Use This Content

Embed Size (px)

Citation preview

  • 7/26/2019 Use This Content

    1/136

    CONTENTS

    CHAPTER TITLE PAGE

    NO.

    1. INTRODUCTION 1

    1.1. Oral drug delivery system 1-8

    1.2. Classification of sustained release polymeric drug

    delivery systems

    8-13

    1.3. Fabrication of oral controlled release delivery

    employs various methods

    13-18

    1.4. Matrix systems 19-21

    1.5. Approaches for preparation of matrix dosage forms 21-23

    1.6. Mechanism of drug release from matrix devices 23-29

    2. NEED AND OBJECTIVES 30-31

    3. PLAN OF WORK 31-33

    4. LITERATURE REVIEW 34-41

    4.1. Drug Profile 42-44

    4.2. Excipients Profile 45-63

    5. MATERIALS AND METHODS 64

    5.1. Excipient Used 65

    5.2. Equipments Used 66-74

    5.3. Preformulation Study 74-74

    5.4. Compression of Powder Blends into Tablets 74-79

    5.5. Evaluation of Sustained Release Matrix Tablets 79

    5.6. Stability Study 80

  • 7/26/2019 Use This Content

    2/136

    6. RESULTS AND DISCUSSION 80

    6.1. Preformulation Parameters 80-93

    6.2. Evaluation of Sustained Release Matrix Tablets 93-117

    6.3. Stability Study 117-

    119

    7. SUMMARY AND CONCLUSION 120

    8. FUTURE PROSPECTS 121

    9. BIBLIOGRAPHY 122-

    127

  • 7/26/2019 Use This Content

    3/136

    LIST OF FIGURES

    FIGURE

    NO.

    CONTENTS PAGE

    NO.

    1.1 A hypothetical plasma concentrationtime profile fromconventional multiple and single doses of sustained and

    controlled delivery formulations.

    4

    1.2 Schematic representation of diffusion controlled drug release

    reservoir.

    11

    1.3 Schematic representation of diffusion controlled drug releasematrix system.

    11

    1.4 Drug delivery from environmentally pH sensitive releasesystems. 13

    1.5 Hydrogel formation in Reservoir Systems. 14

    1.6 Drug Release from a Matrix Tablets 15

    1.7 Parameter definitions for swellable soluble matrix. 16

    5.1 Schematic representation of compatibility studies 69

    5.2 Steps in traditional Wet granulation techniques 71

    6.1 max observed for Telmisartan in 0.1N HCl. 80

    6.2 max observed for Telmisartan in 0.1N HCl. 81

    6.3 Standard graph of Telmisartan 0.1N HCl. 83

    6.4 Standard graph of Telmisartan in pH 6.8 Phosphate buffer. 84

    6.5 FTIR Spectra of Telmisartan 86

    6.6 FTIR Spectra of Telmisartan and HPMC K100M 86

    6.7 FTIR Spectra of Telmisartan and ethyl cellulose 87

    6.8 FTIR Spectra of Telmisartan and xanthan gum 876.9 DSC Thermal analysis of pure Telmisartan 89

    6.10 DSC Thermal analysis of Telmisartan+ HPMC K100M 89

    6.11 DSC Thermal analysis of Telmisartan + ethylcellulose 90

    6.12 DSC Thermal analysis of Telmisartan + xanthan gum 90

  • 7/26/2019 Use This Content

    4/136

    6.13 Drug release profile of formulation F1 96

    6.14 Drug release profile of formulation F2 97

    6.15 Drug release profile of formulation F3 98

    6.16 Drug release profile of formulation F4 99

    6.17 Drug release profile of formulation F5 100

    6.18 Drug release profile of formulation F6 101

    6.19 Drug release profile of formulation F7 102

    6.20 Drug release profile of formulation F8 103

    6.21 Drug release profile of formulation F9 104

    6.22 Drug release profile of formulations containing HPMC K100Mpolymer

    105

    6.23 Drug release profile of formulations containing Ethylcellulosepolymer

    105

    6.24 Drug release profile of formulations containing Xanthan gumpolymer

    106

    6.25 Drug release profile of all nine formulations (F1 to F9) 107

    6.26 Best fit model (Peppas) of formulation F1 111

    6.27 Best fit model (Zero-order) of formulation F2 111

    6.28 Best fit model (Zero-order) of formulation F3 112

    6.29 Best fit model (Zero-order) of formulation F4 113

    6.30 Best fit model (Peppas) of formulation F5 113

    6.31 Best fit model (Zero-order) of formulation F6 114

    6.32 Best fit model (Zero-order) of formulation F7 114

    6.33 Best fit model (Zero-order) of formulation F8 115

    6.34 Best fit model (Zero-order) of formulation F9 116

    6.35 Comparisons of hardness for formulation F3 with initial anddifferent periods of stability

    118

    6.36 Comparisons of drug content for formulation F3 with initial anddifferent periods of stability

    118

    6.37 Comparisons of drug release profile for formulation F3 withinitial and different periods of stability

    119

  • 7/26/2019 Use This Content

    5/136

    LIST OF TABLES

    TABLE

    NO.

    CONTENTS PAGE

    NO.

    1.1 Drug properties suitable for sustained release 6-8

    1.2 Characteristics of major classes of sustained release drug delivery system 17-19

    1.3 The characteristics of various types of matrices 19-21

    1.4 Methods for preparation of matrices. 21-23

    5.1 List of excipients with source 64

    5.2 List of equipments with model/make 65

    5.3 Composition of Telmisartan SR Matrix tablet 70

    5.4Standard values of Carrs index 73

    5.5 Standard values of angle of repose () 74

    5.6 Specifications of % Weight Variation Allowed in Tablets as per Indian

    Pharmacopoeia

    76

    6.1 The solubility of Telmisartan in various solvents 80

    6.2 Data of concentration and absorbance for in Telmisartan 0.1N HCl 82

    6.3 Data of concentration and absorbance for Telmisartan in pH 6.8

    Phosphate buffer.

    83

    6.4 Data for Calibration Curve Parameters 84

  • 7/26/2019 Use This Content

    6/136

    6.5 Percentage purity of pure drug 85

    6.6 Data for characteristic frequencies in FTIR spectrum of

    Telmisartan

    87

    6.7 Data for DSC thermogram parameters 91

    6.8 Flow properties of powder 92

    6.9 Physico-Chemical Characterization of Telmisartan SR Matrix

    Tablets

    94

    6.10 Dissolution data of formulation F1 95

    6.11 Dissolution data of formulation F2 96

    6.12 Dissolution data of formulation F3 97

    6.13 Dissolution data of formulation F4 98-99

    6.14 Dissolution data of formulation F5 99-100

    6.15 Dissolution data of formulation F6 100-

    101

    6.16 Dissolution data of formulation F7 101-

    102

    6.17 Dissolution data of formulation F8 102-

    103

    6.18 Dissolution data of formulation F9 103-

    104

    6.19 Different Kinetic models for Telmisartan SR Matrix Tablets (F1 to F9) 109-110

    6.20 Stability studies of optimized formulation of Telmisartan SR tablet

    (F3)

    117

  • 7/26/2019 Use This Content

    7/136

    LIST OF ABBREVIATIONS USED

    BP - British Pharmacopeia

    CDDS - Controlled drug delivery system

    CDR - Cumulative drug release

    CR - Controlled release

    DE - Dissolution Efficiency

    DSC

    EC

    - Differential Scanning Colorimetry

    - Ethyl Cellulose

    F - Formulation

    FTIR - Fourier Transform Infra-red

    GIT - Gastrointestinal tract

    g - Gram

    HPMC - HydroxyPropyl MethylCellulose

    HCl - Hydrochloric acid

    HBS - Hydro dynamically balanced system

    M - Molarity

    Mg - Miligram

  • 7/26/2019 Use This Content

    8/136

    MRT - Mean residence time

    MMC - migrating myoelectric complex

    MCC - Microcrystalline cellulose

    Mp - Melting point

    MDT - Mean Dissolution Time

    m -

    ml -

    Slope, Units of response

    Milli litre

    N - Normality

    pH - Negative Logaritham of hydrogen ion

    PVP - Polyvinyl pyrolidone

    ppm - parts per million

    RH - Relative humidity

    rpm - Revolutions per minute

    S.D.

    S.R

    SRDDS

    - Standard deviation

    - Sustained Release

    - Sustained Release Drug Delivery System

    t1/2 - Biological half-life

    UV-VIS - Ultraviolet-Visible

    USP - United State Pharmacopoeia

    max - Absorption maximum

    % - Percentage

    g - Microgram

    C - Degree Celsius

  • 7/26/2019 Use This Content

    9/136

  • 7/26/2019 Use This Content

    10/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 1

    1.INTRODUCTION

    ORAL DRUG DELIVERY SYSTEM

    Oral drug delivery is the most widely utilized route of administration among all the routes

    that have been explored for the systemic delivery of drug via pharmaceutical products of

    different dosage form. Oral route is considered most natural, uncomplicated, convenient and safe

    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 fluctuation in the drug concentration may lead to under

    medication or over medication as the cells, valves 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 over medication occurs.

