throbber

`
`Second Edition
`
`Pharmaceutical Formulation
`
`
`Development of Peptides
`and Proteins
`
`by
`Edited
`Lars Hovgaard
`Sven Frokjaer
`
`Marco van de Weert
`
`CRC Pren ls an Imprint of the
`
`
`Taylor & Francis Group, an lnformo bu1iness
`
`Bausch Health Ireland Exhibit 2009, Page 1 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Cl�C Press
`Taylnr & rrnncls Group
`60UO Broken Sound Parkway NW, Suite 300
`Boca Riuon, FL 33487-2742
`
`� 2013 by Taylc,u·&. Francis Group, LLC
`CRC Press Is an Imprint of Taylor& Francis Group, .:an lnforma business
`
`No claim to original U.S. Govcrnmeni works
`
`Printed In the United States of America nn acid-free paper
`Version Date: 20121003
`
`International Standard Book Number; 978-lA398-5388•7 (Hardback)
`
`This book contains lnfotmallon obhlned from authcntk and hlshly l'egarded sourct-s. Rl'asonable efforts
`have been made to publish reliable datu. and information, bul thc author -and publtsher cannot assume
`responsibility for the validity of all nmteri:ils nr the consequences of their use. The authors 11nd publishers
`have attempt<'d 10 trace the copyright holders ofa11 material reproduced irl thil!ii publication anti iipologiie to
`copyright holders lf permission to publish in this form has nol been obtaint'd, 1£ any copyrighl materi;il has
`not been acknowlt'dged please write and let us know so we marrecltfy In any future reprint.
`
`Except as permitted under U.S. CCtpyright Law, no parLofthi9 book maybe reprinted, reproduceJ, tr.msmit­
`ted, or utilized in any form by any t>leclronic, mechllnical, or other means, now known or hNearter invented,
`including photocopying, mlcro{llmlng, and recording, or In any information storitge or retrieval system,
`with()ut written permission from the publishers,
`
`For permission to photocopy or use material decrronfcally from this work, plt'ase 3:CCt'SS www.copyriglu.
`222 Rosewood com (http:llwww.copyright.com/) or contad the Copyright Clearance Center, Inc, (CCC),
`
`
`
`Drwe, Danvers, MA 01923, 978-750•8400. CCC is a nOl•for.proflt organi7.ation that provides licenses and
`registration for a variety of users. For organ11:at1ons I hat have been granted a photocopy license by the CCC,
`
`a scparatesyslem ofpaymenl has been arnnsed.
`
`Trademark Notice: Product or corporate names maybe trademarks or regiSlered trade:marks, and are used
`only ror identirication and explanation without Intent to infringe.
`
`Library o(Congress Cataloging-ln•Publkation D11ta
`
`Pharmaceutical formulation dcvelopmenl o( peptides and proteins/ edllcrs, Lars
`Hovgaard, Sven Frukjaer. Marco van de Weert. -- 2nd ed.
`p.;cm.
`Includes bibliographical references and index.
`ISBN 978· 1-1398·5388•7 (hardcover: alk. paper)
`I. Hovgaard, Lars, 1962- ll. Frokfrer, Sven, 1947- 111. Weert, Marco van de, 1973-
`[DNI.M: 1. Peptide Blosynthtsis. 2. Chemistry, Pharmaceutical--methods. 3. Protein
`Biosynthesis. QU 681
`
`615.1'9--dc23
`
`2012030490
`
`Visit the Taylor & Francis Web site 11t
`http://www.taylorandfrancis.com
`
`and t.he CRC Press Web site at
`http://www.crcpreu.com
`
`Bausch Health Ireland Exhibit 2009, Page 2 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Contents
`
`Prcfnce, .......... ,., ................................ , .• , •...•.•.•.•.••.••• ,, •• ,-,,, .. ,, .. , ........... , .. ,. .........
`vii
`
`
`
`
`
`
`
`
`
`Ed it ors., .... ,, ... ,., ...... , ....................... �.-·�·-·······.,., ............ , .... ,, .... ,, ............. , ................ ,, ix
`
`
`
`
`
`
`
`Contributors ............. , .•...... , ............ , ............... , ........ , ....... , ............. , ........................... xi
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Chapter I Pcplide Synthesis .................................................................................. I
`
`Knud J. Jensen
`
`Chapter 2 Protein E.xpression ... .......................................................................... 17
`
`
`
`Na1111i Din
`
`Chapter 3 Protein Puriticmion ................................
