throbber
Pharmaceutical
`Dissolution Testing
`
`
`
`© 2005 by Taylor & Francis Group, LLC© 2005 by Taylor & Francis Group, LLC
`
`MYLAN EXHIBIT 1032
`
`

`

`Pharmaceutical
`Dissolution Testing
`
`Edited by
`
`Jennifer Dressman
`Johann Wolfang Goethe University
`Frankfurt, Germany
`
`Johannes Krämer
`Phast GmbH
`Homburg/Saar, Germany
`
`
`
`© 2005 by Taylor & Francis Group, LLC© 2005 by Taylor & Francis Group, LLC
`
`

`

`Published in 2005 by
`Taylor & Francis Group
`6000 Broken Sound Parkway NW, Suite 300
`Boca Raton, FL 33487-2742
`
`© 2005 by Taylor & Francis Group, LLC
`
`No claim to original U.S. Government works
`Printed in the United States of America on acid-free paper
`10 9 8 7 6 5 4 3 2 1
`
`International Standard Book Number-10: 0-8247-5467-0 (Hardcover)
`International Standard Book Number-13: 978-0-8247-5467-9 (Hardcover)
`
`This book contains information obtained from authentic and highly regarded sources. Reprinted material is
`quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts
`have been made to publish reliable data and information, but the author and the publisher cannot assume
`responsibility for the validity of all materials or for the consequences of their use.
`
`No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,
`mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and
`recording, or in any information storage or retrieval system, without written permission from the publishers.
`
`For permission to photocopy or use material electronically from this work, please access www.copyright.com
`(http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood Drive,
`Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration
`for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate
`system of payment has been arranged.
`
`Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only
`for identification and explanation without intent to infringe.
`
`Library of Congress Cataloging-in-Publication Data
`
`Catalog record is available from the Library of Congress
`
`Taylor & Francis Group
`is the Academic Division of T&F Informa plc.
`
`Visit the Taylor & Francis Web site at
`http://www.taylorandfrancis.com
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`1
`
`Historical Development of
`Dissolution Testing
`
`JOHANNES KRA¨ MER, LEE TIMOTHY GRADY,
`and JAYACHANDAR GAJENDRAN
`
`Phast GmbH, Biomedizinisches Zentrum,
`Homburg/Saar, Germany
`
`INTRODUCTION
`
`Adequate oral bioavailability is a key pre-requisite for any
`orally administered drug to be systemically effective. Dissolu-
`tion (release of the drug from the dosage form) is of primary
`importance for all conventionally constructed, solid oral
`dosage forms in general, and for modified-release dosage
`forms in particular, and can be the rate limiting step for the
`absorption of drugs administered orally (1). Physicochemi-
`cally, ‘‘Dissolution is the process by which a solid substance
`enters the solvent phase to yield a solution’’ (2). Dissolution
`of the drug substance is a multi-step process involving
`
`1
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`2
`
`Kra¨mer et al.
`
`heterogeneous reactions/interactions between the phases of
`the solute–solute and solvent–solvent phases and at the
`solute–solvent interface (3). The heterogeneous reactions that
`constitute the overall mass transfer process may be categor-
`ized as (i) removal of the solute from the solid phase, (ii)
`accomodation of the solute in the liquid phase, and (iii) diffu-
`sive and/or convective transport of the solute away from the
`solid/liquid interface into the bulk phase. From the dosage
`form perspective, dissolution of the active pharmaceutical
`ingredient, rather than disintegration of the dosage form, is
`often the rate determining step in presenting the drug in
`solution to the absorbing membrane. Tests to characterize the
`dissolution behavior of the dosage form, which per se also
`take disintegration characteristics into consideration, are
`usually conducted using methods and apparatus that have
`been standardized virtually worldwide over the past decade
`or so, as part of the ongoing effort to harmonize pharmaceuti-
`cal manufacturing and quality control on a global basis.