    It is a reasonable assumption that drug concentration at the site of action is related to drug

    plasma level and that, in the great majority of cases, the intensity of effect is some function of

  • 7/26/2019 Use This Content

    11/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 2

    drug concentration at the target site. The objective of the most therapeutic regimens is to rapidly

    raise the plasma concentration to the required level and then to hold it constant for the desired

    duration of treatment .The extent to which this situation can be achieved depends on many

    factors, including the minimum effective concentration of the drug, the level at which side effects

    occur, the dose administered, the rate of drug release from the dosage form, the rate of

    elimination and the frequency of dosing. If the dose size and frequency of administration are

    correct, therapeutic steady state levels of the drug can be achieved rapidly and maintained by

    the repetitive administration of conventional oral dosage forms.

    Traditionally, patient is expected to take medication during the daytime hours. Hence,

    plasma levels are likely to fall to subtherapeutic levels overnight. Moreover the following major

    deficiencies of conventional dosage forms can reduce the patients compliance to dose regimen.

    Inconvenience and/or difficult use of drugs with very short duration of action or biological half-

    life, thus needs frequent dosing.

    Potential for peak-valley plasma levels, leading to toxicity and side effects and incomplete

    therapy.

    Instances of adverse effects, forgetfulness, and inconvenience of dosage forms.

    Need for large systemic concentrations in order to achieve adequate concentration at target site

    or action.

    Potential variations in oral absorption due to variations in GIT pH profile, presence and type of

    food and transit time in gut.

    These above mentioned major deficiencies of drug therapy based on repetitive

    administration of conventional single oral dosage form, In order to overcome the drawbacks of

  • 7/26/2019 Use This Content

    12/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 3

    conventional drug delivery system, 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.

    Thus, various modified drug products have been developed to release the active drug from

    the product at a controlled rate. The term controlled release drug products was previously used to

    describe various types of oral extended-release dosage forms, including sustained release,

    sustained action, prolonged action, slow release, long action, and retarded release.

    The USP/NF presently recognizes several types of modified-release dosage forms,

    1.Extended-release dosage forms (E.g. sustained release dosage forms, controlled release dosage

    forms)

    2.Delayed release dosage forms (e.g. enteric coated tablets)

    Modified-release dosage forms:

    It is defined as one for which the drug-release characteristics of time course and/ or

    location are chosen to accomplish therapeutic or convenience objectives not offered by

    conventional dosage forms such as solutions, ointments or compressed tablets and capsules.

    Extended-release dosage form:

    It is defined as the one that allows at least a twofold reduction in frequent dosing

    compared to the drug presented in a conventional form (e.g., a solution or an immediate release

    dosage form).

    Sustained- release dosage forms:

    It is defined as any drug or dosage form modification that prolongs the therapeutic

    activity of the drug. It provides prolonged but not necessarily uniform release ofdrug.

  • 7/26/2019 Use This Content

    13/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 4

    The United States Pharmacopoeia has adopted the term extendedrelease whereas the

    British Pharmacopoeia has adopted the term slow release. United States Food and Drug

    Administration has adopted the term Prolonged release. However the literature survey indicates

    that the most widely used terms today are sustained release & controlled release.

    Figure 1.1: A hypothetical plasma concentrationtime profile from conventional multiple

    and single doses of sustained and controlled delivery formulations.

    Potential advantages of sustained release systems1:

    1. Avoid patients complianceproblems.

    2. Employ less total drug

    a) Minimize or eliminate local side effects.

    b) Minimize or eliminate systemic side effects.

    c) Obtain less potentiation or reduction in drug activity with chronic use.

  • 7/26/2019 Use This Content

    14/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 5

    d) Minimize drug accumulation with chronic dosing.

    3.Improve efficiency in treatment

    a)

    Cures or controls condition more promptly.

    b) Improves control of condition i.e., reduced fluctuation in drug level.

    c) Improves bioavailability of some drugs.

    d) Make use of special effects, e.g. sustained-release aspirin for morning relief of

    arthritis by dosing before bed time.

    4.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. The time required for health care personnel to dispense and administer the

    drug and monitor patient is also reduced.

    Disadvantages:

    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) Of drugs normally administered in varying strengths.

    6) Reduced potential for dosage adjustment Stability problems.

  • 7/26/2019 Use This Content

    15/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 6

    7) More rapid development of tolerance and counseling.

    8) Need for additional patient education and counseling.

    Properties of drugs relevant to sustained release formulations:

    The drug properties that influence the incorporation of the drug into a sustained release

    dosage form can be classified as:

    1) Physicochemical properties

    2) Biological properties

    Physicochemical properties are those that can be determined by in vitro experiments. Biological

    properties are those that result from typical pharmacokinetic studies of absorption, distribution,

    metabolism, and elimination characteristics of drugs.

    Table 1.1: Drug properties suitable for sustained release2:

    Property Explanation

    1) Physicochemical properties

    Dose Size If dose is greater than 0.5 g it is a poor candidate for a sustained

    release system since the product size will be exceptionally large.

    Aqueous solubility Extremes in aqueous solubility will be undesirable. For drugs with

    low water solubility, they will be difficult to incorporate in sustained

    release mechanism. The lower limit is 0.1 mg/ml. Drugs with very

    high solubility are equally difficult to incorporate. pH dependant

    solubility will be another problem.

  • 7/26/2019 Use This Content

    16/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 7

    Partition Coefficient Drugs that are highly lipid soluble or very water-soluble i.e. extremes

    in partition coefficient, will show either low flux in tissue or rapid flux

    followed by accumulation in the tissues. Both cases are undesirable.

    The value of K at which optimum activity observed is 1000/1 in

    octanol/water.

    Drug Stability As sustained release systems are designed to release their contents

    over much of the length of GI tract, drugs, which are unstable in

    environment of intestine, are poor candidates for sustained release.

    2) Biological Properties

    Absorption Drugs that are slowly absorbed or absorbed with variable absorption

    rate are poor candidates for sustained release systems. Lower limit on

    absorption rate constant is 0.25 h1

    Distribution Drugs with high apparent volume of distribution, which in turn

    influences rate of elimination, are poor candidates.

    Metabolism As long as location, extent, and rates of metabolism are known and the

    rate constant(s) for the processes are not too large, successful

    sustained release systems can be developed.

    Duration of action The biological half-life and hence duration of action plays a major

    role. Drugs with biological half-life less than 2 hrs should not be used.

    At the other extreme a drug with half-life of greater than 8 hrs have

  • 7/26/2019 Use This Content

    17/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 8

    inherently sustained action.

    Therapeutic range Drugs with narrow therapeutic range requires precise control over the

    blood levels of the drug, placing a constraint on sustained release

    dosage form.

    CLASSIFICATION OF SUSTAINED RELEASE

    POLYMERIC DRUG DELIVERY SYSTEMS3,4

    :

    The various sustained release polymeric systems can be classified, depending upon the

    mechanism controlling the drug release, as follows:

    A.

    Chemical ly controll ed systems

    i)Biodegradable systems

    ii)Drugpolymer conjugate

    B.Di ff usion-controll ed systems

    i)Membrane-reservoir systems

    ii)Matrix systems

    C.Dissolu tion controll ed system

    i)Encapsulation/ reservoir type

    ii)Matrix systems

  • 7/26/2019 Use This Content

    18/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 9

    A.Diffusion controlled systems:

    Basically diffusion process shows the movement of drug molecules from a region of higher

    concentration to one of lower concentration. This system is of two types:

    a)Membrane-Reservoir type:

    A core of drug surrounded by polymer membrane, which controls the release rate, characterizes

    reservoir devices.

    Advantages:

    Zero order delivery is possible; release rate varies with polymer type.

    Disadvantages:

    Systems must be physically removed from implant sites.

    Difficult to deliver high molecular weight compounds.

    Increased cost per dosage unit, potential toxicity if system fails.

    Ficks first law of diffusion describes the diffusion process.

    J= -D dc/dx.

    D = diffusion coefficient in area/time

    dc/dx = change of concentration c with distance x

    b)Matrix type:

    Matrix system is characterized by a homogenous dispersion of solid drug in a polymer

    mixture.

    Advantages:

  • 7/26/2019 Use This Content

    19/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 10

    Easier to produce than reservoir or encapsulated devices, can deliver high molecular

    weight compounds.

    Disadvantages:

    Cannot provide zero order release, removal of remaining matrix is necessary for

    implanted system.

    Higuchi has derived the appropriate equation for drug release from this system,

    M = k t1/2

    M = amount of drug released per unit area,

    K= constant

    B.Dissolution Controlled Systems:

    a)Reservoir type:

    Drug is coated with a given thickness coating, which is slowly dissolved in the content

    of gastro intestinal tract. By altering layers of drugs with the rate controlling coats as shown in

    figure no.1.2, 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.

  • 7/26/2019 Use This Content

    20/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 11

    Figure 1.2: Schematic representation of diffusion controlled drug release reservoir.

    b)Matrix type:

    The more common type of dissolution controlled dosage forms as shown in figure

    no.1.3. It can be either a drug impregnated sphere or a drug impregnated tablet, which will be

    subjected to slow erosion.

    Figure 1.3. Systematic representation of diffusion controlled drug release matrix system.

    Advantages:

  • 7/26/2019 Use This Content

    21/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 12

    All the advantages of matrix dissolution system.

    Removal from implant site is not necessary.

    Disadvantages:

    Difficulty to control kinetics owing to multiple processes of release.

    Potential toxicity of degraded polymer must be considered.

    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

    release 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.

    Advantages:

    Zero order release rates are obtainable.

    Preformulation is not required for different drugs.

    Release of drug is independent of the environment of the system.

    Disadvantages:

    System can be much more expensive than conventional counter parts.