`
`
`
`�---·-·······-··
`
`
`
`
`
`
`
`Lars Hnv,gaard, Lar:. S'kriver, and Sven Frokjaer
`
`Chapter 4 CharacLcriza1ion of Therapeutic Peptides and Proteins ... ................. 49
`
`Marco van de Weert and Tudor Arvi,ite
`
`Chapter 5 Chemical Pathways of Peptide and Protein Degr'Jdarion . ................. 79
`
`
`
`Ter1ma J. SiaJuu.m and Christian SchOneic/r
`
`
`
`Chapter 6 Physical Instability of Peptides and Prolcins ................................... 107
`
`
`
`
`
`Marco van de Weert a,id Theodore W Randolph
`
`Chapter 7 Peptide allld Protein Derivatives .................................................. , ....... 131
`
`
`
`
`
`Kristian Strf>mgaard and Thomas H¢eg-Jensen
`
`Chapter 8 Pep1ides and Proteins as Parenteral Solu1ions ................................. 149
`
`
`
`Michael J. Akers tind Michael R. DeFelippis
`
`
`
`Chupter 9 Peptides ilnd Proteins as Parenteral Suspensions: An Overview
`of Design, Development, and Manufacturing Considerations ......... 193
`
`
`
`
`
`Michael I?. DeFelippis and Michael J. Akers
`
`
`
`Bausch Health Ireland Exhibit 2009, Page 3 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`vi
`
`Contents
`
`
`Chapter
`
`10 Rational Design of Solid Protein
`
`
`
`
`
`Formulations···-··················· ....... 239
`
`
`
`
`
`Bingq1um {Stuart) Wang mu/ MichaeJ J. Pikal
`
`Peptide and Protein Drug Delivery Systems for Nonparenteral
`
`
`Chapter ll
`
`
`
`
`
`
`Routes of Administration�-······················ ············-·········· ............... 269
`
`
`
`Ulrik Lyll Rahbek, Frcmti!ek Hubtifek. and Simon Bjerregcwrd
`
`
`Chapter
`
`12 lmmunogenicity of Therapeutic Proteins .......... , ............................. 297
`
`
`
`
`
`Grzegorz Kijanka, Wim Jiskoor, Melody Sauer/Jorn,
`
`
`
`
`Huub Schellekens, and Vera Brinks
`
`of Peptides and Proteins
`Biosimulation
`Chapter 13
`
`.. 323
`
`
`Ti,e S"eborg, Christian Hove Rasm1use.n
`
`
`Morten Colding-Jprgensen
`
`r Erik Mosekilde. and
`
`
`
`
`
`
`
`
`
`Chapter 14 Registration of Peptides and Proteins.
`
`
`
`. ...... 339
`
`Niamh Kinsella
`
`
`
`
`
`Index ...................................................................................................................... 363
`
`
`
`
`
`
`
`Bausch Health Ireland Exhibit 2009, Page 4 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Physical I nstability of
`6
`Peptides and Proteins
`
`
`
`Marco van de Weert and Theodore W Randolph
`
`CONTENTS
`
`lntroduclion ............................................................................................ ..... 107
`6.1
`6.2 Protein Structure ........................................................................................... 108
`6.2.1 Peptides. Polypeptides. and Proteins ................................................ 108
`6.2.2 Protein Structure: Primary. Secondary. Tertiary. and
`Quaternary Structure ........................................................................ 108
`6.3 Prorein Folding: Why Do Proteins Fold? ..................... ............................... 109
`6.3.1 Role of Water and Stabilizing Interactions .. .................................... 109
`6.3.2 The Energy Landscape of a Protein Molecule ................................. 111
`6.4 Pl'Olein Physical Degradation ..................................................................... 114
`6.4.1 Protein Unfolding .......... . ............................................................. 114
`6.4.2 Adsorption .......... _ .....................................
