throbber
Page 1
`
`LUPIN EX. 1026
`Lupin v. iCeutica
`US Patent No. 8,999,387
`
`

`
` Dedication
`
`Disclaimer
`
`The laboratory procedures described herein may involve hazardous material. The authors,
`speaking for themselves, expressly disclaim any liability to users of these lab procedures
`and apparatus for consequential damages of any kind arising out of or connected with
`their use. Items of apparatus and test methodology described in this handbook are in-
`tended to illustrate proper techniques to obtain a quality analysis and are not to be
`considered official and/or required. Official compendia and regulatory requirements are
`to be considered the only authoritative source for such matters.
`
`Copyright © 2004 by Royal A. Hanson
`
`All rights reserved. No part of this book may be reproduced or used in any form or by
`any means whatsoever without the permission of the publisher except in the case of brief
`quotations embodied in critical articles or reviews.
`
`Printed in the United States of America
`2 1 10 9 8 7 6 5 4 3
`
`Library of Congress Control Number 2004115666
`
`Hanson. Royal, and Gray, Vivian
`Handbook of Dissolution Testing/Royal Hanson and Vivian Gray
`Includes bibliographical references and index
`ISBN 0—9761519—0—l (hardcover)
`
`Published by Dissolution Technologies, Incorporated
`9 Yorkridge Trail, Hockessin, DE 19707.
`www_dissolutiontech.com
`
`This, the Third Edition of the Handbook of Dissolution Testing, is
`
`dedicated to the memory of William A. Hanson, Ph.D., founder of
`Hanson Research Corporation. Dr. Hanson was a recognized pioneer
`and longtime leader in the field of dissolution testing.
`Prior to his death at the end of 1994, Dr. Hanson was a constant
`
`contributor to the evolution of test apparatus and procedures. His sig-
`nificant experiments in the field led to his completion of a Doctorate
`in Engineering with a dissertation on the variables encountered in phar-
`maceutical dissolution testing.
`Dr. Hanson supplemented his technical work by writing the Hand-
`book of Dissolution Testing in 1982, followed by a Second Edition in
`1991. As the field of dissolution testing continues to evolve, the authors
`are proud to follow in Dr. Hanson’s giant footsteps in presenting this
`Third Edition of his important book.
`
`
`
`Page 2
`
`

`
`1
`
`Overall
`
`Considerations
`
`()ver the last several decades, the dissolution test has grown from
`a pioneering prototype into a standardized test of pharmaceutical dos-
`age quality assurance. Considering industry’s significant investment in
`drug R&D, formulations, and clinical testing, the dissolution test has
`been proven to provide a simple, cost—efficient, yet rigorous laboratory
`test for drug release characteristics. For this reason alone, it is worthy
`of consideration by developing countries and the worldwide pharma-
`ceutical community.
`The second edition of this Handbook of Dissolution Testing (1991)
`
`was written when the methods, techniques and constraints of dissolu-
`tion testing had been established using Apparatus l (Basket) and Ap-
`paratus 2 (Paddle) as the techniques of choice. These two apparatus
`remain the dominant methods to analyze dissolution of immediate re-
`lease dosage forms. The USP has now added Apparatus 3 (Recipro-
`cating Cylinder), Apparatus 4 (Flow—Through Cell), Apparatus 5 (Pad-
`dle Over Disk), Apparatus 6 (Cylinder), and Apparatus 7 (Reciprocat-
`ing Holder) (l) and the EP has added the flow—through cell, recipro-
`cating cylinder and paddle over disk transdermal apparatus (2). All of
`these apparatus are described in detail in Chapters 3 and 4.
`As this Third Edition goes to press, a collaboration to harmonize the
`USP General Chapters Dissolution <711> and Drug Release <724>
`with the European Pharmacopeia and Japanese Pharmacopeia is nearing
`completion. This effort, led by the International Conference on Har-
`monization (ICH), will include coordination of many aspects of the
`general chapters. For example, equipment dimensions and specifica-
`
`Page 3
`
`

`
`Overall Considerations
`
`3
`
`testing. Disintegration testing was originally proposed in an attempt to
`establish minimum standards.
`J’
`
`In the early stages of pharmaceutical development, the goal was an
`“elegant” tablet: a tablet of beauty that did not chip, deteriorate or
`change color. Of course, the harder" the tablet, the less likely it was to
`chip or fracture during packaging Tor shipment.
`Hardness can be increased by inereasing the binding agent and com-
`pression forces. Unfortunately, hardness can be increased to the point
`where the tablet will not disintegrate in the alimentary tract and where
`the only release of drug will arise from the relatively small surface area
`of the outside of the dosage form. In 1997, an important discovery by
`Ralph Shangraw, et al. (3) showed that many vitamin products con-
`taining folic acid were not meeting the standard of dissolving within
`an hour. This has important implications for the absorption of drugs in
`vivo. Even.today, physicians have observed non-disintegrated tablets
`in patients’ stools.
`The disintegration test was introduced to avoid such problems. The
`apparatus consists of six vertical tubes, each about one inch (25 mm) in
`diameter, arranged in a rack with a 10-mesh screen attached to the under
`surface. The tubes are moved 5.5 cm up and down 30 times/minute while
`submerged in water or simulated intestinal fluid at body temperature (37
`°C). To help the process along, a complicated plastic disc “hammers”
`the tablet during the up-and-down movement (Figure 1-1).
`The disintegration test has been official since 1950 (USP XV), and
`it had enjoyed international acceptance. Various modifications have
`been introduced, one incorporating a l0—mesh screen on the top of the
`tubes to keep capsules from floating out. Also available is a sophisti-
`cated apparatus that registers the time for total tablet disintegration and
`plots results on a digital printer. The tablet is said to pass the test when
`no palpable mass remains on the 10-mesh screen after the designated
`time, typically 30 minutes for ordinary tablets and 60 minutes for en-
`teric-coated tablets.
`
`
`
`The test had been mandatory for oral dosage forms for more than
`40 years, but its elimination and replacement with a universal disso-
`lution standard was encouraged in Pharmacopeial Forum in 1981 (4)
`probably because the disintegration test in general had little statistical
`correlation with bioavailability (5). In the early 1960s,
`the industry
`recognized that the ultimate solution of a drug depends upon two fac-
`tors: the disintegration of the tablet and the deaggregation of the par-
`ticles in the tablet. That process is outlined in Figure 1-2 (and will
`receive greater attention in Chapter 2).
`
`Page 4
`
` 2
`
`HANDBOOK OF DISSOLUTION TESTING
`
`tions, equipment modifications, and use of validated sinkers and auto-
`mated equipment are included (see Chapter 7 for more details).
`
`Why Dissolution Testing?
`
`Besides accommodating the obvious need for meeting the legal re-
`quirements for compendial drugs, dissolution testing is increasingly
`used to test special oral dosage forms now listed in the compendia.
`Those dosage forms include vitamins, herbals, veterinary drugs, chew-
`able tablets, suppositories, suspensions, soft gelatin capsules, creams,
`ointments, gels, transdermal systems, and the vast ever-increasing num-
`ber of extended-release preparations. In addition, there are new product
`types and dosage forms under development, such as chewing gums,
`powders, granules, dispersions, microparticles, stents, liposomes, nano-
`spheres and implants, which may require dissolution testing. The phar-
`maceutical industry will continue to use dissolution testing as a laud-
`able quality control measure for their production operations. They also
`are acutely aware that the worldwide sociological trend is toward ex-
`tension of regulation. In the pharmaceutical business, as in any other,
`it pays to anticipate future developments.
`Dissolution testing, of course, is a regular quality control procedure in
`good manufacturing practices (GMP). Whether or not its numbers have
`been correlated with biological effectiveness, the standard dissolution test
`is a simple and inexpensive indicator of a product’s physical consistency.
`If one batch differs widely from. others in its dissolution characteristics,
`or if the dissolution times of production batches show a consistent trend
`upwards or downwards, it sounds a sure warning that some factor in the
`raw material, formulation, or process is out of control.
`Dissolution data are also useful in the early stages of drug devel— /
`opment and formulation. In the early stages of development, researchers
`may take steps to optimize drug and dosage-form characteristics that
`will influence subsequent bioavailability data.
`
`Disintegration Tests
`
`More than fifty years ago, it was recognized that unless an oral dos-
`age form disintegrated into small aggregates, the body could not absorb
`it efficiently. A unique apparatus was devised to establish a standard
`for disintegration. Its description is still published in USP, but now the
`trend is to omit disintegration specifications and to include dissolution
`
`Page 4
`
`

`
`Overall Considerations
`
`5
`
`Rate measured by the
`disintegration test
`
`
`
`Rate measured by the
`dissolution test
`
`Solid dosage form
`
`A}
`
`
`
`
`
`Disintegration from gross
`tablet size to particles of
`various sizes (depending
`on formulation): <2 mm '
`diameter
`
`Deaggregation: breakdown into
`discrete particles that greatly
`increase surface area, providing
`solid-liquid interface and beginning
`dissolution: <0.25 mm diameter
`
`Figure 1-2 The three stages in the dissolution process.
`
`cies, and compendial standards groups. A prominent pharmaceutical
`manufacturer forcibly brought the matter into focus by a routine in-
`vestigation of identical competitive products. Bioavailability tests in—
`dicated a substantial difference in the performance of the two items,
`which were otherwise pharmaceutically identical according to all then—
`existing tests for physical properties.
`The matter was brought to the attention of compendial committees,
`and a study of various dissolution test methods was inaugurated. At
`that time, the major concern involved life-saving drugs, particularly
`
`Page 5
`
`
`
` 4
`
`HANDBOOK OF DISSOLUTION TESTING
`
`T
`5.5-cm stroke, 30/minute
`
`Media 37 °C
`
`1000—mL Beaker
`
`6 Glass Tubes,
`1-inch diameter
`
`Plastic Disks
`(when specified)
`Tablet
`
`10—Mesh Screen
`
`Figure I -1 USP disintegration testing apparatus. (See USP <70]>
`Disintegration.)
`'
`
`From the pattern shown in the diagram it is apparent that the dis-
`solution rate includes the time factor resulting from the disintegration
`process. It also becomes obvious that dissolution testing includes dis— ‘
`integration time and, therefore, that if dissolution data become man-
`datory, then disintegration information becomes superfluous.
`During the period 1990-1995, many USP disintegration tests were
`replaced with dissolution tests, and the disks were removed from
`<70l> Disintegration USP General chapter. Disks may still be called
`for in individual USP monographs.
`
`Early Dissolution Test Development
`In the early 1960s, problems regarding the bioavailability of drugs
`were receiving more and more attention from industry, regulatory agen-
`
`Page 5
`
`

`
`Overall Considerations
`
`7
`
`
`
`Simple
`Drive Motor
`
`‘
`
`Lab Apparatus
`stand
`
`
`
`
`
`Prototype
`Basket-mixer
`
`1-L Resin Vessel
`
`Dosage
`
`Hot Plate
`
`Figure 1-3 An early primitive dissolution tester. (Note basket-propel
`ler assembly.)
`
`Method 3 (not to be confused with today’s Apparatus 3) was intro-
`duced for indomethacin capsules because of a pre—established database.
`That method used a modification of the existing disintegration equip-
`ment (described above) and has not been extended to any other drug.
`This method has since been eliminated from the USP.
`
`As initially specified, the primary dissolution method included the
`rotating basket in essentially the same dimensions as now. It featured
`a 0.25-inch (6.35 mm) shaft, a 37 °C water bath, and a standard 1000-
`mL resin flask similar to the one now specified but with a flat or con-
`cave indention at the apex of the bottom. Any speed-controlled stirring
`device able to operate at 100 rpm : 4% could be used, although other
`speeds were allowed.
`
`Page 6
`
`
`
` 6
`
`HANDBOOK OF DISSOLUTION TESTING
`
`those with a narrow range between ineffective and toxic blood levels.
`Digoxin is an example, because the physician must establish in a ti-
`trated dosage the effective but nontoxic dosage levels for each patient.
`If the patient switches brand sources of this medication, it naturally
`follows that the second brand must closely approximate the first in its
`ability to sustain therapeutic blood levels. Studies suggested that this
`ability could be correlated to some degree with dissolution rate char-
`acteristics.
`Coordination of the studies was assigned to William Mader, director
`of the Drug Standards Laboratories (later evolved into the USP labo-
`ratory) in Washington, D.C., USA, which operated alongside the com-
`pendial groups. Mader took on a formidable job; evaluation and selec-
`tion of a new and official test procedure affecting the pharmaceutical
`and medical professions, regulatory agencies, and drug manufacturers
`constitutes an awesome and agonizing task. Everyone in industry, gov-
`ernment, and academia who suggested methodology seemed to have a
`private ax to grind. Through that period, Bill Hanson’s experimental
`machine shop worked closely with Mader, supplying him with a variety
`of baskets and stirring devices that would easily have challenged the
`imagination of Rube Goldberg. (An example of one early prototype
`design is shown in Figure 1-3.)
`Each device had to be tested and evaluated on its ability to discrim-
`inate between subtle variations in dissolution characteristics, on the
`repeatability of results from test to test and on its adaptability as com-
`pletely as possible to existing, standard laboratory apparatus. The latter
`criterion was most difficult to assess, because it is always possible to
`design sophisticated tests and equipment that are more functional than
`existing hardware. Nevertheless,
`the cost of dissolution testing ulti-
`mately is borne by the (already) overburdened consumer, and the test
`cost—effectiveness in terms of overall health care costs must remain a
`consideration.
`Helping Mader with his project at the Drug Standards Laboratory
`was Dr. Tim Grady, who later took over as director when Mader retired.
`This pioneering dissolution research continued, and the variables af-
`fecting consistent and repeatable results were studied for more than ten
`years.
`The task had fallen to Mader to forge the first widely accepted dis-
`solution test procedure, and it was published in NF XIII in 1970. Dis-
`solution test methods were made a part of the specification for five
`drugs: indomethacin capsules, and acetohexamide, methandrostenolone,
`methylprednisolone, and sulfamethoxazole tablets.
`
`Page 6
`
`

`
`Overall Considerations
`
`9
`
`evaluation of test results. Compendial requirements for the use of the
`calibrators ensured that the alignment and geometric specifications of
`the stirring apparatus would be more rigidly followed; ultimately, even
`the wide ranges allowed with the cgalibrators cannot be met unless the
`equipment is properly aligned.
`_'_‘:'*‘J
`USP continues its research and evaluation of dissolution test technique
`and methodology to this day.
`'I‘he3'i.most recent collaborative study is
`published in the Pharmacopeial Forum (14), The USP Dissolution Cal-
`ibrator Tablet Collaborative Study—An overview of the 1996 process.
`
`Calibrators
`USP now specifies the calibration of dissolution equipment using
`disintegrating and non-disintegrating calibrator tablets (prednisone and
`salicylic acid) available through their headquarters at 12601 Twinbrook
`Parkway, Rockville, MD 20852, USA. The performance—value ranges
`for calibrating equipment are not outlined in the compendium, but they»
`appear in certificates that come with the calibrator tablets. The allowed
`ranges are wide, and it is a point of discussion for those involved in
`establishing the ranges; the debate about calibrator tablets continues.
`The PhRMA (Pharmaceutical Research and Manufacturers of America)
`Dissolution committee undertook a study to examine mechanical cali-
`brations as a possible replacement for calibrator tablets (9). USP, along
`with other experts and equipment manufacturers, continues to examine
`the use of calibrator tablets and Ways to ensure the equipment is work-
`ing properly. There will be much work in the area in the future.
`European scientists are less than enthusiastic about the use of cali-
`brators. One report points out the difficulties of non-uniforrnity and
`instability of calibrator tablets and argues that strict geometry, close
`tolerances, and a regular check of the dynamic parameters of dissolu-
`tion equipment are the only reliable means of ensuring standardized
`performance (15).
`A regular check of equipment with USP calibrators in order to de-
`termine drift or gradual failure in performance is usually a necessary 1
`routine in GMP laboratories. This includes situations where equipment
`is moved and reinstalled in another lab. Calibrator tests also carry the
`
`added benefit of “system suitability”; that is, calibrators provide a mon-
`itor of not only the experimental conditions but the analytical technique
`as well.
`
`It has been suggested that in-house calibrators should be used as
`standards and should be run simultaneously with each dissolution test
`
`Page 7
`
`itJ1
`11
`EA
`
`5 E
`
` 8
`
`HANDBOOK OF DISSOLUTION TESTING
`
`Development of Dissolution Testing Standards
`Once the dissolution test became official, information from industri-
`al, academic, and regulatory laboratories expanded the literature. A
`series of collaborative tests were conducted for various drugs and the
`relative standard deviation (coefficient of variance) in the results from
`different laboratories turned out to be wider than expected.
`Repeatability of analytical results from laboratory to laboratory and
`within narrow limits is essential where the specified dissolution rate for
`a drug is set at a minimum percent dissolved in a stated time and where
`the product in question dissolves just a few percentage points above
`that minimum. For example, prednisolone tablets have a minimum dis-
`solution rate of 80% dissolved in 30 minutes. If the manufacturer and
`
`outside laboratories or both report numbers in the 84-88% range but
`the regulatory agency reports numbers in the 77-79% range, a serious
`legal problem develops (that is, when 84-88% pass but 77-79% does
`not).
`Clearly, when two laboratories are within i 5 absolute percentage
`points but are above and below the limits, precise definitions and con-
`trol of all equipment and analytical Variables are necessary. Problems
`of this sort have actually occurred with both U.S. and Canadian regu-
`latory agencies. During the 1970s and later, a vast amount of research
`was performed to determine the effect of outside variables. Summaries
`were published and serve as a valuable reference for analysts conduct-
`ing dissolution tests (6-11).
`A significant step toward standardizing dissolution methods occurred
`with the introduction of calibrator tablets. Salicylic acid tablets dem-
`onstrated remarkably consistent and stable dissolution characteristics,
`(12), but they are non-disintegrating dosage forms. A large batch of a
`prednisone/lactose formulation was prepared to serve as a disintegrating
`counterpart to the salicylic acid tablets. Several laboratories then par-
`ticipated in a collaborative study to determine dissolution rates of both
`of these tablets in tests using identical protocols.
`The results of these studies provided a foundation to define and con-
`trol Variable inputs to the current dissolution protocol (13). A two-
`sigma range was established as the first crude criterion for calibrating
`equipment and methods. The ranges allowed may now seem inordi-
`nately wide, but they were a place to start.
`Publication of those results stimulated more extensive research into
`
`dissolution technique and instrumentation. The compendia added re-
`strictions on vibration and dissolved gas and expanded the statistical
`
`Page 7
`
`

`
`Overall Considerations
`
`11
`
`rotating basket method has enjoyed more than 30 years of extensive
`testing for numerous dosage forms. It has been modified, and the of-
`ficial method (Apparatus 1) is described in Chapter 3.
`The basket method represents ~an§13ttempt to constrain the position of
`the dosage form in order to provide the maximum probability of a
`consistent solid—liquid interface. This method has several disadvantages:
`a tendency for gummy substances‘?-to clog the basket screen, extreme
`sensitivity to dissolved gases in the’ dissolution fluid, inadequate flow
`rates when particles leave the basket and float in the medium, and
`constmction difficulties when automated methods are attempted.
`Paddle Method. Originally developed by Poole (19),
`the paddle
`method was modified through the work of scientists at US Food and
`Drug Administration (FDA)’s National Center
`for Drug Analysis
`(NCDA) in St. Louis, Missouri, USA. It consists essentially of a ro-
`tating paddle with a blade of specific dimensions conforming to the
`inside radius of a round-bottomed flask. This method (Apparatus 2)
`overcomes many of the disadvantages of the rotating basket, but it
`requires careful precision in the geometry of the paddle and flask and
`suffers from unacceptable variations in dissolution data following even
`minor changes in paddle orientation. Its convenience for automated
`systems, however, is its strong point.
`Flow-Through Methods. Proposed by many but most extensively
`studied by Langenbucher (20), flow-through methods involve constrain-
`ing the dosage form in a cell and pumping dissolution fluid through
`that cell.
`'
`
`Langenbucher studied the geometry of the cell and the variables
`introduced by variable and/or pulsating flow rates. Although the system
`has not been used extensively in the United States, its obvious advan-
`tages in maintaining sink conditions for drugs with low solubility, but
`high dosage amounts, which saturate in volumes more than 25% of
`specified media volume, suggest benefits. The method also has the add-
`ed advantage of inherent adaptability to automated sampling tech-
`niques, The flow-through method is now official in the United States
`(Apparatus 4) and the European Pharmacopeia, and may be considered
`for drugs that impose saturation problems with the basket or paddle
`methods.
`
`Reciprocating Cylinder Apparatus. The reciprocating cylinder is now
`listed in USP as Apparatus 3 for extended—release dosage forms. This is
`a unique system that “floats” deaggregated particles in a reciprocating
`stream. It has advantages of lower volume of solvent; ease of pH change;
`reduced dwell time to avoid degradation; ease of automated sampling;
`
`Page 8 A
`
`.«as-,_.1m...,...«z.-:.......
`
`
`
`
`
`10
`
`HANDBOOK OF DISSOLUTION TESTING
`
`(16). Such a method provides the maximum assurance against inad-
`vertent equipment malfunction, but practical difficulties arise because
`USP specifies increments of six tests and most commercial dissolution
`equipment provides only six vessels. Manufacturers using this tech-
`nique have run stations of 12 vessels, placing their in-house calibrator
`in at least two. Dissolution test stations with seven or eight stirrers have
`become commercially available.
`
`Methods of Dissolution Testing
`
`Many schemes have been reported for the determination of disso-
`lution rate. A detailed review of these is inappropriate for this publi-
`cation. In sum, though, all methods involve providing a renewable sol-
`id—liquid interface between the dosage form and the dissolution fluid,
`ensuring that this can be defined and controlled and is thus repeatable-
`which is more easily said than done! The controlled flow of fluid over
`a solid introduces requirements of maintaining the surface of the solid
`in a position exposed to a non-accelerating flow——quite a requirement
`for a tablet that moves about because of the similarity of its density to
`that of the liquid or for a tablet that disintegrates into various buoyant
`particles that float in the stream.
`Most of the variations in dissolution methods have been devised to
`bring such variables under control. Unfortunately, some of the methods
`are applicable only to unique dosage forms and become unsatisfactory
`with others. The only methods that will be discussed in detail in this
`manual are those that are already established in the compendia or are
`likely to receive serious consideration as official methods in the next
`few years. Some excellent methods must be omitted, but that is no
`reflection on their probable value or unique characteristics. For various
`reasons, it is unlikely they will be seriously considered as official meth-
`ods in the near future. The spin—filter method of Shah and co—workers
`(17), for example, solves a number of the problems of dissolution test-
`ing, largely by avoiding the clogging of the filters used in automated
`sampling. It seems unlikely, however, that their spin filter method will
`ever be universally accepted in the compendia because of cost, com-
`plexity, and the unrealistic manufacturing tolerances required to avoid
`vibration and other disturbances; considerations that are added to ob-
`
`vious problems of maintenance and operation.
`The methods to be discussed are briefly described below.
`Basket Method. Originally proposed by Pernarowski (18) and mod-
`ified to become the first official method adopted in USP in 1970, the
`
`Page 8
`
`

`
`Overall Considerations
`
`13 -
`
`- Reciprocating. A sophisticated development from the old disinte-
`gration test, reciprocating agitation moves the dosage form up and
`down in a succession of media. These apparatus are illustrated in Fig-
`ures 3-4 and 4-6. This is used as
`drive in USP Apparatus 3 and 7.
`- Fluid Movement. Known as a_,’flow-through system, this agitation
`holds the dosage form relatively stationary while a controlled stream
`of media is pumped past it. A schematic is illustrated in Figure 3-5.
`This is used as the drive in USP Apparatus 4.
`- Percutaneous Absorption. This agitation is used to study the re-
`lease or transfer of a dosage form from or through membranes. Typical
`applications are topical and transdermal formulations. This apparatus is
`illustrated in Figures 4-7 and 4-84.
`
`Defining an Apparatus
`
`The apparatus used in a specific dissolution test or method includes
`an agitation method as described above, and details of dosage form
`mounting within the system. Other parameters of the system are also
`defined,
`including temperature, flow rates, dissolution medium, and
`constraints on environmental input variables, such as external vibration.
`In this handbook, each apparatus is described in as much detail as
`current compendia have established. Solid dosage form methods are
`described in Chapter 3 and special dosage forms in Chapter 4.
`
`Use of Dissolution Test Data
`Dissolution testing is a useful method for: ensuring the bioequiva—
`lence of different batches of solid dosage forms when a rank—ord_er
`correlation between dissolution characteristics and bioavailability has
`been established (this is most likely in situations where dissolution is
`rate limiting in the dissolution/absorption system for the drug-—see
`Chapter 2 for further discussion); monitoring formulation and manu-
`facturing processes by quality control personnel; and establishing the
`intrinsic dissolution rate (see Chapter 2), which is somewhat useful in
`screening compounds being considered for new drug applications
`(NDA’s).
`Finally and most basically, the FDA requires dissolution tests in reg-
`ulatory filings for all oral dosage forms and many other dosage forms
`as well. This is true of the regulatory agencies in most other industrial
`countries. It is likely that dissolution testing will increase and will be-
`come more complicated and more global in the future.
`
`Page 9
`
`
`
`12
`
`HANDBOOK OF DISSOLUTION TESTING
`
`correlation with the database from the now-discarded rotating bottle ap-
`paratus; and impressive correlations with bioavailability data (21).
`Transdermal Dissolution Testing. Apparatus for this testing is dis-
`cussed in Chapter 4. Several methods are now in the USP for these
`dosage forms, and the vertical diffusion cell is under current consid-
`eration for semisolid formulations and topicals. Chapter 4 also discusses
`the testing of other novel and special dosage forms.
`
`Classification of Dissolution Techniques
`
`At present, USP accepts the following methods of dissolution testing:
`(See Chapters 3 and 4 for a detailed discussion on each apparatus.)
`
`USP Apparatus Number
`
`Suitable for
`
`Agitation Method
`
`1 (Basket)
`
`2 (Paddle)
`
`Solids, Beads
`
`Rotating Stirrer
`
`Solids, Suspensions
`
`Rotating Stirrer
`
`3 (Reciprocating Cylinder)
`
`Solids, Beads
`
`Reciprocation
`
`4 (Flow-Through Cell)
`
`§,:1viV‘if;r]:°fn‘i:;1antS
`
`Fluid Movement
`
`5 (Paddle Over Disk)
`
`Transdermal Patches
`
`Rffiatiflg stirrer
`
`6 (Cylinder)
`
`Transdermal Patches
`
`Rotating; Stirfef
`
`7 (Reciprocating Holder)
`
`§$indSSdenfla1Pat°heS’
`
`Reciprocation
`
`To these may be added the following noncompendial method:
`
`Percutaneous Absorption
`(Diffusion Cell)
`
`Topicals, Transdermals,
`Implants
`
`Fluid Movement
`
`Agitation Methods
`Each dissolution technique offers a unique means to provide a con-
`trolled flow of solvent over the surface of the solute, i.e. the solid-
`liquid interface. Four agitation methods can be used:
`- Rotating Stirrer. This is based on the original dissolution test drive.
`It rotates the dosage form or stirs the medium in which the dosage form
`is held. Many variations are commercially available as illustrated in
`Figures 3-2, 3-3, 4-4 and 4-5. This is used as the drive in current USP
`Apparatus 1, 2, 5 and 6.
`
`Page 9
`
`

`
`14
`
`HANDBOOK OF DISSOLUTION TESTING
`
`Role of USP in Dissolution Testing
`
`° General Chapters:
`<701> Disintegration
`<711> Dissolution
`
`<724> Drug Release
`<1087> Intrinsic Dissolution
`<1088> In Vitro and In Vivo Evaluation of Dosage Forms
`
`<1090> In Vivo Bioequivalence Guidances
`<1092> Dissolution Procedure: Development & Validation (pro-
`posed)
`<1225> Validation of Compendial Methods
`- Official Monographs for Drug Dosage Forms
`- USP Calibrator Tablets and Reference Standards
`- Co-sponsors Workshops with American Association of Pharmaceu-
`tical Scientists (AAPS) and FDA
`0 Training Courses, Annual Meetings, Open Committee Meetings and
`Forums
`
`Role of FDA in Dissolution Testing
`
`- Performs Off Shelf Testing
`- Performs Validation of NDA Methods
`
`0 Performs Inspections
`- Orders Recalls
`
`° Issues Product Approvals
`- Enforces USP
`- Publishes Guidances
`
`- Co-sponsors Workshops with AAPS, FIP and USP
`- Forms Task Forces
`- Researches and Evaluates New Methods and Technology
`
`Overall Considerations
`
`15
`
`References
`
`(1) “USP Dissolution General Chapter <711> and USP Drug Re-
`lease General Chapter <724>i-’V, 2004, United States Pharmaco-
`peia and National Formulary, Ignited States Pharmacopeial Con-
`venfion,Inc, 27miEdfljon,23Q3-2312.
`
`(2) “2.9.3. Dissolution Test for Solid Dosage Forms”, 2002, Euro-
`pean Pharmacopoeia, European Directorate for the Quality of
`Medicines of the Council of Europe, 4* Edition, Supplement 4.4,
`3247-3250.
`
`(3) Hoag, SW, Ramachandruni, H and Shangraw, RF, “Failure of Pre-
`scription Prenatal Vitamin Products to Meet USP Standards for
`Folic Acid Dissolution”, 1997, Journal of the American Phar-
`maceutical Association, 37(4), 397-400.
`
`(4) “USP Policy on Dissolution Standards”, 1981, Pharmacopeial
`Forum, 7, 1225-1226.
`
`(5) Wagner, J and Pernarowski, M, “Biopharmaceutics and Relevant
`Pharmacokinetics”, 1971, Drug Intelligence Publications.
`
`(6) Cox, D and Wells, C, “In Vitro Dissolution of Digitoxin Tablets”,
`1978, Internal Document, National Center of Drug Analysis.
`
`(7) Hanson, W, “Solving the Puzzle of Random Variables in Disso-
`lution Testing”, 1977, Pharmaceutical Technology, 1(5), 30-41.
`
`(8) Thakker, D, Naik, N, Gray, V and Sun S, “Fine Tuning of Dis-
`solution Apparatus”, 1980, Pharmacopeial Forum, 6(2), 177-
`185.
`
`(9) PhRMA Subcommittee on Dissolution Calibration: Brune, S,
`Bucko, J, Emr, S, Gray, V, Hippeli, K, Kentrup, A, Whiteman, D,
`Loranger, M and Oates, M, “Dissolution Calibrator: Recommen-
`dations for Reduced Chemical Testing and Enhanced Mechanical
`Calibration”, 2000, Pharmacopeial Forum, 26(4), 1149-1166.
`
`(10) Cox D and Furman, W, “Systematic Error Associated with Ap-
`paratus 2 of the USP Dissolution Test: Effects of Physical Align-
`ment of the Dissolution Apparatus”, 1982, Journal of Pharma-
`ceutical Sciences, 71(4), 451-452.
`
`
`
`
`
`Hamilton, JF, Moore, TW and Kerner, CM, “Reproducibility of
`Dissolution Test Results”, 1995, Pharm

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