throbber
PHARMACEUTICAL
`DOSAGE FORMS
`
`Tablets
`
`SECOND EDITION, REVISED AND EXPANDED
`
`In Three Volumes
`VOLUMEl
`
`EDITED BY
`
`Herbert A. Lieberman
`H.H. Lieberman Associates, Inc.
`Consultant Services
`Livingston, New Jersey
`
`Leon Lachman
`Lachrnan Consultant Services
`Westbury, New York
`
`Joseph B. Schwartz
`Philadelphia College of Pharmacy and Science
`Philadelphia, Pennsylvania
`
`MARCEL DEKKER, INC.
`
`New York and Basel
`
`1
`
`EX 1014
`IPR of U.S. Pat. No. 7,829,595
`
`

`
`Library of Congress Cataloging-in-Publication Data
`
`Pharmaceutical dosage forms- -tablets I edited by Herbert A. Lieberman.
`Leon Lachman, Joseph B. Schwartz. - - 2nd ed. , rev. and expanded.
`p.
`cm.
`Includes index.
`ISBN 0-8247-8044-2 (v .. 1 : alk. paper)
`1. Tablets (Medicine) 2. Drugs--Dosage forms.
`I. Lieberman,
`II. Lachman, Leon.
`Ill. Schwartz, Joseph B.
`Herbert A.
`[DNLM: 1. Dosage Forms.
`2. Drugs- -administration & dosage. QV
`785 P535]
`RS201.T2P46 1989
`615'.191--dcl9
`DNLM/DLC
`for Library of Congress
`
`89-1629
`CIP
`
`Copyright © 1989 by MARCEL DEKKER, INC. 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 photo(cid:173)
`copying, microfilming, and recording, or by any information storage
`and retrieval system, without permission in writing from the publisher.
`
`MARCEL DEKKER, INC.
`270 Madison Avenue, New York, New York 10016
`
`Current printing (last digit):
`10 9 8 7 6 5 4 3 2 1
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`Prei
`
`Severa
`Forms;
`have t
`facturE
`vised
`n
`livery
`tablet
`is sug1
`intesti
`tabletE
`release
`chew al
`descril
`In
`the se
`tablets
`ad di tic
`drug l
`suranc
`Tl
`second
`on sul:
`the ph
`Formul
`been r
`There
`of imp•
`Tl:
`tion ar
`previo
`
`2
`
`

`
`Cenfignis
`
`"iireface
`Contributors
`Contents of Pharmaceutical Dosage For-ms: Tablets, Second Edition
`Volumes 2 and 3
`Contents of Pharmaceutical Dosage Forms: Parenteral Medicatiens,
`Volumes 1 and 2
`Contents of Pharmaceuticcd Dosage For-ms: Disperse Systems,
`Volumes I and 2
`
`Chapter 1.
`
`Preformulafion Testing
`Deedatt A. Wadke, Abu T. M. Serufuddin, and
`Harold Jacobson
`I .
`Introductien
`31. Grganoleptic Properties
`111. Parity
`IV. Particie Size, Shape, and Surface Area
`V.
`Soiability
`V1. Dissulution
`VII. Parameters Affecting Absorption
`V111. Crystal Properéies and Poiymorphism
`IX. Stability
`X. Miscelianeous Pmperties
`XI. Examples of Preformuiation Studies
`References
`
`Chapter 2. Tablet Formulation and Design
`Garnet E. Peck, George J. Buley, Vincent E. Mcflurdy,
`and Gilbert 3. Banker
`
`Introduction ,
`1.
`P:-eformulation Studies
`II.
`EH. A Systematic and Modern Approach to
`Tablet Product Design
`.
`IV. Tablet Components and Additives
`V. Regulatory Requirements for Excipients in
`the United States
`’
`VI . References
`
`3
`
`

