throbber
Pharmaceutical
`Preformulation and
`Formulation
`
`
`
`
`
`
`A Practical Guidefirom Candidate Drug
`Select toComme aphase Form
`~~Ga
`
`MYLAN EXHIBIT 1022
`
`

`

`1408_FM 6/10/03 10:46 AM Page i
`
`PHARMACEUTICAL
`PREFORMULATION
`AND
`FORMULATION
`
`A Practical Guide from
`Candidate Drug Selection to
`Commercial Dosage Form
`
`Mark Gibson
`Editor
`
`IHSfi Health Group
`
`An IHSfi GROUP Company
`
`Your Enterprise Solution to (cid:31)
`Global Healthcare Knowledge
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`disclaimer Page 1 Monday, June 9, 2003 12:22 PM
`
`Library of Congress Cataloging-in-Publication Data
`
`Catalog record is available from the Library of Congress
`
`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 authors and the publisher cannot assume responsibility for the validity of all materials
`or for the consequences of their use.
`
`Neither this book nor any part may be reproduced or transmitted in any form or by any means, electronic or mechanical,
`including photocopying, microfilming, and recording, or by any information storage or retrieval system, without prior
`permission in writing from the publisher.
`
`The consent of CRC Press LLC does not extend to copying for general distribution, for promotion, for creating new works,
`or for resale. Specific permission must be obtained in writing from CRC Press LLC for such copying.
`
`Direct all inquiries to CRC Press LLC, 2000 N.W. Corporate Blvd., Boca Raton, Florida 33431.
`
`Trademark Notice:
`Product or corporate names may be trademarks or registered trademarks, and are used only for
`identification and explanation, without intent to infringe.
`
`Visit the CRC Press Web site at www.crcpress.com
`
`© 2004 by Interpharm/ CRC
`
`No claim to original U.S. Government works
`Library of Congress Card Number 1-57491-120-1
`Printed in the United States of America 1 2 3 4 5 6 7 8 9 0
`Printed on acid-free paper
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 97
`
`4
`
`Biopharmaceutical Support in
`Candidate Drug Selection
`
`Anna-Lena Ungell
`Bertil Abrahamsson
`AstraZeneca
`Mölndal, Sweden
`
`Adminstration via the oral route has been, and still is, the most popular and convenient route
`for patient therapeutics. However, even though it is the most convenient route, it is not the
`simplest route, as the barriers of the gastro-intestinal (GI) tract are in many cases difficult to
`circumvent. The main barriers of the GI tract to systemic delivery are the environment in the
`stomach and intestinal lumen, the presence of different enzymes, the physical barrier of the
`epithelium and the liver extraction. These barriers are of functional importance for the or-
`ganism in controlling intake of water, electrolytes and food constituents and still remain a
`complete barrier to harmful organisms such as bacteria, viruses and toxic compounds.
`Generally, drug absorption from the GI tract requires that the drug is brought into solu-
`tion in the GI fluids and that it is capable of crossing the intestinal membrane into the sys-
`temic circulation. It has therefore been suggested that the drug must be in its molecular form
`before it can be absorbed. Therefore, the rate of dissolution of the drug in the GI lumen can
`be a rate-limiting step in the absorption of drugs given orally. Particles of drugs, e.g., insolu-
`ble crystalline forms or specific delivery systems such as liposomes, are generally found to be
`absorbed to a very small extent. The cascade of events from release of the drug from its dosage
`form, i.e. dissolution of the drug in the gut lumen, interactions and/or degradation within the
`lumen and the uptake of its molecular form across the intestinal membrane into the systemic
`circulation, is schematically shown in Figure 4.1. For rapid and effective design and develop-
`ment of new drug products, methods for drug absorption are required that describe the dif-
`ferent steps involved before and during the absorption process. The need for such specific
`
`97
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 98
`
`98
`
`Pharmaceutical Preformulation and Formulation
`
`Figure 4.1 Drawing showing the different steps in the absorption process including the
`dissolution of the compound from the solid dosage form, interactions with the dissolved
`material in the gastro-intestinal lumen and the uptake of the compound through the
`epithelial membrane.