`the
`The history of dissolution testing in terms of
`evolution of the apparatus used was reviewed thoroughly by
`Banakar in 1991 (2). This chapter focuses first on the pharma-
`copeial history of dissolution testing, which has led to manda-
`tory dissolution testing of many types of dosage forms for
`quality control purposes, and then gives a detailed history
`of two newer compendial apparatus, the reciprocating cylin-
`der and the flow-through cell apparatus. The last section of
`the chapter provides some historical
`information on the
`experimental approach of Herbert Strieker’s group. His scien-
`tific work in combining permeation studies directly with a dis-
`solution tester, is very much in line with the Biopharmaceutic
`Classification System (BCS), but was published more than
`two decades earlier than the BCS (4) and can therefore be
`viewed as the forerunner of the BCS approach.
`
`FROM DISINTEGRATION TO DISSOLUTION
`
`Compressed tablets continue to enjoy the status of being the
`most widely used oral dosage form. Tablets are solid oral
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`3
`
`dosage forms of medicinal substances, usually prepared with
`the aid of suitable pharmaceutical excipients. Despite the
`advantages offered by this dosage form, the problems asso-
`ciated with formulation factors remain to some extent enig-
`matic to the pharmaceutical scientist.
`In the case of
`conventional (immediate-release) solid oral drug products,
`the release properties are mainly influenced by disintegration
`of the solid dosage form and dissolution of drug from the dis-
`integrated particles. In some cases, where disintegration is
`slow, the rate of dissolution can depend on the disintegration
`process, and in such cases disintegration can influence the
`systemic exposure, in turn affecting the outcome of both bioa-
`vailability and bioequivalence studies. The composition of all
`compressed conventional tablets should, in fact, be designed
`to guarantee that they will readily undergo both disintegra-
`tion and dissolution in the upper gastrointestinal (GI) tract
`(1). All factors that can influence the physicochemical proper-
`ties of the dosage form can influence the disintegration of the
`tablet and subsequently the dissolution of the drug. Since the
`1960s, the so-called ‘‘new generation’’ of pharmaceutical
`scientists has been engaged in defining, with increasing
`chemical and mathematical precision, the individual vari-
`ables in solid dosage form technology, their cumulative effects
`and the significance of these for in vitro and in vivo dosage
`form performance, a goal that had eluded the previous
`generation of pharmaceutical scientists and artisans.
`As already mentioned, both dissolution and disintegra-
`tion are parameters of prime importance in the product
`development strategy (5), with disintegration often being
`considered as a first order process and dissolution from drug
`particles as proportional to the concentration difference of
`the drug between the particle surface and the bulk solution.
`Disintegration usually reflects the effect of formulation and
`manufacturing process variables, whereas the dissolution
`from drug particles mainly reflects the effect of solubility and
`particle size, which are largely properties of the drug raw
`material, but can also be influenced significantly by proces-
`sing and formulation. It is usually assumed that the dissolu-
`tion of drug from the surface of the intact dosage form is
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`4
`
`Kra¨mer et al.
`
`negligible, so tablet disintegration is key to creating a larger
`surface area from which the drug can readily dissolve. However,
`tablet disintegration in and of itself may not be a reliable indica-
`tor of the subsequent dissolution process, so the tablet disinte-
`gration tests used as a quality assurance measure may or may
`not be a an adequate indicator of how well the dosage form will
`release its active ingredient in vivo. Only where a direct
`relationship between disintegration and dissolution has been
`established, can a waiver of dissolution testing requirements
`for the dosage form be considered (6).
`Like disintegration testing, dissolution tests do not prove
`conclusively that the dosage form will release the drug in vivo
`in a specific manner, but dissolution does come one step
`closer, in that it helps establish whether the drug can become
`available for absorption in terms of being in solution at the
`sites of absorption. The period 1960–1970 saw a proliferation
`of designs for dissolution apparatus (7). This effort led to the
`adoption of an official dissolution testing apparatus in the
`United States Pharmacopeia (USP) and dissolution tests with
`specifications for 12 individual drug product monographs in
`the pharmacopeia. These tests set the stage for the evolution
`of dissolution testing into its current form.
`
`DISSOLUTION METHODOLOGIES
`
`The theories applied to dissolution have stood the test of time.
`Basic understanding of these theories and their application
`are essential for the design and development of sound dissolu-
`tion methodologies as well as for deriving complementary
`statistical and mathematical techniques for unbiased dis-
`solution profile comparison (3).
