throbber
Second Edition
`
`Pharmaceutical Formulation
`Development of Peptides
`and Proteins
`
`Edited by
`Lars Hovgaard
`Sven Frokjaer
`Marco van de Weert
`
`CRC Press is an imprint of the
`Taylor 6t Francis Group, an lnforma business
`
`Bausch Health Ireland Exhibit 2009, Page 1 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`CRCr> ress
`Taylor & hancls Group
`60UU Broken Sound Parkway NW. Suite 300
`Ro a Raton, FL 33487-2742
`
`© 2013 by Taylor & E' rancis Group, LLC
`CRC Press is an lmprlnl o(T-aylor& Francis Group.a n Inrorma business
`
`No claim to origina l U.S. Governmen l works
`
`Printed In the United States or America on ucld-free paper
`Version Da te: 2012 1003
`
`lntermllional Standard Rook Number: 978-l -4398-5388-7 (Hardback)
`
`This book co ncains information obt ained from aut hcnrlc and high ly t'egarded sou rces. Reasonable efforts
`hnve been madt to publish reliable data and informa tio n , bul the author -a nd pubHsher cannot assumt>
`responsibilit y fo r the validity of a ll materials or lhe conseque nces of t hei r u~e. The a ut ho rs and pub lishers
`have anempt<'d 10 t race the copyright holde rs or all material reproduced i11 lhis publlcatlon an<( :.ipologiie to
`copyright holders lf pN mi~slon l0 publish in thi s form ha~ not been obtained. If nny copyrigh1 materi.11 has
`nol been ac lcnowledgcd please wrlle ::\nd let us know so we may rectify In a ny futu re repri nt.
`
`Except as permitted under U.S. Copyright Law, no parl oft hi11 book maybe repri nted, reproduced, t r,rnsm it(cid:173)
`ted. or utilized in any form by any electronic, mechan ical, or other mea ns, now known or hereafler invented,
`including photocopy ing. mlcmWming, a nd re~ording, or in any in formation storage or retrieval sys tem,
`without writcen permission from the publishers,
`
`For permiuion to photocopy or use materia l electronkally from t his work. please access www.c.:opyright.
`com (htt p://www.copy right.com/) or contact the Copyrigh t C learance Center, Inc. (CCC), 222 Rosewood
`Dr,vc, Danvers, MA 019'23, 978-750•8400. CCC is a not-for.profit o rga nizat ion that provides licenses and
`regist n1tion fo r a Vilricty of users. For o rganh.at1o ns that have been gra nted a pholoc:opy license by th.e CCC,
`a separate system of payment has bcC'n a rra nged.
`
`Tndemark Notice: Product oc corporate names maybe tredcma rks or registered trademarks, and :ire used
`on ly fo r Identification and explanation W'ilhou l lncenl to infri nge.
`
`Libr:iry of Congress Catalnging-in -Puhlkalion Data.
`
`Pharmaceutical form ulation development of peptides a nd proteins/ ed itors, Lars
`Hovgaard, Sven Frokjaer, Ma rco va n de Weert. -- 2nd ed.
`p. ;cm.
`Incl udes bibliographica l referenc,esand index.
