throbber
Modern
`Pharmaceutics
`Fourth Edition, Revised and Expanded
`
`edited by
`Gilbert $. Banker
`University of lowa
`lowa City, fowa
`
`Christopher T. Rhodes
`University of Rhode Island
`Kingston, Rhode island
`
`MARCEL
`
`a
`
`Marcer Dexxer, Inc.
`
`DEKKER
`
`New York « Baser
`
`
`
`Hopewell EX1065
`Hopewell v. Merck
`IPR2023-00480
`
`Hopewell EX1065
`Hopewell v. Merck
`IPR2023-00480
`
`1
`
`

`

`ISBN: 6-8247-0674-9
`
`This book is printed on acid-free paper.
`
`Headquarters
`Marcel Dekker, Inc.
`270 Madison Avenue, New York, NY 10016
`tel: 232-696-9000; 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
`itp://www.dekker.com
`
`The publisher offers discounts on this book when ordered in bulk quantities. For more information, write to Special Sales/
`Professional Marketing at the headquarters address above.
`
`Copyright © 2602 hy 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 photocopying, microfilming, and recording, or by any information storage andretrieval system, without permission in
`writing from the publisher.
`
`Currentprinting (ast digit):
`109 8765423242
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`2
`
`

`

`Chapter 10
`
`Tablet Dosage Forms
`
`Mary Kathryn Kottke
`
`Cubist Pharmaceuticals, Inc., Lexington, Massachusetts
`Edward M. Rudnic
`
`Advancis Pharmaceutical Corp., Gaithersburg, Maryland
`
`I.
`
`INTRODUCTION
`
`During the past four decades, the pharmaceutical in-
`dustry has invested vast amounts of time and moneyin
`the study of tablet compaction. This expenditure is
`quite reasonable when one considers how valuable
`tablets, as a dosage form, are to the industry. Because
`oral dosage forms can be self-administered by the pa-
`tient,
`they are obviously more profitable to manu-
`facture than parenteral dosage forms that must be
`administered, in most cases, by trained personnel. This
`is reflected by the fact that well over 80% of the drugs
`in the United States that are formulated to produce
`systemic effects are marketed as oral dosage forms.
`Compared to other oral dosage forms, tablets are the
`manufacturer’s dosage form of choice because of their
`relatively low cost of manufacture, package, and
`shipment;
`increased stability and virtual tamper re-
`sistance (most
`tampered-with tablets either become
`discolored or disintegrate).
`
`Il. DESIGN AND FORMULATION
`OF COMPRESSED TABLETS
`
`A. General Considerations
`
`in con-
`The most common solid dosage forms
`temporary use are tablets, which may be defined as
`
`unit forms of solid medicaments prepared by com-
`paction. Most consist of a mixture of powders that are
`compacted in a die to produce a single rigid body. The
`most common types of tablets are those intended to be
`swallowed whole and then disintegrate and release
`their medicaments in the gastrointestinal tract (GIT).
`A less common type of tablet that is rapidly gaining
`popularity in the United States is formulated to allow
`dissolution or dispersion in water prior to adminis-
`iration. Ideally, for this type of tablet all ingredients
`should be soluble, but frequently a fine suspension has
`to be accepted. Many tablets of this type are for-
`mulated to be effervescent, and their main advantages
`inelude rapid release of drug and minimization of
`gastric irritation.
`Some tablets are designed to be masticated {i.c.,
`chewed). This type of tablet is often used when ab-
`sorption from the buceal cavity is desired or to en-
`hance dispersion prior to swallowing. Alternatively, a
`tablet may be intended to dissolve slowly in the mouth
`(e.g., lozenges) so as to provide local activity of the
`drug. A few tablets are designed to be placed under the
`tongue (i.¢., sublingual) or between the teeth and gum
`(i.c., buccal} and rapidly release drug into the blood-
`stream. Buceal or sublingual absorption is often
`desirable for drugs liable to extensive hepatic meta-
`bolism by the first-pass effect
`(e.g., nitroglycerin,
`
`287
`
`3
`
`

