throbber
Encyclopedia of
`Pharmaceutical Technology
`
`Second Edition
`Volume 3
`Ped-Z
`Pages 2045-3032
`
`edited by
`James Swarbrick
`President
`PharmaceuTech, Inc., Pinehurst, North Carolina
`and
`Vice President for Scientific Affairs, aaiPharma, Inc.
`Wilmington, North Carolina, U.S.A.
`and
`James C. Boylan
`Pharmaceutical Consultant
`Gurnee, Illinois, U.S.A.
`
`MARCEL
`
`DEKKER
`
`MARCEL DEKKER, INC.
`
`NEW YORK • BASEL
`
`MYLAN EXHIBIT 1013
`
`

`

`Cover Art: Leigh A. Rondano, Boehringer angelheim Pharmaceuticals, Inc.
`
`ISBN: Volume 1:
`Volume 2:
`Volume 3:
`Prepack:
`
`0-824 7-2822-X
`0-824 7-2823-8
`0-824 7-2824-6
`0-8247-2825-4
`
`ISBN: Online:
`
`0-8247-2820-3
`
`This book is printed on acid-free paper.
`
`Headquarters
`Marcel Dekker, Inc.
`270 Madison Avenue, New York, NY 10016
`tel: 212-696-9000; fax: 212-685-4540
`
`Eastern Hemisphere distribution
`Marcel Dekker AG
`Hutgasse 4, Postfach 812, CH-4001 Basel, Switzerland
`tel: 41-61-261-8482; fax: 41-61-261-8896
`
`World Wide Web
`http://www.dekker.com
`
`Copyright© 2002 by Marcel Dekker, Inc. except as occasionally noted on the opening page of each article. All Rights Reserved.
`
`Neither this book nor any part may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying,
`microfilming, and recording, or by any information storage and retrieval system, without permission in writing from the publisher.
`
`Current printing (last digit)
`
`10 9 8 7 6 5 4 3 2 1
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`

`

`TABLET FORMULATION
`
`Larry L. Augsburger
`University of Maryland, Baltimore, Maryland
`
`Mark J. Zellhofer
`University Pharmaceuticals of Maryland, Inc., Baltimore, Maryland
`
`INTRODUCTION OBJECTIVES
`OF TABLET FORMULATION
`
`The best new therapeutic entity in the world is of little
`value without an appropriate delivery system. Tableted
`drug delivery systems can range from relatively simple
`immediate-release formulations to complex extended- or
`modified-release dosage forms. The most important role of
`a drug delivery system is to get the drug "delivered" to the
`site of action in sufficient amount and at the appropriate
`rate; however, it must also meet a number of other
`essential criteria. These include physical and chemical
`stability, ability to be economically mass produced in a
`manner that assures the proper amount of drug in each and
`every dosage unit and in each batch produced, and, as far
`as possible, patient acceptability (for example, reasonable
`size and shape, taste, color, etc. to encourage patients to
`take the drug and thus comply with the prescribed dosing
`regimen).
`The discovery of new therapeutic entities always
`initiates excitement, but the contributions of the
`formulation specialist are either not well understood or
`are often taken for granted and thus remain "unsung."
`However, the drug and its delivery system cannot be
