throbber
Parenteral Medications
`"OIIIIIIB 1
`Second Edition, Revised and Expanded
`
`Edited by Kenneth E. Avis,
`Herbert A. liebermnn, and lean lnchmun
`
`
`
`
`v a
`
` a
`7.3
`_£__=
`:5
`.§
`
`50
`
`'
`L V_
`\
`L
`
`'—
`
`.
`
`E 40—
`
`_
`
`30L
`
`II—
`
`E
`
`a
`
`C 8
`
`
`
`;
`a
`
`20
`
`1
`
`0o
`
`
`
`_
`
`L
`.
`2
`
`._.
`
`L— .L
`_
`4
`
`i
`
`l
`6
`
`'
`
`l
`
`o
`- 5;
`’1‘
`_ _L
`
`"P W
`
`1
`3
`
`{Ed
`
`
`
`Time (hours)
`
`
`
`AstraZeneca Exhibit 2103 p. 1
`InnoPharma Licensing LLC v. AstraZeneca AB IPR2017—00904
`
`

`

`Parenteral Medications
`ttolumet
`Serond Edition, Revised and Expanded
`
`Edited by
`
`Kenneth E. Avis
`
`The University of Tennessee
`Memphis, Tennessee
`
`Herbert A. liebermnn
`
`H.H. Lieberman Associates, inc.
`Consultant Services
`
`Livingston, New Jersey
`
`leon larhmun
`
`Lachman Consultant Services
`
`Wesrbunt, New York
`
`Marcel Dekker, Inc.
`
`New York I Basel - Hong Kong
`
`AstraZeneca Exhibit 2103 p. 2
`
`

`

`Library of Congress C'atatoging - “1— Publication Data
`
`Pharmaceutical dosage forms, parenteral medications I edited by
`Kenneth E. Avis, Herbert A. Lieberman, and Leon Leohman. -- 2nd ed. .
`rev. and expanded.
`p.
`cm.
`
`Includes bibliographical references and index.
`ISBN 0-3243—8576-2 (v. 1 : elk. paper)
`1. Parenteral solutions.
`2. Pharmaceutical technology.
`Kenneth E.
`II. Lieberman, Herbert A.
`III. Leohman. Leon.
`[DNLM: 1. Infusions. Parenteral.
`WB 354 P536]
`RSZDIJ‘STPéS 1992
`615'. 19--dc20
`DNLMIDLC
`
`2. Technology, Pharmaceutical.
`
`I. Avis,
`
`for Library of Congress
`
`91 -38063
`CIP
`
`This book is printed on acid—free paper.
`
`Copyright © 1992 by MARCEL DEKKER, INC. All Rights Reserved
`
`Neither this book nor any part may be reproduced. or transmitted in any form
`or by any means; electronic or mechanical, includingphotoeopying. micro-
`filming, and recording, at by any information storage and retrieval system,
`without permission in writing from the publisher.
`
`MARCEL DEKJCER, INC.
`270 Madison Avenue, New York, New York 10016
`
`Current printing (last_ digit):
`10 9 S 7 6 5 4 3 2 ]
`
`PRINTED IN TEE UNITED STATES OF AMERICA
`
`AstraZeneca Exhibit 2103 p. 3
`
`

`

`Contents
`
`Preface
`Contributors
`
`Contents of Pharmaceutical Dosage Forms: Parenteral Medications,
`Second Edition, Revised and Expanded, Volumes 2 and 3
`Contents of Pharmaceutical Dosage Forms: Tablets, Second Edition.
`Revised and Expanded, Volumes 1-3
`Contents of Pharmaceutical Dosage Forms: Dispense Systems,
`Volumes 1 and 2
`
`E.E:-
`
`xiii
`
`xv
`
`xvii
`
`Chapter 1 The Parenteral Dosage Form and Its Historical Development
`
`1
`
`Kenneth E. Arts
`
`1. The Dosage Farm
`11.. History of Parenteral Medications
`Appendix A: Glossary of Terms
`Appendix 3': Highlights in the History of
`Parenteral Medications
`References
`
`Chapter 2
`
`Parenteral Drug Administration: Routes. Precautions,
`Problems, Complications, and Drug Bel-Wary Systems
`
`Richard J. Dame. Michael J. Alters. and
`Salvatore J. Tums
`
`Introduction
`I. General Indications for Parenteral
`
`Administration of Drugs
`ll. Pharmaceutical Factors Affecting Parenteral
`Administration
`
`Specific Routes of Administration
`III.
`IV . Distribution of Parenterally Administered Agents
`
`1
`4
`12
`
`19
`15
`
`1'?
`
`1’?
`
`18
`
`19
`
`21
`39
`
`vii
`
`AstraZeneca Exhibit 2103 p. 4
`
`