    Quality control is more extensive than most conventional tablets.

    F.pH

    independent formulations:

    A buffered controlled release formulation is prepared by mixing a basic or acidic drug with

    or more buffering agents, granulating with appropriate pharmaceutical excipients and coating

  • 7/26/2019 Use This Content

    22/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 13

    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.

    Figure 1.4. 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

    FABRICATION OF ORAL CONTROLLED RELEASE DELIVERY EMPLOYS

    VARIOUS METHODS1,3,4

    :

    Hydrophilic matrix, Plastic matrix, Barrier resin beads, Fat embedment, Repeat action, Ion

    exchange resin, Soft gelatin depot capsules and Drug complex.

    Hydrophilic matrix system:

    Drug delivery technologists usually tend to consider all hydrophilic delivery systems as

    hydrogels. Hydrogels are hydrophilic macro molecular networks that after swelling maintain

    their shape due to permanent links. The very high water content and special surface properties of

  • 7/26/2019 Use This Content

    23/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 14

    swollen form give them the ability to stimulate natural tissues. They have been used in controlled

    drug delivery because of their good tissues compatibility and easy manipulation of swelling level

    and their by solute permeability.

    Hydrogel Based Drug Delivery Systems are classified as:

    Diffusion controlled release system:

    a)Reservoir System:

    Figure 1.5. Hydrogel formation in Reservoir Systems

    It consists of polymeric membrane surrounding a core containing the drug. The rate-limiting step

    for drug release is diffusion through the outer membrane.

    b)Matrix system:

    The drug is dispersed throughout the three dimensional structure of the hydrogel. Release

    occurs due to diffusion of the drug throughout the water filled pores.

    Types:

    Swellable

    Non-swellable

  • 7/26/2019 Use This Content

    24/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 15

    Figure 1.6: Drug release from a matrix tablet

    Swellable controlled release systems:

    During the release life of swellable matrix system, three fronts are generally expected.

    The swelling front, the boundary between the still glassy polymer and its rubbery:

    1. State.

    2. The diffusion front, the boundary in the gel layer between the solid as yet undisclosed,

    drug and the dissolved drug.

    3. The erosion front, the boundary between the matrix and the dissolution medium.

  • 7/26/2019 Use This Content

    25/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 16

    The measurement of front positions gives the possibility to determine three important

    parameters related to the behavior of the matrix i.e. the rate of water uptake, the rate of drug

    dissolution and the rate of matrix erosion associated with the movements of the swelling

    front, diffusion front and erosion front respectively. These parameters are strictly linked to

    the drug release kinetics from matrix.

    Figure 1.7: Parameters definitions for swellable soluble matrix

    Ds- Solvent diffusion co-efficient in the drug/polymer matrix.

    Dd-Drug diffusion co-efficient in the swollen polymers.

    CsDrug solubility at the drug core interface ( R ).

    C*- Drug volume fraction at the gel /solution interface.

    CPolymer volume fraction at R.

    CdPolymer volume fraction at S.

    CcpPolymer volume fractioning in the glassy core.

    CcdDrug volume fraction in the glassy core .

  • 7/26/2019 Use This Content

    26/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 17

    Non- swellable controlled release systems.

    The systems are hollow containing an inner core of the drug surrounded in a water

    insoluble polymer membrane. The polymer can be applied by coating or by wet granulation

    technique. The drug release mechanism across the membrane involves its portioning into the

    membrane with subsequent release into the surrounding fluid by diffusion.

    Table No. 1.2: Characteristics of major classes of sustained release drug delivery system3

    Type of System Mechanism of drug

    release

    Advantages Limitations

    A) Chemically controlled systems

    1.Biodegradable

    system

    Matrix-forming polymer

    contains hydrolytically or

    enzymatically labile

    bonds and drug is

    uniformly dissolved or

    dispersed in this matrix.

    Erosion of polymer by

    hydrolysis or enzymatic

    cleavage.

    Erosion

    process has a

    direct effect on

    drug release

    Difficult to achieve

    targeted drug delivery

    system.

    2. Drug-Polymer

    Conjugates

    Drug molecules

    chemically bonded to a

    polymer backbone. The

    Provides the

    possibility for

    sustaining the

    Improper attachment of

    drugs to carriers can alter

    rate of drug excretion

  • 7/26/2019 Use This Content

    27/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 18

    drug is released through

    hydrolytic or enzymatic

    cleavage of these bonds

    drug release

    over a

    prolonged

    period

    from body

    B. Diffusion-controlled systems

    1. Membrane-

    Reservoir

    System

    Follows either a solution

    diffusion mechanism or

    an osmotic mechanism

    Osmotic pressure

    can control the

    drug release.

    Observed only in non-

    porous membranes

    2. Matrix system Drug release from matrix

    system can occur by any

    of these mechanisms:

    Diffusion, Erosion or

    Swelling.

    One of the least

    complicated

    approaches to

    manufacture the

    sustained release

    dosage forms

    1.first-order release

    behavior with

    continuously

    diminishing release

    rate.

    2.Increasing

    diffusional resistance

    and decreasing area at

    the diffusion front.

    C. Dissolution controlled system

    Encapsulation Drug particles are

    covered with layer of

    polymers. As polymer

    Drug can be

    delivered when

    needed.

    Immediate release

    cannot be achieved.

    High cost of coating

  • 7/26/2019 Use This Content

    28/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 19

    erodes drug gets released. increased.

    MATRIX SYSTEMS1,5,6

    :

    Matrix system is formulated in such manner as to make the contained drug available over

    an extended period following administration. A typical controlled release system is designed to

    provide constant or nearly constant drug levels in plasma with reduced fluctuations via slow

    release over an extended period of time. In practical terms, an oral controlled release should

    allow a reduction in dosing frequency as compared to when the same drug is presented as a

    conventional dosage form.

    The following table (Table No. 1.3) summarizes the characteristics of various types of

    matrix systems.

    Table 1.3: The characteristics of various types of matrices are:

    Slow eroding matrix Hydrophilic matrix Plastic matrix

    Mechanism of The portion of the drug Two competing The drug is

    drug release intended to have mechanisms: Fickian granulated

    sustained action is diffusional release & with an inert

    combined with lipid or relaxation release. plastic material

    cellulosic material and such as

    processed into granules polyethylene,

    that can be placed into polyvinyl

    capsules or tableted. acetate or

  • 7/26/2019 Use This Content

    29/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 20

    When these granules are

    combined with granules

    of drug prepared without

    the special lipid or

    cellulosic excipients, the

    untreated portion

    provides the immediate

    drug effect, and the

    treated portion provides

    the prolonged effect.

    polymethacryla

    te, and the

    granulation is

    compressed

    into tablets

    Example of

    drug

    candidates

    SLOW-K (Summit), a

    KCl sugarcoated

    sustained release tablet

    containing KCl in a wax

    matrix.

    Sparingly soluble

    drug

    Gradumet

    (Abbott)

    Matrix

    characteristics

    Insoluble, erodible Hydrophilic Insoluble, inert

    Material Carnauba wax, Stearyl

    alcohol, Stearic acid,

    Polyethylene glycol,

    Castor wax, Polyethylene

    Methylcellulose

    (400cps, 4000cps),

    Hydroxyethylcellulo

    se, HPMC, Sodium

    Polyethylene,

    polyvinyl

    chloride,

    methyl

  • 7/26/2019 Use This Content

    30/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 21

    glycol monostearate, CMC, Sodium acrylates-

    Triglycerides alginate, methacrylate

    Galactomannose copolymer,

    Carboxypolymethyle ethyl cellulose.

    ne

    Advantages of the matrix systems: (Robinson J.R. and Lee V.H.L., 1987)

    Easy to manufacture.

    Versatile, effective, low cost. Can be made to release high molecular weightcompounds.

    Reproducible release profile.

    Since the drug is dispersed in the matrix system, accidental leakage of the total drug component

    is less likely to occur, although occasionally, cracking of the matrix material can cause

    unwanted release.

    Disadvantages of the matrix systems: (Robinson J.R. and Lee V.H.L., 1987)

    The remaining matrix must be removed after the drug has been released

    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.

    APPROACHES FOR PREPARATION OF MATRIX DOSAGE FORMS:

    There are many approaches for preparing matrices for sustained drug delivery.

    Table No. 1.4 summarizes common approaches for the same7.

  • 7/26/2019 Use This Content

    31/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 22

    Method Features Advantages Limitations

    Melt

    Granulati

    on

    Granules prepared

    by melting all the

    ingredients together

    and then screening

    the congealed mass

    through appropriate

    mesh.

    Useful for enhancing

    solubility and hence

    dissolution of poorly

    water soluble drugs

    and for modifying

    release

    characteristics of

    drugs delivered

    transdermally.

    1. No need of

    solvents.

    2. Fewer processing

    steps.

    3.No need of high

    compression.

    4.Uniform dispersion

    of fine particles

    occurs.

    5.Stability at varying

    pH and moisture

    levels

    1.High energy

    requirement.

    2.Not suitable for

    thermosensitive

    materials including low

    melting binders.

    3.Not suitable for

    blends containing high

    melting binders,

    thermosensitive drugs

    and/ or additives.

    Direct Drug embedded 1. More economic 1. Segregation of

    Compressi matrices prepared by due to fewer steps, particles.

    on direct compression of less equipments &2. Not suitable for

    blend of drug andless labour.

  • 7/26/2019 Use This Content

    32/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 23

    additives and release

    retardant

    2. Less time

    consuming.

