throbber
.AM
`
`.hxE.AIn
`B.m
`MAIA V. BRACCO
`
`IPR PETITION
`
`
`
`
`MAIA Exhibit 1013
`MAIA V. BRACCO
`IPR PETITION
`
`

`

`Pharmaceutical Formulation
`Development of Peptides
`and Proteins
`
`Edited by
`SVEN FROKJAER AND LARS HOVGAARD
`
`
`
`
`

`

`First published 2000 by Taylor & Francis
`I I New Feller Lane, London EC4P 4EE
`
`Simultaneously published in the USA and Canada
`by Taylor & f-rancis Inc
`325 Chestnut Street, 8'h r-loor, Philadelphia P /\ 19 106
`
`Ta_1•/o!-_i\Frw1l'is is a11 i11111ri11t o( the foy/or & Fra11cis Gro1111
`.·'
`(© 200(/lor & Francis Limited
`Typeset in Times by Graphieraft Limited, 1 long Kong
`Printed and bound in Great Britain by TJ International Ltd, l'adstow, Cornwall
`
`/\II rights reserved. No part of this book may be reprinted or
`reproduced or utilised in any form or by any electronic, mechanical,
`or other means, now known or hereafter invented, including
`photocopying and recording, or in any information storage or
`retrieval system, without permission in writing from the publishers.
`
`Every effort has been made to ensure tlwt the advice and
`information in this book is true and accurate at the time or going to
`press. I lowevcr, neither thr.: publisher nor the authors can accept
`:my legal responsibility or liability for any errors or omission:-; that
`may be mallc. In the casr.: or drug a<lministrntion, any mr.:dic,11
`procedure or the use or technical equipment mentioned within this
`book, you arc strongly advised to consult the manufoctun:rs'
`guidelines.
`
`British l,ihrC11y Cawlog11i11g in />11hlicatio11 /)ata
`/\ catalogue record for this book is available from the British Library
`
`/,ih,w:I' ii/" Co11grr1ss Catalogi11g ill /'11hlirntio11 Data
`/\ catalog record for this book has been requested
`
`ISBN 0-748-40745-6
`
`
`
`
`

`

`Contents
`
`List offig11rcs
`List of tahh·s
`Confrihutors
`Pre/i1ce
`
`Peptide Synthesis
`
`Bernard A. Moss
`
`Introduction
`1.1
`1.2 Chemical synthesis of peptides
`1.2.1 Solution and solid phase peptide synthesis
`1.2.2 Large-scale peptide synthesis
`1.3 Concluding remarks
`Relcrenccs and additional sources
`
`,-': .,•'
`
`2 Basics in Rccomhinant DNA T echnology
`
`/
`
`Nanni Din and Jan Enghcrg
`
`~ ----
`'
`- -1-
`..:.-1.
`~ _i '
`
`\ :
`'T
`
`2.1
`2.2
`
`2.3
`
`2.4
`
`Introduction
`General methods in gene technology
`2.2.1 DNA cloning tools
`2.2.2 Cloning of cDNA
`2.2.3 PCR cloning and DNA database mining
`Expression of recombinant proteins
`2.3. 1 Transcription. translation amt protein
`modi !ications
`2.3.2 Choice of expression system
`Protein design
`2.4. 1 Protein variants
`2.4.2 Protei n chimeras
`2.4.3 Epitopc display libraries
`
`This.mat@ria l w as.<opi@d
`at the NLM and m a y b-e
`Subject US-Copyright Lilw.s
`
`page xi
`XIII
`xv
`xvii
`
`I
`2
`4
`6
`10
`10
`
`12
`
`12
`13
`13
`15
`16
`18
`
`18
`19
`22
`22
`24
`24
`
`V
`
`
`
`
`