`

`288
`
`Kottke and Rudnic
`
`testosterone). Recently, a lozenge on a stick, or “lol-
`lipop,” dosage form of fentanyl was developed for
`preoperative sedation in pediatric patients (Oralet®)
`and breakthrough cancer pain in adults (Actiq®).
`Active ingredient is released from the lozenge into the
`bloodstream from the oral nrucosa.
`There are now many types of tablet formulations
`that provide for the release of drug te be delayed or
`control the rate of the drug’s availability. Some of
`these preparations are highly sophisticated and are
`rightly referred to as complete “drug-delivery sys-
`tems.” Since the concepts of controlled drug delivery
`are the subjects of Chapter 15, the strategies of these
`systems will not be discussed here, However, solid
`dosage formulators must be aware of the various op-
`tions available to them.
`For example, when prolonged release of a water-
`soluble drug is
`required, water-insoluble materials
`must be co-formulated with the drug. if the dose of the
`drugis high and it exhibits poor compactibility, purely
`hydrophobic agents, such as waxes, will exacerbate the
`inability of the material to form a compact. In such
`cases, formulators need to turn to other types of water-
`insoluble materials, such as polymers, to achieve both
`drug release and tableting goals.
`Some tablets combine sustained-release and rapid
`disintegration characteristics. Products such as K-
`Dur® (Key Pharmaceuticals) combine coated po-
`tassium chloride crystals in a rapidly releasing tablet.
`In this particular instance, the crystals are coated with
`éthylceliulose, a water-insoluble polymer, and are then
`incorporated into a rapidly disintegrating micro-
`crystalline cellulose (MCC) matrix. The purpose ofthis
`tablet is to minimize GI ulceration, commonly en-
`countered by patients treated with potassium chioride.
`This simple but elegant formulation is an example of a
`solid dosage form strategy used to achieve clinical
`goals.
`Thus, the single greatest challenge to the tablet
`formulator is in the definition of the purpose of the
`formulation and the identification of suitable materials
`to meet development objectives. In order to do this
`properly, the formulator must know the properties of
`the drug, the materials to be co-formulated with the
`drug, and the important aspects of the granulation,
`tableting, and coating processes.
`Pharmaceutical compressed tablets are prepared by
`placing an appropriate powder mix, or granulation, in
`a metal die on a tablet press. At the base of the die is a
`lower punch, and above the die is an upper punch.
`When the upper punch is forced down on the powder
`mix (single station press) or when the upper and lower
`
`punches squeeze together (rotary or multiple station
`press), the powder is forced into a tablet. Despite the
`fact that powder compaction has been observed for
`hundreds of years, scientists still debate the exact me-
`chanisms behind this phenomenon.
`Perhaps the most significant factor in the tableting
`of materials for use as drug products is the need to
`produce tablets of uniform weight. This is achieved by
`feeding constant volumes of homogeneous material to
`the dies. Such an approach is necessary because direct
`weighing at rates commensurate with modern tablet
`press operation is impossible. This requirement im-
`mediately places demands on the physical character-
`istics of the feed and on the design of the tablet press
`itself.
`In the case of the former, precompression
`treatment of the granniation is one of the most com-
`mon ways of minimizing difficulties arising from this
`source.
`The great paradox in pharmaceutical tabletingis the
`need to manufacture a compact of sufficient mechan-
`ical strength to withstand the rigors of processing and
`packaging that is also capable of reproducibly releas-
`ing the drug. In most cases, the release of the drug is
`produced by the penetration of aqueous fluids into the
`fine residual pore structure of the tablet and the con-
`tact of these fluids with components that either swell or
`release gases.
`if any,
`The selected precompression treatment,
`markedly affects the manufacture of tablets. In parti-
`cular, one must determine whether a mixture of pow-
`dered ingredients is to be tableted directly or if an
`intervening wet granulation step is to be introduced.
`This decision is influenced by many factors, including
`the stability of the drug to heat and moisture: the flow
`properties of the granulation; and the tendency of the
`granulation to sepregate. At the present time there are
`also two conflicting considerations that tend to play a
`major role in this choice. These are the reluctance to
`change methods employed traditionally by the com-
`pany versus the economic advantages of omitting
`complete stages in the production sequence,
`In wet granulation,
`the componenis of the for-
`mulations are mixed with a granulating liquid, such as
`water or ethanol, to produce granules that will readily
`compress to give tablets. Wet granulation methods
`predominate in the manufacture of existing products,
`while the trend for new products is to use direct
`compression procedures. Although many steps are
`eliminated when using direct compression, some for-
`mulators have found that wet granulated products are
`more robust and able to accommodate variability in
`raw materials and tableting equipment. Thus, for some
`
`4
`
`