`separated. The general design criteria for tablets are given
`as follows
`
`1. Optimal drug dissolution and, hence, availability from
`the dosage form for absorption consistent with intended
`use (i.e., immediate or extended release).
`2. Accuracy and uniformity of drug content.
`3. Stability, including the stability of the drug substance,
`the overall tablet formulation, disintegration, and the
`rate and extent of drug dissolution from the tablet for an
`extended period.
`4. Patient acceptability. As much as possible, the finished
`product should have an attractive appearance, includ(cid:173)
`ing color, size, taste, etc., as applicable, in order to
`maximize patent acceptability and encourage compli(cid:173)
`ance with the prescribed dosing regimen.
`
`Encyclopedia of Phannaceutical Technology
`Copyright © 2002 by Marcel Dekker, Inc. All rights reserved.
`
`5. Manufacturability. The formulation design should
`allow for the efficient, cost-effective, practical pro(cid:173)
`duction of the required batches.
`
`That tablets can be formulated to uniquely meet these
`criteria accounts for their emergence as the most prevalent
`oral solid dosage form. Although several different types of
`tablets may be distinguished, they are mostly made by
`compression, intended to be swallowed whole and
`designed for immediate release. This paper presents a
`systematic approach to the design and formulation of
`immediate-release compressed tablets.
`
`MODERN TABLET FORMULATION DESIGN
`AND MANUFACTURE
`
`Tablet dosage forms have to satisfy a unique design
`compromise. The desired properties of rapid or controlled
`disintegration and dissolution of the primary constituent
`particles must be balanced with the manufacturability and
`esthetics of a solid compact resistant to mechanical attrition.
`Excipients are critical to the design of the delivery
`system and play a major role in determining its quality and
`performance (1). They may be selected to enhance
`stability (antioxidants, UV absorbers), optimize or modify
`drug release ( disintegrants, hydrophilic polymers, wetting
`agents, biodegradable polymers), provide essential man(cid:173)
`ufacturing technology functions (binders, glidants, lubri(cid:173)
`cants), enhance patient acceptance (flavors), or aid in
`product identification (colorants). Thus a tablet formu(cid:173)
`lation is not a random combination of ingredients, but
`rather a carefully thought out, rational formulation
`designed to satisfy the above criteria.
`A long list of possible excipients is available to the
`formulation scientist, but certain external factors such as
`cost, functional reliability, availability, and international
`acceptance govern their selection. For example, although
`the official compendia provide standards for identity and
`purity of excipients, monographs may not provide tests to
`
`2701
`
`