`
`Solid dosage form Drug in solution Absorbed drug
`Uptake/absorption
`Dissolution
`
`Interactions
`Enzymatic degradation
`
`methods is determined by the information on the rate-limiting step in the cascade of events
`(e.g., solubility, permeability or metabolic instability limited). The results from these methods
`act as a guide to a more efficient discovery process in which resources are given to optimising
`structures that lead to the selection of a good drug candidate with well-defined pharmacoki-
`netic and physicochemical properties. A method now available is multivariate analysis for
`analysing large data sets. Screening and optimisation of several parameters in parallel, e.g.,
`permeability, metabolic stability, solubility, potency, duration and toxicity, represent a grow-
`ing area for rationalising drug discovery using multivariate statistical models (Eriksson et al.
`1999). The importance of this is obvious: There is no point in using resources to increase the
`potency of an oral drug candidate if the drug is not predicted to be orally bioavailable. The
`consideration of biopharmaceutical properties in the selection of candidate drugs has also
`been shown in a recent survey, based on statistics published by the Pharmaceutical and Re-
`search Manufacturers of America (PhRMA), to be the most common reason for terminating
`drug development projects in the clinical phase.
`The dissolution rate and/or the aqueous solubility of the drug will also affect the outcome
`of studies using biological methods, in very early phases of screening. If not dissolved in the
`test system, low solubility drugs will not appear on the receiver side/blood side of a membrane
`or will show incomplete absorption in vivo. Consequently, the drug will be considered a low
`permeability drug and be discarded as being of no potential use as a systemically active drug.
`The situation is even more complex, since there are also mechanistic membrane processes that
`can give the same result. Such processes include drug efflux systems that transport the drug
`from inside the epithelial cell to the lumen of the intestine [e.g., efflux proteins (Hunter and
`Hirst 1997)] or metabolism during transport and adhesion to plastics in the test system (Table
`4.1). The evaluation of the reason for low transport is therefore crucial for the design of
`proper screening procedures.
`In the drug discovery process, the selection of a suitable candidate drug is the milestone
`for continuing into a costly development and clinical phase. Some optimal absorption criteria
`from a biopharmaceutical point of view are shown below:
`
`• High permeability coefficient (determined using in vitro assays such as Caco-2 cell
`monolayers, Ussing chambers, intestinal perfusions, etc.; see below) throughout the
`GI tract [Extended Release (ER) formulation]
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 99
`
`Biopharmaceutical Support in Candidate Drug Selection
`
`99
`
`Table 4.1
`Suggested reasons for low permeability values
`during transport studies with in vitro models.
`
`Adhesions to plastics
`Low solubility
`Complexation with ions in the buffer
`Metabolism in the lumen or in the intestinal segment
`Low activity/viability of the tissue (active transport)
`Analytical problems (analytical response limited)
`Large UWL (unstirred water layer) or mucus layer (unstirred models)
`True low permeability
`
`•
`
`Passive diffusion–directed transport or known mechanism for carrier-mediated
`transport or interaction
`
`• High solubility in aqueous media and over a wide pH range (e.g., pH 1–7).
`
`•
`
`•
`
`No degradation/metabolism in intestinal luminal fluids, intestinal homogenates
`and/or microsomal preparations from the intestine and liver (i.e., low first-pass
`metabolism)
`
`Complete absorption in the GI tract in vivo in several animal species
`
`These criteria are usually difficult to achieve, and the relationship between the in vitro effect of
`the drug (potency/concentration needed), therapeutic effect and index (acceptable variation in
`plasma concentration from safety and efficacy point of view) and the rate and extent of ab-
`sorption must therefore be evaluated carefully for each project and drug. Furthermore, the
`physicochemical characteristics (e.g., the ability of the drug to be formulated into a relevant
`delivery system) of the drug as determined in preformulation studies also guide the selection
`of a potential drug candidate.
`The biopharmaceutical information gathered in the candidate drug selection process re-
`garding the characteristics of the drug molecule (e.g., dissolution, solubility, stability in fluids
`at the site of administration, enzymatic stability, membrane transport and bioavailability) is
`also very useful as input to the subsequent formulation development. This information is im-
`portant, for example,
`
`•
`
`•
`
`•
`
`•
`
`to determine suitable formulation types and technologies,
`
`to set biopharmaceutical targets for formulation development,
`
`to define initial biopharmaceutical test methods and studies needed to reach the
`targets and
`
`as background data for interpretation of different studies used in the development of
`a formulation.