`

`289
`
`The effect of particle size on the compaction char-
`acteristics of two model sulfonamide drugs, one ex-
`hibiting brittle fracture and the other being compressed
`chiefly by plastic deformation, has been reported [3]. In
`particular,
`it was shown that the tensile strength of
`tablets made from the brittle material were more sen-
`sitive to the drug’s particle size than that of tablets
`made from the plastically deforming material. In ad-
`dition, larger granules possess better fiow, while small
`ageregates deform during compaction (¢.g., spray-
`dried lactose) [6].
`reducing the
`An alternative approach aimed at
`segregation tendencies of medicaments and excipients
`involves milling the former to a small particle size and
`then physically absorbingit uniformly onto the surface
`of the larger particles of an excipient substrate. By
`these means “ordered,” 2s opposed to ‘‘random,”
`mixing is realized and dissolution is enhanced as a
`result of the fine dispersion [7].
`
`Moisture Content
`
`One of the mostsignificant parameters contributing to
`the behavior of many tablet formulationsis the level of
`moisture present during manufacture as well as that
`residual in the product. In addition to its role as a
`granulation fluid and its potentially adverse effects on
`stability, water has some subtle effects that should not
`be overlooked. For example, there is increasing evi-
`dence fo suggest that moisture levels may be very cri-
`tical in minimizing certain faults, such as lamination,
`that can occur during compression. Moisture levels can
`also affect the mechanical strength of tablets and may
`act as an internal lubricant. For example, Fig.
`| illus-
`
` 1
`
`3
`
`7
`5
`Hardness (kg)
`Fig. 1 The effect of moisture content on the compactibility
`of anhydrous beta lactose tablets. rom Ref. 8.)
`
`3)
`
`11
`
`qablet Dosage Forms
`
`companies, the trend isreverting to the formulation of
`ablets by wet granulation.
`
`B. Desirable Properties of Raw Materials
`Most formulations are composed of one or more
`medicaments plus a variety of excipients. Irrespective
`of the type Of tablet, general criteria for these raw
`materials are necessary. Jn order to produce accurate
`reproducible dosage forms, it is essential that each
`component be uniformly dispersed within the mixture
`and that any tendency for component sepregation be
`minimized, In addition, the processing operations de-
`mand that the mixture be both free-flowing and co-
`hesive when compressed.
`
`Particle Size
`
`In general, the tendencies for a powder mix to segre-
`gate can be reduced by maintainingsimilarparticle size
`distribution, shape, and, theoretically, density ofall the
`ingredients. Flow properties are enhanced by using
`regular-shaped, smooth particles with a narrow size
`distribution together with an optimum proportion of
`“fines” (particles 50 ym). If such conditions cannot be
`met,
`then some form of granulation should be
`considered.
`Particle size distribution, and hence surface area of
`the drug itself, is an important property that has re-
`ceived considerable attention in the literature. For
`many drugs, particularly those whose absorption is
`limited by the rate of dissolution, attainment of ther-
`apeutic levels may depend upon achieving a small
`particle size [1]. In fact, it has been suggested that for
`such drugs, standards for specific surface areas and the
`number of particles per unit weight should be devel-
`oped. However,
`the difficulty in handling very fine
`powders, as well as the possibility of altering the ma-
`terial in other ways, has shifted the emphasis towards
`producing an optimum, rather than a minimum, par-
`ticle size, For instance, several researchers have found
`that decreasing particle size produces tablets of in-
`Creased strength that also have 4 reduced tendency for
`lamination [2-5]. This is probably due to the mini-
`mization of any adverse influences that 2 particular
`crystal structure may have on the bonding mechanism.
`On the other hand, samples of milled digoxin crystals
`Prepared by a number of size-reduction techniques
`have been reported to elicit different equilibrium 80-
`lubilities [1]. This suggests that the method of grinding
`i well affect the dissolution behavior of certain
`
`Tugs,
`
`5
`
`