`.... ............................... 117
`6.4.3 Protein Aggregation .......................................................................... 118
`6.4.3.1 Aggregation Mechanisms and Kinetics ............................. 119
`6.4.3.2 Fibrillation: A Special Case of Pro<cin Aggregation ......... 120
`6.4.4 Protein Precipitation ......................................................................... 121
`6.5 Stabilization Strategies ................................................................................. 122
`6.6 Concluding Remarks .................................................................................... 125
`Reference.<...........
`. ............................................................................... .... 126
`
`6.1 INTRODUCTION
`
`The biological function of peptides and prolcins is highly dependent on their three­
`dimensional structure. Chrmgcs in that slructurc. whic.:h may arise due to chemical
`or physical processes, may alter or abolish that runction, or even result in Loxicity.
`Thus. it is of importance that a pharm:1ceu1ical formultttion of therapeutic peptides
`and proteins retains the no1·ma.l (native) structure or those peptides or proteins, or
`1hr11 any changes are fully reversible upon administration to the patient.
`A major difference between proteins and low molecular weight drugs is the com­
`plexity of the three-dimensional structure and concomirnnt sensitivily toward exter­
`nal .;tress factors. The three-dimensional structure of proteins is mostly held together
`by noncovolent interactions. such as hydrogen bonds, salt bridges, and van dcr Waals
`fore�. Any stress factor may alter these noncovalent 1ntemcl1ons, possibly leading to
`new inlrn- or in1ermolecular interactions which may not be reversible upon removing
`the stress factor.
`
`107
`
`Bausch Health Ireland Exhibit 2009, Page 5 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`
`
`108 Pharmaceutical Formulation Development of Peptides and Proteins
`
`
`
`
`
`In this chapter. we will discuss the noncovalent intcraclions that result in the for•
`
`
`
`
`
`
`
`
`
`mation of the specific thrcc•dimensional fold of most proteins, the most important
`
`
`
`
`
`
`stress factors that may cause changes in that protein fold. and the resulling physical
`
`
`
`
`
`
`instability of the protein. It should be noied that we will discuss this physical instabil­
`
`
`
`
`ity as a separate issue to the chemical instability discussed in Chapter
`
`5. In reality, the
`
`
`
`
`
`two are highly imerdepcndern, as chemical destabilization may lead to physical desia.
`
`
`
`
`bilization, and vice versa. This chapter will end with potential stabilization strategics
`
`
`
`
`
`
`to prevent physical instability of proteins in pharmaceutical formulations. Several of
`
`
`
`
`lhese strategics will be discussed with more specific ex1c1mples in other chapters,
`
`
`
`
`
`
`Throughout the chapter, a number of semantic issues wil.l be discussed, which arc of
`
`
`
`
`
`
`importance when reading the literature. The commonly used terminology within the
`
`
`
`
`
`
`field of protein structure, folding, and stability is not always strictly defined. and defini­
`
`
`
`
`
`
`tions may differ over time and depending on the context. Unfortunately, the definitions
`
`
`
`used in a particular scientific paper are often not explicit, which may lead lo confusion
`
`
`
`When the reader is insufficienlly aware of 1he different descrip1ions that are in Use.
`
`6.2 PROTEIN STRUCTURE
`
`
`
`ANO PROTEINS 6.2.1 PEPTIDES, POLYPEPTIDES,
`
`
`
`All peptides, polypeptides, and proteins are considered condensation polymers of
`
`
`
`
`
`
`
`
`
`
`
`
`amino acids, rcsuhing in a linear backbone uf altcrnaling urnidc, C-C, and C-N
`
`
`
`
`
`
`bonds. However, the distinction hctween peptide, polypeptide, and protein is rather
`
`
`
`
`diffuse. One may find at least three different and partly overlapping descriptions,
`
`
`
`
`
`
`
`rather than definitions, of the difference between peplide and protein alone. The cur­
`
`rently mos! common description refers to any peptide chain of more than 50 amino
`
`
`
`
`
`
`
`acids as a protein. Others refer to peptides as proteins whenever the peptide has
`
`
`
`
`
`
`a biological function. This could then even include several simple dipeptidcs (i.e ..
