymers used in matrix tablet:

    Hydrogels: Polyhydroxyethylemethylacrylate

    (PHEMA), Cross-linked polyvinyl alcohol (PVA),

    Cross-linked polyvinyl pyrrolidone (PVP),

    Polyethylene oxide (PEO), Polyacrylamide (PA)

    Soluble polymers: Polyethyleneglycol (PEG),

    polyvinyl alcohol (PVA), Polyvinylpyrrolidone

    (PVP), Hydroxypropyl methyl cellulose (HPMC)

    Biodegradable polymers: Polylactic acid (PLA),

    Polyglycolic acid (PGA), Polycaprolactone (PCL),

    Polyanhydrides, Polyorthoesters

    Non-biodegradable polymers: Polyethylene vinyl

    acetate (PVA), Polydimethylsiloxane (PDS),

    Polyether urethane (PEU), Polyvinyl chloride

    (PVC), Cellulose acetate (CA), Ethyl cellulose

    (EC)

    Mucoadhesive polymers: Polycarbophil, Sodium

    carboxymethyl cellulose, Polyacrylic acid,

    Tragacanth, Methyl cellulose, Pectin

    Natural gums: Xanthan gum, Guar gum, Karaya

    gum, Locust bean gum

    Components of matrix tablets: (ME. 2005)

    These include:

    Active drug

    Release controlling agent(s): matrix

    formers

    Matrix Modifiers, such as channelling

    agents and wicking agents

    Solubilizers and pH modifiers

    Lubricants and flow aid

    Supplementary coatings to extend lag time

    further reduce drug release etc.

    Density modifiers (if required)

  • Drug Delivery Systems - A Review

    24

    Mechanism Of Drug Release From Matrix

    Tablet:

    Drug in the outside layer exposed to the bathing

    solution is dissolved first and then diffuses out of

    the matrix. This process continues with the

    interface between the bathing solution and the solid

    drug moving toward the interior. It follows that for

    this system to be diffusion controlled, the rate of

    dissolution of drug particles within the matrix must

    be much faster than the diffusion rate of dissolved

    drug leaving the matrix. Derivation of the

    mathematical model to describe this system

    involves the following assumptions:

    a) A pseudo-steady state is maintained during drug

    release,

    b) The diameter of the drug particles is less than

    the average distance of drug diffusion through the

    matrix,

    c) The bathing solution provides sink conditions at

    all times.

    The release behaviour for the system can be

    mathematically described by the following

    equation:

    dM/dh = Co. dh - Cs/2 (1)

    Where, dM = Change in the amount of drug

    released per unit area

    dh = Change in the thickness of the zone of matrix

    that has been depleted of drug

    Co = Total amount of drug in a unit volume of

    matrix

    Cs = Saturated concentration of the drug within the

    matrix.

    Additionally, according to diffusion theory:

    dM = ( Dm. Cs / h) dt........................... (2)

    Where, Dm = Diffusion coefficient in the matrix.

    h = Thickness of the drug-depleted matrix

    dt = Change in time

    By combining equation 1 and equation 2 and

    integrating:

    M = [Cs. Dm (2Co Cs) t] (3)

    When the amount of drug is in excess of the

    saturation concentration then:

    M = [2Cs.Dm.Co.t] 1/2 (4)

    Equation 3 and equation 4 relate the amount of

    drug release to the square-root of time. Therefore,

    if a system is predominantly diffusion controlled,

    then it is expected that a plot of the drug release vs.

    square root of time will result in a straight line.

    Drug release from a porous monolithic matrix

    involves the simultaneous penetration of

    surrounding liquid, dissolution of drug and

    leaching out of the drug through tortuous interstitial

    channels and pores.

    The volume and length of the openings must be

    accounted for in the drug release from a porous or

    granular matrix:

    M = [Ds. Ca. p/T. (2Co p.Ca) t] 1/2

    . (5)

    Where, p = Porosity of the matrix

    t = Tortuosity Ca = solubility of the drug in the

    release medium

    Ds = Diffusion coefficient in the release medium.