`

`viii
`
`Contents
`
`V'. Precautions. Problems. Hazards, and
`Complication Associated with Parenteral Drug
`Administration
`
`VI. Methods and Devices for Drug Delivery Systems
`VII.
`Summary
`References
`
`_
`
`Chapter 3 Biopharmaceutics of Injectable Medications
`
`So! Motola
`
`I.
`
`Introduction
`
`II. Physioochemical and Physiological Factors
`Affecting Drug Absorption by Injection: An
`Overview
`
`111. Application of Pharmacokinetics to Biopharmr
`centic Investigations: Pharmacokinetic Models
`"IV. Examples of BiopharmaeeuticlPharmaeokmetio
`Principles
`V. Regulatory Considerations for Bioequivalence
`Studies
`
`VI. Bioequivolence Study of Two Injeotable Forms
`of the Same Drug
`Summary
`References.
`
`VII.
`
`Chapter 4
`
`Preformulation Research of Parenteral Medications
`
`So! Morale and Shreamm N. Aghorkor
`
`I .
`
`Introduction
`
`II. Drug Substance Physieochemical Properties
`III. Accelerated Stability Evaluation
`IV. General Modes of Drug Degradation
`V. Preformulatlon Studies for Proteins and Peptides
`VI.
`Preformulation Screening of Parenteral
`Packaging Components
`Summary
`VII.
`VIII. Preformulation Worksheet
`
`References
`
`Chapter 5
`
`Formulation of Small Volume Parenterals
`
`Patrick P. DeLuca and James C. Boylon
`
`I .
`
`Introduction
`
`Formulation Principles
`II.
`III. Container Effect-e. on Formulation
`1V. Stability Evaluation
`V.
`Process Effects
`References
`
`41
`
`49
`56
`5'?
`
`59
`
`59
`
`60
`
`7'?
`
`9%
`
`108
`
`109
`111
`112
`
`115
`
`115
`
`115
`140
`150
`154
`
`158
`183
`163
`
`169
`
`1'?3
`
`17 3
`
`174
`22?"
`234
`244
`245
`
`AstraZencca Exhibit 2103 p. 5
`
`

`

`Contents
`
`ix
`
`Chapter 6
`
`Formulation of Large Volume Parenterals
`
`Levit J. Demorest and Jeffrey G. Hamilton
`
`I.
`II.
`III.
`IV.
`V.
`
`Introduction ‘
`
`Concepts of Formulation
`Formulation Development
`Sqution Quality
`Summary
`References
`
`Chapter 7
`
`Parenteral Products of Peptides and Proteins
`
`YuHChong John Wang
`
`I.
`II.
`III.
`IV.
`
`V.
`
`Introduction
`
`
`Characteristics of Proteins and Peptides
`Formulation Principles
`Compatibility with Packaging Components and
`Infusion Sets
`Formulation of Market Products
`References
`
`Chapter 8
`
`Sterile
`
`Diagnostics
`
`Leif E.
`
`Olsen
`
`Introduction
`
`II.
`III.
`IV.
`
`VI.
`VII.
`
`Diagnostic Products Defined
`Sterile Diagnostics
`Definitions
`
`Aseptic Manufacturing Considerations
`Validation Program
`Conclusion
`References
`
`Chapter 9
`
`Glass Containers for Parenterals
`
`R. Poul Abendroth and Robert N. Clark
`
`I.
`II.
`III.
`
`IV .
`V .
`VI .
`VII.
`VIII.
`
`Introduction
`The Nature of Glass
`
`United States Pharmacopeia Glassware
`Classifications
`The Manufacture of Glass Containers
`Chemical Performance
`Mechanical Performance
`
`The Container and Closure as a System
`Quality Assurance
`References
`
`249
`
`249
`250
`273
`280
`281
`281
`
`283
`
`283
`284
`302
`
`310
`312
`317
`
`321
`
`323.
`321
`322
`325
`330
`351
`359
`359
`
`351
`
`381
`381
`
`362
`369
`375
`380
`380
`382
`384
`
`AstraZeneca Exhibit 2103 p. 6
`
`

