throbber
Pharmaceutics
`
`The Science of Dosage Form Design
`
`Edited by
`Michael E. Aulton BPharm PhD FAAPS MAPharmS
`Professor of Pharmaceutical Technology,
`School of Pharmacy,
`De Montfort University,
`Leicester, UK
`
`SECOND EDITION
`
`/,&\ CHURCHILL
`~~ LIVINGSTONE
`
`:u:
`
`EDINBURGH LONDON NEW YORK OXFORD PHILADELPHIA ST LOUIS SYDNEY TORONTO 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 1 of 66
`
`

`

`CHURCHILL LIVINGSTONE
`An imprint of Elsevier Science Limited
`
`© Harcourt Publishers Limited 2002
`© Elsevier Science Limited 2002. All rights reserved.
`
`No part of this publication may be reproduced, scored in a
`retrieval system, or transmitted in any form or by any means,
`electronic, mechanical, photocopying, recording or otherwise,
`without either the prior permission of the publishers or a
`licence permitting restricted copying in the United Kingdom
`issued by the Copyright Licensing Agency, 90 Tottenham
`Court Road, Lo.ndon WIT 4LP. Permissions may be sought
`directly from Els:evier's Health Sciences Rights Department
`in Philadelphia, USA: phone: (+ I) 215 238 7869, fax: (+!)
`2 I 5 238 2239, e-mail: healtl1permissions@elsevier.com. You
`may also complete your request on-line via the Elsevier
`Science homepage (http://www.elsevier.com), by selecting
`'Customer Support' and then ' Obtaining Permissions'.
`
`First published I 988
`Second Edition ,1002
`Reprinted 2002 twice
`
`Standard edition ISBN O 443 05517 3
`
`International Student Edition ISBN O 443 05550 5
`Reprinted 2002 twice
`
`British Library Cataloguing in Publication Data
`A catalogue record for this book is available from the British
`Library
`
`Library of Cong,ress Cataloging in Publication Data
`A catalog record for this book is available from the Library of
`Congress
`
`Note
`Medical knowledge is constantly changing. As new
`information becomes available, changes in treatment,
`procedures, equipment and the use of drugs become
`necessary. The editor, contributors and the publishers have
`taken care to ensure that the information given in this text is
`accurate and up to date. However, readers are strongly
`advised to confirm. that the information, especially with
`regard to drug usage, complies with the latest legislation and
`standards of practice.
`
`,,;1w1 ~ournals and mu_ltimedia
`
`p your source for books,
`
`on t:he health sciences
`www.elsevierhec1lth.com
`
`Printed in Spain
`
`The
`put,lishe(s
`policy rs to use
`paper manulactured
`from suslalnable forests
`
`I
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 2 of 66
`
`

`

`Contents
`
`What is 'Pharmaceutics'? xiii
`
`1. The design of dosage forms
`Peter York
`
`PART ONE
`Scientific principles of dosage form
`design 13
`2. Dissolution and solubility 15
`M ichael A ulton
`
`3. Properties of solutions 33
`Michael Au/ton
`
`4. Rheology 41
`Ch,·is M arriou
`
`5. Surface and interfacial phenomena 59
`J ohn Fell
`6. Disperse systems 70
`David A uwood
`
`7. Kinetics and product stability 101
`J ohn Pugh
`
`8. Pharmaceutical preformulation 113
`James Wells
`
`PART TWO
`Particle science and powder
`technology 139
`9. Solid-state properties 141
`Graham Buckton
`
`10. Particle-size analysis 152
`John Staniforth
`
`11 . Particle-size reduction 166
`J ohn Sianiforth
`
`12. Particle-size separation 174
`John Siam/ orth
`
`13. Mixing 181
`Andrew Twitchell
`
`14. Powder flow 197
`J ohn Staniforth
`
`PART THREE
`Biopharmaceutical principles of drug
`delivery 211
`15. Introduction to biopharmaceutics 213
`M arianne A shford
`
`16. The gastrointestinal tract - physiology and
`drug absorption 217
`Marianne Ashford
`
`17. Bioavailability - physicochemical and
`dosage form factors 234
`M arianne Ashford
`
`18. Assessment of biopharmaceutical
`properties 253
`M arianne Ashford
`
`19. Dosage regimens 275
`Swart Proudfoot, (updated by John Colleu)
`20. Modified-release peroral dosage form 289
`John Co/leu, Chris M oreton
`
`PART FOUR
`Dosage form design and
`manufacture 307
`21. Solutions 309
`Michael Bil/any
`
`22. Clarification 323
`Andrew Twitchell
`
`23. Suspensions and emulsions 334
`M ichael Bil/any
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 3 of 66
`
`