`ISBN 978 -1-•1398-5388-7 (hardcover: alk, paper)
`I. Hovga.ard, Lars) 1962 - II. Frukjrer, Sven, 1947• LI L \Veert, Marco va n de, 1973 -
`fDNL.M: L (>cptidc Blosynthesis, 2, Chemistry, Pha rmaceutical -- melhod.s, 3. Protei n
`Biosynthesis. QU 68J
`
`615.1'9- dc23
`
`2012030490
`
`Visit the Taylor & Francis Web site 11 1
`http://www.taylora ndfranci s.com
`
`and th e CRC Press Web site a t
`http://www.c rcpress.com
`
`Bausch Health Ireland Exhibit 2009, Page 2 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`Contents
`
`Preface .............. , ........................................................................ , .. ,., .......................... vii
`Editors ................... ................... _ .............................................................................. ix
`Contributors .............................................................................................................. xi
`
`Chapter 1 Peptide Synthesis .................................................................................. I
`
`Knud J. Jensen
`
`Chapter 2 Protein Expression .............................................................................. 17
`
`Nanni Din
`
`Chapter 3 Protein Purification ........................................................... _ .............. 35
`
`!.ars Havgaard. !.ars Skriver, and Sven Frokjaer
`
`Chupter 4 Clrnractcrization of Therapeutic Peptides and Proteins .................... .49
`
`Marco van de Weer/ and Tudor Arvlltte
`
`Chapter S Chemical Pathways of Peptide and Protein Degradation ................... 79
`
`Teru11a J. Sia/,aan and Chris/ian Schorreic/1
`
`Chapter 6 Physical Instability of Peptides and Proteins ................................... 107
`
`Marco van de Weer/ a11d Theodore W. Randolph
`
`Chapter 7 Peptide and Protein Derivatives .................................................... 131
`
`Kristian Str<Jmgaard and Thomas l/9eg•Je11sen
`
`Chapter 8 Peptides and Proteins as Parenteral Solutions ................................ 149
`
`Michael J. Akers and Michael R. DeFelippis
`
`Chapter 9 Peptides and Proteins as Parenteral Suspensions: An Overview
`of Design, Development, and Manufacturing Considerations ......... 193
`
`Michael R. DeFelippis and Michael J. Akers
`
`Bausch Health Ireland Exhibit 2009, Page 3 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`vi
`
`Contents
`
`Chapter IO Rational Design of Solid Protein Formulations ···~··· .. ········· .. ··· ....... 239
`Bingq11t111 (Stuarr) Wang tmd Michael J. Pikal
`
`Chapter U Peptide and Protein Drug Delivery Systems for Nonparcmeral
`Routes of Administration ....................................... .......................... 269
`
`Ulrik Lyu Rahbek, Frm,tisek Hubtilek, and Simon Bjerregcwrd
`
`Chapter 12 lmmunogenicity ol'Therapeutic Proteins ......................................... 297
`
`Grzegorz Kijanka, Wim Jiskoo1, Melody Suuerbon,.
`Huub Srhel/ekens, and Vern Brinks
`
`Chapter 13 Biosimulation of Peptides and Proteins ........................................... 323
`
`Tue S¢eborg. Chris1ian Hove Ras11111sse11, Erik Mosekilde. and
`Morten Coldi11g-J¢rge11se11
`
`Chapter 14 Registration of Peptides and Protein~ ............................................. 339
`
`Niamh Kinsella
`
`Index ...................................................................................................................... 363
`
`Bausch Health Ireland Exhibit 2009, Page 4 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`6 Physical Instability of
`Peptides and Proteins
`
`Marco van de Weert and Theodore W. Randolph
`
`CONTENTS
`
`Introduction .................................................................................................. 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 Struciurc .............................................. , .......................... 108
`6.3 Protein 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 Protein Physical Degradation ...................................................................... 114
`6.4. I Protein Unfolding ........... ................................................................ 114
`6.4.2 Adsorption ........................................................................................ l 17
`6.4.3 Protein Aggregation .......................................................................... 118
`6.4.3.1 Aggregation Mechanisms and Kinetics ............................. 11 9
`6.4.3.2 Fibrillation: A Special Case of Protein Aggregation ......... 120
`6.4.4 Protein Precipitation ......................................... ................................ 121
`6.5 Stabilization Strategies ................................................. ................................ 122
`6.6 Concluding Remarks .................................................................................... 125
`References...............................
`................... ...........
`. .......... 126
`
`6.1
`
`INTRODUCTION
`
`The biological funct ion of peptides and proteins is highly dependent on their three·
`djmcnsionaJ structure. Changes in Lha.L structure, which may arise due to chemical
`or physical processes, may alter or abolish that function , or even result in toxicity.
