throbber
Pharmaceutics
`"IE Sliiflllllfl [ll llllflflflfl [lll‘lll llflSiflll
`
`,
`
`;
`
`; Ediftédby'M E AuIth ,
`
`>
`
`I
`
`i.
`
`1.
`
`AstraZeneca Exhibit 2094 p. 1
`InnoPharma Licensing LLC V. AstraZeneca AB IPR2017-00904
`Fresenius-Kabi USA LLC V. AstraZeneca AB IPR2017-01910
`
`

`

`CHURCHILL LIVINGSTONE
`
`Medical Division of Longmém Group UK Limited
`Distributed in the United States of America by
`Churchill Livingstone 1110., 650 Avenue of the Americas,
`New York, 10011, and associated companies, branches
`and representatives throughout the world.
`
`© Michael Aulton 1988
`
`All rights reserved. No part of this publication may
`be reproduced, stored in a retrieval system, or
`transmitted in any form or by any means, electronic,
`mechanical, photocopying, recording, or otherwise,
`without the prior permission of the publishers
`(Churchill Livingstone, Robert Stevenson House, 1-3
`Baxter’s Place, Leith Walk, Edinburgh EH1 3AF'), or
`a Licence permitting restricted copying in the United
`Kingdom issued by the Copyright LiCensing Agency Ltd,
`90 Tottenham Court Road, London, WlP 9HE.
`
`First published 1988
`Reprinted 1989
`Reprinted 1990
`Reprinted 1991
`Reprinted 1992
`
`ISBN D-LiLiB-UBELiB-fi
`
`British Library Cataloguing in Publication Data
`Pharmaceutics: the science of dosage form
`design.
`1. Pharmaceutics
`I. Aulton, Michael E.
`615’.19
`RS403
`
`Library of Congress Cataloging in Publication Data
`Pharmaceutics: the science of dosage form design.
`Replaces: Cooper and Gunn’s tutorial pharmacy.
`6th ed. 1972.
`
`2. Drugs
`
`Includes bibliographies and index.
`1. Drugs — Design of delivery systems.
`— Dosage forms. 3. Biopharmaceutics.
`4. Pharmaceutical technology.
`5. Chemistry,
`Pharmaceutical.
`6. Microbiology, Pharmaceutical.
`I. Aulton, Michael E.
`2. Chemistry,
`[DNLM: l. Biopharmaceutics.
`Pharmaceutical.
`3. Dosage Forms.
`4. Technology,
`Pharmaceutical.
`5. Microbiology, Pharmaceutical.
`QV 785 P5366]
`RS420.P48
`1987
`
`615.5’8
`
`86—25888
`
`Printed in Hong Kong
`CPP/OS
`
`The
`publisher’s
`policy is to use
`paper manufactured
`from sustainable forests
`
`AstraZeneca Exhibit 2094 p. 2
`
`

`

`Contents
`
`
`
`1
`Preface
`Contributors
`Acknowledgements
`About this book
`.
`1 The demgn Of dosage forms
`PART ONE Physicochemical
`principles of pharmaceutics
`2 Rheology and the flow of fluids
`3 Solutions and their properties
`4 Surface and interfacial phenomena
`5 Solubility and dissolution rate
`6 Disperse systems
`7 Kinetics and Stablhty testing
`PART TWO Biopharmaceutics
`8 Introduction to biopharmaceutics
`9 Factors influencing bioavailability
`10 Assessment of bioavailabilit
`11 Dosage regimens
`"
`PART THREE Drug delivery systems
`.
`12 Packs for pharmaceutical products
`.
`13 Preformulatlon
`.
`14 Solutions
`.
`15 SuspenSlons
`.
`16 EmuISions
`17 Powders and granules
`18 Tablets
`19 Capsules
`.
`20 Therapeutic aerosols
`21 Parenteral products
`22 Topical preparations
`23 Suppositories and pessaries
`
`-
`
`1
`
`15
`17
`38
`50
`62
`81
`119
`129
`131
`135
`174
`191
`213
`215
`223
`254
`269
`282
`_ 300
`304-
`322
`341
`359 '
`381
`412
`
`PART FOUR Pharmaceutical
`vii
`ix microbiology
`Xi
`24 Fundamentals of microbiology
`xiii
`’25 The action of physical and chemical
`agents on micro-organisms
`26 Principles of sterilization
`27 Microbiological Contamination and
`preservation of pharmaceutical
`preparations
`28 Pharmaceutical applications of
`microbiological techniques
`PART FIVE Pharmaceutical
`technology
`29 Materials of fabrication and corrosion
`30 Heat transfer and the properties of
`steam
`31 Filtration
`32 Mixing
`33 Particle size analysis
`34 Particle Size “dual?“
`35 Particle Size separation
`36 Powder flow
`.
`37 Granulation
`_
`.
`.38 Drying
`.
`39 Tableting
`.
`40 Tablet coating
`.
`41 Encapsulation
`.
`.
`42 DeSlgn and operation of clean rooms
`.
`.
`.
`.
`,
`43 Sterilization practice
`,
`44 P k .
`t hnol
`ac aging ec
`Index
`
`,
`
`.
`
`ogy
`
`423
`425
`
`452
`472
`
`479
`
`49-1
`
`509
`511
`
`525
`538
`550
`564
`581
`591
`600
`
`616
`
`629
`647
`669
`678
`
`686
`700
`712
`
`725
`
`AstraZeneca Exhibit 2094 p. 3
`
`