    3.No batch-to-batch

    variation.

    4.Useful for moisture

    sensitive and

    thermosensitive

    drugs.

    drugs having large

    doses.

    3.Development of

    static charges during

    blending.

    4.Difficulty in uniform

    distribution of color.

    Wet Granules containing 1. Less energy is 1. Solvents are

    Granulatidrug are prepared required. required.

    onusing adhesive

    2. Useful for 2. High compression is

    properties ofthermosensitive required.

    binders.drugs.

    MECHANISM OF DRUG RELEASE FROM MATRIX DEVICES3,5

    1.Dissolution controlled release:

    Controlled release oral products employing dissolution as a rate limiting step are in

    principle simplest to prepare. Even if the drug has a rapid rate of dissolution it is possible

    incorporate it into a tablet with a carrier, which has a slow rate of dissolution.

  • 7/26/2019 Use This Content

    33/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 24

    We assume the dissolution process where the rate of diffusion from the surface to the

    bulk of the solution through an unstirred liquid fill is the rate limiting step. In this case the

    dissolution process at study would be described by the Noyes-Whitney equation:

    dc/dt = kDA(Cs-C)1

    dc/dt is the dissolution rate

    KD is the dissolution rate constant

    Cs is the saturation solubility of the drug and

    C is the concentration of the drug in the bulk of the solution.

    In relation diffusion expression kD equals to D/V.I.

    From the above expression it can be seen that rate of dissolution i.e, availability is

    approximately proportional to the solubility of the drug in the dissolution media (Cs) provide

    constant surface area and diffusion path length are maintain.

    This equation predicts constant dissolution rate as long as enough drug is present to

    maintain Cs constant and provided surface area does not change. For spherical particles the

    change in surface area can be related to the weight of the particle and substituted in the diffusion

    equation to give an expression that related dissolution to the weight remaining (W).

    W01/3- W1/3 = K1 D.2

    W0 is the initial weight.

    K1

    D is the cube root dissolution expression.

    The above equation described dissolution rate of spherical particles when surface area

    and diffusional path are changing.

    The common forms of dissolution controlled formulations for into two categories:

  • 7/26/2019 Use This Content

    34/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 25

    a) Encapsulation dissolution control.

    b) Matrix dissolution control.

    Encapsulation dissolution control:

    These methods generally involve coating individual particles of drug with a slow

    dissolving material. The coated particles can be directly compressed into tablets as in space tabs

    or placed in capsules as in spansule products. Since the time required for dissolution of the coat

    is a function of thickness and aqueous solubility, one can obtain repeat or sustained action by

    employing a narrow or a wide spectrum of coated particles of varying thickness respectively.

    Matrix dissolution control:

    Those methods involve compressing the drug with a slowly dissolving carrier into a

    tablet form. Here the rate of drug availability is controlled by the rate of penetration of

    dissolution fluid into the matrix. This in turn can be controlled by porosity if the tablet matrix,

    the presence of hydrophobic additives and the wettability of the tablet and particle surface.

    1.DIFFUSION CONTROLLED RELEASE:

    Diffusion controlled release products are basically of two types

    a) Encapsulated diffusion control

    b) Matrix diffusion control

    Encapsulated diffusion control:

    In this system water insoluble polymeric material encases a core of drug. Drug

    will partition into the membrane and exchange with the fluid surrounding the particles or tablet.

    The release rate is given by equation.

  • 7/26/2019 Use This Content

    35/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 26

    dm/dt = ADKC/I..(3)

    A is the area

    D is the diffusional coefficient

    K is the partition coefficient of the drug between the membrane and drug core

    I is the diffusional path length

    C is the concentration difference across the membrane.

    An important parameter in the above equation is the partition coefficient which is defined

    as the concentration, of the drug in the membrane over the concentration of the drug in the core.

    If the partition coefficient is high, the core will be depleted of the drug release rate all the terms

    in the right hand side of the above equation must be held constant.

    Methods to develop reservoir type devices include press coating, air suspension coating

    techniques. Microencapsulation process is a commonly used procedure to coat the drug articles

    to be incorporated.

    Matrix diffusion control:

    In this type a solid drug is dispersed in an insoluble matrix and the rate of release of drug

    is independent if the rate of diffusion and not on the rate of solid dissolution. The equation

    describing drug release from this system has been derived by Higuchi.

    Q = [ (DE/T) (2A-ECs) Cst] ......... 4

    Q is the diffusion coefficient of drug released per unit area at timet

    D is the diffusion coefficient of drug in release medium

    E is the porosity of matrix

  • 7/26/2019 Use This Content

    36/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 27

    Cs solubility of drug in the release medium

    T is the tortuosity of matrix

    A is the concentration of drug in the tablet expressed as gm/ml

    The assumptions made in above equation are:

    1. A pseudo steady state is maintained during release

    2. A >> Cs i.e., excess solute is present

    3. C = 0 in solution at all times (perfect sink)

    4. Drug particles are much smaller than matrix

    5. The diffusion coefficient remains constant

    6. No interaction occurs between drug and matrix.

    For the purpose of data treatment the above equation is usually reduced to Q = Kt1/2

    Where K is a constant so that the plot of amount drug released versus the square root of

    time should be linear if the system is diffusion controlled.

    Depending on the properties of the matrix and the polymer system, deviation from

    constant release can occur. For example in the case of insoluble matrix, the drug may have to

    diffuse through tortuous correction factor is added to the release equation.

    The most popular drug delivery system has been matrix system containing uniformly

    dissolved or dispersed drug such as tablets and granules, because of its ease of fabrication.

    However the release behavior is inherently first order in nature with continuously diminishing

    release rate for all three standard geometries: slab, cylinder and sphere. This is the result of

    increase in diffusional resistance and a decrease in effective area at the diffusion front as drug

  • 7/26/2019 Use This Content

    37/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 28

    release proceeds. Use of zero order delivery systems will optimize the therapy by maintaining

    dug concentration for prolonged periods.

    Methods for altering the kinetics of the drug release from the inherent first order behavior

    to zero order have involved the use of geometry factors, erosion, dissolution, control, swelling

    mechanisms, non-uniform drug loading and matrixmembrane combinations.

    Swelling phenomena are generally encountered in both hydrophilic and hydrophobic

    matrices during release of the entrapped water soluble drugs in aqueous environment. The

    release of water soluble drugs from initially dehydrated hydrogels generally involves the

    simultaneous absorption of water and desorption of drug via a swelling controlled diffusion

    mechanism.

    In case where the sorption process is completely governed by the rate of the polymer

    relaxation the so called case 11 transport, characterized by linear time dependence in both the

    amount diffused and the penetrating swelling front position results. In most system the

    intermediate situation, which is often termed non- Fickian or anomalous diffusion will prevail

    wherever the rates of diffusion and polymer relaxation are comparable.

    By Korsemeyer and Peppas equation one can express the fraction released Mt/M as a

    power function of timet for the short time period.

    Mt/M = K1tn

    Where K is a constant characteristic of the system and n is an exponent characteristic

    of the mode of transport. For n = 0.5, the drug release follows the well known Fickian diffusion

    mechanism. For n > 0.5 non- Fickian or anomalous diffusion behavior is generally observed. The

  • 7/26/2019 Use This Content

    38/136

    INTRODUCTION

    Department of Industrial Pharmacy, NIPS Page 29

    special case of n= 1 describes a case 11 transport mechanism; the drug release from such devices

    having constant geometry will be constant (zero order).

    Usually non Fickian release is observed till the polymer chains rearrange to an

    equilibrium state. Once the hydrogel matrix is significantly hydrated, drug release becomes

    Fickian. The relative importance of relaxation and diffusion can in principle be estimated with

    the Deborah number, De which is the ratio of the characteristic relaxation time to a

    characteristic diffusion time ( = L2/D) where L is length.

    De = /

    When De

  • 7/26/2019 Use This Content

    39/136

    AIM AND OBJECTIVE

    Department of Industrial Pharmacy, NIPS Page 30

    2.AIM AND OBJECTIVE

    The basic goal of therapy is to achieve a steady state blood or tissue level that is

    therapeutically effective and non-toxic for an extended period of time. Sustained

    release drug delivery systems, with an aim of improved patient compliance, better

    therapeutic efficacy, less side effects and reduced dosage regimen with less toxicity

    for treatment for many acute and chronic diseases.

    Anti hypertensive drugs are used for the treatment of hypertension. Telmisartan has

    been demonstrated to be superior to previous peptide receptor antagonists and

    angiotensin converting enzyme (ACE) inhibitors because of its enhanced specificity,

    selectivity and tolerability.

    Matrix tablets are very useful in the field of healthcare for sustained release dosage

    regimen.

    Keeping this in view, the present investigation has been aimed at designing suitable

    sustained release matrix tablets using polymers like HPMC K100M, Ethyl cellulose,

    Xanthan Gum.

    The major objectives of the investigation are as follows:

    Preparation of calibration curve.

    To perform compatibility studies by using IR spectroscopy.

    To perform preformulation studies like flowing properties and bulking density for

    powders of drug and polymers.

    To evaluate prepared formulations for physical parameters like weight variation,

    friability and hardness etc.

  • 7/26/2019 Use This Content

    40/136

    AIM AND OBJECTIVE

    Department of Industrial Pharmacy, NIPS Page 31

    To formulate matrix tablets of Telmisartan by wet granulation method by using different

    polymer like Hydroxypropyl methylcellulose K100M (HPMC K100M), Ethyl cellulose,

    Xanthan Gum.