`

`CONTENTS
`
`24. Powders and granules 360
`M alcolm Summers
`
`25. Granulation 364
`Malcolm Summers, Michael A ulton
`
`26. Drying 379
`Michael Aulwn
`
`27. Tablets and compaction 397
`Goran Alderbom
`
`28. Coating of tablets and
`multiparticufates 441
`J ohn Hogan
`
`29. Hard gelatin capsules 449
`Brian J ones
`
`30. Soft gelatin capsules 461
`Keith Hutchison, Josephine Ferdinando
`
`31. Pulmonary drug delivery 473
`Kevin Taylor
`
`32. Nasal drug delivery 489
`Peter Tay lor
`
`33. Transdermal drug delivery 499
`Bn·an Bany
`
`34. Rectal and vaginal drug delivery 534
`J osef Tukker
`
`35. Delivery of pharmaceutical proteins 544
`Daan Crommelin, Ewoud van Winden
`Albert M ekking
`
`36. Packs and packaging 554
`Dixie Dean
`
`37. Pharmaceutical plant design 571
`Michael Aulton, Andrew Twitchell
`
`38. Heat transfer and the properties and use
`of steam 586
`Andrew Twitchell
`
`PART FIVE
`Pharmaceutical microbiology 597
`39. Fundamentals of microbiology 599
`Geoff Hanlon
`
`40. Pharmaceutical applications of
`microbiological techniques 623
`Norman Hodges
`
`41 . The action of physical and chemical agents
`on microorganisms 643
`Geoff H anlon, Norman Hodges
`
`42. Microbiological contamination and
`preservation of pharmaceutical
`products 658
`Malcolm Parker, Norman Hodges
`Index 669
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 4 of 66
`
`

`

`8P
`
`harmaceutical preformulation: the
`physicochemical properties of drug substances
`
`James Wells
`
`CHAPTER CONTENTS
`
`The concept of preformulation 114
`
`Spectroscopy 114
`
`Solubility 115
`Aqueous solubility 115
`Intrinsic solubility (C0) 115
`pKj, from solubility data 116
`Salts 116
`Solvents 118
`Partition coefficient (K\) 119
`Solvent solubility 119
`Methodology and structure activity
`prediction 119
`Choice of non-aqueous solvent (oil) 119
`Structure-activity relationships 120
`Dissolution 122
`Intrinsic dissolution rate 122
`Measurement of intrinsic dissolution rate 123
`Common ion effect 123
`
`Melting point 124
`Techniques 124
`Capillary melting 124
`Hot-stage microscopy 124
`Differential scanning calorimetry and thermal
`analysis 124
`Polymorphism 124
`Pseudopolymorphism (solvates) 125
`True polymorphism 126
`Crystal purity 126
`Solubility 126
`
`Assay development 127
`UV spectroscopy 128
`Molecular weight 128
`pKa 128
`Thin-layer chromatography 128
`High-performance liquid chromatography
`(HPLC) 128
`
`Normal-phase HPLC 129
`Reverse-phase HPLC 129
`
`129
`
`Drug and product stability
`Temperature 130
`Order of reaction 130
`Hydrolysis 130
`The influence of pH 130
`Solvolysis 131
`Oxidation 131
`Chelating agents 131
`Photolysis 131
`Solid-state stability 132
`Hygroscopicity 132
`Stability assessment 132
`
`Microscopy 132
`Crystal morphology 133
`Particle size analysis 133
`
`Powder flow properties
`Bulk density 133
`Angle of repose 134
`
`133
`
`134
`
`Compression properties
`Plastic material 134
`Fragmentation 135
`Elastic material 135
`Punch filming (sticking) 136
`
`Excipient compatibility 136
`Method 136
`Interpretation 136
`
`Conclusions 138
`
`References 138
`
`Bibliography 138
`
`113
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 5 of 66
`
`