`Thus, it is of importance that a pharmaceutical formulation of therapeutic peptides
`and proteins retains the normal (native) structure of those peptides or proteins, or
`that any changes are fully reversible upon administration to the patient.
`A major difference between proteins and low molecular weight drugs is the com(cid:173)
`plexity of the three-dimensional structure and concomitant sensitivity toward cxtcr.
`nal stress factors. The threc•dimensional structure of proteins is mostly held log.ether
`by noncovalent interactions, such as hydrogen bonds, salt bridges, and van der Waals
`forces. Any stress factor may alter these noncovalent interactions. possibly leading to
`new inlrn• or intermolecular 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 - IPR2022-01104
`
`

`

`108
`
`Pharmaceutical Formulation Develo1pment of Pe ptides and Proteins
`
`In this chapter, we will discuss the noncovalent ;n1eractions that result in the for(cid:173)
`mation of the specific three-dimensional fold of most proteins, the most important
`stress factors that may cause changes in that protciin fold, and the resulting physical
`instability of the protein. It should be noted that we w ill discuss this physical instabil(cid:173)
`ity as a separate issue to the chemical instability discussed in Chapter 5. In reality, the
`two are highly interdepcndem, as chemical destabilization may lead to physical desta(cid:173)
`bilization, and vice versa. This chapter will end with potential stabilization strategics
`to prevent physical ins1ability of proteins in pharmaceutical formu lations. Several of
`these strategics will be discussed with more specific examples in other chapters.
`Throughout the chapter, a number of semantic issues wil I be discussed, which arc of
`importance when reading the literature. The commonly used terminology within the
`fie ld of protein structure, folding, and stability is not always strictly defined. and dcfini•
`Lions may differ over lime and depending on the cont,ext. Unfortunately, the definitions
`used in a particular scientific paper are often not cxpl.icit, which may lead to confus.ion
`when the reader is insufficiently aware of the different descriptions that are in use.
`
`6.2 PROTEIN STRUCTURE
`
`6.2.1
`
`PEPTLDES, POLYPEPTIDES, AND PROTEINS
`
`All peptides, polypeptides, and proteins are considered condensation polymers of
`amino acids, resulting in a linear backbone of alternating amide, C- C, and C-N
`bonds. However, the distinction between peptide, polypeptide, and protein is rather
`diffuse. One may find at least three different and partly overlapping descript ions,
`rather than definitions, of the difference between peptide a nd protein alone. The cur(cid:173)
`rently most common description refers to any peptide chain of more than 50 amino
`ac ids 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 linked together), which can have a, biological function. Finally, the
`absence or presence of a well-defined tertiary struc1:ure has been used to distinguish
`peptides from proteins. Also, this distinction is not without problems; there arc pro(cid:173)
`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 structures, such as glucagon (forming trimers) (Formisano ct al .. 1977),
`which involves lhc formation of a defined three-dimensional structure. The term
`"polypeptide" generally overlaps with that of "peptide" and "protein." The reader
`may thus encounter a ll three terms used in conn,ection with the same biological
`compound. For practical purposes, we have used the first definition, calling every
`compound with more than 50 amino acids a protein.
`
`6.2.2 PROTEIN STRUCTURE: PRIMARY, 5ECONDARll,
`TERTIARY, AND Q uATERNARV STRUCTURE
`
`T he three-dimensional structure of proteins is often subdivided into four types of
`Structure, referred to as the primary, secondary, tertiary, and quaternary structure.
`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 - IPR2022-01104
`
`

`

`Physical Ins tability of Peptides a nd Prote ins
`
`109
`
`Intra- or intcrchain c ross-links are also prevalent, usually through cyslcinc residues
`(forming a cystine or S-S bridges).
`The secondary structure refers to the folding of this backbone into specific struc(cid:173)
`tures, which are defined by the bond angles and hydrogen bonding pattern of the
`amide bond. The secondary structure can roughly be subdivided into four classes:
`helic al struct ures 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" struc tures.