`

`a:
`
`Contents
`
`Chapter 10 Use of Plastics for Parenteral Packaging
`
`John M. Anes. Robert S. Nose, and
`Charles H. White
`
`I.
`II.
`III.
`IV.
`V.
`VI.
`
`Introduction
`Fundamentals
`Fabrication Processes
`
`Important Criteria for Selection of Plastics
`Plastics Used in Parenteral Packaging
`Quality Assurance of Parenteral Containers
`References
`
`Chapter 11 Elastomeric Closures for Parenterals
`
`Edward J. Smith and Robert J. Nash
`
`II
`
`II.
`III.
`IV.
`V.
`VI .
`VII.
`VIII.
`IX .
`
`XI.
`
`Elaetomeric Parenteral Packaging Components:
`A Physical Description
`Physical Description of Rubber
`Types of Rubber Used in Parenteral Packaging
`Closure Design
`Rubber Compdunding
`Vulcanization Process
`Closure Manufacture and Control
`
`Closure Design Qualification
`Regulatory Considerations
`Interaction of Drug Formnlations with
`Rubber Closures
`
`Contemporary Closure-Related Issues
`References
`
`Chapter 12 Parenteral Products in Hospital and Home Care
`Pharmacy Practice
`
`John W. Levchuk
`
`1.
`II.
`
`III.
`IV.
`
`V.
`VI.
`VII.
`
`Introduction
`
`The Preparation of Sterile Dosage Forms in the
`Hospital and in Home Care
`Dispensing and Compounding Processes
`Technology of Sterile Compounding in the
`Hospital Pharmacy
`Clinical Supply and Use of Sterile Products
`Quality Assurance
`Conclusion
`
`Appendix: Abbreviated Sequence for Preparing a
`Series of Extemporaneously Compounded LV.
`Admixtures
`References
`
`Index
`
`387
`
`387
`. 389
`398
`407
`422
`439
`443
`
`445
`
`445
`450
`451
`462
`463
`470
`477
`494
`503
`
`505
`50'?
`503
`
`513
`
`513
`
`513
`524
`
`532
`547
`552
`562
`
`56 3
`566
`
`569
`
`AstraZencca Exhibit 2103 p. 7
`
`

`

`4 P
`
`reformulation Research of
`Parenteral Medications
`
`Sol Motola"
`
`Whitehall Laboratories, Hammonton, New Jersey
`
`Shreeram N. Agharkar
`
`Bristol—Myers Squibb Company. Syracuse. New York
`
`I.
`
`INTRODUCTION
`
`Preformulaiion research relates to pharmaceutical and analytical investigations
`that both pressed and so
`.
`. ormu a on eve went e
`a
`r
`sage
`
`forms. Expenmen s are designed to generate
`a a
`aracterizing speci c,
`pharmaceutically important. physicochemical preperties of the drug substance
`and its combination with selected solvents, excipients, and packaging com—
`ponents. These studies are carried out under stressed conditions of tempera-
`ture, light, humidity, and oxygen in order to accelerate and detect potential
`reactions. Taking into account early pharmacological and biopharmaceutlcal
`data, preformulation studies yield key information necessary to guide the term—
`ulator and analyst toward the development of an elegant, stable dosage form
`with good bioavailability. Prior to development of the clinical and marketed
`dosage form, preformulation studies yield basic knowledge necessary to de-
`velop suitable formulations for toxicological use.
`Due to important research leads in a highly competitive field, rapid pro—
`gress is essential, and clinical studies should be initiated as soon as possible.
`Thus an expeditious preformulation program {i.e. , one typically taking 6 to
`10 weeks to complete) is generally required.
`If clinical program acceleration
`is desired, it may be necessary to streamline studies and develop crucial de—
`cision-making data in shorter time periods. Should interim results indicate
`that a more stable or more soluble drug form is needed, expansion of the
`original program will be necessary. Additionally, areas of particular interest
`may arise, such as the elucidation of a reaction mechanism or the investigation
`of unusual solubility phenomena. Such studies may be of prime importance
`and are often addressed either initially or as second-phase preformulation
`studies, depending on their potential impact on the overall program.
`
`*C'ur'r'ent affiliation: Wyeth-Ayerst Laboratories, Radnor, Pennsylvania
`
`115
`
`AstraZeneca Exhibit 2103 p. 8
`
`

`

`115
`
`Metals and Aghar'lcar
`
`The general subject of preformuletion research has been described in de-
`tail by several investigators [1- 5] and is in wide use throughout the pharma-
`ceutical industry. These presentations have dealt mainly with studies designed
`for solid dosage forms. Some specific applications have been made to certain
`areas of parenteral interest [6,7] . The objective of this chapter is to outline
`methods used in developing preformuletion data necessary to characterize
`significant physicochemical properties of new drugs important to a paren-
`teral formulation development program.
`
`ll. DRUG SUBSTANCE PHYSICOCHEMICAL PROPERTIES
`
`Typical physicochemical properties of drug substances that either characterize
`or may exert significant influence on the development of a parenteral formula-
`tion are listed in Table 1.
`
`A. Molecular structure and Weight
`
`These are the most basic characteristics of. a drug ubstance and are among
`the first items to be known. From the molecular structure the investigator
`can make initial judgments regarding potential properties and functional group-
`reactivities, as described in Section IV.
`
`B . Color
`
`Color is generally a function of a drug's inherent chemical structure relating to
`a certain leVel of unsaturation. Color intensity relates to the extent of conju—
`gated unsaturation as Well as the presence of chromophores such as ~NH2, -N03,
`and -CO- (ketonel, which intensify color. Some compounds may appear to have
`color although structurally saturated. Such a phenomenon can often be due to
`the presence of minute traces of highly unsaturated, intensely colored. impurities
`andior degradation products. Thesse substances may be prone to increased
`color formation under stress conditions of heat, oxygen. and light. A signifi-
`cant color change can become a limiting factor to the shelf life of s parenteral
`product even before a significant change in chemical stability is noted.
`
`Table l Physicochemical Properties of Drug Substances
`
`Molecular structure and weight
`
`Solubility
`
`Color
`
`Odor
`
`pH solubith profile
`
`Polymorphism potential
`
`Melting point
`
`Solvate formation
`
`Thermal analytical profile
`
`Absorbance spectra
`
`Particle size and shape
`
`Light stability
`
`Hygrosco'picity potential
`
`Thermal stability
`
`Ionization constant
`
`pH stability profile
`
`Optical activity
`
`Asnaancca Exhibit 2103 p. 9
`
`