`

`l3
`5‘ I Wells and M E Aulton
`
`
`Preformulation
`
`__________________—_.__———————-—————-———
`
`THE CONCEPT OF PREFORMULATION
`
`1 SPECTROSCOPY
`
`'
`
`2 SOLUBILITY
`
`Intrinsic solubility (Co)
`pKa from solubility data
`Salts
`
`Solvents
`
`Partition coefficient (Kw)°
`Cosolvent solubility
`Methodology and structure activity prediction
`Choice of non-aqueous solvent (oil)
`Structure-activity relationships
`Dissolution
`
`Intrinsic dissolution rate
`
`Measurement of intrinsic dissolution rate
`
`Common ion effect
`
`3 MELTING POINT
`
`Capillary melting
`Hot stage microscopy
`Differential scanning calorimetry and differential
`thermal analysis
`Polymorphism
`Pseudopolymorphism
`True polymorphism
`Crystal purity
`Solubility
`
`4 ASSAY DEVELOPMENT
`
`U.v. spectroscopy
`Molecular weight
`
`pKa
`Mixtures
`
`Thin layer chromatography (t.l.c.)
`High performance liquid chromatography
`(h.p.l.c.)
`
`Normal phase h.p.l.c.
`Reserve phase h.p.l.c.
`
`5 DRUG AND PRODUCT STABILITY
`
`Temperature
`Order of reaction
`
`Hydrolysis
`The influence of pH
`
`Solvolysis
`Oxidation
`
`Chelating agents
`Photolysis
`Solid-state stability
`
`Hygroscopicity
`Stability assessment
`
`6 MICROSCOPY
`
`Crystal morphology
`Particle size analysis
`
`7 POWDER FLOW PROPERTIES
`
`Bulk density
`Angle of repose
`
`8 COMPRESSION PROPERTIES
`
`Interpretation of the results from the compression
`scheme
`
`Plastic material
`
`Fragmenting material
`Elastic material
`
`Punch filming (sticking)
`
`9 EXCIPIENT COMPATABILITY
`
`Method
`
`Interpretation
`
`10 CONCLUSIONS
`
`223
`
`AstraZeneca Exhibit 2094 p. 4
`
`