`

`290
`
`trates the effect of moisture content on the cempact-
`ibility of anhydrous lactose [8]. As the moisture con-
`tent increases, it is adsorbed by the lactose, thereby
`converting it from the anhydrousto the hydrous form.
`During this transformation, the B-form oflactose most
`probably changes to the o-form and thus produces
`changes in compactibility.
`Accelerated aging and crystal transformation rates
`have also been traced to high residual moisture con-
`tent. Andoet al. studied the effect of moisture content
`on the crystallization of anhydrous theophylline in
`tablets [9]. Their results also indicate that anhydrous
`materials convert to hydrates at high levels of relative
`humidity. In addition, if hygroscopic materials {e.g.,
`polyethylene glycol 6000) are also contained in the
`formulation, needle-like crystals form at
`the tablet
`surface and significantly reduce the release rate of the
`theophylline.
`In many products it seems highly probable that
`there exists a narrow range of optimum moisture
`contents that should be maintained. More specifically,
`the effect of moisture on MCC-containingtablets has
`been the subject of an investigation that demonstrates
`the sensitivity of this important excipient to moisture
`content [10]. These researchers found that differences
`exist in both the cohesive nature and the moisture
`content to two commercial brands of MCC. A very
`useful report on the equilibrium moisture content of
`some 30 excipients has been compiled by a collabora-
`tive group of workers from several pharmaceutical
`companies and appears in the Handbook af Pharma-
`ceutical Excipients [11,12].
`
`Crystalline Form
`
`Selection of the most suitable chemical form of the
`active principle for a tablet, while not strictly within
`our terms of reference here, must be considered. For
`example, some chloramphenicol esters produce little
`clinical response [13]. There is also a significant dif-
`ference in the bioavailability of anhydrous and hy-
`drated forms of ampicillin [14]. Furthermore, different
`polymorphic forms, and even crystal habits, may have
`a pronounced influence on the bioavailability of some
`drugs due to the different dissolution rates they exhibit.
`Such changes can also give rise to manufacturing
`problems. Polymorphism is, of course, not restricted to
`active ingredients, as shown, for example, in an eva-
`fuation of the tableting characteristics of five forms or
`sorbitol [15].
`Many drugs have definite and stable crystal habits.
`Morphological changes rarely occur in such drugs as
`
`Kottke and Ruduic
`
`Table 1 Some Drugs That Undergo Polymorphic Transi-
`tion When Triturated
`
`Numberof
`Number of
`polymorphs
`polymorphs
`Drug
`before trituration
`after trituration
`
`
`Barbitone
`Caffeine
`Chiorpropamide
`Clenbuterol HCI
`Dipyridamole
`Maprotiline HC!
`Mebendazole
`Nafoxidine HC]
`Pentobarbitone
`Phenobarbitone
`Sulfabenzamide
`
`Source: Ref. 16.
`
`2
`2
`3
`2
`2
`3
`4
`4
`3
`2
`2
`
`I
`1
`2
`3
`i
`1
`5
`3
`2
`I
`i
`
`the formulation process is scaled up. However, some
`drugs exhibit polymorphism or have different identifi-
`able crystal habits. Chan and Doelker reviewed a
`number of drugs that undergo polymorphic transfor-
`mation when triturated in a mortar and pestle [16].
`Some of their conclusions are listed in Table 1 and ii-
`lustrated in Fig. 2.
`In addition, a number of re-
`searchers have concluded that both polymorph and
`crystal habit
`influence the compactibility and me-
`chanical strength to tablets prepared from poly-
`morphic materials
`[16-21]. York compared the
`compressibility of naproxen crystals that had been
`spherically agglomerated with different solvents and
`found that significant differences existed between the
`various types of agglomerates (see Fig. 3) [21]. Other
`investigators have found that, in some instances, there
`is a correlation between the rate of reversion to the
`metastable form during dissolution and the crystal
`growthrate of the stable form [22]. These polymorphic
`changes may have a profound effect on tablet perfor-
`mance in terms of processing, in vitro dissolution and
`in vive absorption. Thus, formulators of solid dosage
`forms must be aware of a subject compound’s pro-
`pensity for polymorphic transition so that a rational
`approach to formulation can be followed.
`
`Hiestand Tableting Indices
`
`Materials that do not compress well produce soft ta-
`Diets. In addition, brittle crystalline materials will yield
`brittle tablets. Hiestand was the first pharmaceutical
`scientist to quantify rationally the compaction prop-
`erties of pharmaceutical powders [23-28]. The results
`
`6
`
`