`
`Cozlients
`
`13}
`
`Ccmprsssed Tablets by Wet Granulation
`Fred J. Butndeiin
`
`i. Properties of Tablets
`IL E‘o:°mulation 01‘ Tablets
`Hi. Tablet Manufacture
`EV . Gramzlation
`V.
`Ex-nignients and Fcxtcmulation
`V1. Multiiayer Tablats
`VIE.
`Prolonged Reiease ’.'i.‘abie‘:‘_s
`VIE}. Manufacturing Problems
`References
`
`Chapter =1.
`
`Compressed ’£‘a¥:>Ee*z3 by Direct Compression
`Rcziph Shflngraw
`E.
`1r:'ttoducti<m and History
`ii. Advantages and Disadvantages of the We‘:
`Granuiation Process
`:11. The Diracbcorsrpression ?1:'0r:ess
`IV. Direct-Compression Filler Binders
`V.
`Factors in Fax-muiation Development
`‘VI. Morphalogy of Direcbflompression Fillers
`Vii. Coprocessed Active Ingredients
`VIII.
`l\-iudiflcation and Entagration of Direct-
`Cempression and Granulation Processes
`Future -sf i)irect—Comp:-essien Tableting
`Formulations for Direct Compression
`Glossary of Traée Names and Manufacturers
`References
`
`Chapter 5.
`
`Compre.=;.=si1>n—Coated and Layer Tablets 7
`William C. Gunsel and Robert G. Buss!‘
`
`;
`1. Compression Coating
`I1.
`Formulations {Compression Coating)
`HI.
`Iniay Tablets
`IV. Layer Tablets
`V.
`Formuiations (Layer)
`References
`
`Chapter 6. Effervescent Tablets
`Raymond Mohrle
`I .
`Introduction
`H. Raw Materials
`Hi. Prqeessing
`IV. Maxpzfaeturing Operations
`V. Tablet Evajeuation
`
`4
`
`

`
`Contents
`
`Chapter 7.
`
`VI.
`VII .
`VIII.
`
`Effervescent Stability
`Effervescent Formulations
`References
`
`Special Tablets
`James W. Canine and Michael J’. Pilcai
`I.
`II.
`III.
`
`Drug Absorption ThI‘0ugi1_'th9 Oral Niucosa
`Molded Sublingual Tablets
`Special Problems with Molded Nit:-oglycerin
`Tablets
`
`IV.
`V.
`VI.
`V1}.
`VIII.
`IX.
`
`Compressed Sublingual Tablets
`Buccal Tablets
`Vaginal Tablets
`Rectal Tablets
`flispensing Tablets
`Tablets for Miscellaneous Uses
`References
`
`Chayter 8.
`
`Chewable Tablet S
`
`Robert W. Menzies, Aloysius O. Anuebonam, and
`Juhan B. Duruwalu
`I.
`Introduction
`I1.
`Formulation Factors
`III.
`i~‘ormuiati_on Techniques
`IV.
`Excipients
`V.
`Flavoring
`Colorants
`VI.
`VII.
`Manufacturing
`VIII.
`Evaluation of Chewable Tablets
`IX.
`Summary
`References
`
`Chapter 9.
`
`Medicated Lozenges
`David Peters
`
`I.
`13.
`III.
`IV.
`V.
`VI.
`VII.
`VIII.
`EX.
`X.
`X].
`
`Hard Candy Lozenges
`Processing
`Formulations (Hard Candy Lazenges)
`Centaz-«Filled Hard Candy Lozenges
`Formuiations (Center Filled Lozenges)
`Packaging
`Chewy or Caramel Base Medicated Tablets
`Formulations {Chewy Based Confections)
`Compressed Tablet Lozenges
`Manufacturing: Compression Sequence
`Typical Formulations (Compressed-Tablet
`Lozenges)
`
`5
`
`

`
`XII. Quality Central Proeedure$
`References
`
`Suggested Reading
`
`Ccmtenfs
`
`56'?
`576
`S80
`
`6
`
`