    T = Diffusional path length For pseudo steady

    state,

    the equation can be written as:

    M = [2D.Ca .Co (p/T) t]

    .. (6)

    The total porosity of the matrix can be calculated

    with the following equation:

    p = pa + Ca/ + Cex / ex

    (7)

    Where, p = Porosity

    = Drug density

    pa = Porosity due to air pockets in the matrix

    ex = Density of the water soluble excipients

    Cex = Concentration of water soluble excipients

    For the purpose of data treatment, equation 7 can

    be reduced to: M = k. t 1/2 ..

    (8)

    Where, k is a constant, so that the amount of drug

    released versus the square root of time will be

    linear, if the release of drug from matrix is

    diffusion-controlled. If this is the case, the release

    of drug from a homogeneous matrix system can be

    controlled by varying the following parameters:

    Initial concentration of drug in the matrix

    Porosity

    Tortuosity

    Polymer system forming the matrix

    Solubility of the drug.

    Effect Of Release Limiting Factor On Drug

    Release:

    The mechanistic analysis of controlled release of

    drug reveals that partition coefficient; diffusivity;

    diffusional path thickness and other system

    parameters play various rate determining roles in

    the controlled release of drugs from either capsules,

    matrix or sandwich type drug delivery systems.

    A. Polymer hydration: It is important to study

    polymer hydration/swelling process for the

    maximum number of polymers and polymeric

    combinations. The more important step in

    polymer dissolution include

    absorption/adsorption of water in more

  • Drug Delivery Systems - A Review

    25

    accessible places, rupture of polymer-polymer

    linking with the simultaneous forming of

    water-polymer linking, separation of

    polymeric chains, swelling and finally

    dispersion of polymeric chain in dissolution

    medium

    B. Drug solubility: Molecular size and water

    solubility of drug are important determinants

    in the release of drug from swelling and

    erosion controlled polymeric matrices. For

    drugs with reasonable aqueous solubility,

    release of drugs occurs by dissolution in

    infiltrating medium and for drugs with poor

    solubility release occurs by both dissolution of

    drug and dissolution of drug particles through

    erosion of the matrix tablet.

    C. Solution solubility: In view of in vivo

    (biological) sink condition maintained actively by

    hem perfusion, it is logical that all the in vitro drug

    release studies should also be conducted under

    perfect sink condition. In this way a better

    simulation and correlation of in vitro drug release

    profile with in vivo drug administration can be

    achieved. It is necessary to maintain a sink

    condition so that the release of drug is controlled

    solely by the delivery system and is not affected or

    complicated by solubility factor.

    D. Polymer diffusivity: The diffusion of small

    molecules in polymer structure is energy activated

    process in which the diffusant molecules moves to

    a successive series of equilibrium position when a

    sufficient amount of energy of activation for

    diffusion Ed has been acquired by the diffusant is

    dependent on length of polymer chain segment,

    cross linking and crystallinity of polymer. The

    release of drug may be attributed to the three

    factors viz,

    i. Polymer particle size

    ii. Polymer viscosity

    iii. Polymer concentration.

    i. Polymer particle size: Malamataris

    stated that when the content of hydroxyl

    propyl methylcellulose is higher, the

    effect of particle size is less important on

    the release rate of propranolol

    hydrochloride, the effect of this variable

    more important when the content of

    polymer is low. He also justified these

    results by considering that in certain areas

    of matrix containing low levels of

    hydroxyl propyl methylcellulose led to

    the burst release.

    ii. Polymer viscosity: With cellulose ether

    polymers, viscosity is used as an

    indication of matrix weight. Increasing

    the molecular weight or viscosity of the

    polymer in the matrix formulation

    increases the gel layer viscosity and thus

    slows drug dissolution. Also, the greater

    viscosity of the gel, the more resistant the

    gel is to dilution and erosion, thus

    controlling the drug dissolution.

    iii. Polymer concentration: An increase in

    polymer concentration causes an increase

    in the viscosity of gel as well as

    formulation of gel layer with a longer

    diffusional path. This could cause a

    decrease in the effective diffusion

    coefficient of the drug and therefore

    reduction in drug release. The mechanism

    of drug release from matrix also changes

    from erosion to diffusion as the polymer

    concentration increases.