`

`Tablet Dosage Forms
`
`291
`
`350
`
`30
`
`Zz 2
`
`10
`
`E:R
`
`0
`
`50
`
`undergo plastic transformation to produce a suitable
`tablet. The third index, the brittle fracture index (BFI),
`is a measure of the brittleness of the material and its
`compact, Table 2 lists these indices for a number of
`drugs and excipients. For most materials, the strength
`of the tablet is a result of competing processes. For
`example, erythromycin is a material known forits
`tendency to cap and laminate when tableted. On the
`basis of its BI value, one might expect relatively good
`bonding. However, the very high strain index asso-
`ciated with this drug appears to overcomeits bonding
`abilities. MCC, on the other hand, has very high strain
`index, but its bonding index is exceptionally high and
`compensates for this effect.
`250
`450
`Other investigators have evaluated the potential for
`Pressure (MPa}
`—w— Lower Surface
`these indices. In their studies, Williams and McGinnity
`~e— Middle Region
`have concluded that evaluation of single-material sys-
`oood
`a~wa—- Upper Surlace
`tems should precede binary or tertiary powder systems
`Side
`(29}. A full discussion of compaction mechanisms is
`piven later in this chapter.
`
`Fig. 2 Percentage of caffeine “form A” transformed vs.
`applied pressure, (From Ref. 16.)
`
`of this work are three indices known as the Hiestand
`Tableting Indices. The strain index (SDis a measure of
`the internal entropy, or strain, associated with a given
`material when compacted. The bonding index (BD is a
`measure of the material’s ability to form bonds and
`
`15
`
`oh Qo
`
`on
`
`Hardness{kq)
`
`“500
`
`7000
`
`3000 350
`
`2500
`2000
`1500
`Pressure {ibs}
`—e- Control
`—w Hexanot
`— Octarat
`—o— Tolusne
`
`Intrinsic compressibility of nonagglomerated na-
`Fig. 3
`proxen (control) and of naproxen that has been spherically
`agglomerated with different solvents. (From Ref. 21.)
`
`Variability
`
`The effect of raw material variability on tablet pro-
`duction [2,30,31] and suggestions for improving ta-
`bleting quality of starting materials [21] has been the
`subject of several pubiications. Table 3, whichlists the
`characteristics of different
`sources of magnesium
`stearate, clearly illustrates the variability of this ma-
`terial [32]. Phadke and Eichorst have also confirmed
`that significant differences can exist between different
`sources, and even different lots, of magnesium stearate
`[33]. Given the fact that the effectiveness of magnesium
`stearate is due, in large part, to its large surface area,
`these variations should not be overlooked. In addition,
`studies assessing raw material variability emphasize the
`need for physical as well as chemical testing of raw
`materials to ensure uniformity of the final product.
`
`Purity
`Raw material purity, in general, must also be given
`careful attention, Apart from the obvious reasonsfor a
`high level of integrity, as recognized by the regulatory
`requirements, one should be aware of more subtle
`implications that are perhaps only just beginning to
`emerge, For instance, small proportions of the im-
`purity acetylsalicylic anhydride have been shown to
`reduce the dissolution rate of aspirin itself (see Fig. 4)
`134].
`Another area of interest is that of microbiological
`contamination of solid dosage forms, which is thought
`to arise chiefly from raw materials rather than the
`
`7
`
`