`
`end Jacobson
`
`Preformulation Testing
`
`s
`
`observing the melting point, especially with a hot-stage microscope. More
`quantitative information can be obtained by using quantitative differential
`scanning calorimetry or by phase-rule solubility analysis.
`As important to a compound's chemical characteristic are its physical
`ones. Crystalline form (including existence of solvates) is of fundamental
`importance, and for complete documentation of the compound X-ray powder
`diffraction patterns for each batch is desirable. This is simple to execute
`and provides useful information for later comparison and correlation to
`other properties.
`
`IV. PARTICLE SIZE, SHAPE, AND SURFACE AREA
`
`Various chemical and physical properties of drug substances are affected
`by their particle size distribution and shapes. The effect is not only on
`the physical properties of solid drugs but also, in some instances, on their
`biopharmaceutical behavior. For example, the bioavailability of griseofulvin
`and phenacetin is directly related to the particle size distributions of these
`drugs [ 3, 41 .
`It is now generally recognized that poorly soluble drugs
`showing a dissolution rate-limiting step in the absorption process will be
`more readily bioavailable when administered in a finely subdivided state
`than 8s a coarse material. Very fine materials are difficult to handle [5];
`but many difficulties can be overcome by creating solid solution of a material
`of interest in a carrier, such as a water-soluble polymer. This represents
`the ultimate in size reduction, since in a (solid) solution, the dispersed
`material of interest exists as discrete molecules or agglomerated molecular
`bundles of very small dimensions indeed.
`Size also plays a role in the homogeneity of the final tablet. When
`large differences in size exist between the active components and excipients,
`mutual sieving ( demixing) effects can occur maldng thorough mixing diffi(cid:173)
`cult or, if attained, difficult to maintain during the subsequent processing
`steps. This effect is greatest when the diluents and active raw materials
`are of significantly different sizes. Other things being equal, reasonably
`fine materials interdisperse more readily and randomly. However, if materials
`become too fine, then undersirable properties such as electrostatic effects
`and other surface active properties causing undue stickiness and lack of
`flowability manifest. Not only size but shape too influences the flow and
`mixing efficiency of powders and granules.
`Size can also be a factor in stability: :fine materials are relatively more
`open to attack from atmospheric oxygen, heat, light, humidity, and inter(cid:173)
`acting exipients than coarse materials. Weng and Parrott [ 6] investigated
`influence of particle size of sulfacetamide on its reaction with phthalic an(cid:173)
`hydride in 1: 2 molar compacts after 3 hr at 95°C. Their data, presented
`in Table 2, clarly demonstrate greater reactivity of sulfacetamide with de(cid:173)
`creasing particle size.
`Because of these significant roles , it is important to decide on a desired
`size range, and thence to maintain and control it. It is probably safest to
`grind most new drugs having particles that are above approximately 100 µm
`in diameter.
`If the material consists of particles primarily 30 µm or less in
`diameter, then grinding is unnecessary, except if the material exists as
`needles-where grinding may improve flow and handling properties, or if
`the material is poorly water-soluble where grinding increases dissolution
`rate. Grinding should reduce coarse material to, preferably, the 10- to
`
`rimental drug.
`
`'£ chromato(cid:173)
`anses due to
`ypieally, the
`graphic proce-
`response due
`1 shows an
`:ermination
`detector.
`on time of
`M?aks are due
`l. Thus, II
`1).8515 =
`
`In
`
`lated procedure
`In
`TLC.
`e to impurities
`. of the main
`ities [1,2].
`iSP are not
`~. • molecular
`s assumed to
`analysis re(cid:173)
`'JJY preparation
`;t always un-
`
`m.ia1 and
`? a qualitative
`resence of sol(cid:173)
`!Cterizing the
`appearance
`be indicative
`ersted by
`
`---------
`
`7
`
`