`

`SCIENTIFIC PRINCIPLES OF DOSAGE FORM DESIGN
`
`THE CONCEPT OF PREFORMULATION
`
`Table 8.1 Frequency distribution of dosage form
`types manufactured in the UK
`
`Frequency (%)
`
`46
`
`16
`
`15
`
`13
`
`3 3 2 1 1
`
`Dosage form
`
`Tablets
`
`Liquid oral
`Capsules
`
`Injections
`Suppositories and pessaries
`Topicals
`
`Eye preparations
`
`Aerosols (inhalation)
`
`Others
`
`Independent of this pharmaceutical profiling
`(Table 8.2), analysts will generate data (Table 8.3)
`to confirm structure and purity, and this should be
`used to complement and confirm pharmaceutical
`data. Their greater training and knowledge in
`analysis will assist in the identification of suitable
`stability-indicating assays by high-performance
`liquid chromatography (HPLC).
`
`SPECTROSCOPY
`
`The first step in preformulation is to establish a
`simple analytical method. Most drugs absorb light in
`the ultraviolet wavelengths (190-390 run) as they are
`
`Almost all new drugs are marketed as tablets, cap-
`sules or both (Table 8.1). Although only a few are
`marketed as an injection (25% of those marketed as
`tablets) the intravenous route is always required
`during early toxicity, metabolic, bioavailability and
`clinical studies to provide a precise drug and dose
`deposition. Other dosage forms may be required
`(Table 8.1) but these are drug specific and depend to
`a large extent on the successful development of
`tablets, capsules and injections.
`Prior to the development of these three major
`dosage forms, it is essential that certain fundamen-
`tal physical and chemical properties of the drug
`molecule and other derived properties of the drug
`powder are determined. This information dictates
`many of the subsequent events and approaches in
`formulation development. This first learning phase is
`known as prefor mutation.
`A recommended list of the information required
`in preformulation is shown in Table 8.2. This is
`assembled, recognizing the relative importance and
`probable existence of only limited quantities of new
`bulk drug (mg rather than g). Investigators must be
`pragmatic and generate data of immediate rele-
`vance, especially if the likely dosage forms are
`known.
`Two fundamental properties are mandatory for a
`new compound:
`1. Intrinsic solubility (C0),
`2. Dissociation constant (pKJ.
`
`Table 8.2 Preformulation drug characterization
`
`Test
`
`Method/function/characterization
`
`Spectroscopy
`Solubility
`aqueous
`pKa
`salts
`solvents
`partition coeff K^
`dissolution
`Melting point
`Assay development
`Stability (in solution and solid state)
`Microscopy
`Powder flow
`bulk density
`angle of repose
`Compression properties
`Excipient compatibility
`
`114
`
`Simple UV assay
`Phase solubility, purity
`Intrinsic solubility, pH effects
`Solubility control, salt formation
`Solubility, hygroscopicity, stability
`Vehicles, extraction
`Lipophilicity, structure activity
`Biopharmacy
`DSC - polymorphism, hydrates, solvates
`UV, TLC, HPLC
`Thermal, hydrolysis, oxidation, photolysis, metal ions, pH.
`Morphology, particle size
`Tablet and capsule formulation
`
`Tablet and capsule formation
`Excipient choice
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 6 of 66
`
`