`

`224 DRUG DELIVERY SYSTEMS
`
`THE CONCEPT OF PREFORMULATION
`
`Almost all new drugs which are active orally are
`marketed as tablets, capsules or both (Table 13.1).
`Although only a few drug compounds are eventu-
`ally marketed as an injection (25% of those drugs
`marketed as tablets), an injection, particularly by
`the intravenous route,
`iS always required during
`early toxicity, metabolic, bioavailability and
`clinical studies in order to guarantee precise drug
`and dose deposition. Other dosage forms may be
`required (Table 13.1) but these are usually drug
`specific andoften depend to a large extent on the
`successful development of tablets, capsules and
`injections.
`Prior to the development of these three major
`dosage forms with a new drug candidate,
`it
`is
`
`Table 13.1 Frequency distribution of dosage form types
`manufactured in the UK (Data Sheet Compendium)M
`
`
`
`Dosage form Frequency (%)H
`
`45.8
`Tablets
`16.0
`Liquid oral
`15.0
`Injections
`13.0
`Capsules
`3.3
`Suppositories and pessaries
`3.0
`Topicals
`1.8
`Eye preparations
`1.2
`Aerosols (inhalation)
`0.3
`Others
`——————_——_*_._
`
`that certain fundamental physical and
`essential
`chemical properties of the drug molecule and
`other derived properties of the drug powder are
`determined. This information will dictate many of
`the subsequent events and possible approaches in
`formulation development. This
`first
`learning
`phase is known as preformulation.
`A suggested list of information required in
`preformulation is
`shown in Table 13.2.
`It
`is
`assembled in a logical order recognizing the relative
`importance of the data and probable existence of
`only limited quantities of bulk drug (mg rather
`than g) at this stage of its development. Investi-
`gators Should also be pragmatic and only generate
`data which is of immediate relevance, especially
`if the likely dosage forms are known.
`However, a knowledge of two fundamental
`properties is mandatory for a new compound:
`
`intrinsic solubility (C0),
`1
`2 dissociation constant (pKa).
`
`These will immediately determine (a) the need and
`(b) the possibility of making more soluble salts of
`the drug to eliminate solubility-related poor bio-
`availability, particularly from solid dosage forms.
`Independent of this pharmaceutical profiling
`(Table 13.2), analysts will need to generate data
`(Table 13.3) to support the assay of existing and
`
`Table 13.2 Preformulation drug characterization
`
`M T
`
`Method/function/characterization ‘
`est
`References/BibliographyW
`l Spectroscopy
`Simple u.v. assay
`Dalglish (1969)
`
`2 Solubility
`aqueous
`pKa
`salts
`solvents
`partition coeff K3,
`dissolution
`
`3 Melting point
`
`Phase solubility, purity
`Intrinsic solubility, pH effects
`Solubility control, salt formation
`Solubility, hygroscopicity, Stability
`Vehicles, extraction
`Lipophilicity, structure activity
`Biopharmacy
`
`DSC ——- polymorphism, hydrates, solvates
`
`4 Assay development
`
`u.v., t.l.c., h.p.l.c.
`
`5 Stability (in solution
`and in solid State)
`
`Thermal, hydrolysis, oxidation, photolysis,
`metal ions, pH
`
`Mader (1954); Higuchi and Connors (1965)
`
`Albert and Serjeant (1984)
`Berge, et al. (1977)
`YalkOwski and Roseman (1981)
`Leo, et al. (1971)
`Swarbrick (1970)
`
`Wendlandt (1974); Haleblian (1975),
`Haleblian and McCrone (1969)
`
`Jaffe and Orchin (1962); Bristow (1976)
`
`Mollica et al. (1978); Connors et al. (1979)
`
`6 Microscopy
`7 Powder flow
`
`Morphology, particle Size
`
`McCrone et al. (1978)
`
`(a) bulk density
`(b) angle of repose
`
`Tablet and capsule formulation
`Tablet and capsule formulation
`
`8 Compression properties
`
`Aids excipient choice
`
`Neumann (1967)
`Neumann (1967)
`
`DeBoer et a1. (1978); Jones (1981)
`
`9 Excipient compatibility
`
`Preliminary screening by DSC, confirmation by
`t.l.c.
`M
`
`Smith (1982)
`
`AstraZeneca Exhibit 2094 p. 5
`
`

`

`Table 13.3 Analytical preformulation
`
`
`
`Attribute Test
`
`Identity
`
`Purity
`
`Assay
`
`Nuclear magnetic resonance (n.m.r.)
`Infra red spectroscopy (i.r.)
`Ultraviolet spectroscopy (u.v.)
`Thin layer chromatography (t.l.c.)
`Differential scanning calorimetry (DSC)
`Optical rotation, where applicable
`
`Moisture (water and solvents)
`Inorganic elements
`Heavy metals
`Organic impurities
`Differential scanning calorimetry (DSC)
`Titration
`
`Ultraviolet spectroscopy (u.v.)
`High performance liquid chromatography
`(h.p.l.c.)
`
`Quality
`
`Appearance
`Odour
`Solution colour
`
`pH of slurry (saturated solution)
`
`Melting point
`
`new bulk material. Although their data meet a
`different need, it can often be. used to complement
`and confirm pharmaceutical data. Their greater
`training and knowledge in analysis will assist, for
`example, in the identification of suitable stability-
`indicating assays by ultraviolet spectroscopy (u.v.)
`or high performance liquid chromatography
`(h.p.l.c.) and the screening of incompatibilities by
`thin layer chromatography (t.l.c.).
`
`1 SPECTROSCOPY
`
`The first step in preformulation is to establish a
`simple. analytical method so
`that
`all
`future
`measurements can be quantitative. Most .drugs
`absorb light
`in the ultraviolet wavelengths
`(190—390 nm) since they are generally aromatic
`and/or contain double bonds. Confirmation of
`synthetic structure is, of course, the responsibility
`of the synthetic chemist. The acidic or basic
`nature of the molecule can be predicted from the
`functional groups in its structure (Perrin et al.,
`1981). This will
`indicate suitable solvents
`to
`ensure solution of either the ionized or undis—
`sociated species. This is important since the ionic
`status of a drug can alter the shape of the u. v.
`spectrum by increasing absorption or changing the
`wavelengths (bathochromic (red) or hypsochromic
`(blue) shifts) at which maxima, minima or both
`occur.
`
`PREFORMULATION
`
`225
`
`the new drug
`spectrum of
`Once the u.v.
`molecule is established, it is possible to choose an
`analytical wavelength (often kmax) that is suitable
`to quantify the amount of drug in a particular
`solution. Excitation of the molecule in solution
`
`causes a loss in light energy and the net change
`from the intensity of the incident light (Io) and the
`transmitted light
`(I)
`can be measured. The
`amount of light absorbed by a solution of drug is
`proportional to the concentration (C) and the path
`length of the solution (I) through which the light
`source has passed. Equation 13.1 is
`the well-
`known Beer—Lambert
`law where e is the molar
`
`extinction coefficient.
`
`Absorbance (A) = loglo (IQ/I) = eCl
`
`(13.1)
`
`in pharmacy to quote the
`is usual
`it
`However,
`specific absorption coefficient (E } 22/3,, or E i for short)
`where the pathlength is
`1 cm and the solution
`concentration is 1% w/v (10 mg ml'l) since doses
`and concentrations are generally in metric units.
`
`2 SOLUBILITY
`
`the
`When a preformulation programme begins,
`availability of drug is always limited and the
`preformulation scientist may only have 50 mg.
`Thus,
`it
`is important that
`the best use of this
`limited bulk is made to support the continuing
`efforts to the synthetic chemists and the biologists
`pursuing activity and toxicity screens. Further-
`more, because the compound is new, the quality
`is invariably poor,
`so that a large number of
`impurities may be present and often the first crys-
`tals come down as a metastable polymorph (see
`Section 3 of this chapter and Chapter 5). Accord-
`ingly, if nothing else is measured, the solubility
`and pKa must be determined. These control all
`future work. The solubility dictates the ease with
`which formulations
`for
`intravenous injection
`studies in animals are obtained. The pKa allows
`the informed use of pH to maintain solubility and
`to choose salts should they be required to achieve
`good bioavailability from the solid state (Chapter
`9) and improve stability (Chapter 7) and powder
`properties (Chapter 36).
`Kaplan (1972) suggested that unless acompound
`has
`an aqueous
`solubility in excess of
`1%
`(10 mg mlrl) over the pH range 1—7 at 37 °C,
`
`AstraZeneca Exhibit 2094 p. 6
`
`