`

`292
`
`Kottke and Rudunic
`
`Table 2 Hiestand Compaction Indices for Some Drugs and
`Excipients
`
`Table 3 Average Particle Data for Different Sources of
`Magnesium Stearate
`
`Material
`
`Strain
`Brittle
`Bonding
`index fracture index index
`
`Aspirin
`Caffeine
`Croscarmellose sodium NF
`Dicalcium phosphate
`Erythrorayein dihydrate
`Hydroxypropyl cellulose
`Ibuprofen
`A
`B
`c
`Lactose USP
`Anhydrous
`Hydrous Fast-Flo
`Hydrous beited
`Hydrous spray process
`Spray-dried
`A
`B
`Mannitol
`A
`B
`Methenamine
`Methyl cellulose
`Microcrystalline
`cellulose NF
`Avicel PH 102 (coarse)
`Avicel PH 101 (fine)
`Povidone USP
`Sorbitol NF
`Starch NF
`Corn
`Pregelatinized
`Pregelatinized compressible
`Modified (starch 1500}
`Sucrose NF
`A
`B
`Cc
`
`Source: Refs, 23-28,
`
`15
`13
`2.7
`13
`19
`1.6
`
`1.9
`1.8
`2.7
`
`0.8
`0.4
`0.6
`0.6
`
`0.6
`6.5
`
`0.8
`0.5
`1.6
`4.5
`
`4.3
`3.3
`1.7
`0.9
`
`0.4
`18
`1.2
`1.5
`
`1.0
`0.8
`0.5
`
`0.16 i
`6.34
`2.19
`0,02
`3.79
`0.15
`1.43
`0.98
`2.13
`0.04
`2.10
`
`0.05
`0.57
`0.45
`
`0.27
`0.19
`0.12
`0.45
`
`0.18
`0.12
`
`0.19
`0.15
`0.98
`0.06
`
`0.04
`0.04
`6.42
`6.16
`
`0.26
`6,14
`6.02
`0.27
`
`0.35
`0.42
`0.53
`
`0.98
`1.51
`1.21
`
`1.40
`1.70
`2.16
`2.12
`
`1.47
`1.81
`
`2.18
`2.26
`0.84
`3.02
`
`2.20
`2.37
`3.70
`1.70
`
`248
`2.02
`2.08
`2.30
`
`1.45
`1,79
`1.55
`
`Ibrahim and Olur-
`manufacturing process [35,36].
`inaola monitored the effects of production, environ-
`ment and methed of production, as weil as microbial
`quality of starting materials, on the microbial load
`during various stages of tablet production [35]. Al-
`though high levels of contamination were present
`during the wet granulation process, these levels were
`significantly reduced during the drying process. The
`investigators also found that products derived from
`
`Size
`Surface
`Pore tadius
`Source
`(um)
`area (m”/g)
`(A)
`
`United States
`1.5-3.2
`13,4
`50
`Great Britain
`2.1-5.2
`12.2
`68
`Germany
`41-69
`7A
`6
`
`Italy 36 5.5-9.1 4.6
`
`
`Source: Ref. 32.
`
`natural origins, such as gelatins and starch, can be
`contaminated heavily.
`
`Compatibility
`
`One final area that should be considered when choos-
`ing the excipients to be used in the tablet formulation is
`that of drug-excipient interactions. There is stil! much
`debate as to whether excipient compatibility testing
`should be conducted prior to formulation [37-39].
`These tests most often involve the trituration of smail
`amounts of the active ingredient with a variety of ex-
`cipients. Critics of these small-scale studies argue that
`their predictive value has yet to be established and
`indeed do notreflect actual processing conditions [37].
`Instead,
`they suggest a sound knowledge of the
`chemistry of the materials used in conjuncture with
`“mini-formulation” studies as a preferable method for
`investigation of drug-excipient interactions.
`
` Y= 1.9054 -03035tx F42 = 0.978
`
`DissolutionRate
`
`% Acetylsallcyile Anhydride
`
`Fig. 4 Effect of acetylsalicylic anhydride impurity on the
`dissolution rate of aspirin tablets. (From Ref. 34.)
`
`8
`
`

`

`Tablet Dosage Forms
`
`C. Tablet Components
`
`233
`
`Intact Tablet
`
`SS
`
`DAUG IN
`
`BLOODSTREAM
`
`Coarse
`
`o 2esoRep,Patties