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 100
`
`100
`
`Pharmaceutical Preformulation and Formulation
`
`Thus, a well-performed drug substance characterisation minimises the risk of a subopti-
`mal final formulation as a result of neglecting important biopharmaceutical prerequisites for
`a certain drug substance. Furthermore, such information also allows an efficient development
`process based on science, while trial and error approaches are avoided.
`The ideal model for the biopharmaceutical assessment of drug transport, metabolism and
`dissolution should have certain characteristics, i.e., should represent the main physiological or
`physicochemical barrier as relevantly as possible to the human in vivo situation. No single
`method can represent all barriers and at the same time give information about the mecha-
`nisms underlying the absorption process. Furthermore, no single method can provide all the
`information needed, from the synthesis of a series of compounds in the screening phase (dis-
`covery) to the development of the specific formulation intended for human use. Many differ-
`ent methods have been developed over the last 20 years for use in different phases of drug
`discovery and development. This chapter will deal with some of these techniques to gain a
`basic knowledge of drug absorption. Also, it will give a description of related methods, and the
`functional use of the information provided by these methods, to aid in the selection of a can-
`didate drug and the development of formulations intended for use in humans.
`
`DRUG DISSOLUTION AND SOLUBILITY
`
`Drug dissolution is a prerequisite for oral absorption. Thus, a drug that is not fully dissolved
`cannot be completely absorbed through the GI epithelium. It is thus extremely important to
`understand drug dissolution and solubility in aqueous media, both in early drug discovery
`studies and as a prerequisite for the subsequent formulation development. More specifically,
`drug dissolution/solubility data give important information that provides answers to the fol-
`lowing biopharmaceutical questions during the discovery phase:
`
`• Will the drug absorption be limited by the drug dissolution/solubility?
`
`• Will the drug dissolution/solubility limit the bioavailability to an extent that endan-
`gers the clinical usefulness of the drug?
`
`• Which types of vehicles are needed in preclinical studies to provide the desired drug
`exposure?
`
`•
`
`Should the substance form be changed to improve dissolution (e.g., salt, polymorph,
`particle size)?
`
`After the choice of a candidate drug, solubility and dissolution data are used for guidance
`in the following:
`
`•
`
`•
`
`Should dissolution rate-enhancing principles be applied in the formulation develop-
`ment (e.g., wetting agents, micronisation, solubilising agents, solid solutions, emul-
`sions and nanoparticles)?
`
`In the case of modified release formulations, which formulation principles are suit-
`able and which release mechanisms can be expected?
`
`• Which test conditions should be used for in vitro dissolution testing of solid formu-
`lations?
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 101
`
`Biopharmaceutical Support in Candidate Drug Selection
`
`101
`
`It should be emphasised that dissolution is the dynamic process by which a material is dis-
`solved in a solvent and is characterised by a rate (amount dissolved per time unit), while sol-
`ubility is the amount of material dissolved per volume unit of a certain solvent. Solubility is
`often used as a short form for “saturation solubility”, which is the maximum amount of drug
`that can be dissolved at equilibrium conditions. Finally, the term intrinsic solubility is some-
`times used as well, which is the solubility of the neutral form of a proteolytic drug.
`Theoretically, the dissolution rate is most often described by the Noyes-Whitney equation
`(equation 1):
`
`D A
`dm

`)
`(
`Cs Ct
`-
`dt
`h
`where D is the diffusion coefficient of the drug substance in a stagnant water layer around
`each drug particle with a thickness h, A is the drug particle surface area, Cs is the saturation
`solubility and Ct is the drug concentration in the bulk solution. If the drug concentration in
`the bulk (Ct) in equation 1 is negligible as compared to the saturation solubility (Cs), the dis-
`solution rate is not affected by Ct. This state is denoted a “sink condition” and is often assumed
`to be the case in vivo, owing to the continuous removal of a drug from the intestine due to the
`absorption over the intestinal wall. A, Ct and h in equation 1 will be time dependent, whereas
`the other variables are constants at a certain test condition. The surface area (A) of a dissolv-
`ing particle will be constantly reduced by time (provided that no precipitation occurs); the
`thickness of the diffusion layer (h) is dependent on the radius of the particle size; and the bulk
`solution will increase toward its maximum when the total amount has been dissolved. In ad-
`dition, no solid drug powder is monodisperse, i.e., the starting material will consist of a dis-
`persion of different particle sizes with different surface areas (A). Extensions of equation 1
`have therefore been derived that take into account some or all of these factors. A full review of
`such equations and underlying assumptions, and a presentation of some other less used the-
`ories for dissolution, can be found elsewhere (Abdou 1989). A modification of equation 1 was
`recently presented that takes into account all time-dependent factors that can be useful for
`predictions of the dissolution rate (Hintz and Johnson 1989).