    To ascertain the release mechanics and kinetics of drug release from compressed matrix

    tablets.

    To perform stability studies as per ICH guidelines.

    BASICS FOR DRUG SELECTION AND DOSAGE SELECTION.

    For many drugs, the optimal therapeutic response is observed only when adequate

    blood levels are achieved and maintained with minimum variations, (the matrix

    tablets will give more consistent blood levels).

    Drugs with biological half-life 2-4 hrs are viewed as suitable candidate for sustained

    release dosage form. So it is suitable to deliver this drug in SR dosage form.

    Generally a lower daily dose of Telmisartan potassium may be sufficient for long-

    term administration (40 mg).

    Maintenance of dose is usually 40mg per day.

  • 7/26/2019 Use This Content

    41/136

    Department of Industrial Pharmacy, NIPS Page 32

    PLAN OF WORK

    3.PLAN OF WORK

    Literature Survey

    Selection of drug candidate

    Procurement of Drug and Excipients

    Characterization of Telmisartan

    - Melting Point

    - UV

    - IR

    - DSC

    Preparation of granules

    (Wet granulation method)

    Evaluation of granules

    Tests for compatability Tests for bulk properties

    -Infra Red Spectroscopy - Angle of repose,

  • 7/26/2019 Use This Content

    42/136

    Department of Industrial Pharmacy, NIPS Page 33

    PLAN OF WORK

    -Differential scanning colorimetry - Bulk density

    Compression of granules obtained by wet

    granulation method into tablets

    Evaluation of Tablets

    -Thickness and Diameter

    -Weight variation

    -Hardness

    -Drug content

    -

    Friability

    -In-Vitro Release Studies

    Data Compilation and

    Analysis of result.

    -Tapped density

    -Flow Rate,

    -Carrsindex

  • 7/26/2019 Use This Content

    43/136

    Department of Industrial Pharmacy, NIPS Page 34

    LITERATURE SURVEY

    4.LITERATURE SURVEY

    Saptarshi D., et al. An attempted was to formulate the oral sustained release metformin

    hydrochloride matrix tablets by using hydroxyl methyl cellulose polymer (HPMC) as rate

    controlling factor and to evaluate drug release parameters as per various release kinetic models.

    It is observed that the basic goal of therapy in the development of metformine hydrochloride

    release dosage form is to increase bioavailability; reduce risk of hospitalization, deliver drug at a

    near constant rate for approximately 12hrs; independent of food intact and gastrointestinal pH.

    The dry granulation technique was used to compress the tablet as powder showed the poor

    flowability; less cohesiveness during direct compression and due to moisture sensitivity and

    tendency to hydrolyte, wet granulation technique was not selected for the present work8.

    Dhirendra K, et al. The aim of the present study was to formulate once daily sustained

    release matrix tablets of Stavudine to increase therapeutic efficacy, reduce frequency of

    administration and improve patient compliance. The sustained release tablets were prepared by

    direct compression and formulated using different drug:polymer ratios, formulations such as F1

    to F15. Hydrophilic polymers like Hydroxypropyl methylcellulose (HPMC), Carboxy methyl

    cellulose (CMC) and starch 1500 were used. Compatibility of the drug with various excipients

    was studied. The compressed tablets were evaluated and showed compliance with

    pharmacopoeial standards9.

    Madhusmruti K, et al. The aim of the present work was to develop sustain release

    matrix formulation of Propanolol hydrochloride and investigate the effects of both hydrophilic

    and hydrophobic polymer on in-vitro drug release. Matrix tablets were prepared by direct

    compression method using different concentrations of Hydroxypropyl methylcellulose (HPMC)

    and Ethyl Cellulose (EC). Prepared formulations were subjected to various studies like hardness,

  • 7/26/2019 Use This Content

    44/136

    Department of Industrial Pharmacy, NIPS Page 35

    LITERATURE SURVEY

    friability, thickness, %drug content, weight variation, dynamic of water uptake and erosion etc.

    Tablets were subjected to in-vitro drug release in 0.1N HCl (pH 1.2) for first 2 hours followed by

    phosphate buffer (pH 6.8) remaining time10

    .

    Jayaprakash S., et al . The sustained release (SR) tablets of Ambroxol Hydrochloride

    were prepared by Wet granulation method. The effect of hydrophilic matrices on the behavior of

    Ambroxol hydrochloride using different polymers and their combinations. The prepared tablets

    were evaluated for physical characteristics such as Hardness, Thickness, Friability, Weight

    Variation, Content uniformity and in-vitro release behavior. The drug release from the optimized

    formulation was found to follow zero order kinetics11.

    Umesh D.S, et al . The objective of the present study was to develop once daily

    sustained release tablets of Aceclofenac by wet granulation using carboxypolymethylene

    polymer. The drug excipient mixtures were subjected to preformulation studies while the tablets

    were subjected to physicochemical studies, in vitro drug release, stability studies and validation

    studies12

    .

    Raghavendra R.N.G, et al. The main objective of the present work was to develop

    sustained release matrix tablets of water soluble Tramadol hydrochloride using different

    polymers viz. Hydroxypropyl methylcellulose (HPMC) and natural gums like karaya gum (KG)

    and carrageenan gum (CG). Varying ratios of drug and polymer like 1:1 and 1:2 were selected

    for the study. After fixing the ratio of drug and polymer for control the release of drug upto

    desired time, the release rates were modulated by combination of two different rates controlling

    material and triple mixture of three different rate controlling material13

    .

  • 7/26/2019 Use This Content

    45/136

    Department of Industrial Pharmacy, NIPS Page 36

    LITERATURE SURVEY

    Anton S et al. The objective of the present work was to develop sustained release matrix

    tablets of Ondansetron Hydrochloride(5mg) formulated employing Hydroxy propyl Methyl

    Cellulose(HPMC), polymer and the sustained release behavior of the tablets was investigated.

    Tablets were prepared by wet granulation methods14

    .

    Parasuram R.R, et al . The aim of the present study was to design an oral sustained

    release matrix tablet of Glipizide and to optimize the drug release profile using response surface

    methodology. The tablets were prepared by the wet granulation method using HPMC K100 and

    Eudragit L 100 as matrix forming polymers. A central composite design for 2 factors at 3 levels

    each was employed to systematically optimized drug release profile. HPMC K100 and Eudragit

    L 100 were taken as the independent variables. The dependent variables selected were % of drug

    released in 2 h (release 12 h)15.

    Kalyani C, et al. The objective of the study was to design oral sustained release matrix

    tablets of Zidovudine using Hdroxypropyl methylcellulose (HPMC)K 4M, Guar Gum and Ethyl

    Cellulose as the retardant polymers and study the effect of various formulation factors such as

    polymer proportion, polymer type and effect of filler type on the in-vitro release of drug. Matrix

    tablets were by Wet granulation method and prepared tablet were evaluated weight variation,

    %friability, hardness, thickness and in-vitro dissolution studies. All the granules and

    formulations showed compliance with pharmacopeial standards.In-vitro release studies revealed

    that the release rate decrease with increase polymer proportion and hydrophobic polymers retard

    the drug release more than hydrophilic polymers16

    .

    Tapan K.P, et al . The aim of the current study was to design an oral sustained release

    matrix tablet of Metformin HCl and to optimize the drug release profile using response surface

    methodology. Tablets were prepared by non-aqueous wet granulation method using HPMC K

  • 7/26/2019 Use This Content

    46/136

    Department of Industrial Pharmacy, NIPS Page 37

    LITERATURE SURVEY

    15M as matrix forming polymer. A central composition design for 2 factors at 3 levels each was

    employed to systematically optimize drug release profile. HPMC K 15M (X1) and PVP K 30

    (X2) were taken as the independent variables. The dependent variable selected were % of drug

    release in 1 hr (rel1hr), % of drug release in 8 hr (rel8hr), and time to 50% drug release (t50%)17

    .

    Basak S. C et al . Monolithic matrix tablets of Ambroxol Hydrochloride were formulated

    as sustained release tablets employing Hydroxypropyl methylcellulose polymer, and the

    sustained release matrix tablets containing 75 mg Ambroxol hydrochloride were developed using

    different drug polymer ratios of Hydroxy propyl Methyl Cellulose. Tablets were prepared by

    direct compression. Formulation was optimized on the basis of acceptable tablet properties and

    in vitro drug release18

    .

    Varshosaz J et al . The objective of this work was to develop matrix sustained release

    tablets of highly water-soluble Tramadol HCl using natural gums Xanthan [X gum] and Guar [G

    gum] as cost-effective, nontoxic, easily available and suitable hydrophilic matrix systems

    compared with extensively investigated hydrophilic matrices (ie, Hydroxypropyl methylcellulose

    [HPMC]/Carboxymethyl cellulose [CMC] with respect to in-vitro drug release rate) and

    hydration rate of the polymers. Matrix tablets of Tramadol (dose 100 mg) were produced by

    Direct compression method19

    .

    Muhammad K.S et al . The present study was conducted to investigate the low viscosity

    grades of Hydroxypropyl methylcellulose(HPMC), and ethyl cellulose(EC), in sustaining the

    release of water insoluble drug, Naproxen from the matrix tablets. Both HPMC and EC were

    incorporated in the matrix system separately or in combinations by wet granulation technique. In

    vitro dissolution studies indicated that EC significantly reduced the rate of drug release

  • 7/26/2019 Use This Content

    47/136

    Department of Industrial Pharmacy, NIPS Page 38

    LITERATURE SURVEY

    compared to HPMC in 12hr testing time. But, no significant difference was observed in the

    release profiles of matrix tablets made by higher percentages of EC20

    .