`The three-dimensiona l alignment of the secondary structural elemenls 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 tertiary structures can be described by commonly appearing architectures
`such as barrels or alpha-he lix bundles (Orengo and Thornton. 2005).
`Some proteins exist under physiological conditions as specific multimcric pro(cid:173)
`teins linked through noncovalent interac tions. This multimerization is known as the
`quaternary struc ture of a protein. Examples of proteins with a quaternary struc ture
`include hemoglobin, alpha-crystallin, and HIV-I protease. In general, the biologi(cid:173)
`cal function of such multimeric proteins depends on I.his multimerization, but some
`proteins may also form specific (and reversible) multimers that arc not biologically
`active. Perhaps the best known example of the latter is insulin; insulin forms dimers
`and hcxamers at elevated concentration, especially in the presence of certain diva(cid:173)
`lent metal ions. but is only active as a monomer (Uvcrsky et al .. '2003).
`The ultimate fold of the protein is usually referred 10 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(cid:173)
`gest there are multiple "native" struc tures. l'urthermorc, artificially created proteins
`(e.g., fusion proteins created by genetic engineering techniques) may assemble into
`well-defined folds but have unknown levels of func tio n. It may therefore be easier to
`describe the protein structure under physiological conditions (in terms of pH, ionic
`strength, etc.) as the native Slructure. The mechanism of protein folding is discussed
`in the following section.
`
`6.3 PROTEIN FOLDING: WHY DO PROTEINS FOLD?
`
`6.3.1 ROLE OF WATER AND STABILIZING INTERACTIONS
`
`The observation that most proteins are folded into a specific structure 111 simple
`aqueous solutions suggests that fol ding is a thermodynamically favorable process.
`Many decades of research have been aimed at elucidating why. and how. proteins
`fold (Anfinseo, 1973). Altho ugh there arn still several limitations, it is now possible
`to 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(cid:173)
`ing forces for folding, starting with a protein in the gas phase hcfore moving to the
`more complex situation of a protein in solUlion.
`For a single protein molecule in the gas phase, there are four fundamental forces
`to take into account. The first is the entropy of the amino acid chain. which tends
`
`Bausch Health Ireland Exhibit 2009, Page 7 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`11 0
`
`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. Jn
`contrast, hydrogen bonding and van der Waals forces favor folding. Electrostatic
`interactions may either favorordisfavor folding, depcndingon the sign ofthc charges.
`Experiments with peptides in the gas phase have shown that folding can be spontane(cid:173)
`ous (Chin et al., 2006); hence, at least in some circumstances the entropy loss upon
`foldi ng can be overcome by the enthalpy gain from electrostatic interactions, hydro(cid:173)
`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, these gas-phase
`experiments offer only limited insights to the understanding of protein folding under
`solution conditions. The high dielectric constant of water means that the strength of
`electrostatic interactions is significantly reduced, and therefore is a much less imper(cid:173)
`Lant driving force for folding. if at all. Moreover, the peptide chain now has the ability
`to form hydrogen bonds with water, as well as to interact with water molecules through
`van der Waals forces. Thus, intramolecular interactions like van der Waals forces and
`hydrogen bonds also are 1101 immediately apparent driving forces for folding.
`And yet, proteins do fold in water, Ao important d riving force of this foldi ng 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, fou r 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 a.nd two s lightly fu rther 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. In essence, any solute 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 mailer of accounting: as long as there is a negative
`change in Gibbs free energy for the system as a whole upun dissolution of the solute.
`the compound will d issolve. Thus, the negative energetic contribution by distorting
`the dynamic water network needs to be counterba lanced by the positive contribution
`of the solute dissolving, which includes increased eruropy of the solute upon dis(cid:173)
`solution as well as hydrogen bonding and van der Waals interactions with the water
`molecules. Due 10 the ability to form hydrogen bonds and significant van der Waals
`interactions, polar (hydrophilic) compounds dissolve to a much larger extent in water
`than non polar (hydrophobic) compounds.