`Basic theoretic considerations and experimental methods regarding solubility are re-
`viewed in more detail in Chapter 3.
`The present chapter focuses on aspects of drug solubility/dissolution of specific relevance
`for biopharmaceutical support in candidate drug selection and preformulation. These aspects
`include solubility in candidate drug screening, physiological aspects of test media, solubility
`of amphiphilic drugs and substance characterisation prior to solubility/dissolution tests.
`
`˚ ¸`
`
`=
`
`(1)
`

`
`(cid:136) fl(cid:152)
`
`Aspects of Solubility in Candidate Drug Screening
`
`Although drug solubility is an important factor in drug absorption in the GI tract, it has not
`been extensively screened for as a barrier to absorption. Drug solubility should, however, be
`complementary to models predicting drug permeability through the lipid membrane. Solu-
`bility as a high-throughput screening (HTS) parameter has therefore been discussed rather in-
`tensively. The importance of solubility as a selective tool during early screening of hundreds
`of compounds, to choose a drug with a potential to be absorbed in vivo in humans, has not
`been fully evaluated, however. Several drugs that are very useful in the clinical situation have
`very low water solubility. For example, candesartan cilexetil, an effective and well-tolerated
`antihypertensive drug, has a water solubility of about 0.1 ␮g/mL. On the other hand, more
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 102
`
`102
`
`Pharmaceutical Preformulation and Formulation
`
`soluble drugs will minimise the risk of failure during the subsequent development phase and
`may avoid delays, increased costs or discontinuation of the project.
`Another aspect of solubility is seen during screening for good pharmacokinetic proper-
`ties of candidate drugs. The HTS systems or in vitro assays are the critical point for most drugs
`insoluble in water. This means that if the drug is not soluble in the buffer solution used in the
`in vitro system, it cannot be properly experimentally evaluated. The most common negative
`effect of this is that the concentration needed to induce transport across the epithelial mem-
`brane in the in vitro model is too low to be detected on the receiver side (see Table 4.1). For
`this reason, vehicles known to increase the solubility of sparingly soluble compounds are used
`(see section “Vehicles for Absorption Studies”). However, since these vehicles are based on sur-
`factant systems, toxic effects may be seen on the membrane (Oberle et al. 1995), and the per-
`meability values obtained may be overestimated. New methods are now available for screening
`large numbers of compounds in small volumes (e.g., the Nephelometer [BMG labtechnolo-
`gies GmbH]). This method is based on turbidimetric determinations and is therefore not an
`exact tool. It can, however, contribute substantially as a first estimate of solubility of sparingly
`soluble compounds and make it possible to understand the results of the screening methods
`and to design specific experiments using vehicles.
`
`Determinations of Drug Dissolution Rate
`
`The dissolution rate, rather than the saturation solubility, is most often the primary determi-
`nant in the absorption process of a sparingly soluble drug. Experimental determinations of
`the dissolution rate are therefore of great importance. The main area for dissolution rate stud-
`ies is evaluation of different solid forms of a drug (e.g., salts, solvates, polymorphs, amor-
`phous, stereoisomers) or effects of particle size. The dissolution rate can either be determined
`for a constant surface area of the drug in a rotating disc apparatus or as a dispersed powder in
`a beaker with agitation.
`The rotating disc method is in most cases the technique of choice for determining the drug
`dissolution rate of drug substances. Compressed discs of the pure drug without any excipients
`are placed in a holder (see Figure 4.2a, b). The disc is immersed in a dissolution medium and
`rotated at a high speed (e.g., 300–1000 rpm). The disc may be centrally or excentrally
`mounted to the stirring rod. The dissolution process is preferably monitored by on-line meas-
`urements of the dissolved drug. A more detailed description of the application of the rotating
`disc method in a preformulation programme can be found elsewhere (Niklasson et al. 1985).