`
`6
`
`Wadke, Serajuddin, and Jacobson
`
`Influence of Particle Size
`Table 2
`on Conversion of Sulfacetamide
`
`Particle size of
`sulfacetamide
`(µm)
`
`128
`
`164
`
`214
`
`302
`
`387
`
`% Conversion
`±SD
`
`21.54 ± 2. 74
`19. 43 ± 3.25
`
`17.25 ± 2.88
`
`15.69 ± 7. 90
`
`9.34 ± 4.41
`
`Source: Modified from Weng, H. ,
`and Parrott, E. L., J. Pharm. Sci.,
`73: 1059 ( 1984). Reproduced with
`the permission of copyright owner.
`
`40- µm range. Once this is accomplished, controlled testing can be per(cid:173)
`formed both for subsequent in vivo studies and for in-depth preformulation
`studies. As the studies proceed, it may become apparent that grinding is
`not required and that coarser materials are acceptable. At that time, it
`is conceptually simpler to omit that step without jeopardizing the information
`already developed. The governing concept is to stage the material so that
`challenges are maximized.
`There are several drawbacks to grinding that may make it inadvisable.
`Some are of lesser importance. For example, there are material losses when
`grinding is done. Sometimes a static electricity buildup occurs, making the
`material difficult to handle. Often, however, this problem, if it exists, may
`be circumvented by mixing with excipients such as lactose prior to grinding.
`Reduction of the particle size to too small a dimension often leads to aggre(cid:173)
`gation and an apparent increase in hydrophobicity, possibly lowering the
`dissolution rate and making handling more troublesome. When materials are
`ground, they should be monitored not only for changes in the particle size
`and surface area, but also for any inadvertent polymorphic or chemical
`transformations. Undue grinding can destory solvates and thereby change
`some of the important characteristics of a substance. Some materials can
`also undergo a chemical reaction.
`
`A. General Techniques for Determining Particle Size
`
`Several tools are commonly employed to monitor the particle size. The most
`rapid technique allowing for a quick appraisal is microscopy. Microscopy,
`since it requires counting of a large number of particles when quantitative
`information is desired, is not suited for rapid, quantitative size determina(cid:173)
`tions. However, it is very useful in estimating the range of sizes and the
`shapes. The preliminary data can then be used to determine if grinding is
`needed. A photomicrograph should be taken both before and after grinding.
`The range of sizes observable by microscopy is from about 1 µm upward.
`
`ilm' 0
`ma~
`~·to'
`
`~· For· a
`~· ~.iii
`~.
`~,,­
`~
`age {H!Ai
`C~nter}.
`the USP'
`The instr
`eriy msp
`~5
`sor m 1i!!!
`Ot'be!
`a:~­
`fue ~
`The latte
`~·~
`as fue. A!
`settling: 5
`general d
`camnm t.
`~
`teme•
`metric•
`ratio of 11
`
`~
`Sieve
`
`~
`
`Du~
`
`Cen~
`
`p~
`
`light SC'llll
`
`.source:
`4:31{1•
`~
`
`8
`
`

`
`. and Jacobson
`
`Preformulation Testing
`
`7
`
`For optical microscopy, the material is best observed by suspending it
`in a nondissolving fluid (often water or mineral oil) and using polarizing
`lenses to observe birefringence as an aid to detecting a change to an amor(cid:173)
`phous state after grinding.
`For a quantitative particle size distribution analysis of materials that
`range upward frOm about 50 µm, sieving or screening is appropriate, al(cid:173)
`though shape has a strong influence on the results. Most pharmaceutical
`powders, however, range in size from 1 to 120 µm. To encompass these
`ranges, a variety of instrumentation has been developed. There are in -
`struments based on lasers (Malvern), light scattering (Royco), light block(cid:173)
`age' (HIAC), and blockage of an electrical conductivity path (Coulter
`Counter). The instrument based on light blockade has been adopted by
`the USP to monitor the level of foreign particulates in parenteral products.
`The instrument will measure particle size distribution of any powder prop(cid:173)
`erly dispersed in a suspending medium. The concentration of sample sus(cid:173)
`pension should be such that only a single particle is presented to the sen(cid:173)
`sor in unit time, thus avoiding coincidence counting.
`Other techniques based on centrifugation and air suspension are also
`available. Most of these instruments measure the numbers of particles, but
`the distributions are readily converted to weight and size distributions.
`The latter way of expressing the data is more meaningful. A number of
`classical techniques based on sedimentation methods, utilizing devices such
`as the Andreasen pipet or recording balances that continuously collect a
`settling suspension, are also known. However, these methods are now in
`general disfavor because of their tedious nature. Table 3 lists some of the
`common techniques useful for measurement of different size ranges [7].
`There are many mathematical expressions that can be used to charac(cid:173)
`terize an average size. These refer to average volumes or weights, geo(cid:173)
`metric mean diameters, and relationships reflecting shapes, such as the
`ratio of an area to a volume or weight factor [ 8] •
`
`Table 3 Common Techniques for
`Measuring Fine Particles of
`Various Sizes
`
`Technique
`
`Microscopic
`Sieve
`
`SE!dimentation
`Elutriation
`Centrifugal
`Permeability
`
`light scattering
`
`Particle size
`(µm)
`
`1-100
`
`>50
`>1
`1-50
`<50
`>1
`0.5-50
`
`Source: Parrott, E. L., Phann. Mfg.,
`4: 31 ( 1985). Reproduced with the
`permission of. copyright owner.
`
`!8i1 be per(cid:173)
`p:reformulation
`st grinding is
`!lat time, it
`the information
`derial so that
`
`t inadvisable.
`isl losses when
`rs • making the
`f it exists, may
`ior to grinding.
`eads to aggre(cid:173)
`Ollifering the
`i materials are
`e particle size
`r chemical
`iereby change
`merials can
`
`lre. The most
`Microscopy,
`1 quantitative
`ize determina(cid:173)
`siaes and the
`if gl"inding is
`after grinding.
`:..m upward.
`
`9
`
`