`

`Table 8.3 Analytical preformulation
`
`Attribute Test
`
`Identity Nuclear magnetic resonance (NMR)
`Infra red spectroscopy (IR)
`Ultraviolet spectroscopy (UV)
`Thin-layer chromatography (TLC)
`Differential scanning calorimetry (DSC)
`Optical rotation, where applicable
`
`Purity
`
`Assay
`
`Quality
`
`Moisture (water and solvents)
`Inorganic elements
`Heavy metals
`Organic impurities
`Differential scanning calorimetry (DSC)
`
`Titration
`Ultraviolet spectroscopy (UV)
`High-performance liquid chromatography (HPLC)
`
`Appearance
`Odour
`Solution colour
`pH of slurry (saturated solution)
`Melting point
`
`generally aromatic and contain double bonds. The
`acidic or basic nature of the molecule can be predicted
`from functional groups (Perrin et al 1981). Using the
`UV spectrum of the drug, it is possible to choose an
`analytical wavelength (often Amax) 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 (/0) and the transmitted light (7) 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 (/) through which the
`light has passed. Equation 8.1 is the Beer-Lambert
`law, where e is the molar extinction coefficient.
`
`In pharmacy it is usual to use the specific absorp-
`tion coefficient E\%
`cm (£]), where the pathlength
`is 1 cm and the solution concentration is 1% w/v
`(10 mg mLr1), as doses of drugs and concentrations
`are generally in unit weights rather than molarity
`(E\ = 10e/MW).
`
`SOLUBILITY
`
`Aqueous solubility
`The availability of a drug is always limited and the
`preformulation scientist may only have 50 mg. As the
`
`PHARMACEUTICAL PREFORMULATION
`
`compound is new the quality is invariably poor, so
`that a large number of impurities may be present and
`often the first crystals come down as a metastable
`polymorph. Accordingly, as a minimum, the solubil-
`ity and pKa must be determined. Solubility dictates
`the ease with which formulations for oral gavage and
`intravenous
`injection studies
`in animals are
`obtained. The pKa allows the informed use of pH to
`maintain solubility and to choose salts required to
`achieve good bioavailability from the solid state
`(Chapter 9) and improve stability (Chapter 7) and
`powder properties (Chapter 13 and 14).
`Kaplan (1972) suggested that unless a compound
`has an aqueous solubility in excess of 1% (10 mg
`mLr1) over the pH range 1-7 at 37°C, potential
`bioabsorption problems may occur. If the intrinsic
`dissolution rate was greater than 1 mg cnr2 mkr1
`then absorption was unimpeded. Dissolution rates
`less than 0.1 mg cm~2 mkr1 were likely to give dis-
`solution rate-limited absorption. This tenfold differ-
`ence in dissolution rate translates to a lower limit for
`solubility of 1 mg mL'1. Under sink conditions, dis-
`solution rate and solubilities are proportional.
`A solubility of less than 1 mg mL"1 indicates the
`need for a salt, particularly if the drug will be
`formulated as a tablet or capsule. In the range
`1-10 mg mLr1 serious consideration should be given
`to salt formation. When the solubility of the drug
`cannot be manipulated in this way (neutral mole-
`cules, glycosides, steroids, alcohols, or where the pKa
`is less than 3 for a base or greater than 10 for an
`acid) then liquid filling in soft or hard gelatin cap-
`sules may be necessary.
`
`Intrinsic solubility (C0)
`to
`An increase in solubility in acid compared
`aqueous solubility suggests a weak base, and an
`increase in alkali a weak acid. In both cases a disso-
`ciation constant (pKJ can be measured and salts
`should form. An increase in 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 sug-
`gests a non-ionizable neutral molecule with no mea-
`surable pKa, and solubility manipulation 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 (C0), ie. the fundemen-
`tal solubility when completely unionized. The solu-
`bility
`should
`ideally be measured
`at
`two
`temperatures:
`
`115
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 7 of 66
`
`