`The dissolution rate is determined by linear regression from the slope of the initial linear
`part of the dissolution time curve. It is often expressed as amount of drug dissolved per time
`and surface area unit (G), e.g., mg/cm2·s, by dividing the rate by the surface area of the disc.
`This dissolution rate is specific for the rotational speed (␻) of the disc and is linearily related
`to the square root of the rotational speed of the disc according to hydrodynamic therories that
`have been experimentally verified (Levich 1962). Thus, if experiments are performed at sev-
`eral rotational speeds, and the dissolution rate at each speed is plotted versus the square root
`of the rotational speed, a linear relationship should be obtained. It should be noted that this
`determination of dissolution rate is still dependent on other experimental hydrodynamic
`conditions, such as positioning of the disc, shape of the vessel and viscosity. An equation has
`therefore been derived that allows for determination of an “intrinsic dissolution rate” (k1) that
`is independent of the drug diffusion in the boundary layer (Niklasson and Magnusson 1985);
`1
`k

`=
`0 5
`¥w .
`G k
`
`R
`
`(2)
`
`+
`
`1 1
`
`
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 103
`
`Biopharmaceutical Support in Candidate Drug Selection
`
`103
`
`Figure 4.2a The rotating disc method: the disc holder and the compressed disc.
`
`Figure 4.2b The rotating disc method: experimental set-up for the rotating disc.
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 104
`
`104
`
`Pharmaceutical Preformulation and Formulation
`
`where k¢ is a proportionality constant. The rotating disc must be mounted eccentrically at a
`certain distance (R) from the center of the stirring rod to perform this evaluation. A plot is
`made between 1/G and 1/(R ⫻ ␻0.5) at different agitation rates, which should yield a straight
`line. The reciprocal of the “intrinsic dissolution rate” (1/k1) is determined by extrapolating the
`line to the y axis. If G is determined at different speeds, and laminar flow along the disc can
`be assumed, other theoretical evaluations can be made of the data, such as determination of
`the diffusion coefficient of the drug in the boundary layer around the solid particles and the
`thickness of the diffusion boundary layer (Levich 1962).
`The main merits of the rotating disc apparatus are the well-defined hydrodynamic con-
`ditions and constant surface area. These reduce the risk of artefacts in dissolution rate deter-
`minations caused by non-ideal test conditions. Furthermore, it is possible to determine an
`intrinsic dissolution rate and to perform other mechanistic evaluations of the dissolution
`process. The main limitation of the method is that it is not suitable for drugs that form frag-
`ile or porous discs, since it is not possible to maintain a constant surface area.
`The drug dissolution rate of powder may be determined by methods such as in a beaker
`with appropriate agitation, as described in the pharmacopoeial methods. The dissolution
`rate determined by such approaches will be method dependent, and it will not be possible
`to derive an intrinsic value of the dissolution rate. The main reason for using this type of
`experimental approach can be understood when the effect of the drug particle size must be
`considered. Experimental errors and uncontrollable variations may occur for hydrophobic
`drugs due to agglomeration in the test medium or due to floating. The use of a wetting agent
`in the test medium (such as a surfactant in concentrations well below the critical micelle con-
`centration [CMC]), may be needed to avoid such undesired effects.
`
`Biopharmaceutical Interpretation of Dissolution/Solubility Data
`
`It is desirable to predict the influence of drug dissolution on oral absorption based on meas-
`urements of dissolution or solubility, both before the selection of a candidate drug, in order
`to obtain a drug molecule with acceptable properties, and in the preformulation phase, to de-
`termine the need for solubility-enhancing formulation principles. The primary variable for
`judgements of in vivo absorption is the dissolution rate rather than the solubility. Drug disso-
`lution will limit the bioavailability when the dissolution rate is too slow to provide complete
`dissolution in the part of the intestine where it can be absorbed. In addition, the drug con-
`centration in the intestinal fluids will be far below the saturation solubility, under the as-
`sumption that “sink conditions” in the GI tract will be obtained due to absorption of the drug.