`

`226
`
`DRUG DELIVERY SYSTEMS
`
`then potential bioabsorption problems may occur.
`He also found that if the intrinsic dissolution rate
`was greater than 1mg cm—2 min—1 then absorp-
`tion was unimpeded. However, dissolution rates
`of less than 0.1 mg cm‘2 min*1 were likely to give
`dissolution rate-limited absorption. This ten-fold
`difference in dissolution rate translates into a
`lower
`limit
`for solubility of
`1 mg ml‘1 since,
`under sink conditions, dissolution rate and solu-
`bilities are proportional (Hamlin er al., 1965).
`A solubility of less than 1 mg ml“l indicates the
`need for a salt, particularly if the drug will be
`formulated as a tablet or capsule. In the range
`1—10 mg ml—1
`serious consideration should be
`given to salt formation. When the solubility of the
`drug cannot be manipulated in this way (as in the
`case of neutral molecules, glycosides, steroids,
`alcohols or where the pK, is less than 3 for a base
`or greater than 10 for an acid) then liquid filling
`in soft gelatin capsules (in a solution in PEG 400,
`glyceryl triacetate or fractionated coconut oil) or
`as
`a paste or
`semisolid (dissolved in oil or
`triglyceride) in a hard gelatin capsule may be
`necessary.
`
`Intrinsic solubility (Co)
`
`An increase in solubility of the new drug in an
`acidic solution compared with its aqueous solu-
`
`bility suggests a weak base, and an increase in
`alkali, a weak acid. In both cases a dissociation
`constant (pK,) will be measurable and salts should
`form. An increase in both acidic and alkaline solu—
`bility suggests either amphoteric or zwitterion
`behaviour; in this case there will be two pKas,-one
`acidic and one basic. No change in solubility
`suggests a non—ionizable, neutral molecule with no
`measurable pKa. Here solubility manipulations
`will require either solvents or complexation.
`When the purity of the drug sample can be
`assured, the solubility value obtained in acid for
`a weak acid or alkali for a weak base can be
`assumed to be the intrinsic solubility (CD). The
`solubility should ideally be measured at
`two
`temperatures:
`(a) 4 or
`5 “C to ensure good
`physical stability and to extend short-term storage
`and chemical stability until more definitive data
`is available and (b) 37 °C to support biopharma—
`ceutical evaluation.
`However, since absolute purity is often in doubt
`for the first few batches of new drug, it is more
`accurate to determine this crucial solubility by use
`of a phase—solubility diagram (Fig. 13.1). The data
`are obtained from a series of experiments in which
`the ratio of the amount of drug to the amount of
`dissolving solvent is varied. Any deviation from
`the horizontal is indicative of impurities which a
`higher drug loading and its inherent impurities
`
`
`
`lSoluhiiity(mgml“)
`
`9
`
`
`Self-association: complexation
`
`or soiubilization by impurities
`
`
`
`
`
`
`Pure—no interaction
`
`Suppression by common ion effect
`or salting out
`
`
`
`1
`
`2
`
`4
`
`Drug /Solvent Phase Ratio
`
`8
`
`Fig". 13.] Effect of drug: solvent ratio on solubility when the drug is iulpure. Assuming the compound is a base and the
`estimate of its solubility in 0.1 M NaOH was 1 mg ml‘l, four solutions of 3 ml should be set up containing 3, 6, 12 and
`Mam-g of drug. These give the phase ratios shown here. 3 ml is the smallest volume which can be manipulated for either
`cenmfugauon or filtration and dilution for u.v. analysis. The vials should be agitated continuously overnight and then the
`concentration in solution determined
`
`AstraZeneca Exhibit 2094 p. 7
`
`