`In the 1960s and 1970s, there was a proliferation of
`dissolution apparatus design. With their diverse design speci-
`fications and operating conditions, dissolution curves
`obtained with them were often not comparable and it was
`gradually realized that a standardization of methods was
`needed, which would enable correlation of data obtained with
`the various test apparatus. As a result,
`the National
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`5
`
`Formulary (NF) XIV and USP XVIII and XIX (8) standardized
`both the apparatus design and the conditions of operation for
`given products. With these tests, comparable results could be
`obtained with the same apparatus design, even when the appa-
`ratus was produced by different equipment manufacturers.
`
`PERSPECTIVE ON THE HISTORY OF
`COMPENDIAL DISSOLUTION TESTING
`
`. . . it would seem that prompt action of certain remedies
`must be considerably impaired by firm compression. ...
`the composition of all compressed tablets should be such
`that they will readily undergo disintegration and solution
`in the stomach. [C. Caspari, ‘‘A Treatise on Pharmacy,’’
`1895, Lea Bros., Philadelphia, 344.]
`
`Tableting technology has had more than a century of
`development, yet the essential problems and advantages of
`tablets were perceived in broad brush strokes within the
`first years. Compression, powder flow, granulation, slugging,
`binders, lubrication, and disintegration were all appreciated
`early on, if not scientifically, at least as important considera-
`tions in the art of pharmacy. Industrial applications of tablet-
`ing were not limited to drugs but found broad application in
`the confectionery and general chemical industry as well. Poor
`results were always evident and, already at the turn of the
`20th century, some items were being referred to as ‘‘brick-
`bats’’ in the trade.
`With the modern era of medicine, best dated as starting
`in 1937, tablets took on new importance. Modern synthetic
`drugs, being more crystalline, were generally more amenable
`to formulation as solid dosage forms, and this led to greater
`emphasis on these dosage forms (9). Tableting technology
`was still largely empirical up to 1950, as is evidenced by the
`literature of the day. Only limited work was done before
`1950, on drug release from dosage forms, as opposed to disin-
`tegration tests, partly because convenient and sensitive
`chemical analyses were not yet available. At that time, disso-
`lution discussions mainly revolved around the question of
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`6
`
`Kra¨mer et al.
`
`whether the entire content could be dissolved and was mostly
`limited to tablets of simple, soluble chemicals or their salts.
`The first official disintegration tests were adopted in
`1945 by the British Pharmacopoeia and in 1950 by the USP.
`Even then,
`it was recognized that disintegration testing
`is an insufficient criterion for product performance, as
`evidenced by the USP-NF statement that ‘‘disintegration does
`not imply complete solution of the tablet or even of its active
`ingredient.’’ Real appreciation of the significance of drug
`release from solid dosage forms with regard to clinical relia-
`bility did not develop until there were sporadic reports of
`product failures in the late 1950s, particularly vitamin pro-
`ducts. Work in Canada by Chapman et al., for example,
`demonstrated that formulations with long disintegration
`times might not be physiologically available. In addition,
`the great pioneering pharmacokineticist John Wagner
`demonstrated in the 1950s that certain enteric-coated pro-
`ducts did not release drug during Gl passage and that this
`could be related to poor performance in disintegration tests.
`Two separate developments must be appreciated in
`discussing events from 1960 onward. These enabled the field
`to progress quickly once they were recognized. The first was
`the increasing availability of reliable and convenient instru-
`mental methods of analysis, especially for drugs in biological
`fluids. The second, and equally important development, was
`the fact that a new generation of pharmaceutical scientists
`were being trained to apply physical chemistry to pharmacy,
`a development largely attributable, at least in the United
`States, to the legendary Takeru Higuchi and his students.
`Further instances in which tablets disintegrated well (in
`vitro) but were nonetheless clinically inactive came to light.
`Work in the early 1960s by Campagna, Nelson, and Levy
`had considerable impact on this fast-dawning consciousness.