`
`8
`
`Wadke, Serajuddin, and Jacobson
`
`Cumulotlve Weight Percentage at the Indicated Size
`99.9
`99 98 915 90 80 70
`:JO 30
`10 15
`2 I 0.15
`
`99.8
`
`60 40 20
`
`100
`80
`60
`
`40
`30
`
`20
`
`-E
`
`::t.
`It.I 10
`~ 8
`6
`
`f/)
`
`4
`3
`
`2
`
`Figure 2 Log probability plot of the size distribution of a sample of tri(cid:173)
`amcinolone acetonide.
`
`A convenient way to characterize a particle size distribution is to con(cid:173)
`struct a log probability plot. Log probability graph paper is commercially
`available, and particle size distributions resulting from a grinding operation
`with no cut being discarded will give a linear plot. An example is illus(cid:173)
`trated in Figure 2 for a powder sample of triamcinolone acetonide. The
`data used in the construction of Figure 2 are presented in Table 4.
`The numbers of particles in Table 4 are converted into weight fractions
`by assuming them to be spheres and multiplying by the volume of a single
`sphere (particle) calculated from the geometric relationship:
`
`where V is the volume and d the particle diameter (using the average
`value of the range given in the first column of Table 4). The result is
`the total volume occupied by particles in each of the size ranges and is
`given in the third column of the table. The volume is directly related to
`a mass term by the reciprocal of the density. However, since the density
`is constant for all particles of a single species and is rarely known accurate(cid:173)
`ly, it is sufficient to use the volume terms to calculate the weight percent(cid:173)
`ages in each size range by dividing the total volume of all the particles
`into the volumes in each range (column 4 of Table 4).
`If densities were
`used, it is obvious that they would cancel out in this calculation. The
`cumulative weight percentage in each size range is shown in the last column.
`Statistical descriptions of distributions most often give a measure of
`central tendency. However, with powders the distributions are skewed in
`the direction of increasing size. This type of distribution can be described
`by the Hatch-Choate equation:
`
`>~,~~:.!~
`·~£-
`
`ka"'· ... -~
`.........
`........
`•
`.'h.._
`~···
`
`Pm- ti
`piiidide a
`shpe· ten
`• lmem' p
`
`s. Oeler
`
`Tire deter
`~a
`Simple -
`
`Size rangii
`(i.U)
`
`!2.5-26..5
`
`18.5-U.t
`
`14.9-18.I
`
`11.8-14-1
`
`9.4-U...&
`
`7.4-5.'t:
`5.9--'f .. ~;1
`
`10
`
`