`

`
`Table 13.4 Potential pharmaceutical salts
`
`
`Basic drugs
`Acidic drugs
`
`
`Anion
`pK"
`“/n Usage“
`Cation
`13K,
`“/0 Usage"
`
`PREFORMULATION 227
`
`Hydrochloride
`Sulphate
`Tosylate
`Mesylate
`Napsylate
`Besylate
`Maleate
`Phosphate
`Salicylate
`Tartrate
`Lactate
`Citrate
`Benzoate
`Succinate
`Acetate
`Others
`
`—-6. 10
`—3.00, +1.96
`— 1.34
`— 1. 20
`0.17 ‘
`0.70
`1.92, 6.23
`2.15, 7.20, 12.38
`3.00
`3.00
`3.10
`3.13, 4.76, 6.40
`4.20
`4.21, 5.64
`4 . 76
`
`Potassium
`Sodium
`Lithium
`Calcium
`Magnesium
`Diethanolamine
`Zinc
`Choline
`Aluminium
`Others
`
`43.0
`7.5
`0.1
`2.0
`0.3
`0.3
`3.0
`3.2
`0.9
`3.5
`0.8
`3.0
`0.5
`0.4
`l .3
`30.2
`
`16.00
`14.77
`13. 82
`12.90
`11.42
`9.65
`8.96
`8.90
`5.00
`
`10.8
`62.0
`1.6
`10.5
`1.3
`1.0
`3.0
`0.3
`0.7
`8.8
`
`* Martindale (1982), The Extra Pharmacopoeia, 28th edition. The Pharmaceutical Press, London.
`
`either promotes or suppresses solubility. In the
`cases where the observed result changes with the
`amount of solvent, the line is extrapolated to zero
`phase ratio, where solubility will be independent
`of solvent level and a true estimate of the intrinsic
`
`the drug. The United States
`solubility of
`Pharmacopoeia uses this method to estimate the
`purity of mecamylamine hydrochloride.
`
`pJKa from solubility data
`
`Seventy—five per cent of all drugs are weak bases
`(20% are weak acids and the remaining 5% are
`non—ionic, amphoteric or alcohols). It is therefore
`appropriate to consider the Henderson—Hasselbalch
`equations for weak bases and acids. These have
`been discussed in Chapter 3 and their bioavail~
`ability consequences in Chapter 9.
`For weak bases
`
`DH = pKa + logloflB] / [BH+])
`
`(13.2)
`
`and for weak acids
`
`PH Z pKa + 10gm([A‘] / [HAD '(13-3)
`
`Equations (13.2) and (13.3) are used:
`
`(a)
`
`to determine the pK, by following changes in
`solubility,
`(b) to allow the prediction of solubility at any pH
`provided that the intrinsic solubility (Co) and
`pK, are known, and
`to facilitate the selection of suitable salt-
`
`(c)
`
`forming cornpounds and predict the solubility
`and pH properties of the salts.
`
`Albert and Serjeant (1984) give a detailed account
`of how to obtain precise pKa values by potentio-
`metry, spectroscopy and conductivity and there is
`further discussion in Chapter 3 of this book.
`
`Salts
`
`in solubility can be
`improvement
`A major
`achieved by selection of a salt. Acceptable phar—
`maceutical salts are shown in Table 13.4 which
`
`also includes their corresponding pK, values (see
`Chapter 3 for further discussion). As an example,
`the consequences of changing chloridiazepoxide to
`various salt forms is shown in Table 13.5.
`
`Table 13.5 Theoretical solubility and pH of salts of
`chlordiazepoxide
`
`Salt
`
`Base
`
`pKa
`
`Salt pH Solubility
`(mg ml")
`
`4. 80
`
`8.30
`
`2.0
`
`Hydrochloride
`Sulphate
`Besylate
`Maleate
`Tartrate
`Benzoate
`Acetate”)
`
`<165‘”
`2.53
`—6.10
`Freely soluble
`2.53
`—3.00
`Freely soluble
`2.53
`0.70
`57.1
`3.36
`1.92
`17.9
`3.90
`3.00
`6.0
`4.50
`4.20
`
`4.76 ——4rl 4.78
`
`
`(1) Maximum solubility of chlordiazepoxide hydrochloride,
`achieved at pH 2.39, is governed by crystal lattice energy
`and common ions.
`
`(23 Chlordiazepoxide acetate may not form; pKI too high and
`close to drug.
`
`AstraZeneca Exhibit 2094 p. 8
`
`