`

`Pre formulation Research
`
`1 1 ‘7
`
`The drug substances color should he recorded by s subjective description,
`as well as by an objective means such as by comparison with standard color
`chips [8] , or by spectrophotometric analysis if the compound‘s color intensity
`in solution is proportinsl to concentration. The American Public Health Assn“
`cistion (APHA) color standards [9} can be used effectively to quantitate
`changes in solution color with time. Visible absorbsnce of APHA color stand~
`srds (diluted appropriately) can be measured spectrophotometrically {10]
`to
`
`monitor more accurately the color of solution test samples. An example plot
`
`is shown in figure 1.
`
`Ci Odor
`
`The odor of a new drug substance should be examined by cautiously smelling
`the headspsce of the drug container which has been previously closed to allow
`volatiles to concentrate. The presence and description of any odor should be
`recorded. The substance may exhibit an inherent odor characteristic of major
`functional groups present (i.e. , sulfurcus or garlic—like for sulfides, sulfoxides,
`or sulfhydryl-contsining compounds, or smmoniacsl as for amines). Alternaw
`tively, a drug may be void of characteristic odor or it may have an odor of
`residual solvent. The presence of a solvent odor should be reviewod with the
`synthesis chemist to determine whether the sample has been sdeQustely dried.
`
`D. Particle Size, Shape, and Crystallinity
`
`The particle size of s weter~soluble drug is not of concern unless it exists in
`large aggregates and an increase in rate of solution is desired to reduce menu-
`factoring time. Under such circumstances milling through on appropriate size
`sieve [11] may be sufficient.
`
`soc
`
`37.5
`
`b
`'U
`I
`b
`
`n
`250 e
`fl
`
`U’
`c
`
`"
`
`125
`
`D
`
`2O
`
`4O
`
`60
`
`30
`
`100
`
`% Certified APHA Color S1ancsrc (500 units)
`
`0.4
`
`0.3
`
`EC
`
`Q g
`
`..
`a:
`.-..
`E
`
`"j
`v 02
`3
`
`5.E
`
`D
`
`3 0.1
`q
`
`0
`
`0
`
`
`Figure 1 Typical visible absorbsnce readings at 4100 nm for certified APHA
`color standard (500 units) at various dilutions versus APHA color number.
`
`AstraZeneca Exhibit 2103 p. 10
`
`

`

`ll 8
`
`Mo talc and A ghorkor
`
`Particle size and shape characteristics can be determined by microscopic
`evaluation using either an optical adorescope, preferably with polarizing at—
`tachments, or by a scanning electron microscope. The morphological charac-
`teristics of the drug substance should be recorded either by a sketch or,
`more accurately. by a photomicrograph which acts as a permanent record for
`comparison with future batches. A good estimate of particle size and particle
`size range can be obtained by viewing several fields of a representative sam-
`ple of drug- substance.
`A polarizing microscope is also used to determine whether a compound is
`crystalline or amorphous. Crystalline materials refract polarized light and are
`thus visible when polarization attachments in the ocular and. objective are
`crossed at a 90° angle (crossed polars”) , whereas amorphous or glassy- sub-
`stances become invisible.
`
`Optical microscopes usually operate at useful magnifications of up to illllthE
`with a resolution limit in the vicinity of 1 mo. The scanning electron micro-
`scope provides magmfications up to 200,000! with a resolution of approximate-
`ly 25 3 to determine detailed particle surface morphology as well as. individ-
`ual particle surface characteristics. Whereas the optical microscope provides
`only a two-dimensional View, the seaming electron microscope adds the dimen-
`sion of depth by tilting- the stage to several angles of view during operation.
`Thus what may appear to be a combination of acicular and plate-shaped struc-
`tures under a polarizing microscope could in reality only be a field of flat.
`plates of various sizes with some on edge, as shown in Figure 2. This ability
`to resolve various shapes helps the investigator determine whether a sample
`is morphologically homogencOus. Mixtures of morphological forms either within
`a sample or between samples could indicate the existence of hydrates, solvstes,
`or polymorphic forms which could later significantly affect properties such as
`solubility, stability. and bioaVaflability.
`
`E . Melting Point
`
`The melting point of a substance is thermodynamically defined as the tempera-
`ture at which the solid and liquid phases are in equilibrium as described in
`Equation ( 1).
`
`ssolid r—Jsliquid
`
`m
`
`A melting-point determination is a good first indicatiOn of purity since the
`presence of relatively small amounts of impurity can be detected by a lowering
`as Well as widening in the melting-point range. Methods for detErmining the
`melting- range or temperature are described in detail for various compounds
`[12] . Any peculiar behavior of a substance undergoing melting, such as dra-
`matic change in volume, melting and recrystallization, gas evolution. color
`change, or other physical change, should he recorded and investigated fur-
`ther. Such behavior could be indicative of significant changes, such as a
`polymorphic transition, desolvation, oxidation, or decarboxylation.
`
`F. Thermal Analytical Profile
`
`During- synthesis and isolation. a sample may have been exposed to changes.
`in the temperature environment which may be exhibited as a thermal profile
`
`AstIaZeneca Exhibit 2103 p. 1 I
`
`