`Most proteins contain a significant amount of nonpolar amino acid residues and
`their dissolution in an aqueous environment would be energetically unfavorable.
`In contrast, the dissolution 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(cid:173)
`den from the aqueous s urroundings. 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(cid:173)
`ing "hiding" of nonpolar amino acids in the core of the protein 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 - IPR2022-01104
`
`

`

`Physical Instability of Peptides and Proteins
`
`salt bridge formation, and van der Waals interactions between tightly packed resi(cid:173)
`dues (Rose and Wolfenden, 1993). Finally, the ultimalc fold may be stabilized by the
`formation of cystines.
`The imrortance of the hydrophobic amino acids for protein folding is also sug(cid:173)
`gested by the relatively conservative changes of the core amino acids for similar pro(cid:173)
`teins across species. That is, even though the overall amino acid sequence may be
`significantly differenl for a given protein isolated from various species, the differences
`in the (usually hydrophobic) amino acids forming the core of the protein are usually the
`smallest (Mirny and Shak.bnovich, 2001), and mutations io these amino ucids ure more
`likely to yield an inactive protein (Guo et al., 2004). As a result, even proteins with a
`mere 30- 40% similarity io amino acid sequence can yield very similar protein folds.
`Considering the above, it should be no surprise that natively unfolded proteins
`generally do not contain such a core of hydrophobic amino acids. In fact, it is likely
`the absence of a significant amount of hydrophobic amino acids, along with many
`charged residues, that allows these proteins to have liule tertiary fold (IJvcrsky and
`Dunker, 2010). However, they often do have a specific seconda ry structure, which
`suggests that for amino acid chains, the intrachain hydrogen bonding is more favor(cid:173)
`able than hydrogen bonding tu water.
`
`6.3.2 THE ENERGY LANDSCAPE Of A PROTEIN MotECULE
`
`As discussed above, proteins may spontaneously folu in aqueous solution. Thal means
`that the change in Gibbs free energy upon folding is negative, that is, t>G, < 0, and
`thus the change in Gibbs free energy of unfolding is positive (tlG. > 0). However,
`due to the complex interaction 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 examine a simple two-state reversible folding process between a
`protein in its unfolded stale (U) und in a folded stale (N) (Scheme 6.l):
`
`U~N
`
`(Scheme 6. 1)
`
`The change in Gibbs free energy for this folding process can be approximated
`using a modi tied form of the Gibbs-Helmholtz equation (Equation 6.1), in which the
`temperature dependence of t>H and t>S are approximated by a constant difference
`in heat capacity between ·.he native and unfolded stated of the protein, t>C,. In this
`equation, Tm is a temperalure where tlG is zero; 6.H, is the enthalpy change upon
`fold ing at this temperature, and t>C,.r is the change in heat capacity upon folding.
`
`(6.1)
`
`Data on Tm, t>C,, and AH, can he Obtained. for example, using differential scan(cid:173)
`ning calorimetry (DSC)•. Plotting this data using Equation 6.1 will yield a parabola
`
`• No1e lhnl in a typical DSC cxp::r1ment the prorcin i5 folded ut the s1art of thecxpcrimcnt. Thus. the till
`and llCP obtained ar-c those for the unfolding proc~ss.
`
`Bausch Health Ireland Exhibit 2009, Page 9 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`112
`
`Pharmaceut,cal formula1ion Developn1e11t or Peplides and Proteins
`
`40
`
`20 1 :;;-
`J
`<I -20
`
`0
`
`\
`
`- Mr>alobln
`
`- - - -~
`
`--IO -'----~---.-----.,;Lys~oz-'1_m_•-.