`

`SCIENTIFIC PRINCIPLES OF DOSAGE FORM DESIGN
`
`1. 4°C to ensure physical stability and extend
`short-term storage and chemical stability until
`more definitive data are available. The maximum
`density of water occurs at 4°C.This leads to a
`minimum aqueous solubility.
`2. 37°C to support biopharmaceutical evaluation.
`
`However, as absolute purity is often in doubt it is
`more accurate to determine this crucial solubility by
`the use of a phase-solubility diagram (Fig. 8.1). 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 either promotes or suppresses
`solubility. In 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 the true intrinsic solubility of the
`drug. The United States Pharmacopoeia uses this
`method to estimate the purity of mecamylamine
`hydrochloride.
`
`pKa from solubility data
`Seventy-five per cent of all drugs are weak bases;
`20% are weak acids and only 5% are non-ionic,
`amphoteric or alcohols. It is therefore appropriate to
`consider the Henderson-Hasselbalch equations for
`weak bases and acids.
`
`For weak bases:
`
`and for weak acids:
`
`Equations 8.2 and 8.3 can be used:
`1. to determine pKa by following changes in
`solubility
`2. to predict solubility at any pH, provided that the
`intrinsic solubility (C0) and pKa are known
`3. to facilitate the selection of suitable salt-forming
`compounds 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 potentiometry,
`spectroscopy and conductivity.
`
`Salts
`
`A major improvement in solubility can be achieved
`by forming a salt. Acceptable pharmaceutical salt
`counter-ions are shown in Table 8.4. As an example,
`the consequence of changing chlordiazepoxide to
`various salt forms is shown in Table 8.5.
`In some cases, salts prepared from strong acids or
`bases are freely soluble but very hygroscopic. This
`does lead to instability in tablet or capsule formula-
`tions, as some drug will dissolve in its own adsorbed
`films of moisture. It is often better to use a weaker
`acid or base to form the salt, provided any solubility
`requirements are met. A less soluble salt will gener-
`
`Fig. 8.1 Effect of drug: solvent ratio on solubility when the drug is impure. Assuming the compound is a base and the estimate of its
`solubility in 0.1 M NaOH was 1 mg mLr1, four solutions of 3 ml_ should be set up containing 3, 6, 12 and 24 mg of drug. These give the
`phase ratios shown here. 3 ml is the smallest volume that can be manipulated for either centrifugation or filtration and dilution of UV
`analysis. The vials should be agitated continuously overnight and then the concentration in solution determined.
`
`116
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 8 of 66
`
`

`

`PHARMACEUTICAL PREFORMULATION
`
`Table 8.4 Potential pharmaceutical salts
`
`Basic drugs
`
`Anion
`
`Hydrochloride
`
`Sulphate
`
`Mesylate
`
`Maleate
`
`Phosphate
`
`Salicylate
`
`Tartrate
`
`Lactate
`
`Citrate
`
`Succinate
`
`Acetate
`
`Others
`
`pKa
`
`-6.10
`
`-3.00, +1.96
`
`-1.20
`
`1.92,6.23
`
`2.15, 7.20, 12.38
`
`3.00
`
`3.00
`
`3.10
`
`3.13, 4.76, 6.40
`
`4.21,5.64
`
`4.76
`
`Acidic drugs
`
`% Usage
`
`Cation
`
`Potassium
`
`Sodium
`
`Lithium
`
`Calcium
`
`Magnesium
`
`Diethanolamine
`
`Zinc
`
`Choline
`
`Aluminium
`
`43.0
`
`7.5
`
`2.0
`
`3.0
`
`3.2
`
`0.9
`
`3.5
`
`0.8
`
`3.0
`
`0.4
`
`1.3
`
`31.4
`
`Others
`
`PKa
`
`16.00
`
`14.77
`
`13.82
`
`12.90
`
`11.42
`
`9.65
`
`8.96
`
`8.90
`
`5.00
`
`% Usage
`
`10.8
`
`62.0
`
`1.6
`
`10.5
`
`1.3
`
`1.0
`
`3.0
`
`0.3
`
`0.7
`
`8.8
`
`Table 8.5 Theoretical solubility and pH of salts of
`chlordiazepoxide
`
`pKg
`
`SaltpH
`
`Solubility (mg mL-1)
`
`Salt
`
`Base
`
`4.80 8.30
`
`Hydrochloride
`
`-6.10
`
`2.53
`
`Maleate
`
`Tartrate
`
`Benzoate
`Acetate b
`
`1.92 3.36
`
`3.00 3.90
`
`4.20 4.50
`
`4.76 4.78
`
`2.0
`<165a
`57.1
`
`17.9
`
`6.0
`
`4.1
`
`a Maximum solubility of chlordiazepoxide hydrochloride,
`achieved at pH 2.89, is governed by crystal lattice energy
`and common ions.
`b Chlordiazepoxide acetate may not form; pKa of acetate
`too high and too close to that of drug ion.
`
`ally be less hygroscopic and form less acidic or basic
`solutions (Table 8.5). Injections should ideally 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 as a propellant-acid
`reaction will
`corrode the canister.
`From Table 8.5, not only does the intrinsic pH of
`the base solution fall significantly if salt forms are
`produced but, as a consequence,
`the solubility
`
`increases exponentially (Eqns 8.2 and 8.3). This has
`important implications in vivo. A weak base with an
`intrinsic solubility greater than 1 mg mL"1 will be
`freely soluble in the gastrointestinal tract, especially
`in the stomach. However, it is usually better to for-
`mulate with a salt, as it will control the pH of the dif-
`fusion layer (the saturated solution immediately
`adjacent to the dissolving surface, known as the pH
`microenviroment). For example, although chlor-
`diazepoxide base (Cs = 2 mg mL^1 at pHsat 8.3)
`meets the requirements for in vivo 'solubility'
`(Kaplan, 1972); commercial capsules contain chlor-
`diazepoxide hydrochloride (Cs = 1 65 mg roLr1 at
`pHsat 2.53).
`A weak base will have a high dissolution rate in the
`stomach, but as it moves down the gastrointestinal
`tract the pH rises and dissolution rate falls.
`Conversely, a weak acid has minimal dissolution in
`the stomach but becomes more soluble and dissolu-
`tion rate increases down the gut. Paradoxically, as
`dissolution rate increases so absorption falls because
`the drug is ionized.
`The dissolution rate of a particular salt is usually
`much greater than that of the parent drug. Sodium
`and potassium salts of weak acids dissolve much
`more rapidly than do the parent acids, and some
`comparative data are shown in Table 8.6. 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
`
`117
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 9 of 66
`
`

