throbber
PHARMACEUTICAL
`PREFORMUlATION
`AND
`FORMUlATION
`
`A Practical Guide from
`Candidate Drug Selection to
`Commercial Dosage Form
`
`Mark Gibson
`Editor
`
`lnterpharm Press
`
`Denver, Colorado
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`- -
`
`Invitation to Authors
`
`( ~ Interpharm Press publishes books focused upon applied technology and regulatory affairs
`e~~ impacting healthcare manufacturers worldwide. If you are considering writing or con-
`l ~ tributing to a book applicable to the pharmaceutical, biotechnology, medical device,
`diagnostic, cosmetic, or veterinary medicine manufacturing industries, please contact our acquisi-
`-
`tions editor.
`
`Library of Congress Cataloging-in-Publication Data
`
`Pharmaceutical preformulation and formulation : a practical guide from candidate drug
`selection to commercial dosage form I Mark Gibson, editor.
`p.; em.
`Includes bibliographic references and index.
`ISBN 1-57491-120-1 (hard: alk. paper)
`1. Drugs-Dosage forms. I. Gibson, Mark, 1957-
`[DNLM: 1. Drug Compounding. 2. Biopharmaceutics-methods. 3. Chemistry,
`Pharmaceutical-methods. 4. Dosage Forms. 5. Drug Evaluation. QV 778 P53535 2001]
`RS200 .P425 2001
`615' .14-dc21
`
`2001016816
`
`Commissioned in Europe by Sue Horwood of Medi-Tech. Publications, Stonington, England, on
`behalf of IHS® Health Group, Denver, Colorado, USA. General Scientific Advisor: Dr. Guy Wingate,
`UK Quality Manager, Computer Systems Compliance, Secondary Manufacturing, Glaxo Wellcome,
`Barnard Castle, England.
`
`10987654321
`\
`ISBN: 1-57491="!10-Y ...
`Copyright(© 2001,.-'2002 by'lnterpharm Press, An IHS Health GroupTM company.
`All rights reser~d. · -· · -·
`
`All rights reserved. This book is protected by copyright. No part of it may be reproduced, stored in a
`retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying,
`recording, or otherwise, without written permission from the publisher. Printed in the United States
`of America.
`Where a product trademark, registration mark, or other protected mark is made in the text, ownership
`of the mark remains with the lawful owner of the mark. No claim, intentional or otherwise, is made
`by reference to any such marks in this book.
`While every effort has been made by IHS~ Health Group to ensure the accuracy of the information
`contained in this book, this organization accepts no responsibility for errors or omissions.
`
`Interpharm Press
`15 Inverness Way East
`Englewood, CO 80112-5776
`
`303-662-9101
`Phone:
`303-754-3953
`Fax:
`Orders/on-line catalog:
`www.interpharm.com
`
`This.mate.rial w:ascopied
`at the NLM and may he
`Subject US Copyright Law-s
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`7
`
`Biopharmaceutical Support in
`Formulation Development
`
`Berti/ Abrahamsson and
`Anna-Lena Unge/1
`Astra Zeneca
`Molndal, Sweden
`
`The pharmaceutical formulation plays an important role in the delivery of a drug to the body.
`The clinical benefit of a drug molecule can thereby be optimised by delivering the right
`amount at the right rate to the right site at the right time. For example, extended-release (ER)
`formulations have been used for a long time to control the rate of absorption and thereby keep
`drug levels within the therapeutic interval during an entire dosage interval. More examples of
`biopharmaceutical properties that can be provided by oral formulations are given in Table 7.1.
`In the future, the pharmaceutical possibilities for improving clinical utility may be extended
`to include she-specific drug delivery systems that reach systemic targets, such as cancer cells
`and the central nervous system (CNS), or gene delivery to cell nuclei. Such areas of drug de-
`livery are, however, outside the scope for the present chapter.
`In order to achieve the potential clinical benefits that can be provided by a formulation,
`as exemplified in Table 7.1, biopharmaceutical input is needed from the start of preformula-
`tion, through formulation development, to documentation for regulatory applications. The
`main objective is to obtain and verify desirable drug delivery properties for a pharmaceutical
`formulation. The key activities are as follows:
`
`Characterisation of relevant physicochemical, pharmacokinetic/dynamic prerequi-
`sites provided by the drug molecule
`Identification of the relevant biopharmaceutical targets and hurdles in formulation
`development
`
`This material was copie.cf
`at the NLM .and m.ay b.e
`~bj&t US Cojpyright La\vs.