`However, most often, solubility data are more readily available than dissolution rates for a
`drug candidate, especially in early phases when the amount of drug available does not allow
`for accurate dissolution rate determinations. Predictions of in vivo effects on absorption
`caused by poor dissolution must thus often be made on the basis of solubility data rather than
`dissolution rate. This can theoretically be justified by the direct proportionality between dis-
`solution rate and solubility under “sink conditions” according to equation 1. A list of proposed
`criteria to be used to avoid a reduction in absorption caused by poor dissolution is given in
`Table 4.2. These criteria are discussed in further detail in this chapter. A solubility in water of
`ⱖ 10 mg/mL in pH range 1–7 has been proposed as an acceptable limit to avoid absorption
`problems, while another suggestion is that drugs with water solubilities ⬍0.1 mg/mL often
`lead to dissolution limitations to absorption (Kaplan 1972; Hörter and Dressman 1997). It
`should be noted that these limits may be conservative, especially in the context of screening
`and selection for candidate drugs. For example, a drug with much lower solubility, such as
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 105
`
`Biopharmaceutical Support in Candidate Drug Selection
`
`105
`
`Table 4.2
`Proposed limits of drug dissolution on solubility to avoid absorption problems.
`Factor
`Limit
`Reference
`
`Solubility in pH 1–7
`Solubility in pH 1–8 and dose
`
`Water solubility
`
`Dissolution rate in pH 1–7
`
`⬎10 mg/mL at all pH
`Complete dose dissolved in
`250 mL at all pH
`⬎0.1 mg/mL
`
`⬎1 mg/min/cm2 (0.1–1 mg/nm/cm2
`borderline) at all pH
`
`Kaplan (1972)
`Amidon et al. (1995)
`
`Hörter and
`Dressman (1997)
`Kaplan (1972)
`
`felodipine (0.001 mg/mL), provides complete absorption when administered in an appropri-
`ate solid dosage form (Wingstrand et al. 1990). This may be explained both by successful ap-
`plication of dissolution-enhancing formulation principles and by more favourable drug
`solubility in vivo owing to the presence of solubilising agents such as bile acids.
`Another model for biopharmaceutical interpretation based on solubility data is found in
`the biopharmaceutical classification system (BCS) (Amidon et al. 1995). Four different classes
`of drugs have been identified, based on drug solubility and permeability as outlined in Table
`4.3. If the administered dose is completely dissolved in the fluids in the stomach, which is as-
`sumed to be 250 mL (50 mL basal level in stomach plus administration of the solid dose with
`200 mL of water), the drug is classified as a “high solubility drug”. Such good solubility should
`be obtained within a range of pH 1–8 to cover all possible conditions in a patient and exclude
`the risk of precipitation in the small intestine due to the generally higher pH there than in the
`stomach. Drug absorption is expected to be independent of drug dissolution for drugs that
`fulfil this requirement, since the total amount of the drug will be in solution before entering
`the major absorptive area in the small intestine, and the rate of absorption will be determined
`by the gastric emptying of fluids. Thus, this model also provides a very conservative approach
`for judgements of dissolution-limited absorption. However, “highly soluble drugs” are advan-
`tageous in pharmaceutical development since no dissolution-enhancing principles are
`
`Class
`
`I
`II
`III
`IV
`
`Table 4.3
`Biopharmaceutical classification system.
`Solubility
`
`High
`Low
`High
`Low
`
`Permeability
`
`High
`High
`Low
`Low
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 106
`
`106
`
`Pharmaceutical Preformulation and Formulation
`
`needed, and process parameters that could affect drug particle form and size are generally not
`critical formulation factors. Furthermore, if certain other criteria are met, in addition to
`favourable solubility, regulatory advantages can be gained. Bioequivalence studies for bridg-
`ing between different versions of clinical trial material and/or of a marketed product can be
`replaced by much more rapid and cheaper in vitro dissolution testing (Guidance for Industry,
`FDA 1999; Note for Guidance on Investigation, EMEA 1998).