`
`
`
`
`
`two amino acids linkc<l together), which can have a biological function. Finally, the
`
`
`
`
`absence or presence of a wel l -defined tertiary structure has been used to distinguish
`
`
`
`
`
`peptides from proteins. Also, this distinction is not· wi1hout problems; there arc pro·
`
`
`
`
`
`teins that are referred to as "natively unfolded.'' so called because they do not have
`
`
`
`
`a specific tertiary structure. In addition, some "peptides" can form fully reversible
`
`
`
`
`
`
`multimeric stru{'turcs, such as glucagon (forming trimcrs) (Formisano ct al., 1977),
`
`
`
`
`
`whicb involves the formation of a dcli.ned three•dimensional structure. The term
`
`
`
`
`
`
`"polypeptide" generally overlaps with that of '"peptide" and '"protein." The reader
`
`
`
`may thus encounter all three terms used in connection with the same biological
`
`
`
`
`
`
`compound. For practical purposes, we have used the first definition, calling every
`
`
`compound with more than 50 amino acic.ls a protein.
`
`6.2.2 PROTEIN STRUCTURE: PRIMARY, SECONDARY,
`
`
`
`
`TERTIARY, ANO QUATERNARY STRUCTURE
`
`The thrcc•dimensional structure of proteins ls often subdivided into tour types of
`
`
`
`
`
`
`structure, referred to as the primary, secondary, tertiary. and quaternary s1ructurc.
`
`
`
`
`
`
`The primary structure refers to the amino acid sequence within the polymer chain.
`
`Bausch Health Ireland Exhibit 2009, Page 6 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Physical lnslability of Peptides and Proteins
`
`109
`
`Intra- or intcrchain cross-links are also prevalent, usually lhrough cyslcinc residues
`(forming a cysLine or S-S bridges).
`The secondary structure refers 10 the folding of this backbone into specific struc­
`tures, which arc defined by the bond angles and hydrogen bonding pattern of the
`amide bond. The secondary struc.ture can roughly be subdivided into four classes:
`helical structures such as the alpha-helix, pleated structures such as the beta-strand
`and beta-sheet, turn structures such as the beta-turns. and loop structures. The latter
`are often referred to as "random" structures.
`The three-dimensional alignment of the secondary structural clemenls is known
`as the tertiary structure of a protein. This alignment often results in an almost ideal
`close packing of the amino acids, particularly in the core of the protein molecule.
`Protein tcniary structures can be described by commonly appearing architectures
`such as barrels or alpha-helix bundles (Orengo and Thornton. 2005).
`Some proteins exisL under physiological conditions as specific muhimeric pro­
`teins linked through noncovalent interactions. This multimerization is known as lhc
`quaternary structure of a pro1eiu. Examples of proteins with a quaternary structure
`include hemoglobin, aJpha-crystallin, and HIV-I protease. In general, the biologi­
`cal func1ion or such multimeric proteins depends on this multimerization, but some
`proteins may also form specific (and reversible) multimers that arc not biologically
`active. Perhaps the best known example of the laucr is insulin; insulin forms dimers
`and hcxamers al elevated concemration, especially in the presence of certain diva­
`lent metal ions, but is only active as a monomer (Uvcrsky et al., 1003).
`The ullirnate fold of the protein is usually referred to as the "native" structure.
`In principle. the latter refers to the functional structure of the protein. However,
`many proteins change structure during their biological function. which would sug­
`gesl there are multiple "native" slructures. furthermore, artifi.cinlly created proteins
`(e.g., fusion proteins created by genetic engineering techniques) may a�scmble into
`well-defined folds but have unknown levels of function. It may therefore be easier tO
`describe the protein structure under physiological conditions (in terms of pH, ionic
`strength, etc,) as 1hc na1ive structure. The mechanism of protein folding is discussed
`in 1hc following section.
`
`6.3 PROTEIN FOLDING: WHY DO PROTEINS FOLD?
`
`
`
`6,3,1 RotE OF WATER AND STABILIZING INTERACTIONS
`
`The observation that most proteins are folded into a specific structure in simple
`aqueous solu1ions suggests that foldfog is a thermodynamically favorable process.