`
`239
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`240
`
`Pharmaceutical Preformulation and Formulation
`
`Definition of test methods/study designs needed to obtain the biopharmaceutical
`targets in the formulation development and correct interpretation of the study re-
`sults obtained
`Choke of suitable drug form, formulation principles and excipients
`
`In addition, understanding of the physiological processes that may interact with the bio-
`pharmaceutical function of the dosage form is crucial.
`Successful biopharmaceutical input during development can make a significant contri-
`bution to clinical efficiency and tolerability of a drug product. In certain cases, such as poorly
`absorbable drugs or drugs that are degraded in the gut, the biopharmaceutical aspects can
`make the difference between a new useful product or an aborted development programme of
`a potentially very useful drug compound. Additionally, appropriate use of biopharmaceutics
`will also contribute to a time and cost-efficient development process.
`The present chapter is limited to presentations and uses of different biopharmaceutical
`test methods in formulation development, such as
`in vitro dissolution testing,
`bioavailability studies,
`in vitro/in vivo (IVIVC) correlation of drug dissolution,
`use of animal models in in vivo studies of formulations and
`in vivo imaging of formulations by gamma scintigraphy.
`
`This chapter is strongly focussed on oral drug delivery. The relevant principles and meth-
`ods involved in biopharmaceutical characterisation of a drug molecule, mainly applied in the
`preformulation phase, are described in Chapter 4, "Biopharmaceutical Support in Candidate
`Drug Selection':
`
`Table 7.1
`Examples of biopharmaceutical properties of oral dosage forms.
`Biopharmaceutical Target
`Fonnulatron Function
`Increase amount absorbed/
`Dissolution or permeability enhancement
`reduced variability of amount absorbed
`Protection from degradation in Gl tract
`Extended release
`Control rate of absorption
`Pulsed release
`Gastric retention
`Colon release
`Mucoadhesive
`
`Control site of delivery
`
`Th.i:S materia I w.:~s copiei:l'
`at the Ni!vl and m:ay be
`!;ubjed USC.opyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`241
`
`IN VITRO DISSOLUTION
`In vitro dissolution testing of solid dosage forms is the most frequently used biopharmaceuti-
`cal test method in formulation development. It is used from the start of dosage form devel-
`opment and in all subsequent phases. Examples of different purposes of dissolution testing in
`research and development are as follows:
`
`Investigation of drug release mechanisms, especially for ER formulations
`To obtain a predefined target release proftle and robust formulation properties re-
`garding influences of physiological factors (e.g., pH and food) on the drug release
`Generation of supportive data to bioavailability studies as an aid in interpretation of
`in vivo results
`Validation of manufacturing processes
`Investigation of effects of different storage conditions
`Batch quality control ( QC)
`A surrogate for bioequivalence studies
`An in vitro dissolution method for batch QC is always defined for a new solid dosage form
`product. However, this method may not be sufficient for all the different aims of dissolution
`testing that might arise. The choice of dissolution method and test conditions should there-
`fore be adapted to best serve their purpose. For example, simplicity and robustness are crucial
`properties of a QC method; whereas physiological relevance may overrule these factors when
`a method is used for in vivo predictions.
`Standard in vitro dissolution testing models two processes; the release of drug substance
`from the solid dosage form and drug dissolution. Drug release will be determined by formu-
`lation factors such as disintegration/dissolution of formulation excipients or drug diffusion
`through the formulation. Drug dissolution will be affected by the physicochemical substance
`properties (e.g., solubility, diffusivity), solid-state properties of the substance (e.g., particle
`surface area, polymorphism) and formulation properties (e.g., wetting, solubilisation). In
`vitro dissolution testing should thus provide predictions of both the drug release and the dis-
`solution processes itt vivo. Therefore, in most situations, the use of in vitro dissolution will be
`meaningless if the method used does not provide some correlation with in vivo data or re-
`semblance with the physiological conditions in the gastro-intestinal ( GI) tract. In order to
`reach this goal, the choice of dissolution apparatus and test medium should be carefully con-
`sidered. Another important aspect in the development and defmition of a new method is that
`it must be designed and operated in such a way that drug release and dissolution are not sen-
`sitive to minor variations in the operating conditions.