`

`2702
`
`assure their functionality. For instance, the NF monograph
`for Compressible Sugar provides no test for compress(cid:173)
`ibility. The monograph for Lactose USP does not address
`the many particle size and tableting grades meeting
`monograph standards. The NF monograph for Pregelati(cid:173)
`nized Starch refers to grades that are "compressible and
`flowable in character," but provides no specifications or
`tests for these properties. Nor do the monograph tests for
`disintegrants and lubricants necessarily relate to their
`functionality. The need to provide functionality tests or
`tests for properties clearly related to functionality may be
`as important as controlling identity and purity (2). This
`point has been made even more apparent in recent years
`with the emergence of multiple sources of such modem
`excipients as direct-compression filler-binders and the
`various classes of "super" disintegrants.
`A major problem currently being faced by multi(cid:173)
`national firms and others who market in the international
`arena, is the lack of universal acceptability of excipients in
`different countries. The selection of excipients for
`international markets is often a compromise between
`functional efficacy, local restrictions, and cost and
`availability in the countries where the product is to be
`made. In recent years, the globalization of the pharma(cid:173)
`ceutical industry has brought about an intense interest in
`developing harmonized pharmacopeial excipient stan(cid:173)
`dards, Good Manufacturing Practices (GMP) for excipient
`manufacture, and safety evaluation guidelines for new
`excipients to eliminate or avoid trade barriers between
`different countries (3). The International Pharmaceuticals
`Excipients Council (IPEC), which consists of producers,
`users, and pharmaceutical scientists, was launched in 1991
`to assist regulatory authorities in the United States, Japan,
`and Europe with harmonization. The separate organi(cid:173)
`zations later formed in the United States (IPEC-Americas),
`Europe (IPEC-Europe), and Japan (JPEC) are now known
`as TriPEC and include, as of 1993, more than 100 excip(cid:173)
`ient and pharmaceutical firms (3).
`
`PREFORMULATION
`
`The objective of preformulation studies is to develop a
`portfolio of information about the drug substance to serve
`as a set of parameters against which detailed formulation
`design can be carried out. Preformulation investigations
`are designed to identify those physicochemical properties
`of drug substances and excipients that may influence the
`formulation design, method of manufacture, and pharma(cid:173)
`cokinetic-biopharmaceutical properties of the resulting
`product.
`
`Tablet Formulation
`
`Following is a generalized preformulation protocol
`appropriate for tablet dosage forms. For certain tests, it is
`assumed that the drug substance is multisourced (a
`previously new chemical entity whose patent has expired
`and which is available to the generic market) for which a
`USP monograph exists.
`
`Identity and Purity
`
`The study of any drug substance must start with the
`determination of identity and purity. Such tests are
`necessary to identify degradents and contaminants and
`may include organoleptic tests for color, odor, and taste.
`Purity tests can be found in the USP for almost all
`marketed compounds. Alternative methods can be
`employed only if they are validated against the USP
`procedure. Tests other than potency, which can help to
`identify or determine the purity of compounds, are melting
`point, specific rotation, pH, heavy metals, residue on
`ignition, etc. Impurities can occasionally affect stability,
`and metal contamination can catalyze chemical reactions.
`Impurities can also alter the color of drug substances.
`Techniques can be utilized to give a quantitative estimate
`of impurities such as the impurity index (II) and the
`homogeneity index (HI). An ordinary impurity test can be
`found in the USP that estimates impurities by thin-layer
`chromatography (TLC).
`
`Crystal Properties and Polymorphism
`
`Many drug substances appear in more than one
`polymorphic form. The form is determined by certain
`conditions during the crystallization step. Occasionally
`drug substances are precipitated in such a way that
`molecules do not organize themselves in any set pattern,
`resulting in an amorphous powder. It is also possible for
`solids to entrap solvents stoichiometrically to form
`solvates.
`Even though they are chemically identical, the different
`polymorphic forms of a compound are associated with
`different free energies, and, therefore, have different
`physical properties that can impact significantly on
`product performance (4). These include differences in
`solubility and dissolution rate (affecting bioavailability),
`solid-state stability (affecting potency), deformation
`characteristics (affecting compactibility), and particle
`size and shape (affecting powder density and flow
`properties). The form with the lowest energy is more
`stable than the others. Although the other polymorphs are
`thus energetically unfavored, if kept dry, they may persist
`indefinitely and are called "metastable." A metastable
`
`