`
`Preformulation Testing
`
`f =
`
`I:n
`&1n" g
`
`exp
`
`[
`
`-
`
`9
`
`( 1)
`
`where f is the frequency with which a particle of diameter d occurs, and
`n is the total number of particles in a powder in which the geometric mean
`particle size is M and the geometric standard deviation is crg. Equation (1)
`is succinctly discussed by Orr and Dalla Valle [9].
`The two measures M and crg uniquely characterize a distribution, and
`are readily obtained graphically from a log probability plot in which cumu(cid:173)
`lative weight percentage is plotted against the particle si2e (Fig. 2) . The
`geometric mean diameter corresponds to the 50% value of the abscissa, and
`the geometric standard deviation is given by the following ratios, the values
`for which are taken from the graph.
`
`cr = --,-.....,---
`84.13% size
`50% size
`50% size = 15. 87% size
`g
`
`For the example, the values are 8. 2 and 1. 5 µm for the geometric mean
`particle size and its standard deviation, respectively. The latter is also a
`slope term. For particle size distributions resulting from a crystallization,
`a linear plot can often be obtained using linear probability paper.
`
`B. Determination of Surface Area
`
`The determination of the surface areas of powders has been getting in(cid:173)
`creasing attention in recent years. The techniques employed are relatively
`simple and convenient to use, and the data obtained reflect the particle
`
`Table IJ
`Acetonide
`
`Particle Size Distribution of a Ground Sample of Triamcinolone
`
`Size range
`(µm)
`
`No. of
`particles
`
`Volume of
`particles
`-3
`3
`x 10
`(µm )
`
`Weight
`percent
`in range
`
`Cumulative
`weight
`percent
`
`22.5-26.5
`
`18.6-22.0
`
`14.9-18.6
`
`11.8-14.9
`9.4-11.8
`
`7.4-9.4
`
`5. 9-7.4
`
`4. 7-5. 9
`
`3. 7-4. 7
`
`5
`
`54
`
`488
`
`2072
`5376
`
`9632
`
`12,544
`
`12,928
`
`13,568
`
`38
`
`237
`
`1212
`
`2552
`3352
`
`2989
`
`1888
`
`1008
`
`526
`
`0.2
`
`1. 7
`
`8.8
`18.5
`24.3
`
`21.7
`
`13.7
`
`7.3
`
`3.8
`
`100.0
`
`99.8
`
`98.1
`
`89.3
`70.8
`
`46.5
`
`24. 8
`
`11.1
`
`3.8
`
`... : .... :~.:.cc.::~=""·==--------------
`
`>"
`
`"•''•~~bed
`
`11
`
`

`
`iabtet
`
`sanitation see Sesign
`
`Garnet E. Peck
`Purdue University
`West Lafayette,
`Indiana
`
`George J. Baley and
`Vmcem E‘ Mccumy
`The Upjohn Company
`Kalamazoo, Michigan
`
`Giibert S. Banker
`University of Minnesota
`Health Sciences Center
`Minneapolis, Minnesota
`
`I.
`
`iNTRDDUC"{'i0N
`
`The §orm:1lation of solid oral dosage forms, and tablets in particular, has
`undergone rapid change and development over the last several decades with
`the emergence of precompression,
`induced tiie feeding, higlwspeeci ano now
`ultrahigh—speed presses, automated weightmontrol systems,
`the availability
`of many new direct compression materials, and the microprocessor control
`of precompression, compression, ejection forces, as weil as upper punch
`tightness on tablet presses.
`Some of the newer tablet presses have tablet
`rejection Systems that are operated by a computer, Computerwcontroiied
`tablet presses onty require an operator to set up the press at the proper
`tablet weight and thickness (or pressure). The computer can then assume
`complete controi of the run. Still other tabiet presses only require the op-
`erator to provide a product itlentifioatiori code to make tablets within speci-
`fications previously estabiished and stored in the computer memory.
`Most recently, new concepts and fedora} reguiations hearing on bio-
`availability and bioequivalence, anti on validation, are impacting on tablet
`formulation, design, and manufacture.
`Once,
`lavish golcihplatecl pills were manufactured and marketed with
`little knowledge of their pharmacological activity. Appearance and later
`stability of the dosage form were the prime requirements of pharmaceutical
`preparations. The introduction of the fricbie pill denoted in part the reaEi—
`zation that solid medicinais rnust——in some fashionwdisintegrate within the
`body for the patient to benefit from the drug. We now reaiize that disin-
`tegration and dissolution alone do not insure therapeutic activity. As only
`one example of this point, Meyer et el
`[1] presented information on 14
`nitrofurantoin products, which were evaluated both in vitro and in vivo.
`All products tested met USP XVIEI specifications for drug content, disin~
`tegration time, and dissotution rate; however, statisticaliy significant difw
`ferences in bioavailability were observed.
`
`12
`
`