`This chapter will provide some practical considerations for developing and using in vitro
`dissolution methods. Aspects of study design and evaluation of in vitro dissolution data will
`also be discussed. For additional information on in vitro dissolution testing, the "FIP Guide-
`lines for Dissolution Testing of Solid Oral Products, (1997), Handbook of Dissolution Testing
`(Hansson 1991), pharmacopoeias and regulatory guidelines are recommended.
`
`This matl!rial was.co.pied
`atth.e NLMandimay be
`S.ubje.<~:t US C.o.p·yright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`242
`
`Pharmaceutical ?reformulation and Formulation
`
`Dissolution Apparatus
`The most well-established apparatuses are those described in the pharmacopoeias. Four meth-
`ods, mainly intended for oral solid dosage forms, are described in the U.S. Pharmacopeia
`(USP) XXIV: the rotating basket method (USP I), the rotating paddle method (USP II), the
`reciprocating cylinder (USP III) and the flow-through method (USP IV). All of these meth-
`ods, except for the reciprocating cylinder, are also described in the European Pharmacopoeia
`(EP), although the equipment specifications are not identical to those in the USP. These
`methods are schematically presented in Figures 7.1 a-d.
`
`USP I. The dosage form is placed in a cylindrical basket that is covered by a mesh.
`The basket is immersed in the dissolution medium and rotated at a speed of between
`25 and 150 rpm. The standard beaker has a volume of 1 L, but 4 L vessels are also
`available. The mesh size in the basket wall can also be varied.
`USP II. The dosage form moves freely in the same type of glass beaker as used for
`USP I. A paddle is rotated at a speed of 25 to 150 rpm. The dosage form may be
`placed in a steel helix in order to avoid floating.
`USP III. The formulation is placed in a cylindrical glass tube with steel screens in the
`bottom and the top. The mesh size of the tubes may vary. This tube is moved up and
`down in a larger tube that contains the dissolution fluid. The amplitude of the inner
`tube movements is 5-40 dips/min, and the volume of the outer tube is 300 mL. Tubes
`containing 100 mL and 1 L are also available. The inner tube can be moved during
`the dissolution process between different outer tubes, which may hold different dis-
`solution fluids.
`USP IV. The formulation is placed in a thermostated flow-cell. The dissolution fluid
`is pumped through the cell in a pulsating manner at a constant rate, typically be-
`tween 4 and 16 mL/min. Before the inlet flow reaches the formulation, it is passed
`through a bed of glass pellets to create a laminar flow. A filter is placed in the cell at
`the outlet side of the formulation. The cell is available in different sizes/designs, and
`tablet holders are available as an option.
`
`USP XXIV and the EP describe four additional apparatuses mainly intended for trans-
`dermal or dermal delivery: the paddle over disc (USP V, EP), the extraction cell method (EP),
`the cylinder method (USP VI, EP) and the reciprocating holder method (USP VII). A large
`number of other non-compendia! methods have been described. Most of them could be cat-
`egorised as
`
`modified USP methods,
`rotating flask methods (Koch 1980) and
`dialysis methods (El-Arini et al. 1990).
`
`An example of a commercially available (VanKel, Cary, N.C., USA) alternative to the stan-
`dard USP II method is one that consists of a glass vessel that has been modified by introduc-
`ing a peak in the bottom (see Figure 7.2). This modification has been introduced to create
`appropriate stirring in all parts of the vessel and thereby avoid formation of poorly agitated
`heaps of undissolved material.
`
`This m:at~ri a I ~vas copied
`at the N LM and may be
`Subject US Copyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`243
`
`Figure 7.1 Different dissolution apparatuses:
`(a) the rotating basket CUSP I) dissolution apparatus.
`
`(b) The rotating paddle CUSP II).
`
`This mat~H"ial was copied
`at the- NLM and: may !J.e
`:tubject US~pyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`244
`
`Pharmaceutical Preformulation and Formulation
`
`Figure 7.1 continued
`(c) The reciprocating cylinder (USP I II) dissolution apparatus .
`
`.
`
`.
`
`.......... . ~. _, ...... ·~· ·~~ .......... _ .•.... --.. --···· _ ...... - .... -~ ..... ~ .. ---·--· ......