`240
`320
`280
`T(K)
`FIGURE 6.1 Graphical rcpn:sentalion of lhc 1hcrmodyn11mic stobilily of two model pro(cid:173)
`teins 3S u funclion of temperature a!li: denved from lhc mod1fitd G1bbs-Hclmholt1. equation
`(P.qus\lion 6.1). Figure created using numerical d111a from Anjum et al (2000), wilh r~, =
`340.4 K, All,= -343 kl/mol, and AC.,= -J 1.45 kJ/mol for myoglobln in a pH 6.1 buffer:
`7~"' 335.7 K, AH,= - 372 kl/mol, and AC.,,"' -6.52 kJ/mol for lysn,ymc in a pll 4.8 buffer.
`(D:uo Fron, Anjum et al .. Biochim. Biophys. Acea 1476, 2000.)
`
`(Figure 6.1). which is known as the protein stability curve. It hos been observed thar
`many proteins have their highes1 thermodynamic stability around 283 K, indepen(cid:173)
`dent of their melting temperorures (Recs and Robertson, 2001). Thi& is significantly
`below physiological temperatures for many organisms, probably because some struc(cid:173)
`tural llcxibility is required for :ictivity.
`Figure 6.1 shows there are two crossings where AG = 0, suggesting 1ha1 proteins
`can unfold due 10 both increased as well as decrea.ed 1emperntures. The laucr, cold
`denaturation (Privalov, 1990), usually occurs at temperatures below 270 K and 1hus
`is less likely 10 be observed in standard analytical techniques due 10 ,ce formation.
`Funhermore, the kinetics of unfolding slows down with decreased temperature,
`which may result in kine1ic trapping of 1hc protein in its foldled s1ruc1ure. Finally, i1
`is important to no1e that under physiological conditions, the magnitude of lJ.G,..,., is
`relatively smaJJ, typically ca. 10-50 kJ/mol. This 1s a n11her weak stabilizing interac(cid:173)
`tion, con<idering that a typical hydrogen bond conmbures about 5-30 kJ/mol
`The pathway from unfolded to folded state is, for mnny protein•, likely not as sim(cid:173)
`ple as suggested by Scheme 6.J. Unfolded proteins mn)' assume an enormous number
`of conformational states; indeed, a simple calculation shows that in a typical sample
`of unfolded protein molecules, each molecule is likely to be found in u different co11-
`for111a1ional state.• Yet, proteins can spontaneously fold lo their native conformation
`within a second. This suggests that each protein molecule must ntcessarily follow a
`slightly different pathway to the folded state. Thi, complex folding process can be
`conccptuolizcd in terms of a biased random walk, wherein proteins fold via a lnrge
`number of small conformational changes, with the likdihood of any conformational
`clumgc occurring being biased 1oward tho,e rhat lower the overall free energy of the
`protein (Bryngelson el al., 1995). The collection ot all possible cooforma11onal trajec(cid:173)
`tories and associa1ed free energies forms an "tnergy landscape" To belier visualize
`
`• T;ake, ror example, a prot~in of 100 amino acids, and aUow each a.mmo ricld only 1wo diffcren1 coofor(cid:173)
`maIK>fl:S Thr:salrcady yiclds1• = 10-• diffcrcnt potenual conform:ulon&. This exceeds the numbered
`mob:-11.ks of a specific proeein on Earth.
`
`Bausch Health Ireland Exhibit 2009, Page 10 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`Physical Instability of Peptides and Proteins
`
`113
`
`the high-dimensional space represented by this enormous col leclion of conformational
`stales and energies, the energy landscape is often conceptualized as a "folding fun(cid:173)
`nel," wherein the vertical position on the funnel is represemativc of the Free energy
`of a given conformation, and the circumference of the funnel is representative of the
`number of stales having a given free energy (Bryngelson et al., 1995). Thus, the large
`number of ui1foldcd stales would be found al the lop of the funnel, ant! the singular
`native state conformation woultl be found at the funnel boltom (Figure 6.2a).