`

`Tablet Formulation
`
`form may be preferred, particularly for its ability to
`dissolve more rapidly.
`Polymorphic transformation can take place during
`pharmaceutical processing, such as particle size
`reduction, wet granulation, drying, and even during the
`compaction process (5). Tests employed to determine
`crystal properties include differential thermal analysis
`(DTA), differential scanning calorimetry (DSC), and
`X-ray diffraction (4). See also the article Thermal
`Analysis of Drugs and Drug Products by D. Giron in this
`encyclopedia.
`
`Particle Size, Shape, and Surface Area
`
`Probably no characteristics of a drug substance are
`more important than particle characteristics in determin(cid:173)
`ing
`its performance
`in a
`formulation. This
`is
`particularly true in those cases where the drug is a
`poorly soluble nonelectrolyte or a free acid form with
`poor solubility at low pH values. Such drugs are likely
`to exhibit dissolution-rate-limited absorption, and if
`dissolution does not
`take place rapidly enough, a
`therapeutic concentration in the body fluids may never
`be achieved, the peak plasma concentration may be
`significantly delayed, or much of the drug may bypass
`that region of the gastrointestinal (GI) tract where
`absorption
`is best. Particle size reduction (e.g.,
`rnicronization) is often utilized to enhance dissolution
`rate. Small particles present a larger surface area per
`unit weight to the dissolution media and hence dissolve
`more rapidly than large particles. Particle size and
`surface area are two of the most important properties
`determining the solubility rate of a drug and thus
`potentially
`its bioavailability. There are numerous
`examples of bioavailability problems and bioinequiva(cid:173)
`lence due to the inappropriate particle size of the drug
`substance.
`Particle size and shape also play an extremely
`important role in the homogeneity of powder blends and
`the unblending of powders in a mixer. Segregation in
`handling or during the compaction process has a
`significant effect on the content uniformity of the finished
`products. Particle size can also affect the stability of a drug
`substance in that it governs the surface area available for
`oxidation and hydrolysis. Surface area is critical for
`interaction with excipients in tablet dosage forms and can
`greatly affect stability. Methods to determine particle size
`and shape include light microscopy, scanning electron
`microscopy, sieve analysis, and various electronic
`sensing-zone particle counters. Methods available for
`surface area measurement include air permeability and
`various gas adsorption techniques.
`
`2703
`
`Bulk Powder Properties
`
`Bulk powder properties are extremely important in
`pharmaceutical processing (6). Knowledge of the true
`and bulk densities of the drug substance as well as of the
`excipients is extremely useful in
`
`• Providing perspective as to the size of the final tablet
`and the size and type of processing equipment needed,
`• Anticipating problems in the physical mixing of
`powders and the homogeneity of intermediate and
`final products because significant differences in true
`densities can result in segregation,
`• Anticipating problems in flow properties, since that
`property is affected by density, and
`• Identifying differences in different lots and raw
`materials from different suppliers because different
`polymorphic forms can be expected to exhibit different
`true densities.
`
`A comparison of true particle density, apparent particle
`density, and bulk density can provide information on total
`porosity, interparticle porosity, and intraparticle porosity.
`Methods include true particle density measurements via
`helium pycnometry, mercury intrusion porosimetry, and
`poured and tapped bulk density.
`The influence of sorbed moisture on chemical stability
`and the flow and compaction of powders and granulations
`is well established. The moisture content and hygro(cid:173)
`scopicity of excipients is particularly important as total
`product processing as well as finished product stability can
`be affected. Hygroscopicity, moisture-sorption isotherms,
`and equilibrium moisture content can be determined by
`thermogravimetric analysis and Karl Fisher titration
`methods.
`The compactibility of relatively large-dose drug
`substances and formulations is another important property.
`Compactibility is of less concern for smaller-dose drugs for
`which direct compression fillers may be able to compensate
`for a lack of ability to form mechanically strong compacts.
`An instrumented tablet press (7) or compaction simulator
`(8) may be used to assess the relationship between the
`mechanical strength of the compact and the force ( or
`pressure) employed to form the tablet. This relationship is
`the easiest of all compaction measurements to establish and
`provides important information on the ability of the
`material to form practical compacts. Measures of compact
`mechanical strength include hardness ( or crushing force),
`tensile strength, and friability. Other more complex
`studies, more easily and perhaps best done using a
`compaction simulator, include measurement of the work or
`energy of compaction, pressure-density (Athy-Heckel)
`analysis, strain-rate sensitivity, and elastic recovery (9).
`
`--
`
`