`,....:.._:.:.....~'~ ... ~.:.....~ •.. -.. ~~-~--···---·--··-····~·~··· .. -- ~--·~····· .. ····-···--···--·
`......... "~--~~=msm~~
`
`·•
`. .
`
`~
`
`.
`
`... ' . ,..,..,. ...... . . . ..
`
`(d) Flow-through cell (USP IV].
`
`This materia I was cnpied
`at the NLM and m<:~y be
`Siubje~t USC.opyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`245
`
`Figure 7.2 Standard and modified (peak vessel) USP II dissolution apparatuses
`including illustrations of the different flow patterns within the beakers and photographs
`taken at a paddle stirring rate of 100 rpm showing a heap of pellets beneath the paddle
`in the standard method compared to the desirable dispersion of pellets in the modified
`method.
`
`Standard USP vessel
`
`Peak vessel
`
`Conventional
`
`Peali Vessel
`
`The choice of dissolution apparatus will be specific for each formulation, and the follow-
`ing factors should be considered:
`Correlation to i11 vivo data
`Risk for hydrodynamic artefacts
`Regulatory guidelines
`Drug solubility
`Need to change the dissolution medium during dissolution testing
`Ease of operation, in-house know-how and suitability for automation
`
`As a general guideline in the choice of dissolution test apparatus, the simplest and most
`well-established method should be chosen, with respect to both in-house know-how and reg-
`ulatory aspects. In most cases, this is the USP II paddle method or the USP I rotating basket
`method. However, if satisfactory performance cannot be obtained by these methods, others
`should be considered. Primarily, the USP III and USP IV methods, and non-compendia}
`methods could also provide relevant advantages.
`Correlation of the in vitro dissolution to the in vivo dissolution is a crucial property of a
`dissolution test. The major difference in this respect between different apparatus is the
`hydrodynamic conditions. It has been argued for some of the methods, such as the USP IV
`flow-through cell or a rotating flask with baffles, that an in vivo-like situation is created in the
`
`This materi-al wasc.O'pied
`at the NlM and maybe
`!tuhje'Ct US Copyright La\vs
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`246
`
`Pharmaceutical Preformulation and Formulation
`
`vitro test. However, this hypothesis has not been verified by any experimental means for any
`method, and it is clear that no apparatus mimics the full complexity of the motility patterns
`in the GI tract. A recommended approach is therefore to evaluate different apparatuses on a
`case-by-case basis using IVIVC studies (see "Bioavailability Studies': p. 257) to reveal which
`method provides the most desirable results.
`The potential for hydrodynamic artefacts (e.g., floating, clogging of material to screens,
`adhesion to equipment of the formulation or variable flow conditions in the vicinity of the
`formulation due to other reasons) is strongly formulation dependent and thus has to be eval-
`uated for each type of formulation. In order to detect artefacts, careful visual inspection of the
`dissolution test equipment is crucial. Video recordings can be used to aid such investigations.
`The present regulatory guidelines in the United States and Europe propose the use of
`USP I and USP II as the methods of choice. Other methods, both compendia! and non-
`compendia!, could be acceptable, but the rationale for not using USP I and II must be clearly
`stated and supported by experimental data. In generic product development, complete disso-
`lution methods, including the apparatus, are provided for many products in the USP and
`should thus be a first choice in a regulatory context. It should be noted, however, that this is
`not applicable in all cases. A dissolution method that is well functioning for a certain formu-
`lation type may provide high variability, artefacts or poor IVIVC for other dosage forms. Thus,
`in particular, for ER formulations or dosage forms containing dissolution enhancing princi-
`ples, different dissolution tests may be needed for different formulations, although the drug
`substance is the same.
`For sparingly soluble substances, the volume in standard vessel methods may not be suf-
`ficient to dissolve the dose. In this case, the USP IV flow-through method is beneficial, since
`it provides a continuous renewal of the dissolution fluid. However, the maximum flow rate
`will limit the apparent solubility in this procedure. Sufficient solubility will not be obtained
`for a rapidly releasing formulation of a drug with very low solubility in relation to the dose.
`In certain cases, it is desirable to change the dissolution medium during the dissolution
`test. For example, a more physiologically relevant medium is desired with changes of the con-
`ditions (e.g. pH) corresponding to the differences along the GI tract. Both USP III and USP
`IV permit such changes without significant interruptions of the dissolution process.