`

`Preformulotion Research.
`
`119
`
`when the sample is heated between ambient temperature and its melting point.
`When no thermal history exists, the sample will neither absorb nor give off
`heat prior to its melting point. The basic technique used to study this phe-
`nomenon is called differential thermal analysis (DTA). Essentially, the sample
`is heated in the presence of a sensitive thermocouple while a second, balanced
`thermocouple electrically connected in series opposition is heated at the same
`rate in the presence of an inert reference material. The reference substance
`is one that does not undergo thermal transition within the range of tempera-
`tures to be used. The data are plotted on rectilinear paper with the ordinate
`equal to the difference in temperature between sample and reference, AT,
`and the abscissa equal to the temperature T . Although differences in conven-
`tion exist, this treatment will indicate exothermic reactions above and endo-
`thermic reactions below the baseline.
`If a flat signal results. no differential
`change in temperature (aT) occurred between sample and reference. When a
`sample shows a defined exotherm (heat liberation) 0r endotherm (heat absorp-
`tion) resulting- from physical or chemical changes as a function of temperature,
`these phenomena are indicative of phase transitions. Examples of character-
`istic endothermic transitions that can be detected by this technique are:
`fu-
`sion or melting, crystalline structure changes such as polymorphic transitions,
`sublimation. boiling, and desolvation. An. exothermic effect is seen when crys-
`tallization occurs. Examples of thermal analysis profiles of melts with and
`without decomposition are. shown in Figures 3 and 4. Decomposition upon melt-
`ing is noted when the signal drops below the original baseline following the
`melting endotherm.
`A similar process which examines this phenomena is called differential
`Scanning calorimetry (DSC) . With this technique, the area under the output
`curve is directly proportional to the total amount of energy (q) absorbed or
`liberated from the sample. The abscissa is proportional to the rate of heat
`transfer (dqldt) at any given time. The loss of surface moisture and a de-
`composition melt is evident in the DEC curve shown in Figure 5.
`The thermal analytical method used to detect the existence and stability
`of solvated drug molecules is called thermogrovimetric analysis (TGA).
`In
`this technique relative weight loss is studied between the sample and a refer-
`ence during the heating cycle. The reference chosen is one known not to
`undergo weight loss over the temperature range desired. Weight loss can
`occur as. a result of the loss of sample surface moisture or by molecular desol-
`vation or decomposition. Combined DTA and TGA curves for amphotericin B
`showing intermediate phase transition (DTA) , weight loss, and decomposi-
`tion (TGA) are shown in Figure 6. An overview of thermal analytical methods
`described above is available for further information [17] .
`
`G . Hygroscopicity
`
`A compound is hygroscopic if it picks up a significant amount of moisture
`under a specific condition of temperature and humidity. A high degree of
`hygroscopicity may adverser affect the physical and chemical properties of
`a drug substance, making it either pharmaceutically difficult or unsatisfactory
`to work with.
`
`Hygroscopicity studies are usually carried out over a range of humidity
`conditions relevant to the general laboratory and manufacturing areas as" well
`as uncontrolled storage environment. A low-humidity condition can be used
`
`Asuacheca Exhibit 2103 p. 12
`
`