    Yeole P.G. et al . In the present investigation, an attempt has been made to increase

    therapeutic efficacy, reduce frequency of administration, and improve patient compliance, by

    developing sustained release matrix tablets of Diclofenac sodium. Sustained release matrix

    tablets of Diclofenac sodium, were developed by using different drug:polymer ratios, such as

    F1(1:0:12), F2(1:0:16), F3(1:0:20), F4(1:0:24) and F5(1:0:28). Xanthan gum was used as matrix

    former, and microcrystalline cellulose as diluents. Compressed tablets were evaluated for

    uniformity of weight, content of active ingredient, friability, hardness, thickness, in vitro

    dissolution using basket method and swelling index21

    .

    Hosseinali T et al. A sustained release tablet formulation should ideally have a proper

    release profile insensitive to moderate changes in tablet hardness that is usually encountered in

    manufacturing. In the study, matrix Aspirin (acetylsalicylic acid) tablets with ethyl cellulose

    (EC), Eudragit RL100, Eudragit S100 were prepared by direct compression. The release

    behaviors were then studied in two counterpart series of tablets with hardness difference of three

    Kp units, and compared by non-linear regression analysis22

    .

    Raghuram R.K et al . The objective of the present study was to develop once-daily

    sustained- release matrix tablets of Nicorandil, a novel potassium channel opener used in

    cardiovascular diseases. The tablets were prepared by the wet granulation method. Ethanolic

    solutions of ethyl cellulose(EC), Eudragit RL-100, Eudragit RS-100, and poly vinyl pyrrolidone

    were used as granulating agents along with hydrophilic matrix materials like Hydroxy propyl

    Methyl Cellulose(HPMC), sodium carboxy methyl cellulose, and sodium alginate23

    .

  • 7/26/2019 Use This Content

    48/136

    Department of Industrial Pharmacy, NIPS Page 39

    LITERATURE SURVEY

    Chandira M. et al . The present investigation attempt has been made increase therapeutic

    efficacy , reduce frequency of administration and improve patient Compliance, by developing

    sustained release matrix tablets of Zidovudine, Were developed by using drug polymer ratio

    kollidon SR, HPMC k15M, HPMC K100M as matrix former.aii lubricated formulation were

    compressed by direct compression and wet granulation method24

    .

    Krishnaiah Y.S.R. et al. The objective of the present study is to carry out

    pharmacokinetic evaluation of oral controlled release formulation (guar gum-based three layer

    matrix tablets) containing highly soluble Metoprolol tartrate as a model drug. Six healthy

    volunteers participated in the study, and a two way crossover design was followed. The plasma

    concentration of Metoprolol tartrate was estimated by reverse-phase HPLC. The

    pharmacokinetic parameters were calculated from the plasma concentration of metoprolol

    tartrate versus time data25

    .

    Suvakanta D. et al . In this paper were reviewed mathematical models used to determine

    the kinetic of drug release from drug delivery system the quantitative analysis of the values are

    obtained in dissolution/ release rate is easier when mathematical formula used to describe the

    process. The mathematical modeling can optimize to design therapeutic design of therapeutic

    device to yield information on the various efficacy of various release models26

    .

    Ganesan V. et al ., The objective of the study was to develop guar gum matrix tablets for

    oral controlled release of Ambroxol hydrochloride. According to the theoretical release profile

    calculation, a twice daily sustained release formulation should release 19.6 mg of Ambroxol

    hydrochloride in 1 hour like conventional tablets, and 5.2 mg per hour upto 12 hours. Ambroxol

    hydrochloride matrix tablets containing either 30%wt/wt of low viscosity (F-III), 25% w/w

  • 7/26/2019 Use This Content

    49/136

    LITERATURE SURVEY

    Department of Industrial Pharmacy, NIPS Page 40

    medium viscosity (F-VI) or 20 %wt/wt high viscosity (F-IX) guar gum showed sustained

    release27

    .

    Gothi G.D. et al. In the present investigation an attempt was made to reduce the

    frequency of dose administration, to prevent nocturnal heart attack and to improve the patient

    compliance by developing extended release (ER) matrix tablet of metoprolol succinate. The

    effect of concentration of hydrophilic (hydroxypropyl methylcellulose [HPMC K 100 M],

    Xanthan gum) on the release rate of metoprolol succinate was studied28

    .

    Keny R.V. et al . The present study was aimed to develop once daily extended release

    matrix tablets of minocycline hydrochloride, using hydroxyl propyl methyl cellulose either alone

    or in combination with ethyl cellulose as the matrix material in different proportions. The

    formulated tablets were also compared with a marketed product. The results of the dissolution

    study indicate that formulations FC-IV, FC-V, FC-VI, shows maximum drug release upto 24 hr.

    Drug release from matrix occurred by combination of two mechanisms diffusion of tablet matrix

    and erosion of tablet surface which was reflected from Highuchis model and Erosion plot29

    .

    Tabandeh H. et al . A sustained release tablet formulation should ideally have a proper

    release profile insensitive to moderate changes in tablet hardness that is usually encountered in

    manufacturing. In the study, matrix Aspirin (acetylsalicylic acid) tablets with ethyl cellulose

    (EC), Eudragit RL100, Eudragit S100 were prepared by direct compression. The release

    behaviors were then studied in two counterpart series of tablets with hardness difference of three

    Kp units, and compared by non-linear regression analysis30

    .

  • 7/26/2019 Use This Content

    50/136

    LITERATURE SURVEY

    Department of Industrial Pharmacy, NIPS Page 41

    Sasidhar R.L.C et al. In the present investigation an attempt has been made to increase

    therapeutic efficacy, reduced frequency of administration and improved patient compliance by

    developing controlled release matrix tablets of Telmisartan. Telmisartan was formulated as oral

    controlled release matrix tablets by using poly (ethylene oxides) {Polyox WSR 303}.The aim of

    this study was to investigate the influence of polymer level and type if fillers namely lactose

    [soluble filler], microcrystalline cellulose and anhydrous dibasic calcium phosphate [insoluble

    fillers] on the release rate and mechanism of Telmisartan from

    matrix tablets31

    .

    Chopra S et al. The aim of the present research work was to systemically device a model

    of factors that would yield an optimized controlled release tablet dosage form of an anti-

    hypertensive agent, Telmisartan. Independent variables such as the amount of the release

    retardant polymers- Methocel K15M(X1), Methocel K100M(X2) and Sodium carboxy methyl

    cellulose(X3) were optimized using a 3-Factor, 3-level Box-Behnken statistical design. The

    dependent variables selected were the burst release in 15min (Y1), cumulative % release of

    Telmisartan after 60 min. (Y2) and hardness (Y3) of the tablets32

    .

    Shruti C et al. The aim of the present research work was to systemically device a model

    of factors that would yield an optimized sustained release tablet dosage form of an anti-

    hypertensive agent, Telmisartan, using response surface methodology by employing a 3- Factor,

    3-level Box-Behnken statistical design. Independent variables studied were the amount of the

    release retardant polymers-HPMC K15M(X1), HPMC K100M(X2) and Sodiumcarboxy

    methylcellulose(X3). The dependent variables selected were the burst release in 15min.(Y1),

    cumulative % release of Telmisartan after 60 min. (Y2) and hardness(Y3) of the tablets with

    constraints on the Y2= 31-35%33

    .

  • 7/26/2019 Use This Content

    51/136

    Department of Industrial Pharmacy, NIPS Page 42

    DRUG PROFILE

    DRUG PROFILE

    TELMISARTAN34-38

    Telmisartan is in the drug class of angiotensin receptor blockers (ARBs) and is prescribed for the

    treatment of high blood pressure, reducing the risk of heart attack, stroke, or death from

    cardiovascular causes, Telmisartan is an angiotensin II receptor antagonist (ARB) used in the

    management of hypertension. Generally, angiotensin II receptor blockers (ARBs) such as

    Telmisartan bind to the angiotensin II type 1 (AT1) receptors with high affinity, causing

    inhibition of the action of angiotensin II on vascular smooth muscle, ultimately leading to a

    reduction in arterial blood pressure. Recent studies suggest that Telmisartan may also have

    PPAR-gamma agonistic properties that could potentially confer beneficial metabolic effects

    Structure of Telmisartan

    Chemical Name:-

    [4-[[4-methyl-6-(1-methylbenzimidazol-2-yl)-2-propylbenzimidazol-1-yl]methyl] 1,1 biphenyl]2 carboxylic acid.

    Molecular Formula:- C33H30N4O2

    Molecular Weight:- 514.61 DaltonsDescription:- White to off-white crystalline powder.

    Melting range:- Between 265oC - 272

    oC

    Solubility:- Practically insoluble in water, slightly soluble in methanol, sparingly soluble in

    methylene chloride, it dissolves in 1M sodium hydroxide.

    Partial coefficient:- The Octanol/buffer partial coefficient (log P) for Telmisartan is

    approximately 3.20

    Storage & Stability:- Stored in well-closed, light-resistant containers at 5-30C. When stored

  • 7/26/2019 Use This Content

    52/136

    Department of Industrial Pharmacy, NIPS Page 43

    DRUG PROFILE

    under these conditions, Telmisartan generally is stable for 24 months after the date of

    manufacture.