`

`Vl
`
`Co11te111.,·
`
`2.5 Recombinant protein therapeutics - status and future trends
`References
`
`3 Protein Purification
`Lars I lovgaard, Lars Skrivcr and Sven Prokjacr
`3.1
`Introduction
`3.2
`f-ractionation strategics
`3.2.1
`Initial fractionation step
`3.2.2
`Intermediate purification step
`3.2.3 Final polishing step
`3.2.4 The 11nished product
`3.3 Protein stability in downstream processing
`3.3.1 Protein conformation stability
`3.3.2 Protein instability
`3.3.3 Essential process-related parameters
`References
`
`4 Peptide and Protein Characterization
`Miroslav Baudys and Sung Wan Kim
`4.1
`Introduction
`4.2 Chromatogrnphy
`4.2.1 Reversed phase chromatography
`4.2.2
`I lydrophobic interaction chromatography
`4.2.3
`Ion-exchange chromatography
`4.2.4 Size-exclusion chromatography
`4.3 Electrophoresis
`4.3. I Gel electrophoresis
`4.3.2 Two-dimensional gel electrophoresis
`4.3.3 Capillary electrophoresis
`4.4 Structural characterization
`4.4.1 Primary structure
`4.4.2 Mass spectrometry
`4.5 Secondary and tertiary structure
`4.5.1 Absorption and fluorescence spectroscopy
`4.5.2 Circular dichroism spectroscopy
`4.5.3
`Infrared spectroscopy
`4.5.4 Other methods
`4.(> Conclusion
`References
`
`5 Chemical Pathways of Peptide and Protein Degradation
`Chimanlall Goolclrnrran, Mehrnaz Khossravi and
`Roarnlcl T. Borchardt
`5.1
`Introduction
`5.2 1 lydrolytic pathways
`5.2.1 Deamidation or Asn and Gin residues
`5.2.2 Degradation or Asp residues
`
`This materlsl wascoDied
`
`25
`27
`
`29
`
`29
`30
`30
`31
`32
`32
`33
`33
`34
`37
`38
`
`41
`
`41
`43
`43
`46
`47
`47
`48
`48
`50
`50
`51
`52
`53
`55
`56
`57
`58
`59
`60
`60
`
`70
`
`70
`71
`71
`75
`
`
`
`
`

`

`Confellfs
`
`5.2.3 Degradation of N-terminal sequences containing
`rcnultimate Pro residues via dikctopircrazine formation
`5.3 Oxidation pathways
`5.3.1 Autooxidation
`5.3.2 Metal-catalysed oxidation
`5.3.3 Photooxidation
`5.3.4 Strategies to prevent oxidation
`5.4 Other chemical pathways
`r3-Elimination reactions
`5.4.1
`5.4.2 Disulphide exchange reactions
`5.5 Conclusion
`References
`
`6 Physical Stability of Proteins
`.Jens Brangc
`
`I ntro<luction
`6.1
`(i.2 Protein structure
`(>.2.1 Stabilizing interactions
`6.2.2 Role of water in structure and stability
`6.3 Protein destabilization (denaturation)
`6.3.1 Unfolding intermediates (molten globule)
`6.3.2 Temperature-induced changes
`Influence or pl I
`6.3.3
`lnnuencc of pressure
`6.3.4
`6.4 Aggregation and precipitation
`6.4.1 Mechanisms of aggregation
`6.4.2 Precipitation and fibrillation phenomena
`6.4.3 Factors inllllCllcing nggregation and precipiWtion
`6.5 Surface adsorption
`6.6 Solid phase stabi lity
`6.6.1 Lyophil ization-induccd aggregation
`(1.7 Stabilization of protein drugs
`6.7. 1 Stabilization strategics
`References
`
`7 Peptides and Proteins as P;1renteral Suspensions: an Overview
`of Design, Development, mul Man11facl11ring Considerations
`
`Michael R. DeFclippis and Michael J. Akers
`
`7. I
`7.2
`7.3
`
`Introduction and scope
`Rationale for suspension development
`Types of suspensions and particle formation
`In sif11 particle formation
`7.3.1
`7.3.2 Combination of particles and vehicle
`Excipient selection
`7.4
`7 .5 General requirements for suspension products
`Testing and optimiz:1tion of chemical, physical, and
`7.6
`microbiological properties
`
`vii
`
`78
`79
`80
`80
`82
`83
`84
`84
`84
`85
`86
`
`89
`
`89
`90
`92
`93
`94
`95
`98
`98
`99
`99
`100
`102
`105
`106
`107
`107
`107
`108
`109
`
`113
`
`113
`114
`116
`11 (i
`122
`124
`126
`
`127
`
`
`
`
`

`

`VIII
`
`Co11te11/s
`
`7.7
`7.8
`
`7.6.I Chemical properties
`7.6.2 Physical properties
`7.6.3 Microbiological properties
`Techniques !cir characterizing and optimizing suspensions
`Suspension manufacturn
`7.8. l Scale-up
`7.8.2 Manufacturing controls: special considerations for peptide
`and protein suspensions
`7.9 Other related systems
`7.10 Conclusions
`Acknowledgements
`References
`
`8 Pc1>tidcs and Proteins as Parenteral Solutions
`Michael .I. Akers and Michael R. DeFelippis
`8.1 Overview and introduction
`8.2 Optimizing hydrolytic stabi lity
`8.2.1 Uu fl'ers
`8.2.2
`Ionic strength
`8.3 Optimizing oxidative stability
`8.3.l Antioxidants
`8.3.2 Chelating agents
`8.3.3
`Inert gases
`8.3.4 Packaging and oxitlation
`8.3.5 Other chemical stabilizers
`8.4 Optimizing physical stability
`8.4. 1 Dcnaturation
`8.4.2 Protein aggregation
`8.4.3 Adsorption
`8.4.4 Precipitation
`8.4.5 Surfactnnts
`8.4.6 Cyclodcxtri ns
`8.4.7 Albumin
`8.4.8 Other physical complexing/stabilizing agents
`8.5 Optimizing microbiological activity: antimicrobial
`preservatives (APs)
`8.6 Osmolality (tonicity) agents
`8.7 Packaging
`8.8 Processing
`8.9 Conclusion
`References
`
`/
`
`9 Roles of Protein Conformation and Glassy St11tc in the Storni.:e
`Stability of Dried Protein Formulations
`.John F. Carpenter, Lotte Kreilgaard, S. Dean Allison and
`Thcodm·c W. Randolph
`9.1
`Introduction
`9.2
`Infra red spectroscopy to study protein secondary structure
`
`127
`129
`133
`133
`136
`136
`
`136
`138
`139
`139
`139
`
`145
`
`145
`147
`150
`151
`153
`154
`157
`157
`157
`158
`158
`159
`160
`162
`163
`163
`165
`166
`167
`
`167
`170
`170
`171
`171
`172
`
`178
`
`178
`180
`
`
`
`
`