`

`SCIENTIFIC PRINCIPLES OF DOSAGE FORM DESIGN
`
`higher than that of gastric fluid (which is approxi-
`mately 1.5) because of its buffering action. The pH
`is the saturated unbuffered aqueous solution (calcu-
`lated pH in Table 8.6) and the dissolution rate is gov-
`erned by this pH and not the bulk medium 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.
`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 medium.
`With weak bases, their salts dissolve rapidly in the
`stomach but there is no absorption, as the drug is
`ionized and absorption is delayed until the intestine.
`Any undissolved drug, as salt, rapidly dissolves, as the
`higher diffusion layer pH compensates for the higher
`bulk pH, which would be extremely unfavourable to
`the free base. Data for chlordiazepoxide are shown in
`Table 8.5. The maleate salt has a predicted solubility
`of 57 mg mLr1 but, more importantly, reduces the
`pH by 5 units. By controlling diffusion layer pH the
`dissolution rate can increase manyfold, indepen-
`dently of its position in the gastrointestinal tract. This
`is particularly important in the development of con-
`trolled-release products.
`Different salts of a drug rarely change pharmacol-
`ogy, but only physical properties. This statement has
`been qualified to acknowledge that salts do affect the
`intensity of response. However, the salt form does
`change the physiochemical properties of the drug.
`Changes in dissolution rate and solubility affect the
`rate and extent of absorption (bioavailability), and
`changes on hygroscopicity and stability influence
`formulation.
`Consequently each new drug candidate has to be
`examined to choose the most suitable salt, because
`
`each potential salt will behave differently and require
`separate preformulation screening. The regulatory
`authorities also treat each salt as a different chemical
`entity, particularly in the context of toxicity testing.
`
`Solvents
`It is generally necessary to formulate an injection
`even if there is no intention to market. The first-
`choice solvent is obviously water. However, although
`the drug may be freely soluble, it may be unstable in
`aqueous solution. Chlordiazepoxide HC1 is such an
`example. Accordingly, water-miscible solvents are
`used:
`
`1. in formulations to improve solubility or stability
`2. in analysis to facilitate extraction and separation
`(e.g. 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 solvent solubility and stabil-
`ity are unattainable Table 8.7 shows a range of sol-
`vents to fulfil these needs.
`Aqueous methanol is widely used in HPLC and is
`the standard solvent in sample extraction during
`analysis and stability testing. It is often made acidic
`or alkaline to increase solvent power and ensure con-
`sistent ionic conditions for UV analysis. Other phar-
`maceutical solvents are available 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 (i.e. a partition coefficient (log P >1),
`solubility doubles through this series.
`Where bulk is limited and the aqueous solubility is
`inadequate, it is better to measure the solubility in
`
`Table 8.6 Dissolution rates of weak acids and their sodium salts
`
`Drug
`
`pKa
`
`pH (at Cs)
`
`Dissolution rate (mg cm-2 min-1) x 102
`
`Dissolution media
`
`0.1 MHCI(pH 1.5)
`
`Phosphate (pH 6.8)
`
`Salicylic acid
`Sodium salicylate
`
`Benzoic acid
`Sodium benzoate
`
`Sulphathiazole
`
`Sodium sulphathiazole
`
`118
`
`3.0
`
`4.2
`
`7.3
`
`2.40
`
`8.78
`
`2.88
`
`9.35
`
`4.97
`
`10.75
`
`1.7
`
`1870
`
`2.1
`980
`
`<0.1
`550
`
`27
`
`2500
`
`14
`
`1770
`
`0.5
`810
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 10 of 66
`
`