`

`228
`
`DRUG DELIVERY SYSTEMS
`
`In some cases, salts prepared from strong acids
`or bases are freely soluble but also very hygro-
`scopic. This can lead to instability in tablet or
`capsule formulations since some drug will dissolve
`in its own adsorbed films of moisture (the usual
`prerequisite for breakdown) and in the case of a
`weak base, a strongly acid solution may be auto-
`catalytic. Accordingly it is often better to use a
`weak acid or base to form the salt provided any
`solubility requirements are met. A salt which is
`less soluble will also be generally less hygroscopic
`and form less acidic or basic solutions (Table
`13.5). This may also be important in physiological
`terms. Injections should lie in the pH range 3—9
`to prevent vessel or tissue damage and pain at the
`injection site. Oral syrups should not be too acidic
`to enhance palatability. Packaging may also be
`susceptible; undue alkalinity will attack glass, and
`hydrochloride salts should not be used in aerosol
`cans since a propellant-acid reaction will corrode
`the canister.
`
`It is also clear from Table 13.5 that not only
`does the intrinsic pH of the base solution, in this
`example chlordiazepoxide, fall significantly from
`pH 8.3 if salt
`forms are produced but, as
`a
`consequence, the solubility increases exponentially
`(Eqns 13.2 and 13.3). This has important impli-
`cations in viva. A weak base with an intrinsic solu—
`bility greater than 1 mg ml"1 will be freely soluble
`in the gastrointestinal
`tract, especially in the
`stomach. None the less it
`is usually better to
`formulate with a salt since it will control the pH
`of
`the diffusion layer
`(the saturated solution
`immediately adjacent
`to the dissolving surface).
`For example, although chlordiazepoxide base (C8
`= 2 mg ml‘1 at pH 8.3) meets the requirements
`for in viva ‘solubility’ (Kaplan, 1972); commercial
`capsules contain chlordiazepoxide hydrochloride
`(CS = 165 mg ml“1 at pH 2.53).
`A weak base will have a high dissolution rate
`in the stomach, but as it moves down the gastro-
`intestinal tract the pH rises and dissolution rate
`falls. Conversely a weak acid
`has minimal
`dissolution in the stomach but it becomes more
`soluble and its dissolution rate increases as it
`moves down the gut. Paradoxically as dissolution
`rate increases, so absorption falls because the drug
`is ionized.
`
`than the parent drug.
`usually much greater
`Sodium and potassium salts of weak acids dissolve
`much more rapidly than the parent acid and some
`comparative data are shown in Table 13.6. On the
`basis of bulk pH these salts would be expected to
`have lower dissolution rates
`in the stomach.
`However, the pH of the diffusion layer (found by
`measuring the pH of a saturated bulk solution) is
`higher than the pH of gastric fluid (which is
`usually approximately pH 1.5) because of their
`buffering action. The pH approximates
`to a
`saturated unbuffered aqueous solution (calculated
`pH in Table 13.6) and the dissolution rate is
`governed by this pH and not the bulk media pH.
`In the intestine, the salt does not depress the pH,
`unlike the acid which is neutralized, and the
`diffusion layer pH is again raised to promote
`dissolution.
`Since ‘ solubility
`is
`exponentially
`dependent on pH (Eqns 13.2 and 13.3) there is
`a significant increase in dissolution rate over the
`free acid. Providing that the acid forming the salt
`is strong, the pH of the solution adjacent to the
`dissolving surface will be that of the salt, whereas
`for the dissolving free base, it will be the pH of
`the bulk dissolving media. With weak bases, their
`salts dissolve rapidly in the stomach but there is
`no absorption since the drug is
`ionized and
`absorption is delayed until the intestine. There,
`any undissolved drug, as salt, rapidly dissolves
`since the higher diffusion layer pH compensates
`for the higher bulk pH which would be extremely
`unfavourable to the free base. Data for chlordi-
`azepoxide are shown in Table 13.5. The maleate
`
`-
`
`Table 13.6 Dissolution rates of weak acids and their sodium
`salts
`
`
`Drug
`
`pKa
`
`pH
`(at C5)
`
`Dissolution rate
`
`(mg cm'Z min”) X 102
`Dissolution media
`
`
`phosphate
`0.1M HCl
`
`(pH 1.5) (pH 6.8)a“
`
`Salicylic acid
`Sodium salicylate
`
`Benzoic acid
`Sodium benzoate
`
`Sulphathiazole
`Sodium
`
`3.0
`
`4.2
`
`7.3
`
`2.40
`8.78
`
`2.88
`9.35
`
`4.97
`
`1.7
`1870
`
`2.1
`
`980
`
`27
`2500
`
`14
`1770
`
`<0.1
`
`0.5
`
`
`
`10.75 5 50sulphathiazole 810
`
`
`
`
`
`The dissolution rate of a particular
`
`salt
`
`is
`
`Dissolution rate data from Nelson (1958).
`
`AstraZeneca Exhibit 2094 p. 9
`
`