`
`75
`
`Peck, Boley, Mccordy, and Banker
`
`The design of a tablet usually involves a series of compromises on the
`part of the formulator, since producing the desired properties {e.g.,
`rew
`sistance to mechanical abrasion or friability, rapid disintegration and disco-~
`lotion} frequently involves competing objectives. The correct selection and
`balance of excipient materials for each active ingredient or ingredient cora-
`‘oination in a tablet formulation to achieve the desired response (i.e., pro~
`{motion of a safe, effective, and highly reliable product) is not in practice
`2. simple goal to achieve. Add to this fact the need today to develop tabiet
`formulations and processing methods which may be {and must in the future
`be) validated, and the complexity of tablet product design is further in-
`creased in contemporary pharmaceutical development.
`Increased competition
`among manufacturers (brand versus generic, generic versus generic, and
`brand versus brand) has necessitated that products and processes be cost“
`efficient. Thus cost of a raw material or a particular processing step must
`be considered before a finai tablet formulation or manufactoring process is
`seiected.
`Tablet formulation and design may he described as the process whereby
`the formulator insures that the correct amount of drug in the right form
`is delivered at or over the proper time at the proper rate and in the cle~
`sired location, while having its chemical integrity protected to that point.
`Theoreticaiiy, a validated tablet formulation and production process is one
`in which the range in the variation of the component specifications and
`physical properties of the tablet product quaiity properties is known from
`a cause and effect basis.
`It is further known that raw materials specifica-
`tions, at their limits, and when considered as interaction effects of the
`worst possible combinations, cannot produce a product that is out of speci~
`fication from any standpoint. Likewise a validated tablet—manufacturing
`process is one which, when all the operating variables are considered, at
`any extremes which could ever be encountered in practice, and under the
`worst possible set of circumstances, will produce products that are within
`specifications. Total validation of a tabiet product includes all combination
`effects involving formulation, raw materials variables, and processing vari-
`ables, as well as their interaction effects,
`to assure that any system pro-
`duced will be within total product specifications.
`The amount or quantity of a drug which is sufficient to elicit the re-
`quired or desired therapeutic response can be affected by several factors.
`In the case of compendial or official drugs,
`the dosage levels have been
`predetermined. With certain drugs (e.g. , gr-iseofulvin),
`the efficiencjr of
`absorytion has been shown to depend on the particle size and specific sur-
`face area of the drug. By reducing the particle size of such drugs, the
`dosage level may be reduced by one~half or more and still produce the same
`biological response.
`The form in which the drug is absorbed can affect its activity. Most
`drugs are normally absorbed in solution from the gut. Since the absorp-
`tion process for most orally administered drugs is rapid, the rate of solu-
`tion of the drug will be the rate—limiting step from the point of View of
`blood level and activity.
`Thus, we must consider the contribution and influence of the active
`components and nonactive componentsmboth separately and together—to
`measure their impact on the pharmacological response of any tablet system.
`The timing of administration may effect when and how 9. drug will act (and
`to a certain extent where it acts) as will be discussed further in Section
`
`13
`
`

`
`Tablet Formulation and Design
`
`7?’
`
`l’v‘.A. Also, the timing of administration may be crucial in order to reduce
`gastric irritation (uncoated strong electrolytes are often given following
`food};
`to reduce drug interactions with food (formation of insoluble com-
`plexes between the calcium of milk and several antibiotics), reducing their
`bioavailability; or to enhance the solubility and bioavailability of certain
`drugs in foods (notably fats) by their administration with foods (e.g.,
`griseofulviu). Depending on such timing factors plus the relationship and
`rationale of fast, intermediate, or slow drug release as well as other re-
`lease considerations, a particular design and tablet formulation strategy is
`often indicated.
`‘Many excellent review articles have been written on tablet technology,
`including various formulation aspects. Cooper [23 presented a review mono-
`graph on the contributions from 1964 to 1968 in the areas of tablet formun
`lation, processing, quality standards, and biopharrnaceutics. Later, Cooper
`and Rees £3] continued the review and included similar topics covering the
`period 13359 to 1971. Recent book chapters on tablets include those by
`Banker [4] and Sadik [5].
`The present chapter will detail the general considerations of tablet
`product design; will describe a systematic approach to tablet design,
`in-
`eluding the practical use of preformulation data; wfll describe the commonly
`used tablet excipients with particular emphasis on their advantages and liar
`itations or disadvantages; and will present some general tablet formulation
`approaches. Extensive references to the literature should provide the
`reader with directed reading on topics where additional information may be
`obtained. While it is impossible to exhaustively cover as broad a topic as
`tablet formulation and design in one chapter of a book, it is the goal of
`this chapter to cover the major concepts and approaches, including the
`most recent thought bearing on validation, optimization, and programmatic
`methods related to the formulation, design, and processing of compressed
`tablets.
`
`H.
`
`PREFORMULATION STUDVES
`
`The first step in any tablet design or formulation activity is careful conm
`sideratioh of the preformulation data.
`It is important that the formulator
`have a complete physicoohemical profile of the active ingredients available,
`prior to initiating a formulation development activity. Compilation of this
`information is known as preformulation.
`It is usually the responsibility of
`the pharmaceutical chemistry research area to provide the data shown below
`on the drug substances.
`
`temperature, humidity
`light,
`1. Stability (solid state):
`2. Stability (solution):
`excipient~drug stability (differential thermal
`analysis or other accelerated methods)
`Physicomechonical properties:
`particle size, bulk and tap density,
`crystalline form, compressibility, photomicrographs, melting point,
`taste, color, appearance, odor
`Physicochemical properties:
`solubility and pll profile of solutionl
`dispersion (water, other solvents)
`In vitro dissolution: pure drug, pure drug pellet, dialysis of pure
`drug, absorbability, effect of excipients and surfactants
`'
`
`14
`
`