    E. Thickness of polymer diffusional path: The

    controlled release of a drug from both capsule and

    matrix type polymeric drug delivery system is

    essentially governed by Ficks law of diffusion:

    JD = D dc/dx

    Where, JD is flux of diffusion across a plane

    surface of unit area

    D is diffusibility of drug molecule, dc/dx is

    concentration gradient of drug molecule across a

    diffusion path with thickness dx.

    F. Thickness of hydrodynamic diffusion layer: It

    was observed that the drug release profile is a

    function of the variation in thickness of

    hydrodynamic diffusion layer on the surface of

    matrix type delivery devices. The magnitude of

    drug release value decreases on increasing the

    thickness of hydrodynamic diffusion layer d.

    G. Drug loading dose: The loading dose of drug

    has a significant effect on resulting release kinetics

    along with drug solubility. The effect of initial drug

    loading of the tablets on the resulting release

    kinetics is more complex in case of poorly water

    soluble drugs, with increasing initial drug loading

    the relative release rate first decreases and then

    increases, whereas, absolute release rate

    monotonically increases. In case of freely water

    soluble drugs, the porosity of matrix upon drug

    depletion increases with increasing initial drug

  • Drug Delivery Systems - A Review

    26

    loading. This effect leads to increased absolute

    drug transfer rate. But in case of poorly water

    soluble drugs another phenomenon also has to be

    taken in to account. When the amount of drug

    present at certain position within the matrix,

    exceeds the amount of drug soluble under given

    conditions, the excess of drug has to be considered

    as non-dissolved and thus not available for

    diffusion. The solid drug remains within tablet, on

    increasing the initial drug loading of poorly water

    soluble drugs, the excess of drug remaining with in

    matrix increases.

    H. Surface area and volume: The dependence of

    the rate of drug release on the surface area of drug

    delivery device is well known theoretically and

    experimentally. Both the in vitro and in vivo rate of

    the drug release, are observed to be dependent upon

    surface area of dosage form. Siepman et al. found

    that release from small tablet is faster than large

    cylindrical tablets.

    I. Diluents effect: The effect of diluent or filler

    depends upon the nature of diluent. Water soluble

    diluents like lactose cause marked increase in drug

    release rate and release mechanism is also shifted

    towards Fickian diffusion; while insoluble diluents

    like dicalcium phosphate reduce the Fickian

    diffusion and increase the relaxation (erosion) rate

    of matrix. The reason behind this is that water

    soluble filler in matrices stimulate the water

    penetration in to inner part of matrix, due to

    increase in hydrophilicity of the system, causing

    rapid diffusion of drug, leads to increased drug

    release rate.

    J. Additives: The effect of adding non-polymeric

    excipients to a polymeric matrix has been claimed

    to produce increase in release rate of hydro soluble

    active principles. These increases in release rate

    would be marked if the excipients are soluble like

    lactose and less important if the excipients are

    insoluble like tricalcium phosphate

    .

    Biological Factors Influencing Release from

    Matrix Tablet:

    Biological half-life.

    Absorption.

    Metabolism

    Distribution

    Protein binding

    Margin of safety

    Biological half-life: The usual goal of an oral SR

    product is to maintain therapeutic blood levels over

    an extended period of time. To achieve this, drug

    must enter the circulation at approximately the

    same rate at which it is eliminated. The elimination

    rate is quantitatively described by the half-life

    (t1/2). Each drug has its own characteristic

    elimination rate, which is the sum of all elimination

    processes, including metabolism, urinary excretion

    and all over processes that permanently remove

    drug from the blood stream. Therapeutic

    compounds with short half-life are generally are

    excellent candidate for SR formulation, as this can

    reduce dosing frequency. In general, drugs with

    half-life shorter than 2 hours such as furosemide or

    levodopa are poor candidates for SR preparation.