`

`PREFORMULATION
`
`229'
`
`salt has a predicted solubility of 57 mg 1111‘1 but
`more importantly reduces the pH by 5 units. By
`controlling diffusion layer pH, the dissolution rate
`can increase many fold,
`independently of
`its
`position in the gastrointestinal tract.
`Miller and Holland (1960) stated that different
`salts of a drug rarely change its pharmacology,
`only its physical properties. Wagner (1961) qual-
`ified this statement to acknowledge that salts do
`effect the intensity of the response. The salt form
`does change the physicochemical properties of the
`drug. Changes in dissolution rate and solubility
`affect the rate and extent of absorption (bioavail-
`ability),
`and changes
`in hygroscopicity
`and
`stability influence formulation.
`Consequently each new drug candidate must be
`examined extensively to choose the most suitable
`salt for formulation and because each potential salt
`will behave differently, each requires separate
`preformulation screening.
`
`Solvents
`
`As mentioned in the introduction, it is generally
`necessary to anticipate the formulation of an injec-
`tion even if there is no intention of actually
`marketing such a product. The first choice for a
`solvent
`is obviously water. However, although
`the drug may be freely soluble, some are unstable
`in aqueous solution. Accordingly water-miscible
`solvents must be used (a) as cosolvents in formu-
`lations to improve solubility or stability and (b) in
`analysis to facilitate extraction and separation (e.g.
`in chromatography).
`Oils are used in emulsions, topicals (creams and
`ointments), intramuscular injections and liquid-fill
`oral preparations (soft and hard gelatin capsules)
`when aqueous pH and cosolvent solubility and
`
`stability are unattainable. Table 13.7 shows a
`range of solvents to fulfil
`these needs. Aqueous
`methanol is widely used in h. p.l.c. and is the stan-
`dard solvent in sample extraction during analysis
`and stability testing. It is often made acidic or
`alkaline to increase solvent power and ensure
`consistent ionic conditions for u.v. analysis. Other
`pharmaceutical solvents are available (see Spiegel
`and Noseworthy, 1963) but are generally only
`required in special cases. The most acceptable
`non-aqueous
`solvents pharmaceutically are
`glycerol, propylene glycol and ethanol. Generally
`for a lipophilic drug (Le. one with a partition
`coefficient
`(log P) greater
`than 1),
`solubility
`doubles through this series.
`Where bulk is limited and when the aqueous
`solubility is inadequate, it is useful to measure the
`solubility in aqueous cosolvent mixtures rather
`than in a pure organic solvent. Whereas solubili-
`ties at other levels and their mixtures can be
`predicted, the solubility in pure solvent is often
`inconsistent. Furthermore,
`formulations
`rarely
`demand pure non-aqueous solvent, particularly
`injections. For example, ethanol should only be
`used up to 10% in an injection to prevent haemo-
`lysis and pain at the injection site (Cadwallader,
`1978).
`The reader will find more details on the prop-
`erties of solutions in Chapter 3 and on the formu-
`lation of solutions in Chapter 14.
`
`Partition coefficient (K3,)
`
`(solventzwater quotient of
`Partition coefficient
`drug distribution) has a number of applications
`which are relevant to preformulation.
`(a) Solubility: both aqueous and in mixed
`solvents.
`
`Table 13.7 Recommended solvents for preformulation screening
`Salami
`Dielectric constant (e)
`Solubility parameter (6)
`
`
`
`Application
`
`80
`32
`
`24.4
`14.7
`
`All
`Water
`Extraction, separation
`Methanol
`Dissolution (gastric), basic extraction
`0.1 M HCl (pH 1.07)
`Acidic extraction
`0.1 M NaOH (pH 13.1)
`Dissolution (intestinal)
`Buffer (pH 7)
`Formulation, extraction
`12.7
`24
`Ethanol
`Formulation
`12.6
`32
`Propylene glycol
`Formulation
`16.5
`43
`Glycerol
`
`
`
`35PEG 300 or 400 Formulation
`
`Solvents are considered further in the chapter on the formulation of solutions (Chapter 14).
`
`AstraZeneca Exhibit 2094 p. 10
`
`