`

`Table 8.7 Recommended solvents for preformulation screening
`
`PHARMACEUTICAL PREFORMULATION
`
`Solvent
`
`Water
`
`Methanol
`0.1 MHCI(pH 1.1)
`
`0.1 MNaOH(pH 13.1)
`Buffer (pH 6-7)
`
`Ethanol
`
`Propylene glycol
`
`Glycerol
`
`PEG 300 or 400
`
`Dielectric constant (e)
`
`Solubility parameter (8)
`
`Application
`
`80
`
`32
`
`24
`
`32
`
`43
`
`35
`
`24.4
`
`14.7
`
`12.7
`
`12.6
`
`16.5
`
`All
`
`Extraction, separation
`Dissolution (gastric), basic extraction
`
`Acidic extraction
`
`Dissolution (intestinal)
`Formulation
`
`approaches do not allow easy estimates for the
`behaviour of crystalline solids. For a wide range of
`drugs it is possible to relate solvent solubility and the
`partition coefficient (log K^ = log P). Yalkowsky and
`Roseman (1981) derived the following expression
`for 48 drugs in propylene glycol:
`
`Equation 8.4 can be applied more generally by intro-
`ducing a factor </> to account for the relative solvent
`power of pharmaceutical solvents (see Table 8.8 for
`examples).
`For a wide range of solvents Eqn 8.4 now
`becomes:
`
`Methodology and structure activity prediction
`Choice of non-aqueous solvent (oil) The oilrwater
`partition (.K^) is a measure of the relative lipophilic-
`ity (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.
`
`Table 8,8 Solvent power (<f») of some pharmaceutical
`solvents
`
`Solvent
`
`Relative solvent power (4>)
`
`0.5
`
`1 1 2 4
`
`Glycerol
`Propylene glycol
`
`PEG 300 or 400
`
`Ethanol
`
`DNA, DMF
`
`119
`
`aqueous solvent mixtures rather than in a pure
`organic solvent. Whereas solubilities at other levels
`and their mixtures can be predicted, the solubility in
`pure solvent is often inconsistent because of cosol-
`vent effects. Furthermore, formulations rarely use
`pure non-aqueous solvent, particularly injections.
`For example, ethanol should only be used up to 10%
`in an injection to prevent haemolysis and pain at the
`injection site, and include isotonic salts.
`
`Partition coefficient (K°)
`Partition coefficient (the solvent:water quotient of
`drug distribution) has a number of applications
`which are relevant to preformulation:
`1. Solubility: both aqueous and in mixed solvents
`2. Drug absorption in vivo: applied to a
`homologous series for structure activity
`relationships (SAR)
`3. Partition chromatography: choice of column
`(HPLC) or plate (TLC) and choice of mobile
`phase (eluant).
`
`Solvent solubility
`The relative polarities of solvents can be scaled using
`dielectric constant (e), solubility parameter (8),
`interfacial (y) and hydrophilic-lipophilic balance
`(HLB). The best solvent in any given application is
`one whose polarity matches that of the solute; an
`ideal, fully compatible solution exists when Ssolvent =
`Solute- This can be ascertained by determining solu-
`bility maxima, using a substituent contribution
`approach or the dielectric requirement of the system.
`The most useful scale of polarity for a solute is K%,
`(oil:water partition coefficient), as
`the other
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1061, p. 11 of 66
`
`