`By 1962, sufficient industrial concern had been raised to
`merit a survey of 76 products by the Phamaceutical Manufac-
`turers of America (PMA) Quality Control Section’s Tablet
`Committee. This survey set out to determine the extent of
`drug dissolved as a function of drug solubility and product
`disintegration time. They found significant problems, mostly
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`7
`
`occurring with drugs of less than 0.3% (30 ug/mL) solubility in
`water, and came within a hair of recommending that dissolu-
`tion, rather than disintegration, standards be set on drugs of
`less than 1% solubility.
`Another development that occurred between 1963 and
`1968 that continues to confabulate scientific discussions of
`drug release and dissolution testing was the issue of generic
`drug approval. During this period, drug bioavailability
`became a marketing, political, and economic issue. At first,
`generic products were seen as falling short on performance.
`However later it turned out that the older formulations, that
`had been marketplace innovators, were often short on perfor-
`mance compared to the newly formulated generic products.
`To better compare and characterize multi-source (gen-
`eric) products, the USP-NF Joint Panel on Physiological
`1)
`(Table
`Availability was set up in 1967
`under Rudolph
`Blythe, who already had led industrial attempts at standardi-
`zation of drug release tests. Discussions of the Joint Panel led
`to adoption, in 1970, of an official apparatus, the Rotating
`Basket, derived from the design of the late M. Pernarowski,
`long an active force in Canadian pharmaceutical sciences. A
`commercial reaction flask was used for cost and ruggedness.
`The monograph requirements were shepherded by William
`J. Mader, an industrial expert in analysis and control, who
`directed the American Pharmaceutical Association (APhA)
`Foundation’s Drug Standards Laboratory. William A. Hanson
`prepared the first apparatus and later commercialized a
`series of models.
`The Joint Panel proposed no in vivo requirements, but
`individual dissolution testing requirements were adopted in
`12 compendial monographs. USP tests measured the time to
`attain a specified amount dissolved, whereas NF used the
`more workable test for the amount dissolved at a specified
`time. Controversy with respect to equipment selection and
`methodology raged at the time of the first official dissolution
`tests. As more laboratories entered the field, and experience
`(and mistakes!) accumulated, the period 1970–1980 was one
`of intensive refinement of official test methods and dissolution
`test equipment.
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`8
`
`Kra¨mer et al.
`
`Table 1 USP Timeline from 1945–1999
`
`1945–1950
`1962
`1967
`
`1970
`1971–1974
`
`1975
`
`1976
`
`1977
`1978
`
`1979
`1980
`
`1981
`
`1982
`1984
`1985
`
`1990
`
`1995
`
`1997
`
`1999
`
`Disintegration official in Brit Pharmacon and USP
`PMA Tablet Committee proposes 1% solubility threshold
`USP and NF Joint Panel on Physiological Availability
`chooses dissolution as a test chooses an apparatus
`Initial 12 monograph standards official
`Variables assessment; more laboratories, three
`Collaborative Studies by PMA and Acad. Pharm. Sci
`First calibrator tablets pressed; First Case default proposed
`to USP
`USP Policy—comprehensive need; calibrators Collaborative
`Study
`USP Guidelines for setting Dissolution standards
`Apparatus 2—Paddle adopted; two Calibrator Tablets
`adopted
`New decision rule and acceptance criteria
`Three case Policy proposed; USP Guidelines revised; 70
`monographs now have standards
`Policy adopted January, includes the default First Case,
`monograph proposals published in June
`Policy proposed for modified-release dosage forms
`Revised policy adopted for modified-release forms
`Standards now in nearly 400 monographs; field considered
`mature; Chapter < 724 > covers extended-release and
`enteric-coated
`Harmonization: apparatus 4—Flow- through adopted;
`Apparatus 3 Apparatus 5, 6, 7 fortransdermal drugs
`Third Generation testing proposed—batch phenomenon;
`propose reduction in calibration test number
`FIP Guidelines for Dissolution Testing of Solid Oral
`Products; pooled analytical samples allowed
`Enzymes allowed for gelatin capsules reduction from 0.1 N
`to 0.01 N Hcl
`
`Later, a second apparatus was based on Poole’s use of
`available organic synthesis round-bottom flasks as refined
`by the St. Louis laboratory. Neither choice of dissolution
`equipment proved to be optimal, indeed, it may have been
`better if the introduction of the two apparatus had occurred
`in the reverse order. With time, the USP would go on to offer
`a total of seven apparatuses, several of which were introduced
`primarily for products applied to the skin.