`The assumption of “sink condition” in vivo is valid in most cases when the permeability
`of the drug over the intestinal wall is fast, which is a common characteristic of lipophilic,
`poorly soluble compounds. However, if such a drug is given at a high dose in relation to the
`solubility, Ct (see equation 1) may become significant even if the permeation rate through the
`gut wall is high. If the drug concentration is close to Cs (see equation 1) in the intestine, the
`primary substance-related determinants for absorption are the administered dose and Cs
`rather than the dissolution rate. It is important to identify such a situation, since it can be ex-
`pected that the dissolution rate-enhancing formulation principle will not provide any bene-
`fits and that higher doses will provide only a small increase in the amount of absorbed drug.
`As a rough estimate for a high permeability drug, it has been proposed that this situation can
`occur when the relationship between the dose (mg) and the solubility (mg/mL) exceeds a fac-
`tor of 5,000 if a dissolution volume of 250 mL is assumed (Amidon 1996). For example, if the
`solubility is 0.01 mg/mL, this situation will be approached if doses of about 50 mg or more are
`administered. It should, however, be realised that this diagnostic tool is based on theoretical
`simulations rather than in vivo data. For example, physiological factors that might affect the
`saturation solubility are neglected (described in more detail below).
`In order to predict the fraction absorbed (Fa) in a more quantitative manner, factors
`other than dissolution, solubility or dose must be taken into account, such as regional per-
`meability, degradation in the GI lumen and transit times. Several algorithms with varying de-
`grees of sophistication have been developed that integrate the dissolution or solubility with
`other factors. A more detailed description is beyond the scope of this chapter, but a compre-
`hensive review has been published by Yu et al. (1996). Computer programmes based on such
`algorithms are also commercially available and permit simulations to identify whether the ab-
`sorption is limited by dissolution or solubility (Gastroplus™, Simulations Plus Inc, Lancaster,
`Calif., USA). As an example, Figure 4.3 shows simulations performed to investigate the de-
`pendence of dose, solubility and particle radius on the Fa for an aprotic, high-permeability
`drug.
`
`Physiological Aspects of Dissolution and Solubility Test Conditions
`
`The dissolution of a drug in the gut lumen will depend on luminal conditions, e.g., pH of the
`luminal fluid, volume available, lipids and bile acids and the hydrodynamic conditions pro-
`duced from the GI peristaltic movements of the luminal content toward the lower bowel. Such
`physiological factors influence drug dissolution by controlling the different variables in equa-
`tion 1 that describe the dissolution rate. This is summarised in Table 4.4 adapted from Dress-
`man et al. (1998).
`The test media used for determining solubility and dissolution should therefore ideally
`reflect the in vivo situation. The most relevant factors to be considered from an in vivo per-
`spective are
`
`•
`
`•
`
`pH (for proteolytic drugs),
`
`ionic strength and composition,
`
`

`

`1408_Chapter 04II 6/10/03 10:34 AM Page 107
`
`Biopharmaceutical Support in Candidate Drug Selection
`
`107
`
`Figure 4.3 Simulations of fraction of drug absorbed after oral administration for a high-
`permeability drug (Peff = 4.5 ⫻ 10–4 cm/s) for doses 1–100 mg, water solubilities
`0.1–10 g/mL and radius of drug particles 0.6–60 ␮m. During variations of one variable,
`the others are held constant at the midpoint level (dose 10 mg, solubility 1 ␮g/mL and
`particle radius 6 ␮m).
`
`particle
`radius
`
`dose
`
`solubility
`
`Percent Drugs Absd.
`
`
`
`1
`Dose (mg)
`Solubility ((cid:181)g/mL) 0.1
`Particle radius ((cid:181)m) 0.6
`
`10
`1
`6
`
`100
`10
`60
`
`Table 4.4
`Physicochemical and physiological parameters important to
`drug dissolution in the gastro-intestinal tract.
`Physicochemical
`Parameter
`
`Physiological Parameter
`
`Factor
`
`Surface area of drug (A)
`
`Particle size, wettability
`
`Molecular size
`
`Hydrophilicity, crystal
`structure, solubilisation
`
`Diffusivity of drug (D)
`Boundary layer thickness (h)
`Solubility (Cs)
`
`Amount of drug already
`dissolved (Ct)
`Volume of solvent
`available (Ct)
`
`Surfactants in gastric juice
`and bile
`Viscosity of lumenal contents
`Motility patterns and flow rate
`pH, buffer capacity, bile,
`food components
`Permeability
`
`

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