`Irrespective of the chosen apparatus, the equipment must be set up and handled in a way
`that both minimises the variability of the dissolution and avoids artefacts. The most common
`source to such variability or artefacts is hydrodynamic factors, but unwanted chemical reac-
`tions or temperature shifts could also occur. Alterations of hydrodynamics, as well as changes
`of temperature, can both affect the dissolution of a drug substance and the release of a sub-
`stance from the dosage form. Chemical reactions in the test medium may cause degradation
`of the drug substance or some formulation excipient which may affect the dissolution, or may
`lead to misinterpretation of the results. Examples of different sources to variability for the
`USP apparatuses are summarised below:
`
`USP !-dogging of basket screen, positioning of basket
`USP II-adherence of formulation to the beaker wall, floating, "entrapment" of solid
`material in the stagnant area beneath the paddle, positioning of the paddle
`USP III-floating, adherence to tube wall or bottom screen, clogging of screens, dis-
`appearance of undissolved material through the screens
`USP IV-clogging of filter, variations in flow rate
`
`This material wasc:opie'l:l
`at the NLM and may be
`Subje-ct US Copyright Laws.
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`247
`
`Some other factors that potentially could cause problems are not specific for a certain ap-
`paratus. These general factors include vibrations, variations in agitation rate, impurities due
`to poor cleaning or to trace amounts of metals from dissolution equipment, variations in dis-
`solution fluid components and poor quality of dissolution media components. Another fac-
`tor relevant for lipophilic compounds is migration of the drug substance into fillers and
`plastic material.
`
`Choice of Agitation Intensity
`All compendia! dissolution apparatus can be operated at different agitation intensities. The
`three most outstanding aspects to consider when deciding at which level the tests should be
`performed are
`
`correlation to in vivo data,
`1.
`2. variability of dissolution results and
`3.
`regulatory guidelines and pharmacopoeial recommendations.
`
`The U.S. regulatory agency recommends a stirring rate of 50-100 rpm for USP I and 50-
`75 rpm for USP II.
`The above-proposed agitation intensities should be used if IVIVCs cannot be improved
`or the variability in dissolution data can be improved by other settings. The major problem
`associated with a too low agitation is that solid material is not sufficiently well dispersed,
`which will delay the dissolution. On the other hand, the possibility to discriminate between
`different formulations/batches with different dissolution properties might be lost at a too in-
`tensive agitation.
`Sometimes a dissolution test is performed with the aim to investigate the robustness of
`the release properties towards potential changes of the physiological conditions in vivo. In this
`case, tests at different agitation intensities should be considered to model different intestinal
`motilities. The use of more than one apparatus may also be considered.
`
`Choice of Dissolution Test Media
`The choice of dissolution medium is highly dependent on the purpose of the dissolution
`study, but the following aspects should always be considered:
`
`Correlation to in vivo data
`Resemblance of physiological conditions in the GI tract
`Regulatory and pharmacopoeial recommendations
`Drug solubility and stability properties at different pH values
`Known sensitivity of the formulation function for different medium factors
`
`Attainment of IVIVC is a key aspect in the choice of dissolution test medium. However, it
`is not recommended to choose a test medium based only on correlation to in vivo data. The
`dissolution test medium should also be relevant for the physiological conditions in the GI
`
`This material was copied
`at the NLM and may be
`Subject US Copyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`248
`
`Pharmaceutical Preformulation and Formulation
`
`tract. Components and physicochemical characteristics of the GI fluids that might be consid-
`ered in the choice of dissolution medium were discussed in Chapter 4. The in vivo conditions
`are not static, but the fluids are constantly changing along the GI tract due to absorption of
`water and nutrients, secretions of enzymes, carbonate, salts and bile and digestive processes. It
`is therefore clear that it is not realistic to reconstitute the full complexity of the in vivo condi-
`tions in an in vitro test. An approach has to be taken where the most relevant factors are in-
`cluded, based on knowledge of the solubility of the drug substance and the release mechanism
`of the dosage form. Examples of some dissolution test media that have been proposed to be
`physiologically relevant are given in Table 7.2 (USP 2000; Dressman et al. 1998).