`Many decades of research have been aimed at elucidating why. and how. proteins
`fold (Anfinscn, 1973). Although there arc still several limitations, it is now possible
`lo predict with reasonable accuracy how a protein will fold using computational
`methods (Kryshtafovych and Fidel is, 2009). In this section, we will discuss the driv­
`ing forces for folding, starting wilh a pro1ein in the gas phase hcfore moving to the
`more complex situation of a pro1ein in solmion.
`For a single protein molecule in the gas phase, there arc four fundamental forces
`to take into account. The fir!\l is the entropy of the amino acid chain. which tends
`
`Bausch Health Ireland Exhibit 2009, Page 7 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`110
`
`Pharmaceutical Formulation Development of Peptides and Proteins
`
`to disfavor folding. That is, folding of the amino acid chain into a specific structure
`reduces the degrees of freedom for that chain, which results in a loss of entropy. 1n
`contrast. hydrogen bonding and van der Waals forces favor folding. Electrostatic
`interactions may either favor or disfavor folding, depending on the sign of the charges.
`Experiments with peptides in the gas phase have shown that folding can be spontane­
`ous (Chin et al., 2006): hence, at least in some circumstances the entropy loss upon
`folding can be overcome by the enthalpy gain from electrostatic interactions, hydro­
`gen bonding, and/or van der Waals forces. Often electrostatic interactions are an
`important driving force for the folding process in the gas phase (Chin el al., 2006).
`Because proteins arc typically found in an aqueous environment. 1hcse gas•phasc
`experiments offer only limited insights to the understanding of protein folding under
`solUlion conditions. The high dielectric com;tanl of water means that the strength of
`electrostatic interactions is signiJlCantly reduced, and therefore is a much less impor•
`tant driving force for folding. if at all. Moreover, the peplidechain now has the ability
`to form hydrogen bonds with water. as well as lO interact with water molecules through
`van der Waals forces. Thus, intramolecular interactions like van der Waals forces and
`hydrogen bonds also are not immediately apparent driving forces for folding.
`And yet, proteins do fold in water, An important driving force or this folding is
`the negative effect of solute-water interactions on the interaction between the water
`molecules themselves. Pure water may be viewed as a collection of oxygen atoms
`suspended in a sea of hydrogen atoms. On average, four hydrogen atoms surround
`one oxygen atom in a (imperfect) tetrahedral shape, with two of those hydrogen
`atoms close enough to describe the bond as covalent and two slightly further away,
`forming a hydrogen bond. This is, however. a highly dynamic system, and there
`will be a constant exchange between covalently bound and hydrogen bond-linked
`hydrogen atoms. (n essence, any solulc will negatively affect this dynamic system;
`this is known as the hydrophobic effect (Dill et al., 2005). Whether a solute dissolves
`in water. and how much. is a matlcr of accounting: as long as there is a negative
`change in Gibbs free energy for the system as a whole upon dissolution of the solute ..
`the compound will dissolve. Thus. the negative energetic contribution by distorting
`the dynamic water net work needs to be counterbalanced by the positive contribution
`of the soluce dissolving, which includes increased e1ltropy of the solute upon dis­
`solution as well as hydrogen bonding and van dcr Waals interactions with the water
`molecules. Due to the ability 10 form hydrogen bonds and significant van dcr Waals
`inte1w.:1iuns, polar (hydrophilic) (.:Ornpuumls dissolvt: tu a much huger cxtem in wutt:I'
`than nonpolar (hydrophobic) compounds.
`Most proteins contain a significant amount of nonpolar amino acid residues and
`their dissolution in an aqueous environment would be energetically unfavordble.
`I n contrast, the dissQlution of the polar amino acids would be a favorable process.
`By folding of the amino acid chain such that the hydrophobic amino acids arc hid­
`<len from the aqueous surroundings, a protein significantly reduces the hydrophobic
`effect by the nonpolar residues, while maintaining the positive interaction between
`the polar residues and the water molecules. The hydrophobic effect and the result·
`ing "hiding" of nonpolar amino acids in the core of the prolein is believed to be the
`main driving force for folding (Dill, 1990; Kauzmann. 1959). Further folding and
`specificity of the fold arc then governed by other interactions like hydrogen bonding,
`
`Bausch Health Ireland Exhibit 2009, Page 8 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Phys,cal lnstabohty of Peptides and Proteins
`
`111
`
`salt bridge formation. and van der Waals intcrnctions between l1ght..ly packed resi­
`due.< (Rose ond Wolfenden. 1993). Finally, the ultimote fold mny be stabilized by the
`formation o( cys1incs.