`

`SCIENTIFIC PRINCIPLES OF DOSAGE FORM DESIGN
`
`Many partition solvents have been used. The largest
`database has been generated using n-octanol. The
`solubility parameter of octanol (8 = 10.24) lies
`midway in the range for drugs (8-12), although
`some non-polar (8 < 7) and polar drugs (8 > 13) are
`encountered. This allows measurable results between
`equal volumes of oil and aqueous phases.
`In the shake flask method the drug, dissolved in
`one of the phases, is shaken with the other partition-
`ing 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 (2G)
`and after partitioning (Cw) [the aqueous concentra-
`tion] to give K^ = CSC - CW)/(CJ.
`If the transfer of solute to the oil phase is small,
`ZiCw is small, and any analytical error increases
`error in the estimate of K°,. Indeed, to encourage
`greater aqueous loss (>ACW~) a considerably more
`polar solvent, w-butanol, has been used. Where the
`partition coefficient is high, it is usual to reduce the
`ratio of the oil phase 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 K* = (10 £C - CW)/CW).
`The partition of a polar solute between an inert
`non-polar hydrocarbon e.g. hexane, and water is quite
`different from that of hydrogen bonding solvents such
`as octanol. The behaviour of the weak acid phenol
`(pKa = 1 0) and weak base nicotine (pKa = 3.1) is
`worthy of note. For phenol, K°aan°l = 29.5, whereas
`^hexane = Q.ll. The acidic solvent chloroform sup-
`presses partition (K° = 2.239), whereas ethyl acetate
`and diethyl ether are more polar. The basic behaviour
`of the solvents give higher K%, values. With solvents
`capable of both hydrogen donation and acceptance
`(octanol, nitrobenzene and oleyl alcohol), K%, is inter-
`mediate. For nicotine the behaviour is reversed, and
`the hydrogen donor (acidic) solvent chloroform parti-
`tions most strongly K%, - 77.63), even though the
`neutral solvent nitrobenzene, which is marginally
`more lipophilic (logP= 1.87 against 1.96 for chloro-
`form), gives similar values for both phenol and nico-
`tine. Clearly both solute and solvent characteristics
`are important.
`In general, polar solvents are advocated to corre-
`late biological activity with physicochemical proper-
`ties. Solvents less polar than octanol, measured by
`water solvency, have been termed hyperdiscriminat-
`ing, whereas more polar solvents such as butanols
`and pentanols, are hypodiscriminating. This concept
`refers to the discriminating power of a partitioning
`solvent within a homologous series. With w-butanol
`
`120
`
`the values of log P tend to be close, whereas with
`heptane and other inert hydrocarbons the differences
`in solute lipophilicities are exaggerated. w-Octanol
`generally gives a range consistent with other physico-
`chemical properties when compared to drug absorp-
`tion in the GI tract. Hyperdiscriminating solvents
`reflect more closely the
`transport across
`the
`blood-brain barrier, whereas hypodiscriminating sol-
`vents give values consistent with buccal absorption
`(Fig. 8.2). In rationalizing the effects of different par-
`titioning solvents, a good correlation was found to
`exist between the solvent water content at saturation
`and solvent lipophilicity.
`Certainly it is imperative to standardize on
`methodology, especially for the solvent. Where solu-
`bility constraints allow, this should be w-octanol,
`especially as the existing data bank is extensive.
`Structure-activity relationships Since the pioneer-
`ing work of Meyer and Overton numerous studies
`on correlating molecular structure and biological
`activity have been reported. These structure-activity
`relationships (SAR) can rationalize drug activity
`and, particularly in modern medicinal chemistry,
`facilitate a scientific approach to the design of more
`effective, elegant structural analogues.
`The application of SAR depends on a sound
`knowledge of the physicochemical properties of each
`new drug candidate in a therapeutic class, and
`preformulation is an essential information source.
`It is assumed in SAR that:
`
`This relationship holds for all polar and semipolar sol-
`vents, but with non-polar solvents (hexane to iso-
`octane) correlations are poor, and this seems to be
`related to water content. Given the importance of
`water, it is imperative that the octanol is saturated
`with the aqueous phase and the aqueous phase with
`octanol prior to any determination, otherwise the par-
`ti

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