    Indication:- For the treatment of hypertension

    Clinical Pharmacology:-Telmisartan is an orally active nonpeptide angiotensin II antagonist that acts on the AT1 receptor

    subtype. New studies suggest that telmisartan may also have PPAR agonistic properties thatcould potentially confer beneficial metabolic effects. This observation is currently being

    explored in clinical trials. Angiotensin II is formed from angiotensin I in a reaction catalyzed by

    angiotensin-converting enzyme (ACE, kininase II). Angiotensin II is the principal pressor agentof the renin-angiotensin system, with effects that include vasoconstriction, stimulation of

    synthesis and release of aldosterone, cardiac stimulation, and renal reabsorption of sodium.

    Telmisartan works by blocking the vasoconstrictor and aldosterone secretory effects of

    angiotensin II.

    Mechanism of Action:-Telmisartan interferes with the binding of angiotensin II to the angiotensin II AT1-receptor by

    binding reversibly and selectively to the receptors in vascular smooth muscle and the adrenal

    gland. As angiotensin II is a vasoconstrictor, which also stimulates the synthesis and release ofaldosterone, blockage of its effects results in decreases in systemic vascular resistance.

    It does not inhibit the angiotensin converting enzyme, other hormone receptors, or ion channels.

    Pharmacokinetic properties;

    Absorption

    Absorption of telmisartan is rapid although the amount absorbed varies. The mean absolutebioavailability for telmisartan is about 50 %. When telmisartan is taken with food, the reduction

    in the area under the plasma concentration-time curve (AUC0-) of telmisartan varies from

    approximately 6 % (40 mg dose) to approximately 19 % (160 mg dose). By 3 hours after

    administration, plasma concentrations are similar whether telmisartan is taken fasting or withFood.

    Linearity/non-linearityThe small reduction in AUC is not expected to cause a reduction in the therapeutic efficacy.There is no linear relationship between doses and plasma levels. Cmax and to a lesser extent

    AUC

    increase disproportionately at doses above 40 mg.

    Distribution

    Telmisartan is largely bound to plasma protein (>99.5 %), mainly albumin and alpha-1 acid

    glycoprotein. The mean steady state apparent volume of distribution (Vdss) is approximately 500

    Biotransformation

    Telmisartan is metabolised by conjugation to the glucuronide of the parent compound. Nopharmacological activity has been shown for the conjugate.

    EliminationTelmisartan is characterised by biexponential decay pharmacokinetics with a terminal eliminateion half-life of >20 hours. The maximum plasma concentration (Cmax) and, to a smaller extent,

    the area under the plasma concentration-time curve (AUC), increase disproportionately with

    dose. There is no evidence of clinically relevant accumulation of telmisartan taken at the

    recommended dose. Plasma concentrations were higher in females than in males, withoutrelevant influence on efficacy.

    After oral (and intravenous) administration, telmisartan is nearly exclusively excreted with the

  • 7/26/2019 Use This Content

    53/136

    Department of Industrial Pharmacy, NIPS Page 44

    DRUG PROFILE

    faeces, mainly as unchanged compound. Cumulative urinary excretion is 99.5%), mainly albumin and a1-acid glycoprotein. Binding is

    not dose-dependent.

    Biot8ransformationMinimally metabolized by conjugation to form a pharmacologically inactive acylglucuronide;

    the glucuronide of the parent compound is the only metabolite that has been identified in humanplasma and urine. The cytochrome P450 isoenzymes are not involved in the metabolism of

    telmisartan.

    Dosage and Administration:The usually effective dose telmisartan is 4080 mg once daily. Some patients may already

    benefit at a daily dose of 20 mg. In cases where the target blood pressure is not achieved,

    telmisartan dose can be increased to a maximum of 80 mg once daily.

    ContraindicationsTelmisartan is contraindicated during pregnancy. Like other drugs affecting the renin angiotensin

    system (RAS), telmisartan can cause birth defects, stillbirths, and neonatal deaths. It should notbe taken by breastfeeding women since it is not known whether the drug passes into the breast

    milk.

    Side effectstachycardia andbradycardia (fast or slow heartbeat), hypotension (low blood pressure), edema

    (swelling of arms,legs, lips, tongue, or throat, the latter leading to breathing problems), and

    allergic reactions.

  • 7/26/2019 Use This Content

    54/136

    Department of Industrial Pharmacy, NIPS Page 45

    EXCIPIENTS PROFILE

    4.2.EXCIPIENTS PROFILE

    Hypromellose (Hydroxy Propyl Methyl Cellulose)

    A.

    Nonproprietary Names

    BP: Hypromellose

    JP: Hydroxypropyl methylcellulose

    PhEur: Hypromellosum

    USP: Hypromellose

    B.Synonyms

    Benecel MHPC; E464; hydroxypropyl methylcellulose; HPMC; Methocel;

    methylcellulose propylene glycol ether; methyl hydroxypropylcellulose;Metolose; Tylopur.

    C.Chemical Name and CAS Registry Number

    Cellulose hydroxypropyl methyl ether [9004-65-3]

    D.Molecular Weight

    Molecular weight is approximately 10 0001 500 000.

    E.Structural Formula

  • 7/26/2019 Use This Content

    55/136

    Department of Industrial Pharmacy, NIPS Page 46

    EXCIPIENTS PROFILE

    Where R is H, CH3, or CH3CH (OH) CH2

    F.Functional Category

    Coating agent; film-former; rate-controlling polymer for sustained release; stabilizing

    agent; suspending agent; tablet binder; viscosity-increasing agent.

    G.Applications in Pharmaceutical Formulation or Technology

    Hypromellose is widely used in oral, ophthalmic and topical pharmaceutical

    formulations. In oral products, hypromellose is primarily used as a tablet binder, in film-coating,

    and as matrix for use in extended-release tablet formulations. Concentrations between 2% and

    5% w/w may be used as a binder in either wet- or dry-granulation processes. High-viscosity

    grades may be used to retard the release of drugs from a matrix at levels of 1080% w/w in

    tablets and capsules. Depending upon the viscosity grade, concentrations of 220% w/w are used

    for film-forming solutions to film-coat tablets. Lower-viscosity grades are used in aqueous film-

    coating solutions, while higher-viscosity grades are used with organic solvents. Hypromellose at

    concentrations between 0.451.0% w/w may be added as a thickening agent to vehicles for eye

    drops and artificial tear solutions.

    H.Description

    Hypromellose is an odorless and tasteless, white or creamy-white fibrous or granular

    powder.

    I.Typical Properties

    Acidity/alkalinity: pH = 5.58.0 for a 1% w/w aqueous solution.

    Ash: 1.53.0%, depending upon the grade and viscosity.

  • 7/26/2019 Use This Content

    56/136

    Department of Industrial Pharmacy, NIPS Page 47

    EXCIPIENTS PROFILE

    Autoignition temperature: 360C

    Density (bulk): 0.341 g/cm3

    Density (tapped): 0.557 g/cm

    3

    Density (true): 1.326 g/cm3

    Melting point: browns at 190200C; chars at 225230C.

    Glass transition temperature is 170180C.

    Moisture content:

    Hypromellose absorbs moisture from the atmosphere; the amount of water absorbed

    depends upon the initial moisture content and the temperature and relative humidity of the

    surrounding air.

    Solubility:

    Soluble in cold water, forming a viscous colloidal solution; practically insoluble in

    chloroform, ethanol (95%), and ether, but soluble in mixtures of ethanol and dichloromethane,

    mixtures of methanol and dichloromethane, and mixtures of water and alcohol. Certain grades of

    hypromellose are soluble in aqueous acetone solutions, mixtures of dichloromethane and propan-

    2-ol, and other organic solvents.

    Viscosity (dynamic):

    A wide range of viscosity types are commercially available. Aqueous solutions are most

    commonly prepared, although hypromellose may also be dissolved in aqueous alcohols such as

    ethanol and propan-2-ol provided the alcohol content is less than 50% w/w.

  • 7/26/2019 Use This Content

    57/136

    Department of Industrial Pharmacy, NIPS Page 48

    EXCIPIENTS PROFILE

    Typical viscosity values for 2% (w/v) aqueous solutions of Methocel (Dow Chemical

    Co.). Viscosities measured at 20C.

    Methocel product USP 28 designation Nominal viscosity (mPa s)

    Methocel K100 Premium LVEP 2208 100

    Methocel K4M Premium 2208 4000

    Methocel K15M Premium 2208 15 000

    Methocel K100M Premium 2208 100 000

    Methocel E4M Premium 2910 4000

    Methocel F50 Premium 2906 50

    Methocel E10M Premium CR 2906 10 000

    Methocel E3 Premium LV 2906 3

    Methocel E5 Premium LV 2906 5

    Methocel E6 Premium LV 2906 6

    Methocel E15 Premium LV 2906 15

    Methocel E50 Premium LV 2906 50

    Metolose 60SH 2910 50, 4000, 10 000

    Metolose 65SH 2906 50, 400, 1500, 4000

    Metolose 90SH 2208 100, 400, 4000, 15000

  • 7/26/2019 Use This Content

    58/136

    Department of Industrial Pharmacy, NIPS Page 49

    EXCIPIENTS PROFILE

    J.Stability and Storage Conditions

    Hypromellose powder is a stable material, although it is hygroscopic after drying.