`

`2704
`
`The Athy-Heckel analysis can provide information on
`deformation mechanism and give an estimate of the mean
`yield pressure of the material (10). A comparison of yield
`pressures determined at different punch speeds can give
`information on the strain-rate sensitivity of the material
`( 11 ). If the major components of the formulation (including
`the drug) are strain-rate sensitive, the tablets produced on a
`high-speed production press may exhibit lamination or
`capping. Excessive elastic recovery may also indicate such
`tablet failure. The Hiestand indices (bonding and brittle
`fracture) may be used to assess the compactibility of
`materials under laboratory conditions (12).
`For the evaluation of flow properties the following test
`methods may be used:
`
`• Angle of repose
`• Minimum orifice diameter,
`• Carr index,
`• Flow rate, and
`• Direct observation of weight variation during tableting
`runs.
`
`The ultimate goal of flow analysis is to identify the
`powder or powder blend that provides the least weight
`variation in the finished tablet. The more fluid the powder
`is, the more efficiently and reproducibly it should fill the
`die cavities of a tablet press. This more efficient and
`reproducible die fill should be reflected in increased tablet
`weights and reduced intertablet weight variation (13).
`
`Solubility and Permeability
`
`In many cases, the rate of dissolution in gastrointestinal
`fluids is the rate-limiting step in absorption. The
`bioequivalence requirements established by the FDA
`define low solubility as " ... <5 mg/mL in water, and slow
`dissolution rate to be <50% in 30 minutes" (14). However,
`the solubility of a drug should be considered together with
`its dose; that is, even a very poorly soluble drug having a
`sufficiently small therapeutic dose may completely
`dissolve under physiological conditions. Thus, Amidon
`et al. (15) have defined a "high solubility" drug as one
`which at the highest human dose is soluble in 250 ml
`(or less) water throughout the physiological pH range
`(1-8) at 37°C. A "low solubility" drug is thus one
`which requires more than 250 ml of water to dissolve
`the largest human dose at any pH within the physio(cid:173)
`logical range. The likelihood of having bioavailability
`problems requires both a consideration of the dose and
`a solubility volume of the drug and its permeability.
`Amidon et al. (15) created a Biopharmaceutics Drug
`Classification System (BCS) based on estimates of these
`two parameters:
`
`Tablet Formulation
`
`1. Class I: High solubility and high permeability
`2. Class II: Low solubility and high permeability
`3. Class III: High solubility and low permeability
`4. Class IV: Low solubility and low permeability
`A jejuna! permeability of at least 2-4 X 10-4 emfs,
`measured in humans by an intubation technique, is
`considered "high permeability." For many substances,
`this permeability corresponds to a fraction absorbed of
`90% or better. The classification system provides a
`logical basis for estimating the risk of bioavailability
`problems. For example, Class I drugs (e.g., propranolol
`HCI, metoprolol tartrate) are expected to exhibit few
`bioavailability problems. On the other hand, Class II
`drugs (e.g., piroxicam) are more
`likely
`to exhibit
`dissolution-rate-limited absorption problems. Class III
`drugs (e.g., atenolol) are more likely to be prone to
`absorption (permeability) rate-limited absorption. Class
`IV drugs (low solubility-low permeability) present
`formidable obstacles to bioavailability. An in vitro-in
`vivo correlation (IVIVC) is expected only in the case of
`Class II drugs. An IVIVC could be expected for Class I
`drugs if the dissolution rate is slower than the gastric
`emptying the rate. With a sufficiently rapidly dissolving
`Class I drug, little or no IVIVC is expected because
`gastric emptying (not dissolution) would be the rate
`limiting step. Little or no IVIVC is expected for Class
`III or Class IV drugs.
`The FDA has adopted the BCS in developing a
`guidance that provides relaxed policies on scale-up and
`postapproval changes of immediate-release oral solid
`dosage forms (SUPAC-IR). For certain changes, require(cid:173)
`ments depend on the drug class, with the most liberal
`policies for Class I drugs, less liberal policies for Classes II
`and III drugs, and the least liberal policies for Class IV
`drugs. First issued as a draft on Nov. 29, 1994 for comment
`(16, 17), a revised version was published in the Federal
`Register on Nov. 30, 1995.
`The intrinsic dissolution rate (IDR) of drugs is
`frequently measured in preformulation tests by the rotating
`disk method or Wood's apparatus (18). An automated IDR
`system, based on a modification of a standard dissolution
`apparatus, allows for attachment to the stirrer of a die in
`which the pure drug has been compressed with the tablet
`face flush with the bottom surface of the die (19). The IDR
`may be used to detect different polymorphs as well as to
`judge the risk of a drug exhibiting dissolution-rate-limited
`absorption. Kaplan (20) suggested that an IDR of higher
`than 1 mg cm - 2min -l indicated that dissolution-related
`absorption problems were unlikely, whereas an IDR lower
`than 0.1 mg cm - 2min -l indicated dissolution-rate-limited
`absorption.
`
`