`Another important aspect in the selection of a dissolution test medium is the need to con-
`sider the saturation solubility of drug in the test medium in relation to the drug dose tested.
`Drug dissolution will depend on the amount of drug in the solution if the dissolved amount
`of drug in the test medium approaches the saturation solubility. This can be understood from
`the Noyes-Whitney equation (see equation 1 in Chapter 4, "Biopharmaceutical Support in
`Candidate Drug Selection>'); the dissolution rate will be· affected by the drug concentration in
`the dissolution medium (Ct) if Ct is not much less than the saturation solubility (Cs). This is
`not a desirable situation since, if Ct controls the rate of dissolution, the test may not be dis-
`criminative for factors related to the formulation performance. Another disadvantage, if Ct is
`significant in relation to Cs, is that the dissolution rate will be dose dependent, and different
`results will be obtained for different strengths of the same formulation. Finally, it can be as-
`sumed, in most cases, that Ct does not affect the in vivo dissolution rate due to the continu-
`ous removal of drug from the GI lumen by the drug absorption process that keeps the drug
`concentrations in the GI tract at a level far below Cs. Consequently, it is desirable to choose a
`
`USP simulated intestinal fluid
`
`Table 7.2
`Examples of dissolution test media including physiological components.
`USP simulated gastric fluid
`2 g/L NaCI
`3.2 g/L pepsin
`0.06 M HCI
`0.05 M KH2P04
`0.015 M NaOH
`10.0 g/L pancreatin
`pH adjusted to 6.8 by HCI or NaOH
`0.01-0.05 M HCI
`2.5 g/L Na Iaury! sulphate
`2 g/L NaCI
`0.029 M KH2P04
`5 mM Na taurocholate
`1.5 mM lecithin
`0.22 M KCI
`pH adjusted to 6.8 by NaOH
`
`Simulated gastric fluid-fasted
`state (Dressman et al. 1998)
`
`Simulated intestinal fluid-fasted
`state (Dressman et al. 1998)
`
`This materia I WCIS co,p-ie<l
`at the NLM am:lmaybe
`Subje<Ct USCopcyright Laws.
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`249
`
`dissolution test method that provides a high enough saturation solubility to avoid dependence
`on Ct. Cs should be at least 3-5 times higher than Ct when the total dose has been dissolved.
`Such conditions have been termed "sink conditions''. In the first instance, maximum volume
`or flow of test medium should be used to obtain sink conditions. Adjustment of the pH to a
`level that provides optimal solubility should be considered for proteolytic drugs without ne-
`glecting the aspects of physiological relevance. For drugs with a very low solubility in relation
`to the administered dose, the above-described approaches will not provide sufficient drug sol-
`ubility in the test media. In those cases, a surfactant should be added to the test medium in
`amounts above its critical micelle concentration (CMC) in order to solubilise the drug. The
`solubility can thereby be increased several hundred orders of magnitude. This approach is also
`favoured due to the occurrence in vivo of drug solubilising micelles formed in the presence of
`bile acids. However, the use in vitro of bile acids in standard methods is not recommended due
`to variations in quality and high costs, and it will still be almost impossible to simulate the in
`vivo complexity. Therefore, synthetic surfactants are the first choice, and sodium lauryl sul-
`phate (SLS) has been especially recommended (Shah et al. 1989). Due to the risk of specific
`interactions between the formulation and SLS (of no in vivo relevance) or poor solubilisation
`capacity for certain drugs, other synthetic surfactants may be considered on a case-by-case
`basis. An example of the first case is illustrated in Figure 7.3 where the in vitro dissolution-
`time proftles of a poorly soluble compound, felodipine, are shown for three different
`hydrophilic matrix ER tablets (A-C) when three different surfactants (SLS, CTAB [cetyl-
`trimethylammonium bromide], Tween) were used to obtain sink conditions (Abrahamsson et
`al. 1994). SLS interacted with the gel forming excipient, which led to much less of a difference
`in drug dissolution rate between the three different tablets, compared to the use of the other
`surfactants. It is also important to realise that attainment of sink conditions does not guaran-
`tee that the in vitro results correlate to the in vivo performance, due to other effects. The dis-
`criminating power of a dissolution test may be lost if the solubility of the drug is too
`favourable in the dissolution medium. For example, an in vitro dissolution method including
`SLS in amounts providing sink conditions was used to test three other felodipine ER tablets.