`The imponance of the hydrophobic om,no oci<b for protein folding is also sug­
`gested by 1he relallvcly conserviJtive. changes of the core amino oc1ds for similar pro­
`That is, even 1hough the overall
`teins ocross species.
`
`ammo acid sequence mny be
`significantly different for a given protein i'°l:ncd from various �pccics, the differences
`in the (usually hydrophobic) amino acids forming the core of the protein are usually the
`smallest (Mirny and Shakbnovich, 2001), and mutations io thc,c amino acids ure more
`likely 10 yield an inactive protein (Guo et al., 2004). As a result, even proteins with a
`mere 30- 40% similarity i11 amino acid sequence Cfln yield very similar protein folds.
`Considering the above, it should be no surprise that natively unfolded proteins
`generally do not contain such n core of hydmphohic amino ncidR. ln fact, it is likely
`the absence of n significant amount of hydrophobic amino ucids, ulong with many
`charged residues, that allows these proteins 10 have little tertiary fold (Uversky and
`Dunker, 2010). However, they often do hove o specific secondary structure. which
`suggests that for amino acid chains, the intrachain hydrogen bonding is more favor•
`able than hydrogen bonding to water.
`
`
`
`6.3.2 TH, ENncv LANDSCAPE o, A PaornN Moucuu
`
`As discussed above, proteins may spontaneously fold ia aqU<."OUS >0Ju1iot1. Thal means
`that the change m Gibbs free energy upon folding is ncgath·e. that is. 6G, < 0, and
`thw. the change in Gibbs free energy of unfolding ,s pos111vc (6G, > 0). However,
`due lo the complex mteraction between protein and solvent, the Gibbs free energy is
`not a simple linear function of temperature (Privalov, 1990; Robertson and Murphy,
`1997). Let us first eJ<.amine a simple two-stale reversible folding procc.s between a
`proteia in ,ts unfolded state (U) and in a folded slate (N) (Scheme 6.1).
`
`U ,= N
`
`(Scheme 6.1)
`
`The change in Gibbs free energy for this folding process con be opproxima1ed
`using a modified form of the Gibbs-Helmholtz equution (Equatiou 6.1), in which the
`tempe1::nure dependence of Alf nnd AS are op11roximated by a constant <liffcrcnce
`in heat capacity between the native and u11folded stated of 1hc 1>roh.�i11, 6.C". In this
`
`equation. T. is a temperalure where AG is tern; OH, i.s the cmhulpy cha11gc upon
`
`folding at this temperature. am.I 6CP., is the change in ht:at capaci1y upon foldfog.
`
`(6.1)
`
`Data on T •• ac •. and 6H, can be obtained. for example, using differential scan­
`ning calorimetry (DSC)•. P1011ing this data using Equation 6.t wtll yield a parabola
`
`NOleltu.t inalyi,teal DSCcxpcr1menl 1hcprocc1n II folckd al lhCstar1 ol lhecxpcnmcnL rhus. the.Ml
`
`
`
`
`and 6C• obtained are those for the ■nfoldi
`qg PfUCCN
`
`Bausch Health Ireland Exhibit 2009, Page 9 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`112
`
`Pharmaceutical Formulation Dev�loprnent of Peptides and Proteins
`
`40
`
`20
`
`�
`
`a
`
`<t -20 -Y - Lysozyme
`
`--40 ��--�--�--�
`240 280
`T(K)
`
`360
`320
`
`stability of two model pro­
`FIGURE 6.1 Graphical representation of the thcrmodyn1mic
`
`
`as derived from the modified Gibbs-Helmholtz equation
`
`
`teins as a function of temperature
`using numerical da1a from Anjum et al. (2000),
`with r.1 ==
`
`(Equation 6.1). Figure created
`340.4 K, 6Hr = -343 kJ/mol, and 6C,., == -11.45 kJ/mol for myoglobin in a pH 6.1 buffer;
`1;., = 335.7 K, 6-Hr= -372 kJ/mol, and 6C11 • .,;::; - 6.52 kJ/mol for lysozymc in a pl·l 4.8 buffer.