`

`Tablet Formulation
`
`Drug-Excipient Compatibility Studies
`
`A knowledge of the interaction of drugs and excipients is
`essential in the initial formulation of a product. It may also
`be necessary later on during processing scale-up, when
`problems arise, to determine if incompatibilities exist
`which affect manufacturing or stability. Drug-excipient
`interactions are often directly related to the moisture
`present in one or another of the components or to the
`humidity to which the formulation is exposed during
`processing or storage. These studies are always carried out
`at accelerated temperature and humidity conditions, even
`though it must be recognized that some interactions are
`physical (melting and volatilization) and not chemical and
`that accelerated aging may not be predictive. Tests for
`excipient-drug interactions are usually conducted on
`blends of the pure drug and excipient in ratios similar to
`those in the final dosage form. For example, excipient-to(cid:173)
`drug ratios are higher for filler-binders than for lubricants
`and disintegrating agents. These studies are often
`performed with the help of a factorial or fractional(cid:173)
`factorial experimental design (21). Powders are physically
`mixed and may be granulated or compacted to accelerate
`any possible interaction. Samples can be exposed in open
`pans or sealed in bottles or vials to mimic product
`packaging. Evaluation of samples includes
`
`1. Visual inspection for changes in color or texture.
`2. Both HPLC and TLC are commonly employed with
`unstressed samples being used as controls. In general,
`only qualitative results are important initially.
`3. Differential thermal analysis is applied and the
`appearance or disappearance of one or more peaks is
`noted. Isothermal microcalorimetry can also be
`employed as well as a thermal activity monitor
`(TAM) technique.
`
`Compatibility studies are essential in characterizing both
`raw materials and finished formulations. It has been argued
`that binary drug-excipient screening studies are inefficient,
`unrealistic, and ignore processing variables. A better
`approach may be to carefully select potential excipients
`based on known chemistry and published compatibility
`data, and perform miniformulation stability studies (22).
`
`Formulation Design
`
`Based on the preformulation information, decisions can be
`made regarding formulation design and process strategy.
`Initial guidance may be provided by the proposed dose.
`Relatively low-dose drugs can often be tableted by direct
`compression, a term that is applied to the process by which
`tablets are compressed directly from blends of the active
`
`I
`'I
`'
`
`2705
`
`ingredient and suitable excipients. No wet or dry granu(cid:173)
`lation is required, although the drug may occasionally be
`sprayed out of solution onto one of the excipients to ensure
`uniform dispersion of drug in very low dosage. Larger
`doses of poorly compactible drugs may be granulated
`prior to tableting. The process steps required and the
`choice of excipients are often governed by other properties
`of the drug.
`
`Analysis of Critical Variables and
`Formulation Development
`
`Based on the analysis of the preformulation data, likely
`excipients are selected and small batches may be
`produced. The number and size of the batches depend on
`the availability of the drug substance. The batches are
`intended to assess the feasibility of the formulation,
`including the types and levels of excipients, as well as the
`process and its operational variables, such as order of
`addition, mixing times, compression force, granulation
`time, etc. The goal is to develop a formulation and process
`that meets the criteria set forth earlier under Objectives.
`
`MANUFACTURE
`
`Traditionally, tablets have been made by granulation, a
`process that imparts two primary requisites to formu(cid:173)
`lations: compactibility and fluidity. Both wet granulation
`and dry granulation (slugging or roll compaction) are used
`(Table 1). Regardless of whether tablets are made by direct
`compression or granulation, the first steps, milling and
`mixing, are the same; the subsequent steps differ.
`The wet massing of powders is typically carried out in
`high-shear mixers prior to wet screening. The wet
`granules are often dried in fluidized-bed equipment,
`enhancing the efficiency of the process. Alternatively, wet
`granulation may be carried out in fluid-bed drier(cid:173)
`granulators in which the liquid phase is sprayed onto
`fluidized powders while the hot air flow dries the granules.
`This process reduces the number of handling steps and the
`time and space needed for granulation; it can be
`automated. The advantages and disadvantages of wet
`granulation are given in Table 2. See also Granulations
`by H.G. Kristensen and T. Schaeffer, Vol. 7 (1st Ed.),
`pp. 121-160, of this encyclopedia.
`Regardless of the granulation method, the comparative
`simplicity of the direct compression process offers obvious
`advantages, such as
`
`1. Economy
`2. Elimination of heat and moisture
`
`