`

`Co11te11ts
`
`9.3 Physical factors affecting storage stability of dried protein
`formulations
`9.4 Summary nm! conclusions
`Acknowledgements
`References
`
`IO Peptide and Protein Drug Delivery Systems for Non-parenteral
`Roules of Administralion
`Mette lngcmann, Sven Frokjaer, Lars llovgaard and
`llcllc Brnndsled
`
`I 0.3
`
`JO.I
`Introduction
`I 0.2 Non-parenteral routes of delivery for peptides and proteins
`10.2.1 Barriers to non-parenteral administration of peptides
`nnd protci ns
`I 0.2.2 General approaches to bypass enzymatic and
`absorption barriers
`formulatioti principles for peptides and proteins
`I 0.3.1 Entrapment and encapsulation
`I 0.3.2 Covalent binding
`Immobilized proteins intended for local effect in the GI tract
`- a case study
`I 0.4.1 0ml enzyme supplementation therapy - phenylalanine
`ammonia-lyasc
`I 0.5 Future rcrspcctives
`I 0.6 Summary
`References
`
`I 0.4
`
`11 Peptide and Protein Derivatives
`Gitte Juel Friis
`11.1
`In troduction
`I 1.2 4-Jmidazolidinonc prodrugs
`11.3 Prodrugs or TRI I
`11.4 Derivatives of desmoprcssin
`11.5 Derivatives of' insulin
`11.6 Cyclic prodrugs
`11.7 Conclusions
`References
`
`12 Chemical and Phannacculical Documcnlalion
`Karen Fich and Deirdre Mannion
`12.1
`Introduction
`12.2 Composition
`12.3 Method or numuf'acture
`12.4 Control of starting materials
`12.4.1 Active substances
`12.4.2 Excipic111s
`12.4.3 Packaging materials
`
`ix
`
`181
`186
`I 8(1
`18(1
`
`189
`
`189
`J 89
`
`191
`
`192
`194
`194
`198
`
`200
`
`200
`202
`203
`203
`
`206
`
`206
`207
`209
`210
`212
`213
`214
`2 15
`
`220
`
`220
`221
`222
`222
`222
`22(>
`226
`
`
`
`
`

`

`X
`
`lntcnncdiatc products
`12.5
`12.(1 Control tests on tile finished product
`12.7 Stability
`12.7.1 Expcrtrcport
`l?e/en:11ces
`
`illdex
`
`Co11te11ts
`
`227
`227
`228
`230
`23 1
`
`232
`
`
`
`
`

`

`8
`
`Peptides and Proteins as Parenteral
`Solutions
`
`MICHAEL J. AKERS AND MICHAEL R. DEFELIPPIS
`Lilly J.:vw,1rc/1 L.1i.J<H,1torie.~, Jndi,m,rf)oli~, lnc/i;in.1, USA
`
`8.1 Overview an<l inlroduclion
`8.2 Oplimizing hydrolylic stability
`8.2. 1 Buffers
`8.2.2
`Ionic slrenglh
`8.3 Optimizing oxidative stability
`8.3.1 Anlioxidants
`8.3.2 Chelating agents
`8.3.3
`Inert gases
`8.3.4 Packaging and oxidation
`8.3.5 Other chemical stabilizers
`8.4 Oplimizing physical stability
`8.4. 1 Denaluralion
`8.4.2 Protein aggregalion
`8.4.3 Adsorption
`8.4.4 Precipitation
`8.4.5 Surfactants
`8.4.6 Cyclodexlrins
`8.4. 7 Albumin
`8.4.8 Other physical complexing/stabilizing agents
`8.5 Optimizing microbiological activity: antimicrobi:il preservatives (A Ps)
`8.6 Osmolality (tonicity} ~•gents
`8.7 Packaging
`8.8 Processing
`8.9 Conclusion
`References
`
`11.1 Overview and introduction
`The purpose or this chapter is to provide praclical guidance to formulation scicntisls
`charged with the development of stable, manuf:.,cturahle. and elegant solution dosage
`forms or peptides and proteins. The chapler wi ll cover !he basics or chemical stabi liiation,
`physical stabilization. and microbiological qualily or proteins and peptides in solution.
`We will place more emphasis on the approaches used to solve protein /peptide solution
`
`145
`
`-145-
`
`
`
`
`