`
`Y8
`
`Peck, Bailey, McCurdy, and Banker
`
`The basic purposes of the prefoi-mulation activity are to provide a
`rational basis for the formulation approaches,
`to maximize the chances of
`success in formulating an acceptable product, and to ultimately provide a
`basis for optimizing drug product quality and performance.
`From a tablet
`formulatox-‘s perspective, the most important preformulation information is
`the drug-excipient stability study. The question then,
`for a new drug, or
`a drug with which the formulator lacks experience, is to select excipient
`materials that will be both chemically and piiysicaliy compatible with the
`drug.
`The question is compounded by the fact that tablets are compacts; and
`while powder mixtures may be adequately stable, the closer physical contact
`of particles of potentially reactive materials may lead to instability. The
`typical preformulation profile of a new drug is usually of limited value to
`the formulator in assuring him or her that particular drug-excipient com-
`binations will produce adequate stability in tablet form. An added problem
`is that the formulator would like to identify the most compatible excipient
`candidates within days of beginning work to develop a new drug into a tab-
`let dosage form rather than to produce a series of compacts, place them on
`stability, and then wait weeks or months for this information.
`Simon [6}, in reporting on the development of preformulation systems,
`suggested an accelerated approach, utilizing thermal analysis,
`to identify
`possibly compatible or incompatible drug-excipient combinations.
`In his
`procedure, mixtures are made of the drug and respective excipient materi~
`ale in a 1:1 ratio and subjected to differ-entiai thermal analysis. A 1:1 ratio
`is used, even though this is not the ratio anticipated for the final dosage
`form,
`in order to maximize the probability of detecting a physical or chemi-
`cal reaction, should one occur. The analyses are made in visual cells, and
`physical observations accompany the thermal analysis. The thermograms
`obtained with the dz-ug—excipient mixtures are compared to thermograms for
`the drag alone and the excipient alone. Changes in the termograms of the
`mixture, such as unexpected shifts, depressions, and additions to or losses
`from peaks are considered to be significant.
`Simon [6] has given an ex-
`ample of the type of information which may be obtained from such a study '
`by the data shown in Figure l. The thermal peak due to the drug alone
`was lost when the thermal analysis was run on the drug in combination with
`the commonly used lubricant, magnesium stearate. This was strong evidence
`for an interaction between these materials.
`It was subsequently confirmed
`by other elevated-temperature studies that the drug did decompose rapidly
`in the presence of magnesium stearate and other basic compounds.
`Simon
`has concluded the differential thermal analysis can aid immensely in the
`evaluation of new compounds and in their screening for compatibility with
`various solid dosage form excipients. The combination of visual and physical
`data resulting from differential thermal analysis of drugs with excipients is
`suggested as a programmatic approach to the very rapid screening of the
`drug‘-excipient combinations for compatibility.
`Following receipt of the pi-eformuiation information, the formuiator may
`prepare a general summary statement concerning the drug and its properties
`relative to tablet formulation. This statement must often also take into ac-
`count general or special needs or concerns of the medical and marketing
`groups for that drug. A typical statement might be as follows.
`Compound X is a white crystalline solid with a pyridine odor and bitter
`taste, which may require a protective coating (film or sugar).
`It displays
`excellent compressing properties

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