`

`2706
`
`Tablet Formulation
`
`Table 1 Typical unit operations involved in wet granulation, dry granulation, and direct compression
`
`Wet granulation
`
`Milling and mixing of drugs
`and excipients
`Preparation of binder solution
`Wet massing by addition of
`binder solution or granulating solvent
`Screening of wet mass
`Drying of the wet granules
`Screening of the dry granules
`Blending with lubricants and disintegrant
`to produce "running powder"
`Compression of tablets
`
`Dry granulation
`
`Direct compression
`
`Milling and mixing of drugs
`and excipients
`Compression into slugs or roll compaction
`Milling and screening of slugs
`and compacted powder
`Mixing with lubricant and disintegrant
`Compression of tablets
`
`Milling and mixing of drugs
`and excipients
`Compression of tablets
`
`3. Optimization of tablet disintegration
`4. Stability
`
`The most obvious advantage of direct compression is its
`greater economy, owing to reduced processing time, less
`equipment and space required, less process validation, and
`lower energy utilization. Generally, only blending and
`compression are required, although prior micronization of
`the drug may be needed. Unlike wet granulation,
`processing does not require heat or moisture, which can
`be detrimental to drug stability. Moreover, direct
`compression avoids the high pressures associated with
`slugging or roll compaction. In addition, disintegration is
`optimized because directly compressed tablets produce
`primary particles upon disintegration, rather than granules,
`which must deaggregate to liberate primary particles.
`Finally, direct compression tablets often exhibit fewer
`long-term problems of chemical stability or changes in
`dissolution.
`
`Although there are many significant advantages of
`direct compression over granulation, there also are
`important limitations:
`
`1. Uniform blending and prevention of unblending of low(cid:173)
`dose drugs
`2. Fillers often are costlier than fillers used in granulation
`3. Physical properties and functional specifications are
`more critical; properties of raw materials must be
`defined and carefully controlled
`4. Limitations in producing colored tablets
`5. Dust problems
`6. Limitations in the dilution capacity of fillet-binders
`7. More sensitive to lubricant softening and overmixing
`than granulations
`
`Limitations in the dilution capacity of excipients can
`make the direct compression of large-dose, poorly
`compactible drugs impractical. Lubrication is often
`
`Table 2 Advantages and disadvantages of wet granulation
`
`Advantages
`
`Disadvantages
`
`Enhances fluidity and compactibility. suitable for high-dose
`drugs with poor flow and/or compactibility
`Reduces air entrapment
`
`Reduces dustiness
`Provides for the addition of a liquid phase (wet granulation)
`suited to dispersion of low-dose drugs in solution to ensure
`content uniformity
`Enhances wettability of powders through
`hydrophilization (wet granulation)
`Permits handling of powders without loss of blend quality
`
`Each unit process brings its own set of complications
`
`The large number of unit processes increases the chances
`of problems
`Difficult to control and validate
`Potential adverse effects of temperature, time, and rate of drying
`on drug stability and distribution during drying
`
`Overall more costly than direct compression in terms of space,
`time, and equipment requirements
`
`

`

`Tablet Formulation
`
`a compromise between the amount and type needed for
`adequate lubrication and their adverse effect on compact(cid:173)
`ibility. Content uniformity is of greater concern in direct
`compression tableting, particularly with low-dose drugs.
`Since the drug is not "locked" into granules, direct
`compression blends are subject to unmixing in subsequent
`processing steps. In addition, drugs are often micronized
`prior to blending to enhance their dissolution rate, and the
`resulting high surface-to-mass ratios may lead to difficulty
`in flowing and mixing due to surface interactions. Another
`important limitation is that unlike granulation, which tends
`to compensate for variability in excipients, direct
`compression is heavily dependent upon reproducible
`properties of the excipients (and the drug). Raw material
`standards must be carefully defined and address function(cid:173)
`ality. Lot-to-lot variations in both the drug and the
`excipients must be avoided. The cost of raw materials and
`their testing is higher in direct compression.
`Thus, direct-compression tableting requires careful
`attention to the choice of excipients, appropriate flow
`properties, and blend homogeneity, and to the interplay of
`formulation and process variables that can affect both
`compactibility and drug dissolution. See also Direct
`Compression Tableting by R.F. Shangraw, Vol.4, (1st Ed.)
`pp. 85-106, of this encyclopedia.
`
`Excipients
`
`The design of the formulation and selection of excipients
`is especially critical in tablet dosage forms. Products can
`vary from a relatively simple aspirin tablet containing
`aspirin and starch to more complex systems that might
`contain fillers, binders, disintegrating agents, glidants,
`lubricants, and coating agents. Modified release introduces
`even more complexity. The appropriate selection of
`excipients and their concentration are clearly critical to
`both the ability to manufacture tablets as well as to their
`performance as a drug delivery system. Since others have
`illuminated the various excipient classes in great detail,
`references are provided in Table 3.
`
`Manufacturability
`
`Excipients function to provide compactibility, lubrication,
`flow properties, disintegration efficiency, wetting, etc. Poor
`choice of excipients may give rise to poor characteristics
`(hardness, appearance), which can be important in
`packaging, storage, and patient acceptance. Problems
`with excipients may arise from variations in source or lot,
`particularly in formulations made by direct compression.
`Examples of excipient problems include variation in
`performance between Hoc cellulose and microcrystalline
`cellulose relative to particle size, flow, and compactibility,
`
`2707
`
`or different polymorphic forms of

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket