throbber
Pharmaceutical
`Dissolution Testing
`
`
`
`© 2005 by Taylor & Francis Group, LLC© 2005 by Taylor & Francis Group, LLC
`
`MYLAN EXHIBIT 1025
`
`

`

`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
`
`

`

`12
`
`Dissolution Method Development:
`An Industry Perspective
`
`CYNTHIA K. BROWN
`
`Eli Lilly and Company, Indianapolis,
`Indiana, U.S.A.
`
`INTRODUCTION
`
`In today’s pharmaceutical industry, dissolution testing is a
`valuable qualitative tool that provides key information about
`the biological availability and/or equivalency as well as the
`batch-to-batch consistency of a drug. Therefore, a properly
`designed dissolution test is essential for the biopharmaceutical
`characterization and batch-to-batch control of the drug pro-
`duct. During drug development, dissolution testing is used
`to select appropriate formulations for in vivo testing, guide
`formulation development activities, and assess stability of
`the drug product under various packaging and storage
`requirements. For the dissolution test to be a useful drug
`351
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`352
`
`Brown
`
`characterization tool, the methodology needs to be able to
`discriminate between different degrees of product perfor-
`mance and thus, the collection of a multi-time point dissolu-
`tion profile is useful. At present, almost all solid oral dosage
`forms require dissolution testing as a quality control check
`before a product is introduced into the market place. For
`the dissolution test to be a useful quality control tool, the
`methodology should be simple, reliable and reproducible,
`and ideally be able to discriminate between different degrees
`of product performance (1).
`Dissolution testing is also used to identify bioavailability
`(BA) problems and to assess the need for further bioequiva-
`lence (BE) studies relative to scale-up and post-approval
`changes (SUPAC), where it can function as a signal of bioine-
`quivalence (2,3). The issuance of the Food and Drug Adminis-
`tration (FDA) guidance document, Waiver of
`In Vivo
`Bioavailability and Bioequivalence Studies for Immediate-
`Release Solid Oral Dosage Forms Based on a Biopharmaceu-
`tics Classification System, allows dissolution testing to be
`used as a surrogate for in vivo BE testing under certain
`circumstances
`(4). The Biopharmaceutics Classification
`System (BCS) is a scientific framework for classifying drug
`substances based on their aqueous solubility and intestinal
`permeability. When combined with the dissolution of the drug
`product, the BCS takes into account three major factors that
`influence the rate and extent of drug absorption from immedi-
`ate-release solid oral dosage forms: dissolution, solubility, and
`intestinal permeability (5). Based on the BCS framework,
`drug manufacturers may request waivers from additional in
`vivo studies (biowaivers) if their drug product meets certain
`criteria. In addition, the FDA’s guidance on BA and BE (6)
`allows biowaivers for additional strength(s) of immediate-
`release as well as modified-release drug products based
`on formulation proportionality and dissolution profile
`comparison.
`These changes in BE requirements that move away from
`the in vivo study requirement in certain cases and rely more
`on dissolution test results, emphasize the significance of
`dissolution test applications. In all cases where the dissolution
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`353
`
`test is used as a BE test, a link with a bioavailable product is
`established. With the advances in dissolution testing and the
`increased understanding of the scientific principles and
`mechanisms of dissolution testing, a clear trend has appeared
`where the dissolution test is not solely a traditional quality
`control test but may also be used as a surrogate to the in vivo
`BE test (7).
`For the dissolution test to be used as an effective drug
`product
`characterization and quality control
`tool,
`the
`method must be developed with the various end uses in
`mind. In some cases, the method used in the early phase
`of product and formulation development could be different
`from the final test procedure utilized for control of the
`product quality. Methods used for formulation screening or
`BA and/or bioequivalency evaluations may simply be
`impractical for a quality control environment. It is essential
`that with the accumulation of experience, the early method
`be critically re-evaluated and potentially simplified, giving
`preference to compendial apparatus and media. Hence, the
`final dissolution method submitted for product registration
`may not necessarily closely imitate the in vivo environment
`but should still test the key performance indicators of the
`formulation.
`To facilitate the development of appropriate dissolution
`tests several regulatory, pharmacopeial, and industrial orga-
`nizations have issued dissolution-related guidelines that
`provide information and recommendations on the develop-
`ment and validation of dissolution test methodology, the
`establishment of dissolution specifications, and the regulatory
`applications of dissolution testing (8–16). This chapter
`describes a systematic approach for the development of a dis-
`solution method. The information is organized and presented
`in sections that follow the chronological sequence of the
`method development process. These include the assessment
`of relevant physical and chemical properties of the drug,
`determination of the appropriate dissolution apparatus, selec-
`tion of the dissolution medium, determination key operating
`parameters, method optimization, and validation of the
`methodology.
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`354
`
`Brown
`
`PHYSICAL AND CHEMICAL PROPERTIES
`
`The first step in the development of a new dissolution test is
`to evaluate the relevant physical and chemical data for the
`drug substance. Knowledge of the drug compound’s physi-
`cal–chemical properties will facilitate the selection of dissolu-
`tion medium and determination of medium volume.
`Some of the physicochemical properties of the active
`pharmaceutical ingredient (API) that influence the dissolu-
`tion characteristics are:
`
`Ionization constants (pKa),
`Solubility as a function of pH,
`Solution stability as a function of pH,
`Particle size,
`Crystal form, and
`Common ion, ionic strength, and buffer effects.
`
`Two key physicochemical API properties to evaluate are
`the solubility and solution-state stability of the drug sub-
`stance as a function of pH. Knowledge of the pKa (or pKa’s)
`is useful because it defines the charge of the molecule in solu-
`tion at any given pH. Ideally, the drug substance’s solubility
`in the dissolution medium should not be the rate-limiting
`factor for the drug substance’s dissolution from the drug
`product. Hence, the dissolution rate should be characteristic
`of the release of the active ingredient from the dosage form
`rather than the drug substance’s solubility in the dissolution
`medium. When adjusting the composition of the medium to
`insure adequate solubility for the drug substance,
`the
`influence of surfactants, pH, and buffers on the solubility
`and stability of the drug substance need to be evaluated.
`The solution-state stability of the API must also be consid-
`ered in the design of a dissolution test because the molecule’s
`stability in various dissolution media may limit the pH range
`over which the drug product’s dissolution can be evaluated.
`Typically, the drug’s solution stability should be determined
`at 37C for 2 hr for immediate-release formulations and twice
`the designated testing time for sustained-release formula-
`tions (17).
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`355
`
`During the initial stages of a drug product’s develop-
`ment, a dissolution test should facilitate the formulation
`development and selection. During this phase of the drug
`development process bioavailability data is usually not avail-
`able. In the absence of BA, the dissolution medium selection
`should be based on the physicochemical properties, the formu-
`lation design, and the intended dose. The BCS provides a good
`framework for determining if the dissolution of the drug will
`be the rate-limiting factor in the in vivo absorption process.
`Hence, the pH solubility of the drug and the intended dose
`are essential parameters to consider early in the dissolution
`method development process.
`Once you have a good understanding of the physical–
`chemical properties of the drug substance, the key properties
`of the dosage form, i.e., type, label claim, and release mechan-
`ism, need to be considered. The most appropriate dissolution
`testing apparatus and dissolution medium can be selected
`based on the physical–chemical properties of the drug sub-
`stance and the key properties of the dosage form. Dosage forms
`can be designed to provide immediate release, delayed release,
`or extended (controlled) release. Determining the type of
`release and anticipated site of in vivo absorption will facilitate
`the selection of dissolution media, testing apparatus, and test
`duration.
`
`DISSOLUTION APPARATUS SELECTION
`
`The choice of apparatus is based on knowledge of the formula-
`tion design and practical aspects of dosage form performance in
`the in vitro test system. Dissolution testing is conducted on
`equipment that has demonstrated suitability, such as described
`in the 2003 United States Pharmacopeia (USP) under the
`general chapters of Dissolution and Drug Release (10,11). The
`basket method (USP Apparatus 1) is routinely used for solid
`oral dosage forms such as capsule or tablet formulations at
`an agitation speed of 50–100 rpm, although speeds of up to
`150 rpm have been used. The paddle method (USP Apparatus
`2) is frequently used for solid oral dosage forms such as tablet
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`356
`
`Brown
`
`and capsule formulations at 50 or 75 rpm. The paddle method
`is also useful for the testing of oral suspensions at the recom-
`mended paddle speed of 25–50 rpm. The reciprocating cylinder
`(USP Apparatus 3) has been found to be especially useful for
`bead-type modified-release dosage forms. The flow-through cell
`(USP Apparatus 4) may offer advantages for some modified-
`release dosage forms, especially those that contain active ingre-
`dients with limited solubility. Additionally, the reciprocating
`cylinder or the flow-through cell may be useful for soft gelatin
`capsules, bead products, suppositories, or poorly soluble drugs.
`By design, both the reciprocating cylinder and the flow-through
`cell allow for a controlled pH change of the dissolution medium
`throughout the test, which allows the apparatus to be easily
`utilized for physiological evaluations of the dosage form during
`development. The paddle over disk (USP Apparatus 5) and the
`cylinder (USP Apparatus 6) have been shown to be useful for
`evaluating and testing transdermal dosage forms. The recipro-
`cating holder (USP Apparatus 7) has been shown to have appli-
`cation to non-disintegrating oral modified-release dosage forms,
`as well as to transdermal dosage forms.
`In general, compendial apparatus and methods should be
`used as a first approach in drug development. To avoid unne-
`cessary proliferation of equipment and method design,
`modifications of compendial equipment or development and
`use of alternative equipment should be considered only when
`it has been proven that compendial set up does not provide
`meaningful data for a given dosage form. In these instances,
`superiority of the new or modified design has to be proven
`in comparison to the compendial design.
`Table 1 outlines the current status of scientific develop-
`ment for the dissolution or release testing from various
`dosage forms and recommends, where possible, the dissolu-
`tion apparatus of ‘‘first choice’’ (13). Refer also to Chapter 2
`for further description of the USP apparatus.
`
`DISSOLUTION MEDIUM SELECTION
`
`For batch-to-batch quality testing, selection of the dissolution
`medium is based, in part, on the solubility data and the dose
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`357
`
`Table 1 Apparatus Recommended Based on Dosage Form Type
`
`Type of dosage form
`
`Release method
`
`Solid oral dosage forms
`(conventional)
`Oral suspensions
`Oral disintegrating tablets
`Chewable tablets
`
`Transdermals—patches
`Topicals—semisolids
`Suppositories
`
`Chewing gum
`
`Powders and granules
`
`Microparticulate formulations
`Implants
`
`Basket, paddle, reciprocating
`cylinder, or flow-through cell
`Paddle
`Paddle
`Basket, paddle, or reciprocating
`cylinder with glass beads
`Paddle over disk
`Franz cell diffusion system
`Paddle, modified basket, or dual
`chamber flow-through cell
`Special apparatus [European
`Pharmacopoeia (PhEur)]
`Flow-through cell (powder/granule
`sample cell)
`Modified flow-through cell
`Modified flow-through cell
`
`range of the drug product in order to ensure that sink condi-
`tions are met. The term sink conditions is defined as the
`volume of medium at least greater than three times that
`required to form a saturated solution of a drug substance. A
`medium that fails to provide sink conditions may be justifi-
`able if it is shown to be more discriminating or if it provides
`reliable data which otherwise can only be obtained with the
`addition of surfactants. When the dissolution test is to indi-
`cate the biopharmaceutical properties of the dosage form, it
`is more important that the test closely simulate the environ-
`ment in the GI tract than necessarily produce sink conditions
`for release. Therefore, it is not always possible to develop one
`dissolution test or select one dissolution medium that ensures
`batch-to-batch control as well as monitoring the biopharma-
`ceutical aspects of the drug product.
`formulations
`The dissolution characteristics of oral
`should be evaluated over the physiologic pH range of 1.2–
`6.8 [1.2–7.5 for modified release (MR) formulations]. During
`method development, it may be useful to measure the pH
`before and after a run to see if the pH changes during the test,
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`358
`
`Brown
`
`especially if the buffer capacity of the chosen medium is low.
`Selection of the most appropriate medium for routine testing
`is then based on discriminatory capability, ruggedness, stabi-
`lity of the analyte in the test medium, and relevance to in vivo
`performance where possible.
`For very poorly soluble compounds, aqueous solutions
`may contain a percentage of a surfactant (e.g., sodium lauryl
`sulfate, Tween 80 or CTAB) that is used to enhance drug
`solubility. The need for surfactants and the concentrations
`used should be justified. Surfactants can be used as either a
`wetting agent or, when the critical micelle concentration
`(CMC) is reached, to solubilize the drug substance. The sur-
`factant’s CMC depends upon the surfactant itself and the
`ionic strength of the base medium. The amount of surfactant
`needed for adequate drug solubility depends on the surfactant
`CMC and the degree to which the compound partitions into
`the surfactant micelles. Because of
`the nature of
`the
`compound and micelle interaction, there is typically a linear
`dependence between solubility and surfactant concentration
`above the CMC. If a compound is ionizable, surfactant concen-
`tration and pH may be varied simultaneously, and the
`combined effect can substantially change the solubility char-
`acteristics of the dissolution medium. Table 2 lists dissolution
`medium selection criteria as defined in regulatory, industry,
`and compendial guidances.
`The BCS describes the classification of compounds
`according to solubility and permeability (6). Biorelevant med-
`ium is a term used to describe a medium that has some rele-
`vance to the in vivo dissolution conditions for the compound.
`Choice of a biorelevant medium is based on a mechanistic
`approach that considers the absorption site, if known, and
`whether the rate-limiting step to absorption is the dissolution
`or permeability of the compound. In some cases, the biorele-
`vant medium will be different from the test conditions chosen
`for the regulatory test and the time points are also likely to be
`different. If the compound dissolves quickly in the stomach
`and is highly permeable, gastric emptying time may be the
`rate-limiting step to absorption. In this case, the dissolution
`test is to demonstrate that the drug is released quickly under
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`359
`
`Table 2 Recommended Dissolution Medium Composition and
`Volume for Rotating Basket or Rotating Paddle Apparatus
`
`Volume
`
`pH
`
`Additives
`
`pH 1–6.8; above pH
`6.8 with
`justification—not
`to exceed pH 8
`
`Enzymes, salts,
`surfactants with
`justification
`
`500–1,000 mL;
`900 mL
`historical;
`1,000 mL
`recommended
`for future
`development
`500–1,000 mL; up
`to 2,000 mL for
`drug with
`limited
`solubility
`
`Buffered aqueous
`solution pH 4–8 or
`dilute acid
`solutions (0.001 N
`HCl to 0.1 N HCl)
`
`Enzymes, salts,
`surfactants
`balanced against
`loss of discrim-
`inatory power;
`enzymes can be
`used for cross-
`linking of gelatin
`capsules or
`gelatin-coated
`tablets
`Determined per
`product
`
`Surfactants
`recommended for
`water poorly
`soluble drug
`products—need
`and amount
`should be
`justified; enzymes
`use need case-by-
`case justification;
`utilized for the
`cross-linking of
`gelatin capsules
`or gelatin-coated
`tablets
`
`Guidance or
`compendial
`reference
`
`Federation
`International
`Pharmaceutique
`(FIP) (23)
`
`United States
`Pharmacopeia
`(USP) (10–12)
`
`World Health
`Organization
`(WHO) (16),
`European
`Pharmacopoeia
`(PhEur) (14),
`Japanese
`Pharmacopoeia
`(JP) (15)
`FDA (8,9)
`
`Determined per
`product
`
`Adjust pH to within
`0.05 units of the
`prescribed valued
`
`500, 900, or
`1,000 mL
`
`pH 1.2–6.8; higher
`pH justified case-
`by-case—in
`general not to
`exceed pH 8
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`360
`
`Brown
`
`typical gastric (acidic) conditions. On the other hand, if disso-
`lution occurs primarily in the intestinal tract (e.g., a poorly
`soluble, weak acid), a higher pH range (e.g., simulated intest-
`inal fluid with a pH of 6.8) will be more appropriate (18).
`The fed and fasted state may also have significant effects
`on the absorption or solubility of a compound. Compositions of
`media that simulate the fed and fasted states can be found in
`the literature (19) (see also Chapter 5). These media reflect
`changes in the pH, bile concentrations, and osmolarity after
`meal intake and therefore have a different composition than
`that of typical compendial media. They are primarily used
`to establish in vitro–in vivo correlations during formulation
`development and to assess potential food effects and are not
`intended for quality control purposes. For quality control
`purposes, the substitution of natural surfactants (bile compo-
`nents) with appropriate synthetic surfactants is permitted
`and encouraged because of the expense of the natural
`substances and the labor-intensive preparation of
`the
`biorelevant media.
`
`KEY OPERATING PARAMETERS
`
`Media: Volume, Temperature, Deaeration
`
`As shown in Table 2, the recommended volume of dissolution
`medium is 500–1000 mL, with 900 mL as the most common
`volume when using the basket or paddle apparatus. The
`volume can be raised to between 2 and 4 L, depending on
`the concentration and sink conditions of the drug, but proper
`justification is expected.
`The standard temperature for the dissolution medium is
`37  0.5C for oral dosage forms. Slightly increased tempera-
`tures such as 38  0.5C have been recommended for dosages
`forms such as suppositories. Lower temperatures such as
`32  0.5C are utilized for topical dosage forms such as trans-
`dermal patches and topical ointments.
`The significance of deaeration of the medium should be
`determined on a case-by-case basis, as air bubbles can inter-
`fere with the test results and act as a barrier to dissolution
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`361
`
`if present on the dosage unit or basket mesh. Additionally, air
`bubbles can cause particles to cling to the apparatus and
`vessel walls. On the other hand, bubbles on the dosage unit
`may increase the buoyancy and lead to an increase in the dis-
`solution rate, or decrease the dissolution rate by decreasing
`the available surface area. Consequently, the impact of med-
`ium deaeration may be formulation dependent, such that
`some formulations will be sensitive to the presence of dis-
`solved air in the dissolution while other formulations will be
`robust. To determine if deaeration of the medium is neces-
`sary, a comparison between dissolution data generated with
`non-deaerated medium vs. dissolution data generated with
`deaerated medium should be performed.
`The following deaeration method is described as a foot-
`note in the 2003 United States Pharmacopeia (USP) under
`the general chapter Dissolution (10). The USP deaeration
`method requires heating of the medium, followed by filtration,
`and drawing of a vacuum for a short period of time. Other
`deaeration methods such as room temperature filtration, soni-
`cation, and helium sparging are described in literature (20,21)
`and are routinely used throughout the industry. The deaera-
`tion method needs to be clearly characterized, since the
`method chosen might impact the dissolution release rate
`(13). It should be noted that dissolution tests using the flow-
`through cell method could be particularly sensitive to the
`deaeration of the medium. Media containing surfactants are
`not usually deaerated after the surfactant has been added
`to the medium because of excessive foaming. In some labora-
`tories, the base medium is deaerated prior to the addition of
`the surfactant.
`
`Sinker Evaluation
`
`Currently, the Japanese Pharmacopoeia (JP) is the only phar-
`macopeia that requires a specific sinker device for all capsule
`formulations. The USP recommends a few turns of a nonreac-
`tive material wire when the dosage form tends to float (12) (see
`Chapter 2 for illustrations of the Japanese and USP sinkers).
`Because sinkers can significantly influence the dissolution
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`362
`
`Brown
`
`profile of a drug product, detailed sinker descriptions and the
`rationale for why a sinker is used should be stated in the writ-
`ten procedure. When comparing different sinkers (or sinkers
`versus no sinkers), a test should be run concurrently with
`each sinker. Each sinker type should be evaluated based on
`its ability to maintain the dosage at the bottom of the vessel
`without inhibiting drug release.
`Sinkers can significantly influence the dissolution profile
`of a drug. Therefore, the use of sinkers should be part of the
`dissolution method validation. If equivalent sinkers are iden-
`tified during the sinker evaluation and validation, the equiva-
`lent sinkers should be listed in the written dissolution test
`procedure. When a dissolution method utilizes a dissolution
`sinker and is transferred to another laboratory, the receiving
`laboratory should duplicate the validated sinker design(s) as
`closely as possible.
`
`Analytical Detection
`
`For determination of the quantitative step in the dissolution
`method,
`information regarding the spectral,
`chromato-
`graphic, electrochemical, and/or chemical characteristics of
`the drug substance should be considered. The quantitative
`method needs to provide adequate sensitivity for the accurate
`determination of the analyte in the dissolution medium. Since
`formulations are likely to change during product develop-
`ment, it is usually advantageous to use high-performance
`liquid chromatography (HPLC) detection procedures. How-
`ever, because of the ease of automation and faster analysis
`time, UV detection methods are more desirable for the routine
`quality control testing of products.
`Filtration of the dissolution sample aliquot is usually
`needed prior to quantitation. Filtration of the dissolution
`samples is usually necessary to prevent undissolved drug
`particles from entering the analytical sample and dissolving
`further. Also, filtration removes insoluble excipients that
`may otherwise cause a high background or turbidity. Prewet-
`ting of the filter with the medium is usually necessary. Filters
`can be in-line, at the end of the sampling probe, or both. The
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`363
`
`pore size can range from 0.45 to 70 mm. The usual types are
`depth, disk, or flow-through filters. However, if the excipient
`interference is high, or the filtrate has a cloudy appearance,
`or the filter becomes clogged, an alternative type of filter or
`pore size may need to be evaluated.
`Adsorption of the drug(s) to the filter needs to be evalu-
`ated. If drug adsorption occurs, the amount of initial filtrate
`discarded may need to be increased. If results are still unsui-
`table, an alternative filter material should be sought. Centri-
`fugation of samples is generally not recommended, as
`dissolution can continue to occur during centrifugation and
`there may be a concentration gradient in the supernatant.
`A possible exception might be compounds that adsorb to all
`common filters.
`
`Sampling Time Points and Specifications
`
`Key operating parameters that may change (or be optimized)
`throughout a product’s development and approval cycle are
`dissolution sampling time points and dissolution limits or spe-
`cifications by which the dissolution results should be evalu-
`ated. The results generated from the dissolution test need to
`be evaluated and interpreted based on the intended purpose
`of the test. If the test is used for batch-to-batch control, the
`results should be evaluated in regard to the established limits
`or specification value. If the test is being utilized as a charac-
`terization test (i.e., biopharmaceutical evaluations, formula-
`tion development studies, etc.)
`the results are usually
`evaluated by profile comparisons.
`For immediate-release dosage forms, the dissolution test
`duration is typically 30–60 min, with a single time point
`specification being adequate in most cases for routine batch-
`to-batch quality control for approved products. Typical speci-
`fications for the amount of active ingredient dissolved,
`expressed as a percentage of the labeled content (Q), are in
`the range of 75–80% dissolved. A Q value in excess of 80%
`is not generally used, as allowances need to be made for assay
`and content uniformity ranges. Since the purpose of specify-
`ing dissolution limits is to ensure batch-to-batch consistency
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`364
`
`Brown
`
`within a range that guarantees comparable biopharmaceuti-
`cal performance in vivo, specifications including test times
`are usually established based on an evaluation of dissolution
`profile data from pivotal clinical batches and confirmatory BA
`batches (8).
`When the test is utilized as a characterization tool (i.e.,
`biopharmaceutical evaluations, formulation development stu-
`dies, etc.) the results are usually evaluated by profile compar-
`isons.
`In this
`case,
`the product’s
`comparability and
`performance are evaluated by collecting additional sampling
`time points. For registration purposes, a plot of the percen-
`tage of the drug dissolved vs. time should be determined.
`Enough time points are to be selected to adequately charac-
`terize the ascending and plateau phases of the dissolution
`curve. According to the BCS referred to in several FDA
`guidance documents, highly soluble and highly permeable
`drugs formulated with rapidly dissolving products need not
`be subjected to a profile comparison if they can be shown to
`release 85% or more of the active ingredient within 15 min.
`For these types of products, a one-point test will suffice. When
`an immediate-release drug product does not meet the rapidly
`dissolving criteria, dissolution data from multiple sampling
`time points ranging from 10 to 60 min or longer are usually
`collected.
`So-called infinity points can be useful during develop-
`ment studies. To obtain an infinity point, the paddle or basket
`speed is increased significantly (e.g., 150 rpm) at the end of
`the run and the test is allowed to run for an extended period
`of time (e.g., 60 min), and then an additional sample is taken.
`Although there is no requirement for 100% dissolution in the
`profile, the infinity point can provide data that may provide
`useful
`information about the formulation characteristics
`during the initial development.
`For an extended-release dosage form, at least three test
`time points are chosen to characterize the in vitro drug-
`release profile for the routine batch-to-batch quality control
`for approved products. Additional sampling times may be
`required for formulation development studies, biopharmaceu-
`tical evaluations, and drug approval purposes. An early time
`
`© 2005 by Taylor & Francis Group, LLC
`
`

`

`Dissolution Method Development: An Industry Perspective
`
`365
`
`point, usually 1–2 hr, is chosen to show that there is little
`probability of dose dumping. Release at this time-point should
`not exceed values expected according to the mechanism of
`release and the intended overall-release profile. An inter-
`mediate time point is chosen to define the in vitro-release pro-
`file of the dosage form, and a final time point is chosen to show
`essentially complete release of the drug. Test times and speci-
`fications are usually established on the basis of an evaluation
`of drug-release profile data. For products containing more
`than a single active ingredient, drug release is to be deter-
`mined for each active ingredient. Extended-release specifica-
`tions are addressed in the USP under the general chapter
`In Vitro and In Vivo Evaluation of Dosage Forms (12) and
`the FDA’s guidance document Extended Release Oral Dosage
`Forms: Development, Evaluation, and Application of In
`Vitro/In Vivo Correlations (9).
`
`METHOD OPTIMIZATION
`
`When human BA data are available from several formulations,
`the dissolution test should be re-evaluated and optimized (if
`needed). The goal of dissolution method optimization is to iden-
`tify in vitro test conditions that adequately discriminate critical
`formulation differences or critical manufacturing variables.
`During the method optimization process, the biostudy formula-
`tions are tested using various medium compositions (e.g., pH,
`ionic strength, surfactant composition). The effect of hydrody-
`namics on the formulations should also be evaluated by varying
`the apparatus agitation speed. If a non-bioequivalent batch is
`discovered during a bioequivalency study and the in vivo
`absorption is dissolution rate limited (BCS Class 2), the dissolu-
`tion methodology should be optimized to differentiate the

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