`Biophy.,. Acta 1476, 2000.)
`(Data from Anjum et al.. Biochim.
`
`(Figure 6.1). which is known as the protein stability curve. It has been observed that
`many proteins have their highest thermodynamic stability around 283 K, indepen­
`dent of their melriog temperatures (Rees and Robertson, 2001). This is significantly
`below physiological temperatures for many orgilnisms, probably because some struc­
`LUral flcxjbility is required for activity.
`Figure 6.1 shows there are two crossings where 6.G = 0, suggesting that proteins
`can unfold due 10 both increased as well as decreased temperatures. The latter, cold
`denaturation (Privalov, 1990), usually occurs at temperatures below 270 K and thus
`is less likely to be observed in standard analytical techniques due to ice formation.
`Furthermore, the kinetics of unfolding slows down with decreased 1emperature,
`which may result in kinetic trapping of the protein in its folded structure. Finally, it
`is important to note that under physiological conditions, the magnitude of 6.Gr.,n;i� is
`relatively small, typically ca. IU-50 kJ/mol. This is a rather weak stabilizing inLerac­
`tion, con.idering that a typical hydrogen bond conrribures about 5-30 k.J/niol
`The pathway from unfolded to folded stare is, for m11ny proteins, likely not as sim•
`pie as suggested by Scheme 6.J. Unfolded proteins may assume an enormous number
`of conformational states; indeed, a simple calculation ,hows thar in a typical sample
`of unfolded protein molecules, each molecule is likely to be found in a different co11-
`fon11ational state.• Yet. proteins can spontaneously fold to their native conformation
`within a second. This suggests that each protein molecule musl necessarily follow a
`slightly different palhway 10 the folded state. This complex folding process can be
`conctplUalizcd in terms of a biased random walk. wherein proteins fold via a large
`number of small conformational changes, with 1hc likelihood of any conformational
`change occurring befog biased toward those that lower the overall free energy of the
`protein (Bryngelson et al., 1995). The collection of all possible conformational trajec­
`tories and associated free energies forms an °energy landscape." To belier visualize
`
`coofM­
`and allow each s.mino ncid only two different
`
`
`
`• Tilke, for example, a protein of 100 amino acids,
`
`matio"s. This already yields 1100 = IQ.'-> different p01:en1ial conformat
`
`This exceeds ,he nurnbtr of
`ions.
`
`molcct1Jcs of 3 specific pro1ein on Earth
`
`Bausch Health Ireland Exhibit 2009, Page 10 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`Physical Instability of Peptides and Protein5
`
`113
`
`the high-dimensional space represented by this enormous collection of conformational
`states and energies, the energy landscape is often conceptualized as a "folding fun­
`ael," wherein the vertical position on the Funnel is represenr:uivc of the free energy
`of a gjven conformatfon, and the circumference of the funnel is reprcscn1a1ive of the
`number uf states having a given free energy (Bryngelson et al., 1995). Thus, the large
`number of unfolded states would be found al the top of lhe funnel, and the �ingular
`native state conformation would be found at the funnel bottom (Figure 6.2a).
`
`(a)
`
`Native ,tate(s)
`
`Amorphous
`aggregates
`Ftbrlls
`(bl
`
`FIGURE 6.2 Energy landscape of o. pro1ein. (a) (See color insert.) An idenlizcd folding
`
`
`
`
`fun­
`
`
`
`
`nel for a single protein molecule. At the high Gibbs free energy end (top of picture), lhc protein
`
`
`can adopt rnany diffcrenl conformations; the width oflhc funnel can be viewed as a
`molecule
`
`
`
`
`
`measure of the conformational entropy. Al the bo!tom of the funnel a singular folded state wi1h
`
`
`
`
`
`very 1im1tcdconformationo.l entropy is present. (b) A morcrealisL1c two-dimensional representa­
`
`
`
`
`
`
`tion of the energy landJ;cape or ::i protein. On the left-hand side, the tokling of a single protein i1;
`
`
`
`
`shown; as depleted, there may be several folds with almost the same low Gibbs free energy. Also,
`
`
`
`
`
`there may be II folding intcrmcdiotc(s) and mi,;foldcd species with higher energy which can he
`In the middle and on the right-hand
`
`
`signific:intly populeted due to kinl!licbarriers.