`

`120
`
`Motels and Aghcrkar
`
`
`
`Figure 2 Microscopic examination of an experimental drug. Views through
`an optical microscope (a) and a scanning electron microscope (b) at various
`stage angles as noted.
`(Courtesy of E. B. Vedas, Merck Frosst Canada. Inc. .
`Pointe Claire-Dowel, Quebec, Canada.)
`
`to determine whether a hydrate will lose water under such storage. Saturated
`solutions of certain salts stored in sealed containers, such as desiccators,
`are used to establish well- defined humidity conditions; examples are shown
`in Table 2.
`
`To carry Out a study. samples of compound are accurately weighed into
`tared containers and placed at various humidity conditions for periods up to
`2 weeks. Weight gain or loss is measured at predetermined intervals until
`equilibrium is reached. An assessment is made regarding the relative weight
`gain as well as color and general flowability. Chemical analysis is often per-
`formed should physical change indicate possible chemical degradation. A hy-
`groscopicity classification is shown in Table 3. Thus from hygroscopicity
`studies the investigator can determine environmental humidity conditions neces—
`sary to maintain initial properties.
`If the drug is very hygroscopic or deter-
`mined to be unstable in the presence of moisture, the drug would have to be
`stored under dry conditions and worked with under low humidity. Close ex—
`amination of the quantity of moisture gained during these experiments is in“
`portant in determining whether hydrate formation is occurring.
`
`AstraZeneca Exhibit 2103 p. 13
`
`

`

`

`

`Match: and Agharkm-
`
`H CONH—Q—Cfla
`’i
`2
`
`'
`
`50
`
`?5
`
`103
`
`125
`
`1
`
`US
`
`200
`
`225
`
`It
`
`122
`
`END!)—0-
`
`«Ir—HO
`
`Figure 3 Structure and differential thermal analysis scan of ergonovine maleate
`with decomposition (USP reference standard, lot L).
`(From Ref. 13.)
`
`cm3
`
`Cl
`
`93"
`
`"92'
`
`
`
`Figure It Structure and differential thermal analysis scan of ketamine.
`Ref. 14.)
`
`("From
`
`AstraZer-leca Exhibit 2103 p. 15
`
`

`

`Preformulatian Research
`
`123
`
`Rmnwaz
`0
`
`0
`
`NH?
`
`NH?
`
`/§Z;§/NH:2
`
`H0
`
`0
`
`0
`
`TEMPERATURE
`
`
`
`
`
`ENDOTHERM‘—‘5—a-EXGTHERM
`
`0
`
`100
`
`200
`
`300
`
`400
`
`TEMPERATURE,
`
`0(3
`
`Figure 5 Structure sand, differential scanning calorimetry scan 0f gentamicin
`sulfate (USP reference standard).
`(From Ref. 15.)
`
`mculr’sec
`
`
`
`1mm
`
`---I—--
`
`'
`
`I
`210
`
`90%
`
`36%
`
`TGA
`
`AMPHQTERICfN B
`
`o
`
`HQ
`
`d0
`
`30
`
`3 20
`
`160
`
`200
`
`240
`
`280
`
`TEMPERATURE 1
`
`“C
`
`Figure 5 Structure, differential thermal analysis, and thermogravimetric anal~
`ysis scan of amphotericin 13.
`(From Ref. 16.)
`
`AstraZeneca Exhibit 2103 p. 16
`
`

`

`1 24
`
`Mo talc and A gh crkor
`
`Table 2 Saturated Salt Solutions for Humidity Control
`
`Percent relative
`Temperature
`
`humidity
`(0C)
`
`Potassium acetate, Kc2H302
`
`Calcium chloride, CsClz-EHZO
`Potassium thiocysnsts, KSCN
`
`Sodium nitrite, NsN02
`
`Sodium acetate, Ns02H302-3H20
`
`20
`
`31
`47
`
`56
`
`76
`
`20
`
`24,5
`20
`
`20
`
`20
`
`
`
`90Zinc sulfate, AnSOévTHZO 20
`
`
`
`Source: Ref. 18.
`
`H . Absorbsnce Spectra
`
`Molecules with structural unsuturation are solo to absorb light within a spo~
`cific frequency range. As mentioned previously, the degree of unsaturation
`coupled with the presence of chromophnrss will influence the extent of absorp-
`tion and whether ultraviolet (409-190 nm) or visible (800400 nm) light will
`be ubsorbccl. The ultraviolet and Visible spectra of compounds in soiution
`or not highly specific; however, they are very suitable for quantitative ans“
`lyticsl work and serve as additional information for compound identification.
`The ultraviolet or visible spoctrum can be oetsrmined by placing approximate-
`ly s 10 to 20 pg ml‘1 solution of the compound in a 1 cm cell, and recording
`the spectrum versus the appropriate solvent blank in the spectral range 190
`to 800 nm. An example of the ultraviolet spectrum of ohlorothiazicls is shown
`in Figure 7. Absorbsncc maxima are evident at 228, 292, and 31!] nm.
`
`Tobie 3 Hygroscopicity Classification
`
`Class IMNonhygroscopic: Essentially no moisture increases occur at relativo
`humiditics below 90%. Furthermore, the increase in moisture content after
`
`storage for 1 week above 90% relative humidity (RH) is less than 20%.
`
`Class IIwSlightly hygroscopic: Essentially no moisture increases occur at
`relative humiditiss below 80%. The increase in moisture content after storage
`for 1 week above 80% RH is less than 40%.
`
`Class Iilwldoderctcly hygroscopic: Moisture content does not increase above
`5% after storage at relative humiditiss below 60%. The increase in moisture
`content after storage for 1 week above 80% RH is less than 50%.
`
`Gloss I V-nVsry hygroscopic: Moisture increase may occur at relative humidi—
`tics as low ss 40 to 509*. The increase in moisture content after storage for
`1 week above 90% RH may exceed 30%.
`
`Source: Ref. 1 9.
`
`AstraZeneca Exhibit 2103 p. 17
`
`