`

`146
`
`Akers and Dc:Felippis
`
`formulation rroblcms than on discussing the nature or degradation mechanisms which arc
`covered elsewhere in this text and in many other excellent publications. We also will
`have some coverage or packaging and mnnufocturing of protein solution dosage forms, in
`the spirit or emphasizing that scientists developing these dosage forms must be equally
`concerned with the formulation, the package, and the manufacturing rroecss.
`There nrc at least 22 protein products on the market, four or which arc stored as ready-to(cid:173)
`usl.! solutions, and the rest or wh ich arc stored as freeze-dried powders, then reconstituted
`into solutions by adding a diluent before adm inistration. Approximately 200 peptides and
`proteins are being studied in the clinic, most of which arc freeze-dried products. It is reason(cid:173)
`able to assume that nearly every one of these pcrtide and protein products, commercial or
`in clinical study, has had to overcome and control stability issues in solution. The type or
`stability issue and the degree of complexity of the degradation mechanism differ from protein
`to protein, but approaches to resolve instabi lity issues in solution arc relatively universal.
`Therl.! arc some basic guidelines lo consider in the development of parenteral solut ions
`of proteins and peptides. These arc summarized as fol lows.
`
`A thorough understanding of the physical anc..l chemical properties of the protein or
`peptide bulk drug substance is necessary. Well-documented analytical techniques arc
`now available for studying these properties in solution. Effects of temperature, pl I,
`shear, oxygen, buffer type and concentration, ionic strength, and protein /peptide con(cid:173)
`centration must be understood. From prcfonnulation studies, protei n/peptide chemical
`and physical degradation pathways will be better understood so that the final formu la(cid:173)
`tion, manufocturing process, and packaging system will be rationally dcvclorcd.
`
`2 The route of admi 11istrat ion must be known in order to select the final dosage form,
`vehicle, volume, and tonicity requirements for the product. For cxamrlc, if the prim(cid:173)
`ary route of' administration is intravenous, the vehicle has lo be water although
`some water-miscible co-solvents can be used. The volume can be limitless (unless an
`antimicrobial preservative is rart of the formulation, in which case the volume is
`limited to 15 ml), and the ton icily docs not necessarily have to be isotonic because the
`injected solution will be raridly diluted. I lowevcr, if the route or administration w ill
`be subcutaneous or intramuscular, then the vehicle can be aqueous or nonaqucous, the
`volumes arc limited (usually no more than 2 ml for subcutaneous, 3 ml for intramus(cid:173)
`cular), and the tonieity or the product needs to be more tightly controlled since the
`product is not quickly or readily diluted. The rntc of injection is also a factor to be
`considered in selection of final formulation ingredients in that some ingredients. includ(cid:173)
`ing the protein/peptide itself~ can be irritating and even cause local inflammatory
`reactions if injected too quickly and/or at too high a concentration.
`
`3 Careful screening and choice of solutes for solubilization, stabilization, preservation,
`and tonicity adjustment must take place. These aspects will be the thrust or this chapter.
`
`4 Potential effects of the manufocturing process on the stability of the protein/peptide in
`the final formulation must he understood. Proteins/peptides cannot withstand terminal
`sterilization techniques (heat, gas, radiat ion) and, thus, must be sterilized by aseptic
`filtr<1tion. The filter used must be qualified so that it does not bind the protein/peptide.
`The effect of !low rate during filtration and filling on solution stability must be
`studied. Also, the effect of shear (mechanical stress) that is cncoumered during manu(cid:173)
`facturing must be known. Time limitations must be established from the time the
`protein solution is compou11tkd unt il it is sterile-filtered in order to avoid any increase
`in cndotoxin levels from whatever the bioburden, however small. may be in the non(cid:173)
`sterile solution. llarwood C'f al. ( 1993) and Nail and Akers (2000) arc excellent
`
`-146-
`
`
`
`
`

`

`PeJJ!ides a11cl JJrvfei11s as parenteral solutiom
`
`147
`
`references that deal thoroughly with all aspects or the manufacturing of sterile protein
`and peptides dosage forms.
`5 Selection of the most compatible container/closure system is tremendously important.
`Formulation scientists must appreciate that the container and closure system is just
`as important as the final solution formulation in assuring long-term stability and
`maintenance of sterility and other quality parameters of the product. Proteins and
`peptiucs a1c well known to atlsorb to glass, so experiments must be designed to study
`this possibility and, if adsorption occurs significantly, additives such as albumin must
`be considered to reduce the adsorption. Glass leachates and particulates arc possible,
`and the formulator must be aware or this. Experiments must be conducted to assure
`elimination of this potential problem. The choice of rubber closure is particularly
`important because of known potential for the closure to leach some or its own ingredi(cid:173)
`ents into a solution, to adsorb components of the protein/peptide formulation, to core
`(rubber particulates) when penetrated by a needle, to generate particulates, and to leak
`due to problems with the fitmcnt on the glass vial or resealability of the elastomer
`after needle penetration. Studies on adsorption of the protein to plastic surfaces will
`be necessary irthe final product will be a plastic container. Even if plastic is not part
`of the primary container, protein-plastic compatibility studies should be done since
`plastic tubing such as silicone or polyvinyl chloride wil l be used in pharmaceutical
`process equipment (e.g. filling machines), and the final dosage fo1111 might be added
`to large-volume parenteral solutions contained in plastic bags.
`6 Studies must be conducted to understand the effects of distribution and storage on the
`stabi lily of the final product. Temperature excursions during shipping, mechanical
`stress, exposure to light, and other simulated shipping and storage conditions must be
`studied. From these studies, appropriate procedures for distribution and long-term
`storage or these relat ively unstable dosage forms can be developed.
`Table 8. 1 provides or summary or the key steps in the development of solution dosage
`forms of peptides and proteins.
`
`n.2 Optimizing hydrolytic stability
`
`Tile effect of solution pH on stability is a very important factor to study in early protein
`solution development. Figure 8.1 schematically depicts expected stability problems of
`proteins as a function or pH. Preformulation stability studies arc conducted very early in
`the product development cycle lo elucidate relative protein solubility and stability over an
`approprintc pl I range (normally pl I 3 to pl I I 0). The relationship of stability and solubil(cid:173)
`ity at various pl-I values usually follows a pattern of higher solubility, lower chemical
`stability; or lower solubility, lower physical stability. Protein solubility is minimum gen(cid:173)
`erally at its isoclectric point. Insulin, for example, has an isoclcctric point of 5.4, and at
`this pl-I it is quite insoluble in water (<0.1 mg/ml). Adjusting the solution pl I to less than
`4 or greater than 7 greatly increases insulin solubility (>30 mg/m l, depending on zinc
`concentration and species source or insulin), but also increases the rate of' dcamidation at
`the pll ranges ([3range <.!I al., 1992b). An example or the effect or pl I on denmidation aml
`polymcrizntion or insulin is shown in Figure 8.2 (Urangc and Langkjaer, 1993). In dosage
`l'onn development, the scientist must flrst determine what pl I range provides acceptable
`solubility of the protein for proper dosage, then determ ine whether this pH rnngc also
`provides acceptable stability. There is usually a give-and-take relationship between solu(cid:173)
`bility and stability, ~ml it is up to the scientist to idcntiry what pll is optimal for both.
`
`-147-
`
`
`
`
`

`

`148
`
`Akers and DeFcli11pis
`
`Table ll.1 Development str.1tegy for protein and peptide parenteral solution dosage forms
`
`Formulation and package development studies
`
`Process studies
`
`Final strategy/objc<.:tivcs
`Development of' final formulation
`
`•
`
`Justification of' choice of excipients, pH,
`specilications
`
`Selection of container/closure
`
`• Extractables
`
`• Container/closure integrity
`• Glass leachates, particulates
`
`Stability and compatibility studies
`•
`
`Effects of light, oxygen. high temperature,
`frce;:ing
`
`•
`
`Interaction of cxcipicnts wi th active
`components
`
`• Long-term stability studies of final
`container formulation in finnl container/
`closure system
`
`• Temperature/shipping excursions
`
`Microbiological characteristics
`
`• Antimicrobial properties
`• Preservative efficacy
`• Endotoxin control
`
`Optimization studies of' cxcipicnts. pl I. other
`possible variations
`Process development
`
`•
`
`Process control (e.g. time, temperature
`during each processing step
`
`• Filter selection/validation
`
`Microbial retention
`- Adsorption
`-
`Extractables
`
`• Effect or terminal stcril ization
`Justification of excess
`•
`• Process vnlidation
`Sterilization of components
`-
`
`/\septic process
`-
`- Cleaning
`-
`Filling
`
`Establishment of' critical process parameters
`Establishment of control strategy
`
`C-tcnninal
`deamidation
`
`Carbonyl-amine
`reactions
`
`Un-ionized protein
`amides
`
`Un-ionized terminal
`carboxylic acid
`
`Un-ionized side-chain
`carboxylic acids
`
`Selectively oxidized
`under acidic conditions
`
`Asp transpcptidation
`
`Ii-elimination
`
`Met oxidation
`
`Deamidation
`
`Peptide cleavage
`
`Various oxidations
`
`Base-catalysed
`
`Ionized cysteinc
`
`to reactive species
`
`Di.saccharidc browning
`
`Acid
`
`Neutral
`pll
`
`Disulphide
`shifting
`
`!3asc
`
`Figure H.1
`
`l'rotein reactions ;is a function of pH (courtesy of Dr Lc>e Kirsch).
`
`-148-
`
`
`
`
`

`

`fl<'plides ontl proteins us p<1rc11terul s11l11tio11s
`
`149
`
`A
`
`100 - . - - - - - - - - - - - - - - - -~
`
`80
`
`Q)
`
`g' 60 -r=
`
`(1)
`(.)
`ci> 40
`Q..
`
`(cid:127) Monodesamido
`~ Didesam ido
`D Spilt product
`
`20
`
`0
`3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5 8.0
`pH
`
`B
`
`20 -.---A-- - -- -- - -- - --
`
`-
`
`-.
`
`15
`
`Q)
`Cl
`
`~ -r= 10
`
`(1)
`0
`'--(1)
`Q..
`
`II Dimer
`(cid:143) Trimer
`(cid:127) Tetramer
`
`5
`
`0
`3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5 8.0
`pH
`
`I lit'( I ol pl I 011 clt'.1micl.11io11 ,rnd polynwri/,1lio11 ol in,ulin: t lwrnic ,11
`Figure U.2
`1r,1mlom1.ilion duling , to1 ,1g<• ll'i C, 12 111011111') ol rl10111holH•dr ,1 I hovi,w in~ulin ( ry,t,11,
`(ll. 7'Y., N.i( I, ll.2'Y., 11hvnol> ,1, .i fun< lion ol pl I. (/\) I rnm,1tio11 ol 1lw hyd1oly,b pwclull,
`111ont><i<•,,1mido .111<1 clidt•,,1111ido i11,uli11, .111d tlw in,u l in ,pl 11 product (!\II /\'l).
`(ll) I rn111<1tion ol < ov,1 lt•11l clinl('1~ ,llld o lig<>lllt'r,. Rt·p1 inl<·d with p1•1111i,,io11 fron1
`ll1 ,111g(• ,ll)d I ,111gkj,t•1 ( 11) 1) I); r.01'il'lllllll I'll'''·
`
`1 lydrnlysis or dcamidation occurs wi th peptides and protei11s containing susceptible
`J\sn anti ( il n amino acids. lhc only 1,,0 amino acid:-. thnt ;1rc primary amines. The sitk(cid:173)
`chain amide linkage in ;i (iln and J\sn residue may underg<1 deamidation to form f'rcc
`carho:xylic acid. Dca111idatio11 can be promoted by a vmic.:ty or factors includi11g high pl I.
`temperature and ionic strenglh (Manning. et ul .. 1989). The rak: of'dc,11nid:.11ion is alTcttcd
`
`-149-
`
`
`
`
`

`

`150
`
`Akers all(/ DeFelippis
`
`by amino acid sequence, particularly lhc amino acid immcdiatcly following the Asn or
`Gin amino acids. Oliyai and Borchardt ( I 994) studied the influence of primary amino
`acid sequence on the degradation of Asp residues under both acidic and alkaline condi(cid:173)
`tions. /\s expected, the rate of intramolccular formation of the cyclic imidc, the first step
`in the hydrolytic degradation pathway (Pntcl and Borchardt, 1990), was most affected by
`the size or the amino acid on the C-tcrminal side of the Asp residue. Dcamidation rates
`for peptides will be highest when Asn is immediately followed by a Gly amino acid since
`Gly has no side-chain, thus no opportunity lo hinder the hydrolysis reaction stcrically.
`C-termi nal substitution of Gly with increasingly more bulky residues (Ser, Val) inhibi ts
`the amount of cyclic imidc produced. I lowcver, with respect lo Asp amide bond hydrolysis
`with adjacent am ino acids either before or after Asp, such structural changes had little or
`no effect.
`For larger protein structures, the effects of adjacent amino acid sequences on the
`dcmnidation rates of Asn and Gi n arc more difficult to estimate simply due to the three(cid:173)
`dimensional complexities of these structures. However, it certainly is intuitive that
`adjacent amino acids and their size will have some effect on J\sn and Gin deamidation
`regardless of the size of the total protein. l'lydrolytic stability or peptides and proteins can
`be minimized, therefore, through one or more of the frillowi11g approaches.
`
`Optimization of ami no acid sequence, i.e. engineering protein structures to remove
`unstable amino acids or insc11 amino acids that sterically hinder J\sn or Gin dcamidation,
`ns long ,is this docs 1101 affect protein activity, potency, toxicity, or any other quality
`attribute.
`
`2 Formulate at optimal solution pl-I . For example, human epidermal growth factor 1- 48
`demonstrates some interesting pH-dependent stabil ity in that at pl! <6 succinimidc
`fo rmation at Asp11 is favoured, while at pH >6 deamidation of /\sn 1 is favoured
`(Scnderoff ct al., 1994 ). The optimal pl-I, therefore, is right a l pl I = 6.
`
`3 Store at low temperatures, alt hough this will always create difficulties during distribu(cid:173)
`tion and long-term storage of the product.
`
`4 Optimize the effects of ionic strength (to be discussed in section 8.2.2).
`
`ll.2. 1 Buffers
`
`Buffers arc used to prevent small changes in solution pH which can affect protein solubi l(cid:173)
`ity and stability. Buflcrs arc composed of sails of ionic compounds, the most common or
`which arc acetate, ci trate, and phosphate. 13uffcr systems acceptable for use in parenteral
`solutions arc listed in Table 8.2.
`The proper selection of buffer type and concentration is determined by performing
`solubility and stability studies as a function of' pl I and buffer species. Normally, the pl I
`or lllaximum solubility is not the pl l or maximulll stability. I Eowcvcr, a pH range that is
`a good compromise between solubi lity and stability can he selected and maintained with
`the proper selection of the appropriate buffer component.
`In the pll range of 7-12, buffer concentration can have a significant effect on the rate
`of deamidation indicating general acid- base catalysis. Generally, dcamidation is much
`slower at acidic pl I than al neutral or alka line pH. /\CTI I dernnidation in the pH range or
`7- 11 is catalysed by increasi ng buffer concentrations, whereas there is no buffer catalysis
`
`-150-
`
`
`
`
`

`

`Peptides a11d proteins as J>are11reral solutions
`
`151
`
`Table 11.2
`
`lluffcrs used in protein formulations
`
`Buffer system
`
`pKa
`
`pl I Range or use
`
`J\cctatc
`Carbonate
`Citrate
`Glutamate
`Glyeinatc
`llistidinc
`Lactate
`Malcate
`!'hosp hate
`Sueeinatc
`Tartratc
`Tris
`
`4.8
`6.4
`3.14. 4.X. 5.2
`2.2. 4.25, 9.()7
`2.4, 9.X
`1.8, 6.0, 9.2
`3.8
`1.92. (1.23
`7.2(pKa2)
`4.2, 5.64
`2.93, 4.23
`6.2 (pKb 7.8)
`
`2.5-6.5
`5.0- 11.0
`3.0- X.0
`X.2- 10.2
`6.5- 75
`(,.2- 7.8
`3.0- 6.0
`2.5- 5.0
`3.0- 8.0
`4.8- 6.3
`3.0- 5.0
`6.8- 7.7
`
`at pl I 5- (1.5 (Patel, I 993). 1 lowcvcr, insulin dcamidation at the A2 I position predomin(cid:173)
`ates at acidic pl I while deamidation at B3 predominates at neutral pll (11range el al.,
`1992b).
`Several potential problems arc associated with using buffers in parenteral solutions.
`For example, it should not be expected that in large-scale manufacturing the compounded
`solution containing the buffer will always result in the exact pH specified. Dilute solu(cid:173)
`tions of strong acids (hydrochloric acid) or bases (sodium hydroxide) arc usually required
`lo 'fine-tune' the final solution pl I. Excessive use or the pl I adjustment solutions may
`alter the buffer capacity and ionic strength or the buffered solution ( Niebergall, 1990).
`Increasing buffer capacity to control pH better could signilicantly increase ionic strength
`which, in turn, may cause increased potential of' pain upon injl:ction due to the increase in
`solution osmolali1y.
`General acid and/or general base buffer catalysis can accelerate 1he hydrolytic degrada(cid:173)
`tion or the protein. An example is shown in Figure 8.3 (Yoshioka ct al., 1993) where the
`inactivation rate of' ~-galactosidase increased with increasing concentrations or phosph(cid:173)
`ate buffer up to 0.5 M, then decreased, presumably related to higher buffer componenls
`causing a reduction in water mobility. Cleland <'f al. ( 1993) cite several examples whcr·c
`the rate of pro1cin dcamidation was markedly dependent on the buffer anion. Capasso
`ct al. ( 1991) compared the dcamidation rate or a small peptide using different hurlers and
`fi.n11HJ that the peptide was most unstable in a phosphate buffer and most stable in Tris
`buffer. Wang et al. ( 1996) found that buffer type and concentration affected aggregation
`or basic fibroblast growth foctor depending 011 pll. At pl I 5, aggregation increased as
`citrate buffer concentration increased. Citrnte buffer at pH 3.7 caused aggregation, whereas
`acetate buffer rit pH 3.8 did not. At pH 5.5- 5.7, phosphate, acetate and citrate buffers all
`showed similar aggregation rates.
`
`8.2.2
`
`Ionic strength
`
`Ionic strength is a measure of the intensity of the electrical field in a solution. It depends
`on the total concentration of' ions in solution and the valence or each ion. The ionic
`strength of a 0.1 M solution of sodium chloride is 0. 1. The ionic strcng1h of a 0.1 M
`
`-151-
`
`
`
`
`

`

`152
`
`Akers and DeFeliJJJJis
`
`en
`C:
`C:
`C'il
`E
`Q) ...
`
`>, -> -(.) <
`
`~ 0
`
`100
`
`50
`
`0
`
`0
`
`100
`Time (min)
`Figure II.]
`lnac:liv.ition of 11-g.ilactosicl.isc in pH 7.4 phosph,1te buffer solution al 50'(, JS
`a function of phosph.ite buffer conccntr;'llion: (6.) l 0, (0) 50, ((cid:143)} 100, ('v) 200, (& ) 500,
`(e ) 700, ((cid:127) ) 900 mM. The concentration of f1-g;ilactosiclase was 0.1 mg/ml. l~crxinted
`with permission from Yoshiok.i et al. (1 'J'JJ); [)Plenum Press.
`
`200
`
`2-----------------,
`--- KCI
`_._ NaCl
`
`1.5
`
`'
`
`\
`
`\
`
`\
`
`\
`
`\
`
`\
`

`~ 1
`~
`~
`..:c
`
`0.5
`
`\
`
`• .... .... .... .... .... .... ..
`
`o-+-------,.---
`0.8
`0
`0.4
`[salt] (molarity)
`
`1.2
`
`Figure 8.4 Effect of salt concentr;ition on r/\/\T solution slaliilily. Reprinted with
`permission from Vcmuri et al. (l <J'fl); ©l'lcnu111 Press.
`
`solution or sodium sulphate is 0.3, because sulphate ions have a valence or 2 added to the
`valence of I for the sodium ions. Ionic strength may have an effect 011 protein stability
`in solution. The Dcbye-l luckcl theory predicts that increased ionic strength would be
`expected to decrease the rnte or degradation or oppositely dwrgcd reactants. and increase
`the rate or degradation or similarly charged reactants.
`Ionic strength will affect the stability of' a protein, but in which direction (increase
`or decrease) di ffcrs with di ffcre nt proteins. r:or example. increasing ionic strength wi II
`increase the stability of recombinant alpha 1 antitryrsin (Yemuri et al., 1993) (figure 8.4 ).
`
`-152-
`
`
`
`
`

`

`PeJ>tides (//Id pmteins (IS p({renteral solutions
`
`153
`
`100
`
`-
`E -Cl
`.s
`-(/) 10
`..c ...
`Ql :c
`::::,
`0 en
`
`\
`
`'\
`
`' ~
`
`/
`
`,L
`
`I
`,,.
`
`.
`~✓
`
`1 ...... -----,.---..------,.---
`4
`5
`7
`6
`pH
`Figure 8.S Effect of ioni<.; stn:.mgth ()11 rbSt solul)ility-pH profiles. Ionic strength ft
`inc:re,1secl with increasing NaCl: ((cid:127) ) p == 0.24; (0 ) p = 0 .12; (e ) p = 0.072; (0) fl= 0.024;
`( "') ~l == 0.0011. Reprinted with permission from D;ivio and Hageman ( 1 <)<J3); ©Plenum
`l'ress.
`
`8
`
`9
`
`10
`
`Conversely, increasing ionic strength will increase the rate or deamidation or human
`growth hormone (Pearl man and l3ewley, 1993) and bovine somatotropin (Davio and
`I Jageman, I 993) (Figure 8.5).
`
`11.3 Optimizing oxidative stability
`
`Proteins containing methionine, cysteine, cystine, histidine, tryptophan, and tyrosine may
`be sensitive to oxidative and/or photolytic degradation depending on the conformation of
`the protein and resultant exposure of these amino acids to the solvent and environmental
`conditions such as presence of oxygen, light, high temperature, metal ions, and variours
`free radical initiators. Oxidation of sulphydryl-containing amino acids (e.g. methionine
`and cystcinc) wi ll lead to disu lphide bond formation and loss or biological :1ctivity. The
`free thiol group that is present in a Cys residue of any nativt: biologically active protein
`not only may oxidize to produce an incorrect disulphide bridge, but also can resu lt in
`other degradation reactions such as alkylntion, addit ion lo double bonds and complcxation
`with heavy metals.
`I lu man growth hormont:, chymoslrypsin, lysosyme, parathyroid hormone, human
`grnnulocyle-colony stimulating factor, insulin-like growth factor I, acidic and basic
`libroblast growth foctors, relax in, the mo11oc

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