`side possible
`
`
`
`
`
`
`energy states are shown for cmcmbles or protein molecules, resulting in various aggregated
`
`
`
`
`
`
`species (oligomers, fibrils, and amorphow aggregates). These may ha� lower Gibbs free energy
`hul may also be populated
`
`
`due to a large kinetic barrier toward refolding.
`
`than the native prolein,
`
`Bausch Health Ireland Exhibit 2009, Page 11 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`114
`
`Pharmaceutical Formulation Development of Peptides and Protei11s
`
`The energy landscape of proteins can probably be better conceptualized as a
`jagged Funnel (Figure 6.2b), where the protein [old, or a subpopulation of protein
`molecules, may be kinetically trapped in local minima. rather than in the lhcrmo­
`dynam.ically lowest energy state. Nore that the aggregated states, in particular the
`fibrillar state, are shown in Figure 6.2b as being equally or even more thermody­
`namically stable than the native state. That is, the protein fold observed for native
`proteins may well be considered a metastable state, with kinetic barriers preventing
`rapid population of the aggregated and/or fibrillar state (Baldwin el al., 20l l ). In
`the human body, various regulatory processes have developed that are designed to
`degrade and eliminate improperly folded proteins, and thus prevent and/or reduce
`the rate of fibril formation. In a pharmaceutical formulation, there are no processes
`thal remove rnisfolded protein, meaning that aggregation and fibri.llation can occur
`for protejns that are not known to aggregate or fibrillate in vivo, or can occur much
`faster than observed in vivo. Aggregation and fibrillation is further discussed in
`Section 6.4.3.
`The jagged funnel depicted in Figure 6.2b should not be seen as static; changing
`the solution conditions will all'er the re1ative magnitudes of 1hc local minima relative
`to the global minimum, possibly resulting in a new global minimum. This may also
`occur upon binding of a protein to a l igand or to its receptor, if this involves signifi­
`cant changes in protein structure. The energy barriers between the various states will
`likely also change, and may either increase or decrease. This will affect the kinetics
`of the physical degradation processes taking place. As a result, even small changes in
`solution conditions can have a major impact on the main degradation route.
`
`
`6.4 PROTEIN PHYSICAL DEGRADATION
`The physical degradation of proteins refers to any loss in bioactivc protein that docs
`not involve formation or breakage of cJ1emical bonds and is sometimes also referred
`to as denaturation. It can be subdivided in four, often interrelated processes: unfold­
`ing, adsorption, aggregaiion, and precipitation.
`
`6.4.1 PROTEIN U NFOLDING
`In the previous section. the spontaneous folding of a protein into a specific three•
`dimensional sLructurc was discussed. Here, we look al the reverse prm:ess: the
`spontaneous unfolding
`of a protein. sometimes also referred to as denaturation. As
`discussed above, under physiological conditions, the most thermodynamically stable
`state for a single protein molecule (usually) is the folded state. Any deviation from
`physiological conditions. for example, a change in temperature, pH, or ionic strength,
`will change the intramolecular interactions within the protein, as well as the inter•
`actions between protein and water. Thus, oac may expect a change in the protein
`folding stability upon changing 1J1e environment of the protein. As long as those
`changes are fully reversible upon removing the stress factor or upon administration
`to the patient, this may appear irrelevant for a therapeutic protein in a formulation.
`However, as will be discussed in more detail in Section 6.4.3, (parliul) unfolding is
`commonly the first step in protein aggregation, which is often irreversible. Moreover,
`
`Bausch Health Ireland Exhibit 2009, Page 12 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01105
`
`

`

`
`
`Physical Instability of Peptides and Proteins
`
`
`
`
`
`115
`
`(partial) unfolding followed by subsequenl refolding may trap the protein in a non•
`
`
`
`
`
`
`
`
`
`
`
`n::i.live and thus inactive conformation. Finally, unfolded proteins are often more sus­
`
`
`
`
`
`
`ceptible to chemical degradation. Protein unfoliling is thus, in general, delr

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