`

`Preformulction Research
`
`125
`
`ABSOEBANCE
`
`220
`
`250
`
`300-
`
`340
`
`WAVELENGTH. nm
`
`Figure 7 Structure and ultraviolet absorbsnce spectrum of chlorothazide.
`(From Ref. 21.)
`
`Relationships- used to compare and quantitate ultraviolet and visible ab-
`sorbance of compounds in solution are shown in Equations (2) and (3).
`
`A
`“Fe.
`
`__A..
`“be
`
`(2)
`
`(3)
`
`In Equation (2) , the quantity :1, called absorptivity. is related to the absorp—
`tion of a compomd of concentration c (in g! 1000 1111) through a sample cell of
`in centimeters and thus has the units liters per gram-centimeter. When the
`concentration is expressed in moles per liter, the absorptivity becomes molar
`ebsorptivity e, and is expressed in leters per mole centimeter.
`Both values should be recorded for each solvent system of interest. For
`quantitative use. either a or e is determined. For a compound in solution the
`molar concentration of an unknown quantity of the same drug can be deter-
`mined by knowing- the absorbence at the wavelength and the cell path length.
`Real-ranging Equation (3) yields Equation (4) .
`
`—_-*}_.
`c—be
`
`(4)
`
`The infrared (IR) spectrum (run between 2.5 and 15 pm) is highly specific
`for each chemical structure, with small structural differences resulting in
`significant spectral changes. Samples can be prepared as a solution. as a
`dispersion in mineral oil (Nujol mull), cr'as a potassium bromide (KB-r) pellet.
`
`Astra-Zeneca Exhibit 2103 p. 18
`
`

`

`12:3
`
`2.5
`100
`
`microns
`
`Motels and Agharkar
`
`3.0
`
`4,0
`
`5.0
`
`6.0
`
`7.0
`
`5.0 9.010
`
`12 I4
`
`80
`
`a O
`
`{L O
`
`M O
`
`Transmittance
`
`accnJ
`
`coma
`
`0
`
`CH,OCCH3
`
`4000
`
`350.0
`
`3000
`
`2500
`
`2000 1800
`
`1600
`
`1400
`
`1200 1000
`
`000 850
`
`Wavenurnber (cl-n")
`
`Figure 8 Structure and infrared spectrum of cefotaxime- in KBr with peaks of
`several functional groups identified.
`(Adapted from Ref. 22.)
`
`After running a spectrum. significant peaks relating to major functional groups
`are identified; spectra of subsequent samples of the same compound are com-
`pared with the original.
`If IR spectral differences are found. the reason for
`and source of change should be investigated. This technique is used to de-
`tect batch-to-batch variations, as an identity test, and for the detection of
`polymorphs and solvates. KBr and Nujol mull spectra of cefotaxime acid are
`shown in Figures 8 and 9, respectively. Peaks corresponding to various func-
`tional groups are identified by letters a—j on the KBr spectrum with assigned
`frequencies listed in Table 4.
`
`I. Solubility
`
`Solubility is of prime importance for developing solutions that can be injected
`either intravenously or intramuscularly.
`In general, solubility is a function
`of chemical structure: salts of acids or bases represent the class of drugs
`having the best chance of attaining- the degree of water solubility desired.
`Other compound classes, either neutral molecules or very weak acid and bases
`which cannot be solubilized in water within the desired pH range, may require
`the use of nonaqueous solvents. A list of such solvents used for solubility
`studies as well as eventual use in products is shOWn in Table 5.
`
`Solubility Measurement
`
`The analytical method used in obtaining solubility measurements may vary
`according to the drug moiety.
`If the drug‘s. structure has unsaturated conju-
`gation, enabling it to absorb Visible or ultraviolet light, spectrophotometric
`analysis can he performed. A predetermined excess of drug is placed into
`suitable ampuls (or flasks) containing a small volume (2-5 ml) for each solvent
`
`AsUacheca Exhibit 2103 p. 19
`
`