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`9
`
`At the time, the biopharmaceutical problems, such as
`with low-solubility drugs, both in theoretical terms and in
`actual clinical failures were already well recognized. The
`objective of the Joint Panel was to design tests which could
`determine whether tablets dissolved within a reasonable
`volume, in a commercial flask. In those days, drugs were often
`prescribed in higher doses, so the volume of the dissolution
`vessels in terms of providing an adequate volume to enable
`complete dissolution of the dose had to be taken into design
`consideration. Over the last 35 years there has been a trend
`to develop more potent drugs, with attendant decrease in
`doses required (with notable exceptions, especially anti-infec-
`tives). For example, an antihypertensive may have been
`dosed at 250 mg, but newer drugs in the same category
`coming onto the market might be dosed as low as 5 mg. Sub-
`sequently, there has been a change in the amount of drug that
`needs to get dissolved for many categories of drugs. Neverthe-
`less, a few monographs (e.g., digoxin tablets) have always pre-
`sented a challenge to design of dissolution tests. The following
`factors exemplify typical problems associated with the devel-
`opment of dissolution tests for quality control purposes:
`
`1. The need to have a manageable volume of dissolution
`medium.
`2. The development of less-soluble compounds as drugs
`(resulting in problems in achieving complete dissolu-
`tion in a manageable volume of medium).
`Insufficient analytical sensitivity for low-dose drugs,
`especially at higher media volumes (as illustrated in
`the USP monograph on digoxin tablets).
`
`3.
`
`It should be remembered that in 1970, when drug-
`release/dissolution tests first became official through the
`leadership of USP and NF, marketed tablets or capsules in
`general simply did not have a defined dissolution character.
`They were not formulated to achieve a particular dissolution
`performance, nor were they subjected to quality control by
`means of dissolution testing. Moreover, the U.S. Food and
`Drug Administration (FDA) was not prepared to enforce
`dissolution requirements or to even to judge their value.
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`10
`
`Kra¨mer et al.
`
`The tremendous value of dissolution testing to quality
`control had not yet been established, and this potential role
`was perceived in 1970 only dimly even by the best placed
`observers. Until the early 1970s, discussions of dissolution
`were restricted to the context of in vivo–in vitro correlation
`(IVIVC) with some physiologic parameter. The missing link
`between the quality control and IVIVC aims of dissolution
`testing was that dissolution testing is sensitive to formulation
`variables that might be of biological significance because
`dissolution testing is sensitive in general to formulation
`variables.
`Between 1970 and 1975, it became clear that dissolution
`testing could also play a role in formulation research and
`product quality control. Consistent with this new awareness
`of the value of dissolution testing in terms of quality control
`as well as bioavailability, USP adopted a new policy in 1976
`that favored the inclusion of dissolution requirements in
`essentially all tablet and capsule monographs. Thomas Med-
`wick chaired the Subcommittee that led to this policy. Due
`to lack of industrial cooperation, the policy did not achieve full
`realization. Nevertheless, by July 1980 the role of dissolution
`in quality control had grown to appeareance in 72 mono-
`graphs, most supplied by USP’s own laboratory under the
`direction of Lee Timothy Grady, and FDA’s laboratory under
`the direction of Thomas P. Layloff. USP continued to
`(Fig.
`1)
`adopt further dissolution apparatus designs
`and
`refine the methodology between 1975 and 1980, as shown in
`Table 1.
`Over the years, dissolution testing has expanded beyond
`ordinary tablets and capsules—first to extended-release and
`delayed-release (enteric-coated) articles, then to transder-
`mals, multivitamin and minerals products, and to Class
`Monographs for non-prescription drug combinations. (Note:
`at the time, ‘‘sustained-release’’ products were being tested,
`unofficially, in the NF Rotating Bottle apparatus).