`

`230
`
`DRUG DELIVERY SYSTEMS
`
`(b) Drug absorption in viva: applied to a homo-
`logous drug series
`for Structure
`activity
`relationships (SAR).
`(c) Partition chromatography: choice of column
`(h.p.l.c.) or plate (t,.l.c.) and choice of mobile
`phase (eluant), see Section 4.
`The measurement of K3, and its use in cosolvent
`
`solubility and structure activity relationships are
`pertinent here, while application in aqueous solu—
`bility prediction is discussed under melting
`point
`(Section 3) and in chromatography in
`Section 4.
`
`Cosoloent solubility
`
`The relative polarities of solvents have been scaled
`using dielectric constant (e), solubility parameter
`(6), interfacial tension (y) and hydrophilic—lipophilic
`balance (HLB), the latter normally being applied
`to non-ionic surfactants in emulsion technology.
`The best solvent
`in any given situation is one
`whose polarity matches that of the solute; an ideal,
`fully compatible solution exists when SW. =
`550mm. This can be ascertained by determining
`solubility maxima, using a substituent contri-
`bution approach (Fedors, 1974). An alternative
`measure is to evaluate the dielectic requirement of
`the system (Paruta & others, 1965).
`However the most practically useful scale of
`polarity or lipophilicity for a solute is its K8, oil:
`water partition coefficient,
`since
`the other
`approaches do not allow easy estimates for the
`behaviour of crystalline solids (solutes).
`For a .wide range of drugs it is possible to relate
`solvent
`solubility and the partition coefficient
`(log K3, # log P). Yalkowsky and Roseman (1981)
`derived the following expression for 48 drugs in
`propylene glycol
`
`log C, = log Cw + f(0.89 log P + 0.03)
`
`(13.4)
`
`Equation 13.4 can be applied more generally by
`introducing a factor (1) to account for the relative
`solvent power of pharmaceutical solvents found in
`practice (Table 13.8), and predicted by comparison
`of interfacial tension against a liquid hydrocarbon,
`tetradecane (Yalkowsky et al., 1975).
`Equation 13.4 now becomes, for a wide range of
`solvents:
`
`Table 13.8 Solvent power ((1)) of some pharmaceutical
`solvents
`
`Solvent
`Relative solvent
`power
`
`
`0.5
`Glycerol
`1
`Propylene glycol (PEG 300 and 400)
`2
`Ethanol
`
`DMA, DMF 4
`
`log C8 = log C0 + f(log q) + 0.89 log P + 0.03)
`(13.5)
`
`Methodology and structure activity prediction
`
`Choice of non-aqueous solvent (oil) The oil:
`water partition (K3,) is a measure of the relative
`lipophilicity (oil-loving) nature of a compound,
`usually in the unionized State (HA or B), between
`an aqueous phase and an immiscible lipophilic
`solvent or oil. Many partition solvents have been
`used (Leo et al., 1971), but the largest data base
`has been generated using n-octanol. This is, aside
`from any scientific argument,
`the major justifi-
`cation to continue its use in preformulation. The
`solubility parameter of octanol (5 = 10.24) lies
`midway in the range for the majority of 'drugs (5
`= between 8 and 12) although some non-polar
`(6 < 7) and polar drugs (6 > 13) are encountered.
`This allows more easily measurable results than in
`inert
`solvents
`(e.g. hydrocarbons) Since
`it
`is
`convenient to partition between equal volumes of
`oil and aqueous phases.
`A typical technique is the shake flask method
`whereby the drug, dissolved in one of the phases,
`is shaken with the other partitioning solvent for
`30 minutes, allowed to stand for 5 minutes and
`then the majority of the lower aqueous phase
`(density of octanol = 0.8258 g ml‘l) is run off
`and centrifuged for 60 minutes at 2000 rpm. The
`aqueous phase is assayed before (2C) and after
`(Cw,
`aqueous
`solubility) partitioning to give
`K3; 2 (2C _ Cw)/Cw)
`Clearly if the transfer of solute to the oil phase
`is small, AC“,
`is small and any analytical error
`increases error in the estimate of K8,.
`Indeed
`to encourage greater aqueous
`loss
`(>ACW)
`a
`considerably more polar solvent, n-butanol, has
`been used. Where the partition coefficient is high,
`it
`is usual
`to reduce the ratio of the oil phase
`
`AstraZeneca Exhibit 2094 p. 11
`
`

`

`PREFORMULATION
`
`231
`
`from 1:1 to 1:4 or 1:9 in order to increase the
`aqueous concentration (Cw) to a measurable level.
`For a 1:9 oil:water ratio K8, = (10 EC — Cw)/
`
`Cw.
`The partition of a polar solute between an inert
`non-polar hydrocarbon e.g. hexane, heptane etc.,
`is quite different to hydrogen bonding solvents
`like octanol
`(Hansch and Dunn,
`1972). The
`behaviour of the weak acid phenol (pK, = 10) and
`weak base nicotine (pK, = 3.1) is worthy of note.
`For phenol, {(3:3‘3“GI = 29.5 whereas Kim“ 2
`0.11. The acidic solvent, chloroform, suppresses
`partition (K3,. = 2.239) whereas although ethyl
`acetate and diethyl ether are more polar, the basic
`behaviour of the solvents give the highest K3,.
`With solvents capable of both hydrogen donation
`and acceptable (octanol,

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