`5
`T3 hr8
`yo So
`gon
`c
`slow dissolution
`
`Biological
`
`Membrane
`
`Conventional solid dosage forms can be divided into
`two classes: those that disintegrate and those that do
`not. Disintegrating dosage forms release dmg by
`breaking down the physical
`integrity of the dosage
`from, usually with the aid of solid disintegrating agents
`or gas-releasing effervescent agents. Nondisintegrating
`tablets are usually made of soluble drugs and ex-
`cipients that will rapidly dissolve in the mouth or GIT
`upon ingestion.
`With the advent of prolonged-release dosage forms,
`some pharmaceutical scientists have begun to regard
`conventional disintegrating dosage forms as “non—
`controlled-release.” This term is a misnomer since,
`with the aid of “super-disintegrants” and other ex-
`cipients, the disintegration of these dosage forms can
`be controlled, both quantitatively and qualitatively.
`Fine
`.
`Moreover, there are still many drugs in which rapid
`Particles
`.
`ney
`atiainment of therapeuticlevels, rather than controlled
`DRUG IN
`Pees
`a oe ‘s rralSOLUTION
`el te
`pee Dee
`release, is required. Analgesics, antibiotics, and drugs
`>
`as
`egetnest
`for the acute treatment of angina pectoris are prime
`tien stwr
`exarnples, These tablets need to be designed so that the
`drug is liberated from the dosage form in such a
`manner that dissolution of the drug is maximized. Very
`often, this means that disintegration of the tablet must
`be followed by granular disintegration (see Fig. 5) to
`promote rapid dissolution and, hence, absorption.
`The ingredients, or excipients, used to make com-
`pressed tablets are numerous. They can be classified by
`their use, or function, as in Table 4. Keep in mind,
`however, that formulations need not contain all the
`types of ingredients listed in this table, Certain ex-
`cipients, such as antioxidants and wetting agents, are
`used only in situations where they are expressly needed
`to assure the stability and solubility of the active in-
`gredients. Other excipients, such as dissolution modi-
`fiers,
`are
`used
`primarily
`in
`controlled-release
`formulations. In fact, by reducing the number of in-
`gredients in a formulation, one will generally be re-
`ducing the number of problems that can arise in the
`manufacturing process. Hence, many formulators ad-
`here to the motto “Keep it simple.”
`Because of the nature of modern pharmaceutical
`systems, formulators have made more complete in-
`vestigations of the materials they use. This interest has
`identified several materials that may have more than one
`use in tableted systems. The type of effect that an ex-
`cipient will produce is often dependent upon the con-
`centration in which it is used. For example, Table 5 lists
`some ‘“multiuse” excipients and the corresponding con-
`centration ranges required for their various applications.
`
`eee
`
`dissolution
`
`
`
`Fig. 5 Absorption of a drug from anintact tablet.
`
`A telatively new class of “co-processed” excipients
`now exists. These excipients are essentially a “pre-
`blend” of two or more excipients that are commonly
`used in conjunction with each other.
`
`Active Ingredients
`
`The dose of the drug to be administered has a pro-
`found effect on the design and formulation of a dosage
`form. Content uniformity and drug stability become
`very important issues when dealing with highly potent
`compounds that are delivered in very small doses {e.¢.,
`oral contraceptives), However, the effect of the drug’s
`properties on the tablet, in this case,
`is minimal. In
`general, as the dosage increases, so does the effect of
`the drug’s attributes on the tablet.
`
`Table 4 Ingredients Used in Tablet Formulation
`
`Active ingredient (drug)
`Fillers
`Binders (dry and wet)
`Disintegrants
`Antifrictional agents
`Lubricants
`Glidants
`Antiadherants
`
`Dissolution modifiers
`Absorbents
`Flavoring agents
`Coloring agents
`Wetting agents
`Antioxidants
`Preservatives
`
`9
`
`