    Compounds with long half-lives, more than 8 hours

    are also generally not used in sustaining form, since

    their effect is already sustained. Digoxin and

    phenytoin are the examples.

    Absorption: Since the purpose of forming a SR

    product is to place control on the delivery system, it

    is necessary that the rate of release is much slower

    than the rate of absorption. If we assume that the

    transit time of most drugs in the absorptive areas of

    the GI tract is about 8-12 hours, the maximum half-

    life for absorption should be approximately 3-4

    hours; otherwise, the device will pass out of the

    potential absorptive regions before drug release is

    complete. Thus corresponds to a minimum

    apparent absorption rate constant of 0.17-0.23h-1 to

    give 80-95% over this time period. Hence, it

    assumes that the absorption of the drug should

    occur at a relatively uniform rate over the entire

    length of small intestine. For many compounds this

    is not true. If a drug is absorbed by active transport

    or transport is limited to a specific region of

    intestine, SR preparation may be disadvantageous

    to absorption. One method to provide sustaining

    mechanisms of delivery for compounds tries to

    maintain them within the stomach. This allows

    slow release of the drug, which then travels to the

    absorptive site. These methods have been

    developed as a consequence of the observation that

    co-administration results in sustaining effect. One

    such attempt is to formulate low density pellet or

    capsule. Another approach is that of bio adhesive

    materials.

    Metabolism: Drugs those are significantly

    metabolized before absorption, either in the lumen

    or the tissue of the intestine, can show decreased

    bioavailability from slower-releasing dosage form.