`

`microns
`
`1
`
`6.0
`7““
`
`7.0
`1
`
`8.0 9.010
`‘1“"1'7
`
`12
`
`14
`
`2.5
`100
`
`3.0
`
`4.0
`-r-—-
`
`5.0
`“1—”
`
`
`
`Transmittance
`
`4000
`
`3500
`
`3000
`
`2500
`
`2000 1000
`
`1600
`
`1400
`
`1200
`
`1000
`
`800 050
`
`Wavenumber (cm“)
`
`Figure 9
`22.)
`
`Infrared spectrum 0f eefotaxime in Nujol mull.
`
`(Adapted from Ref.
`
`Table 1} Infrared Frequency Assignments for Cefotaxime
`
` Frequency (cm'l)
`
`Assignment
`
`0
`
`b
`c
`
`d
`
`0
`
`f
`
`g
`
`11
`i
`
`j
`
`3420
`
`WNHZ
`
`3340 (broad)
`
`—NH, —NI—I2
`
`2940
`1760
`
`1730
`
`1050
`
`1620
`
`1540
`
`“SHOE-12
`—C=O lactam
`
`0
`
`"CEO carboxylie, DwngHB
`
`O
`
`—y3--NH2
`
`0
`—ngH, owN—, "Cmc—
`0
`—g—-N—
`
`13851355
`1180
`
`—O-C0-—CH3
`0:0 in ester
`
`CMO stretching
`1050
`
`Adapted from Ref. 22.
`
`12?
`
`AstraZeneca Exhibit 2103 p. 20
`
`

`

`128
`
`Motoch and Aghorkcr
`
`Table 5 Examples of Nonaqueous
`Solvents Used in the Parenteral
`Product Formulation
`
`Polyethylene glycol 400 and 600
`
`Propylene glycol
`
`Glycerin
`
`Ethyl alcohol
`
`Fixed oils
`
`Ethyl oleate
`
`Benzyl bensoate
`
`Source: Ref. 20.
`
`tested. The ampuls are sealed and placed on a suitable shaker or rotator at a
`controlled temperature (mg. . 25 or 37°C) for several days to attain equilib-
`rium. At selected time internals samples are withdrawn by an appropriate
`means (syringe. pipet), filtered through a 5111311 micrometer-size (example,
`0. 2—0.45 1.1m) filter, and analyzed for drug in solution using the appropriate
`ultraviolet or visible assay methodology. For example, the absorbanoe is read
`versus a solvent blank at a predetermined. Wavelength. Using the appropriate
`Beer's law reference curve as shown in Figure in, the concentration is either
`estimated from the curve or can be calculated using a previously determined
`molecular absorptivity.
`Solubility determination of compounds that do not absorb ultraviolet or
`visible light can be attempted by transferring filtered aliquot solutions onto
`previously tared weighing- pans, evaporating the solvent, and drying to con-
`stant weight under low-temperature conditions.
`
`Due to limits in the amount of new drugs available at the first stages of
`preformulation studies. it is sufficient to determine approximate solubility
`values for highly- solu‘ole compounds.
`In such cases a minimal volume of solv-
`ent is used and fixed amounts of drug added (Le. , 150 lug—1 m1 of solvent).
`Should this still yield an unsaturated solution, a value (3.3-. , >15%J will be
`sufficient to denote high solubility at this stage. Equilibrium solubility can be
`determined when more compound is available, if important for a particular
`solvent.
`
`It is also very important to run solubility determinations. at refrigeration
`temperature (2- 8°C) using solvents demonstrating a high potential for use in
`formulation studies. This is done to establish the range of concentration usable
`within the range 2 to 25°C without risking saturation and crystal growth dur--
`ing stability studies.
`
`pH—Solubility Profile
`
`Compounds. with either acidic or basic functionality will show differences in
`solubility characteristics with changes in soluticIn pH in accord with their ioni—
`zation constants. These differences are often large and important in attaining
`
`Astra-Zoneca Exhibit 2103 p. 21
`
`

`

`
`
`Prefomnulation Research
`
`129
`
`
`
`Absorbance of sample
`
`.
`
`Conaentraunn of sanpie
`
`1.0
`
`0.8
`
`0.6
`
`Absorbance 0.4
`
`0.2
`
`0
`
`0,2
`
`(3.4
`
`0.6
`
`0.8
`
`1.0
`
`Concentration
`
`Figure 10 Hypothetical Beer's law curve relating abaorbance and concentration.
`
`the concentrations desired for formulations. pH~solubility prafiles can be
`established by running equilibrium selubflit'y experiments within the range
`3 t0 4 pH units can both sides of the pKa or gal-(El.
`The relationship between solubility of an acidic drug and pH can be de-
`fined with

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