`Tablets and capsules that became available on the
`market in the above time frame often showed 10–20% relative
`standard deviation in amounts dissolved. The FDA’s St. Louis
`Laboratories results on about 200 different batches of drugs
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`11
`
`Figure 1 Rotating basket method. Source: From Ref. 10.
`
`available showed that variation tend to be greatest for slowly
`dissolving drugs. Newer formulations, developed using disso-
`lution testing as one of the aids to product design, are much
`more consistent. Another early problem in dissolution testing
`was lab-to-lab disagreement in results. This problem was
`essentially resolved when testing of standard ‘‘calibrator’’
`tablets were added to the study design, for which average
`dissolution values had to comply with the USP specifications
`to qualify the equipment in terms of its operation. Every
`calibrator batch produced since the inaugauration of calibra-
`tors has been subjected to a Pharmaceutical Manufactorers of
`America (PMA)/Pharmaceutical Research and Manufacturers
`of America (PhRMA) collaborative study to determine accep-
`tance statistics. Originally, calibrators were adopted to pick
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`12
`
`Kra¨mer et al.
`
`up the influence on dissolution results due to vibration in the
`equipment, failures in the drive chains and belts, and opera-
`tor error. In fact, perturbations introduced in USP equipment
`are usually detected by at least one of the two types of calibra-
`tors (prednisone or salicylic acid tablets). Although the cali-
`brators were not adopted primarily to test either deaeration
`or temperature control, they proved to be of value here, too.
`As a follow-up, the USP developed general guidelines on de-
`aeration early in the 1990s, presently favoring a combination
`of heat and vacuum. In the late 1990s, the number of tests to
`qualify an apparatus was halved. Yet even today, an appara-
`tus can fail the calibrator tablet tests, since small individual
`deviations in the mechanical calibration and operator error
`can combine to produce out of specification results for the cali-
`brator. Thus, the calibrators are an important check on oper-
`ating procedures, especially in terms of consistency between
`labs on an international basis.
`In addition to the increasing interest in dissolution as a
`quality control procedure and aid to development of dosage
`forms, bioavailability issues continued to be raised through-
`out the 1970–1980 period, as clinical problems with various
`oral solid products dissolution and bioavailability continued
`to crop up. Much of the impetus behind the bioavailability
`discussions came from the issue of bioequivalence of drugs
`as this relates to generic substitution. In January 1973,
`FDA proposed the first bioavailability regulations. These
`were followed in January 1975 by more detailed bioequiva-
`lence and bioavailability regulations, which became final in
`February 1977. A controversial issue in these regulations
`proved to be the measurement of the rate of absorption. The
`1975 revision proposal was the first to contain the concept
`of an in vitro bioequivalence requirement, which reflected
`the growing awareness of the general utility of dissolution
`testing at that time.
`A major wave of generic equivalents were introduced to
`the U.S. market following the Hatch–Waxman legislation in
`the early 1970s and ANDA applications to the FDA provided
`the great majority of IVIVC available to USP for non-First
`Case standards setting during the following years.
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`13
`
`From the USP perspective, digoxin tablets became and
`remained the benchmark for the impact of dissolution on bioa-
`vailability. It is a life-saving and maintaining drug, has a low
`therapeutic index, is poorly soluble, has a narrow absorption
`window (due to p-glycoprotein exotransport) and it is formu-
`lated using a low proportion of drug:excipients due to its high
`potency. Correlation between dissolution and absorption was
`first shown for digoxin in 1973. The official dissolution stan-
`dard that followed was the watershed for the entire field. It
`is interesting to note that clinical observations for digoxin
`tablets were made in only few patients. Similarly, the original
`concerns of John Wagner over prednisone tablets were based
`on observations in just one patient. The message from these
`experiences is that decisive bioinequivalences can be picked
`up even in very small patient populations.