`No difference was obtained in vitro for the three tablets, which contained different forms of
`the drug substance, whereas one of the tablets provided almost no bioavailability in vivo due
`to poor dissolution (Johansson and Abrahamsson 1997). Thus, general recommendations of
`the amount and type of solubiliser to be used in an in vitro test medium may be misleading,
`and the test medium should preferably be based on correlation to relevant in vivo data for
`poorly soluble substances.
`Based on the knowledge of the substance solubility, release mechanisms from the dosage
`forms and known interactions with key excipients, certain components may be of special im-
`portance to include or exclude in the dissolution test medium. This has to be considered on a
`case-by-case basis. Two examples are given below.
`
`Example 7
`Hard gelatine capsules have the potential to be cross-linked during storage, which leads to for-
`mation of non-water soluble capsules. However, this does not affect the in vivo dissolution due
`to the presence of enzymes that digest the gelatine. Thus, the presence of pepsin and pancre-
`atin in simulated gastric and intestinal fluids, respectively, may be especially important in the
`dissolution testing of hard-gelatine capsules (Digenis et al. 1994).
`
`This material wasc:opie~f
`at the NLM am:! may be
`5ubcject US Copyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`250
`
`Pharmaceutical Preformulation and Formulation
`
`Figure 7.3 In vitro dissolution-time profiles of a poorly soluble compound, felodipine, for
`three different hydrophilic matrix extended release tablets (A-C) when three different
`surfactants (SLS, CTAB, Tween) were used in the dissolution test medium at levels
`providing "sink conditions·:
`
`a) SLS
`100
`80
`
`-g 60 m
`CD a; 40
`a:
`cf.. 20
`0
`0
`
`m 0
`
`1
`
`2
`
`3
`4
`Hours
`
`5
`
`6
`
`7
`
`8
`
`b) TWEEN
`100
`"C eo
`Q) g) 60
`Q) a;
`a::
`40
`'$. 20
`a
`0
`
`1
`
`2
`
`m c
`
`3
`
`4
`Hours
`
`5
`
`6
`
`7
`
`8
`
`c) CTAB
`100
`~ 80
`m so
`en
`0: 40
`~
`20
`0
`
`0
`
`1
`
`m c
`
`2
`
`3
`
`4
`Hours
`
`5
`
`6
`
`7
`
`8
`
`This material \Vas. copied
`at the NLM a nod may be
`Subject US Copyright Laws
`
`AMN1044
`IPR of Patent No. 7,919,499
`
`

`

`Biopharmaceutical Support in Formulation Development
`
`251
`
`Example 2
`The ionic concentration in the test medium can affect both the drug solubility and the release
`mechanism for modified-release formulations. One example of the latter case is hydrophilic
`gel matrix tablets, a type of ER tablet that forms a gel layer in contact with the GI fluids.
`Solutes will affect the hydration of the gel matrix and, thereby, affect the drug release rate.
`It has been shown for such tablets that the correlation to in vivo data can be completely lost
`by use of inappropriate ionic compositions in the test medium (Abrahamsson et al.l998a). For
`ER formulations with osmotically driven drug release, a decreased drug release rate approach-
`ing no release will occur for high ionic concentrations in the test medium, and misleading in
`vitro results may be obtained if a relevant ionic composition is not used (Lindstedt et al. 1989).
`The concern for variability in dissolution results is of special significance when setting up
`a specification test method to be used for batch release but may also be considered for other
`tests. In order to reduce variability in dissolution results due to the test medium, the quality
`aspects of the dissolution media components that could affect the drug dissolution and release
`must be identified, and appropriate qualities of the components should be defmed. This is es-
`pecially important for the use of surfactants to provide micellar solubilisation in the test
`medium (Crison et al. 1997). Another potential source of variability is impurities in the com-
`ponents that may alter the solubility or catalyse degradation of labile drugs. It is also impor-
`tant to see that the dissolution test medium is stable, i.e., that the components are not
`degraded or precipitated during the dissolution test period. This is of no concern for plain
`buffer systems but is more relevant for complex media including physiological components.
`Dissolved air in the dissolution medium could, under certain circumstances, be located as
`air bubbles on the surface of the dosage form or released solid material. This will clearly affect
`the dissolution process by

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