  • Drug Delivery Systems - A Review

    27

    Hence criteria for the drug to be used for

    formulating Sustained-Release dosage form is,

    Drug should have law half-life (

  • Drug Delivery Systems - A Review

    28

    DRUGS USED CATEGORY METHOD USED POLYMER USED

    Zidovudine Anti-viral Direct Compression HPMC-K4M, Carbopol-934, EC

    Venlafexine Anti-depressant Wet Granulation Beeswax, Carnauba wax

    Domperidone Anti-emetic Wet Granulation HPMC-K4M, Carbopol-934

    Alfuzosin Alfa-adrenergic

    Agonist

    Direct Compression HPMC-K15M, Eudragit-RSPO

    Minocycline Antibiotic Wet Granulation HPMC-K4M, HPMC-K15M, EC

    Ibuprofen Anti-

    inflammatory

    Wet Granulation EC, CAP

    Metformin HCL Anti-diabetic Direct Compression HPMC-K100M, EC

    Propranolol HCL Beta-adrenergic

    blocker

    Wet Granulation Locust bean gum, HPMC

    Furosemide Anti-diuretic Direct Compression Guar gum, Pectin, Xanthan gum

    Acarbose Anti-diabetic Direct Compression HPMC, Eudragit

    Aceclofenac Anti-

    inflammatory

    Wet Granulation HPMC-K4M,K15M,

    K100M,E15,EC, Guar gum

    Ambroxol HCL Expectorent,

    Mucolytic

    Direct Compression HPMC-K100M,

    Aspirin Anti-

    inflammatory

    Direct Compression EC, Eudragit-RS100, S100

    Diclofenac Na Anti-

    inflammatory

    Wet Granulation Chitoson, EC, HPMCP, HPMC

    Diethylcarbamazepine

    citrate

    Anti-filarial Wet Granulation Guar gum, HPMC-E15LV

    Diltiazem Ca+2 channel

    blocker

    Direct Compression HPMC-K100M, HPMC-K4M,

    Karaya gum, Locust bean gum,

    Sod.CMC

    Enalpril meleate ACE inhibitor Direct Compression HPMC-K100M,HPMC K4M,

    Flutamide Anti-androgen Direct Compression HPMC-K4M, Sod.CMC, Guar gum,

    Xanthan gum

    Indomethacin Anti-

    inflammatory

    Direct Compression EC, HPMC

    Chlorphenarimine

    meleate

    H1 antagonist Melt-extrusion Xanthan gum,Chitoson

    Itopride HCL Prokinetic agent Direct Compression HPMC-K100M, HPMC-K4M, EC

    Losartan potassium Anti-hypertensive Direct Compression HPMC-K100M, HPMC-K4M,

    Eudragit-RSPO

    Metoclopromide Anti-emetic Direct Compression /

    Wet Granulation

    HPMC, CMC, EC, SSG

    Miconazole Anti-fungal Direct Compression /

    Wet Granulation

    Pectin, HPMC

    Naproxen Morphine

    antagonist

    Direct Compression HPMC-K100M, HPMC-K15M, PVP

    Nicorandil Ca+2 channel

    blocker

    Wet Granulation HPMC, CMC, EC

    Ondansertan Anti-hypertensive Wet Granulation HPMC-K100M, HPMC-K4M,

    HPMC-K15M

    Phenytoin Na Anti-epileptic Wet Granulation Tragacanth, Acacia, Guar gum,

    Xanthan gum

    Ranitidine HCL H2 antagonist Direct Compression Chitoson, Carbopol-940

    Theophylline Respiratory

    depressant

    Direct Compression Carbopol-934P, HPMC-K100M,

    HPMC-K4M, HPMC-K15M, EC

  • Drug Delivery Systems - A Review

    29

    Osmotic Drug Delivery:

    Introduction (M. 2009)

    Controlled release dosage form are designed to

    release drug in-vivo according to predictable rate that

    can be verified by in-vitro measurement Potential

    development and new approaches to oral controlled

    release dosage form includes,

    1. Hydrodynamic pressure controlled system

    2. Intragastric floating tablet

    3. Transmucosal tablet

    4. Microporous membrane coated tablet.

    Advantages: (Ghosh T. 2011)

    Decrease frequency of dosing.

    Reduce the rate of rise of drug concentration

    in the body.

    Delivery may be pulsed or desired if

    required.

    Delivery ratio is independent of pH of the

    environment.

    Delivery is independent of hydrodynamic

    condition, this suggest that drug delivery is

    independent of G.I. motility.

    Sustained and consistence blood level of

    drug within the therapeutic window.

    Improve patient compliance.

    High degree of in vitro- in vivo correlation

    is obtained in osmotic system.

    Reduce side effect.

    Delivery rate is also independent of delivery

    orifice size within the limit.

    Disadvantage & limitation (Ghosh T. 2011)

    OCODDS have produced significant clinical benefit

    invarious therapeutic areas .Some system have

    enhanced patient compliance, while other has

    minimized the side effect of their active compounds.

    However some limitations of OCODDS have been

    reported.

    Slightly higher cost of good than matrix

    tablet or multiparticulates ion capsule

    dosage form.

    Gastro intestinal obstruction cases have been

    observed with the patient receiving

    Nifedipine GITS tablet.

    Another case was reported for osmosin

    (Indomethacin OROS) which was first

    introduced in the United Kingdom in 1983

    .A few month later after its introduction

    frequent incidences of serious

    gastrointestinal reaction was observed

    leading to osmosin withdrawal. Various

    explanations were given based on the toxic

    effect of KCl used in osmosin.

    Magnetic resonance imaging (MRI) of tablet

    elucidate that nonuniform coating leads to

    different pattern of drug release among the

    batches.

    What Is Osmotic Pressure: (M. 2009)

    Osmosis

    Osmosis can be defined as the net movement of water

    across a selectively permeable membrane driven by a

    difference in osmotic pressure across the membrane.

    It is driven by a difference in solute concentrations

    across the membrane that allows passage of water,

    but rejects most solute molecules or ions. Osmotic

    pressure is the pressure which, if applied to the more

    concentrated solution, would prevent transport of

    water across the semipermeable membrane.

    The first osmotic effect was reported by Abbe Nollet

    in 1748. Later in 1877, Pfeffer performed an

    experiment using semi-permeable membrane to

    separate sugar solution from pure water. He showed

    that the osmotic pressure of the sugar solution is

    directly proportional to the solution concentration

    and the absolute temperature. In 1886, Vant Hoff

    identified an underlying proportiona


Recommended