`

`294
`
`Kottke and Rudnic
`
`Table 5 Some Multiple-Use Excipients for Tablet Formulation
`
`Use
`Excipient/concentration in formula
`
`Wet or dry binder
`Controlled-release coating
`
`Lubricant
`Controlled-release excipient
`
`Wetor dry binder
`Film former
`Controlled-release excipient
`
`Binder
`Disintegrant
`
`Ethylceliulose
`1-3%
`13%
`Glyceryl palmitostearate
`05%
`10-50%
`Hydroxypropylmethy! cellulose (HPMC), low
`viscosity
`2-S%
`2-10%
`5-25%
`Magnesium aluminum silicate
`2-10%
`2-10%
`Microcrystalline cellulose (MCC)
`08%
`0.2-0.5%
`$-20%
`5-20%
`5-95%
`Polyethylene glycol
`0-10%
`G-15%
`5-40%
`Poly(methacrylates)
`2-10%
`$-20%
`10-50%
`5-10%
`Poly(vinyl pyrrolidone) (Povidone, PVP)
`53-15%
`35-10%
`5-30%
`10-35%
`Starch
`315%
`Intragranular binder/disintegrant
`5-25%
`Wet binder
`520%
`Disintegrant
`
`Improve adhesion of film coat to core
`Glidant
`Antiadherant
`Disintepgrant
`Binder/filler
`
`Lubricant
`Thermoplastic filler/binder
`Controlled-release excipient
`
`Film former
`Controlled-release excipient
`Filler
`Coating excipient
`
`Wet binder
`Coating excipient
`Disintegrant
`Controlled-reiease excipient
`
`Sometimes processing can affect the particle mor-
`phology of the active ingredient. This in turn may lead
`to adverse effects on mixing and tableting operations.
`In particular, micronization of a drug may cause
`crystals to change their shape even though poly-
`morphism is not evidenced,
`
`Fillers
`
`An increasing number of drugs are highly potent and
`are thus used in very low dosages. In order to produce
`tablets of a reasonable size G.e., minimum diameter of
`
`3 mm), it is necessary te dilute the drug with an inert
`material. Such diluents should meet importantcriteria,
`including low cost and good tableting qualities. It may
`be possible, in some instances, to combine the role of
`diluent with a different property, such as disintegrant
`or flavoring agent.
`Commonly used fillers and binders and their com-
`parative properties are listed in Table 6. As can be seen
`by this list, both organic and inorganic materials are
`used as fillers and binders. The organic materials used
`are primarily carbohydrates because of their general
`ability to enhance the product’s mechanicalstrength as
`
`10
`
`10
`
`

`

`Tablet Dosage Forms
`
`295
`
`Table 6 Comparative Properties of Some Directly Compressible Fillers*tgegnnn
`
`
`
`
`
`Compactibility Flowability Solubility Disintegration Hygroscopicity LwubricityFiller Stabilityieweee
`Dextrose
`3
`2
`4
`2
`]
`2
`3
`Spray-dried lactose
`3
`5
`4
`3
`1
`2
`4
`Fast-Flo lactose
`4
`4
`4
`4
`J
`2
`4
`Anhydrous lactose
`2
`3
`4
`4
`5
`2
`4
`Emdex (dextrates}
`3
`4

`3
`l
`2
`3
`Sucrose
`4
`3
`5
`4
`4
`i
`4
`Starch
`2
`i
`0
`4
`3
`3
`3
`Starch 1500
`3
`2
`2
`4
`3
`2
`4
`Dicaicium phosphate
`3
`4
`1
`2
`i
`2
`5
`
`5 1 0 2 2 4Avicel (MCC) 5
`
`
`
`
`
`
`
`“Graded on a scale from 5 (good/high) downto 1 (poor/low); 0 means none.
`
`well as their freedom from. toxicity, acceptable taste,
`and reasonable solubility profiles.
`One of the most commonly used carbohydrates in
`compressed tablets is lactose. Work by Bolhuis and
`Lerk [40] and Shangraw et al.
`[6] has demonstrated
`that all lactoses are not equivalent as determined by
`chemical, physiochemical, and functional measure-
`ments. In addition to the various particle-size grades of
`normal hydrous lactose, one can purchase spray-dried
`lactose, which is an agglomerate of a-lactose mono-
`hydrate crystals with up to 10% amorphous material.
`Spray-dried lactose has very good flow properties, but
`its poor compression characteristics require the addi-
`tion of a binder such as MCC. However, one particular
`brand of spherical crystalline/amorphous agglomer-
`ate, Fast-Fio (NF hydrous), possesses superior com-
`pressibility
`and
`dissolution
`characteristics. The
`spherical nature of the crystals make them more
`compressible than spray-dried agglomerates of lactose
`[41]. Anhydrouslactose has also been used as a diluent,
`particularly in direct compression formulations where
`low moisture content is desirable, since it has very
`gocd stability and a reduced tendency to color upon
`aging. Another advantage in the use of anhydrous
`lactose is that its insensitivity to temperature changes
`
`to be reworked with relative ease. Un-
`allows it
`fortunately,
`its flow properties are not particularly
`good and its compressibility is inferior to other forms
`of lactose. Gne mustalso give attention, though,to this
`component’s stability, as aging may adversely affect
`these properties.
`Some other sugars are now being produced in spe-
`cial grades to meet the needs of the pharmaceutical
`industry. Most of these products contain combinations
`of sucrose with invert sugar or modified dexirins and
`are of particular value in the formulation of chewable
`tablets. In addition, crystalline maltose and directly
`co

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