    Solutions are stable at pH 311. Increasing temperature reduces the viscosity of solutions.

    Hypromellose undergoes a reversible solgel transformation upon heating and cooling,

    respectively. The gel point is 5090C, depending upon the grade and concentration of material.

    K.Incompatibilities

    Hypromellose is incompatible with some oxidizing agents. Since it is nonionic,

    hypromellose will not complex with metallic salts to form insoluble precipitates.

  • 7/26/2019 Use This Content

    59/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 50

    Ethyl cellulose:

    Ethyl cellulose is a chain of -anhydro glucose units joined together by acetal linkage.

    It is prepared from wood

    .

    pulp or chemical cotton by treatment with alkali and

    ethylation of the alkali cellulose with ethyl chloride. Commercial ethyl cellulose has an

    ethoxy content of 43-50 %. A 47 % product softens at 140oC and is soluble in ethyl acetate,

    benzene, toluene, xylem and butanol.

    The properties and pharmaceutical applications of ethyl cellulose are as follows:

    Synonyms: Ethocel

    Description: A tasteless, odourless free flowing, white to light tan powder.

    Specific gravity: 1.14

    Softening temperature: 152-1620

    C

    Emperical formula and Molecular weight:

    Ethylcellulose with complete ethoxyl substitution (DS = 3) is C12H23O6 (C12H22O5)n

    C12H23O5 where n can vary to provide a wide variety of molecular weights. Ethylcellulose, an

    ethyl ether of cellulose, is a long-chain polymer of -anhydroglucose units joined together by

    acetal linkages.

    Structural formula:

  • 7/26/2019 Use This Content

    60/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 51

    Functional Category:

    Coating agent; flavoring fixative; tablet binder; tablet filler; viscosity-increasing agent.

    Solubility:

    Insoluble in water, glycerin, propylene glycol, but soluble in varying degrees in organic solvents.

    Viscosity:

    A property of ethyl cellulose such as tensile strength, elongation and flexibility

    depends largely upon the degree of polymerization, which can be measured by viscosity.

    Therefore, within each type-based ethoxyl content there exists low to high viscosity types,

    based on degree of polymerization.

    Stability:

    It is resistant to alkali both dilute and concentrated but more sensitive to acidic

    materials than cellulosic ethers.

    Applications in pharmacy:

    1. Binder in tablets:

    Ethyl cellulose may be dry blended and wet granulated with a solvent such as alcohol.

    Tablets made with ethyl cellulose as binder tent to exhibit poor dissolution and poor drug

    absorption.

    2.

    Coating material for tablets:

    Ethyl cellulose by itself forms a water insoluble film coating. It is commonly used

    with Hydroxypropyl methylcellulose to alter the solubility of the film. Other materials

    may be used for this as well.

  • 7/26/2019 Use This Content

    61/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 52

    3. Coating material for stabilization:

    Ethyl cellulose dissolved in i so propanol i s us ed t o co a t pa rt i c l es of dr u gs

    t o form microcapsules. This type of microcapsules slows dissolution of the drug as

    a function of microcapsule wall thickness.

  • 7/26/2019 Use This Content

    62/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 53

    3. Xanthan Gum

    A.Nonproprietary Names

    BP: Xanthan gum

    PhEur: Xanthani gummi

    USPNF: Xanthan gum

    B.Synonyms

    Corn sugar gum; E415;Keltrol; polysaccharide B-1459;Rhodigel; Vanzan NF;Xantural.

    C.Chemical Name and CAS Registry Number

    Xanthan gum [11138-66-2]

    D.Structural formula

    E.Molecular Weight

    Approximately 2000000.

    F.Functional Category

    Stabilizing agent; suspending agent; viscosity-increasing agent.

  • 7/26/2019 Use This Content

    63/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 54

    G.Description

    Xanthan gum occurs as a cream- or white-colored, odorless, free-flowing, fine powder.

    H.

    Typical Properties

    Acidity/alkalinity: pH = 6.08.0 for a 1% w/v aqueous solution.

    Freezing point: 0C for a 1% w/v aqueous solution.

    Heat of combustion: 14.6 J/g (3.5 cal/g)

    Melting point: chars at 270C.

    Refractive index: n20

    D = 1.333 for a 1% w/v aqueous solution.

    Solubility:

    Practically insoluble in ethanol and ether; soluble in cold or warm water.

    Viscosity (dynamic):

    12001600 mPa s (12001600 cP) for a 1% w/v aqueous solution at 25C.

    I.Applications in Pharmaceutical Formulation or Technology

    Xanthan gum is widely used in oral and topical pharmaceutical formulations, cosmetics,

    and foods as a suspending and stabilizing agent. It is also used as a thickening and emulsifying

    agent. It is nontoxic, compatible with most other pharmaceutical ingredients, and has good

    stability and viscosity properties over a wide pH and temperature range. Xanthan gum gels show

    pseudoplastic behavior, the shear thinning being directly proportional to the shear rate. The

    viscosity returns to normal immediately on release ofshear stress. When xanthan gum is mixed

    with certain inorganic suspending agents, such as magnesium aluminum silicate, or organic

    gums, synergistic rheological effects occur.

  • 7/26/2019 Use This Content

    64/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 55

    J.Stability and Storage Conditions

    Xanthan gum is a stable material. Aqueous solutions are stable over a wide pH range (pH 312),

    although they demonstrate maximum stability at pH 410 and temperatures of 1060C.

    K.Incompatibilities

    Xanthan gum is an anionic material and is not usually compatible with cationic

    surfactants as precipitation occurs. Xanthan gum is compatible with most synthetic and natural

    viscosity increasing agents.

  • 7/26/2019 Use This Content

    65/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 56

    Cellulose, Microcrystalline: (Rowe R.C., et al., 2003)

    A.Nonproprietary Names

    BP: Microcrystalline cellulose JP: Microcrystalline cellulose

    PhEur: Cellulosum microcristallinum USPNF: Microcrystalline cellulose

    B.Synonyms

    Avicel pH; Celex; cellulose gel; Celphere; Ceolus KG; crystalline cellulose; E460;

    Emcocel; Ethispheres; Fibrocel; Pharmacel; Tabulose; Vivapur.

    C.

    Chemical Name and CAS Registry Number

    Cellulose [9004-34-6]

    D.Molecular Weight

    36 000

    E.Structural Formula

    F.Functional Category

    Adsorbent; suspending agent; tablet and capsule diluent; tablet disintegrant.

    G.Description

  • 7/26/2019 Use This Content

    66/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 57

    Microcrystalline cellulose is a purified, partially depolymerized cellulose that occurs as a

    white, odorless, tasteless, crystalline powder composed of porous particles. It is commercially

    available in different particle sizes and moisture grades that have different properties and

    applications.

    H.Typical Properties

    Density (bulk): 0.337 g/cm3;

    Density (tapped): 0.478 g/cm3;

    Density (true): 1.5121.668 g/cm3

    Flowability: 1.41 g/s

    Melting point: chars at 260270C.

    Specific surface area: 1.061.12 m2/g forAvicel PH-101;

    1.211.30 m2/g forAvicel PH-102;

    0.781.18 m2/g forAvicel PH-200.

    1.211.30 m2/g forAvicel PH-102;

    Moisture content:

    Typically less than 5% w/w. However, different grades may contain varying amounts of

    water. Microcrystalline cellulose is hygroscopic. (Avicel pH

    - 102= 5.0)

    Solubility:

    Slightly soluble in 5% w/v sodium hydroxide solution; practically insoluble in water,

    dilute acids, and most organic solvents.

  • 7/26/2019 Use This Content

    67/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 58

    I.Applications in Pharmaceutical Formulation or Technology

    Microcrystalline cellulose is widely used in pharmaceuticals, primarily as a

    binder/diluent in oral tablet and capsule formulations where it is used in both wet-granulation

    and direct compression processes. In addition to its use as a binder/diluent, microcrystalline

    cellulose also has some lubricant and disintegrant properties that make it useful in tableting.

    Uses of microcrystalline cellulose

    Use Concentration (%)

    Adsorbent 2090

    Antiadherent 520

    Capsule binder/diluent 2090

    Tablet disintegrant 515

    Tablet binder/diluent 2090

    J.

    Stability and Storage Conditions

    Microcrystalline cellulose is a stable though hygroscopic material. The bulk material

    should be stored in a well-closed container in a cool, dry place.

    K.Incompatibilities

    Microcrystalline cellulose is incompatible with strong oxidizing agents.

  • 7/26/2019 Use This Content

    68/136

    EXCIPIENTS PROFILE

    Department of Industrial Pharmacy, NIPS Page 59

    TALC :

    Synonyms: French chalk, purified talc, talcum, soapstone.

    Description:

    A very fine, white to grayish white, impalable, odorless crystalline powder,

    unctuous, adheres readily to skin, soft to touch and free from granules.

    Empirical formula: Mg6(Si2O5)4(OH)4.

    Functional category:

    USP: Tablet and/or capsule lubricant, glidant and anti caking agent

    BP: Talc dusting powder

    Others: Antiadherent

    Typical Properties:

    Density: 19-24 Ib/ft

    Solubility: Insoluble in water, organic solvents, cold acids and dilute alkalis.

    Stability and storage conditions:

    Stable, Preserve in a well-closed container.

    Incompatibilities:

    Quaternary ammonium compounds.

    Safety:

    Talc dust exposure in the modern mining process does not appear to be

  • 7/26/2019 Use This Content