`At the time the critical decisions were made, it seemed
`that diminished bioavailability could usually be linked to
`formulation problems. Scientists recognized early that when
`the rate of dissolution is less than the rate of absorption,
`the dissolution test results can be predictive of correlation
`with bioavailability or clinical outcome. At that time, there
`was little recognition that intestinal and/or hepatic metabo-
`lism mattered, an exception being the phenothiazines. So
`the primary focus was on particle size and solubility. Observa-
`tions with prednisone, nitrofurantoin, digoxin and other
`low-solubility drugs were pivotal to decision making at the
`time, since the dissolution results could be directly linked to
`clinical data. Scientists recognized that it is not the solubility
`of the drug alone that is critical, but that the effective surface
`area from which the drug is dissolving also plays a major role,
`as described by the Noyes–Whitney equation, which describes
`the flux of drug into solution as a mathematical relationship
`between these factors.
`In the mid-70s, it was a generally expressed opinion that
`there could be as many as 100 formulation factors that might
`affect bioavailability or bioequivalence. In fact, most of the
`documented problems centered around the use of
`the
`hydrophobic magnesium stearate as a lubricant or use of a
`hydrophobic shellac subcoat in the production of sugar-coated
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`14
`
`Kra¨mer et al.
`
`tablets. At that time, products were also often shellac-coated
`both for elegance and for longer shelf life. In addition, inade-
`quate disintegration was still a problem, often related to
`disintegrant integrity and the force of compression in the
`tableting process. All four of these factors are sensitive to
`dissolution testing. Wherever there was a medically signifi-
`cant problem, a dissolution test was able to show the differ-
`ence between the nonequivalent formulations and this is, in
`general, still true today.
`In addition to the scientific aspects, much of the discus-
`sion around dissolution and bioequivalence really was and
`is a political, social, and economic argument. Because of reluc-
`tance on the part of the pharmaceutical industry to cooperate
`with USP, a default standard was proposed to the USP in
`1975. This proposal called for 60% dissolved at 20 min in
`water, testing individual units in the official apparatus and
`was based on observations by Bill Mader and Rudy Blythe
`in 1968–1970, who had demonstrated that one could start get-
`ting meaningful data at 20 min, consistent with typical disin-
`tegration times in those days. In 1981, a USP Subcommittee
`pushed forward the default condition, resulting in an explo-
`sion in the number of dissolution tests from 70 to 400 in
`1985, a five-fold increase in four years! Selection of a higher
`amount dissolved, 75%, made for tighter data, whilst the
`longer test time, 45 min, was chosen because it gave formula-
`tors some flexibility in product design to improve elegance,
`stability, and/or to reduce friability—in other words, a lot of
`considerations not directly linked to dissolution. Subse-
`quently, industrial cooperation improved, and later the FDA
`Office of Generic Drugs and the USP established a coopera-
`tion, with the FDA supplying both dissolution and bioavail-
`ability data and information to USP.
`Experience has demonstrated that where a medically
`significant difference in bioavailability has been found among
`supposedly identical products, a dissolution test has been effi-
`cacious in discriminating among them. A practical problem
`has been the converse, that is, dissolution tests are sometimes
`too discriminating, so that it is not uncommon for a clinically
`acceptable product to perform poorly in an official dissolution
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Historical Development of Dissolution Testing
`
`15
`
`test. In such cases, the Committee of Revision has been mindful
`of striking the right balance: including as many acceptable
`products as possible, yet not setting forth dissolution specifica-
`tions that would raise scientific concern about bioequivalence.
`
`COMPENDIAL APPARATUS
`
`The USP 27, NF22 (11) now recognizes seven dissolution
`apparatus specifically, and describes them and, in some cases
`allowable modifications, in detail. The choice of the dissolu-
`tion apparatus should be considered during the development
`of the dissolution methods, since it can affect the results
`and the duration of the test. The type of dosage form under
`investigation is the primary consideration in apparatus
`selection.
`
`Apparatus Classification in the USP
`
`Apparatus 1 (rotating basket)
`Apparatus 2 (paddle assembly)
`Apparatus 3 (reciprocating cylinder)
`Apparatus 4 (flow-through cell)
`Apparatus 5 (paddle over disk)
`Apparatus 6 (cylinder)
`Apparatus 7 (reciprocating holder)
`
`The European Pharmacopoeia (Ph. Eur.) has also
`adopted some of the apparatus designs (12) described in the
`USP, with some minor modifications in the specifications.
`Small but persistent differences between the two have their
`origin in the fact that the American metal proces

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