`
`Entropy
`
`(a)
`
`Amorphous
`"ggregatcs
`
`Fibril,
`(bl
`
`FIGURE 6.2 Energy landscape of n protein. (a) (See color insert .) An idcnlizcd folding fun
`nel for a single protein molecule. At the high Gibbs free energy end (top of picture), the protein
`molecule can adopt. many different confonnations: the widlh of the funnel can be viewed as a
`mca5ure of the conformational entropy. At the bottom of the funnel a singular folded state with
`very limllodconformational enlropy is present. (b) A t00rc realistic two•dimcnsional rcprcscnta•
`lion of the energy land~cnpe of a protein. On the left.hand side. the tolding of a single protein i,;;
`shown; as depicted, there may be several folds with almost the same low Gibbs f rec energy. Also,
`there may be a folding iolcrmcdimc(s) and mi,;;foldcd species with higher energy which cnn be
`significantly popul.-tc:c.J due to kim:lic ba1rit:fs. Jn the middle. and on tlH: right•hund side possible
`energy states are shown for ensembles of protein molecules, resulting in various aggregated
`species (oligomers, fibrils1 and amorphous aggregates). Thcso may haw lower Gibbs free energy
`than lhe native protein. hut may also be populated. due to a large kinetic barrier toward refolding.
`
`Bausch Health Ireland Exhibit 2009, Page 11 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01104
`
`

`

`114
`
`Pharmaceutical Formulation Development of Peptides and Proteins
`
`The energy landscape of proteins can probably be better conceptualized as a
`jagged funnel (Figure 6.2b), where 1hc protein fold, or a subpopul ation of protein
`molecules, may be kinetically trapped in loca l minima, rather than in lhc thermo(cid:173)
`dynamically lowest energy state. Note that the aggregated states, in particular the
`fibrillar state, are show n in Figure 6.2b as being equa lly or even more thermody(cid:173)
`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 fibr ill ar state (Baldwin et al., 20ll ). In
`the human body, various regulato ry processes have developed that are designed to
`degrade and eliminate improperly folded proteins, and thus prevent and/or reduce
`the rate of fibri l formation. In a pharmaceutical formulation, there are no processes
`that remove misfolded protein, mea!ling that aggregation and fibrillation can occur
`for proteins that are not known to aggregate or fibri llate in vivo, or can occur much
`faste r 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 alter the relative magnitudes of the local minima relative
`io the global minimum, possibly resulting in a new global minimum. This may also
`occur upon binding of a protein ID a ligand or 10 its receptor, if this involves signi fi(cid:173)
`ca nt changes in protein structure. The energy barriers between the various Slates will
`likely also change, and may either increase or decrease. This will affect the ki netics
`of the physical degradation processes talcing 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 bioactive protein that docs
`not involve formation or breakage of chemical bonds and is sometimes also referred
`to as dena1ura1ion. ll can be subdivided in four, often interrelated processes: unfold(cid:173)
`ing, adsorption , aggregation, and precipitation.
`
`6.4.1
`
`PROTEIN UNFOLDING
`
`In the previous section, the spontaneous folding of a protein into a specific three(cid:173)
`dimensional structure was discussed. Here, we look at the reverse process: the
`spontaneous 1111[0/ding of a protei n, sometimes also referred to as denaturation. As
`discussed above, under physiologica l conditions, the most thermodynamica lly stable
`state for a single protein molecule (usua lly) is the folded state. Any deviation from
`physiological conditions, for example, a change in temperature, pH, or ionic strength,
`will change the intramolecu lar interactions with in the protein, as well as the inter(cid:173)
`actions between protein and water. Thus, one may expect a change in the protein
`fold ing stabi lity upon changi ng !lie environment of the protein. As long as ll1ose
`changes are fully reversible upon removing the stress factor or upon admi nistration
`ID the patient, this may appear irrelevant for a therapeutic protein in a fo rmulation.
`However, a~ wi ll be discussed in more detail in Section 6.4.3, (partial) unfoldiJ1g 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 - IPR2022-01104
`
`

`

`Physical Instability of Peptides and Proteins
`
`115
`
`(partial) unfolding followed by subsequent refolding may trap lhe protein in a non(cid:173)
`native and thus inactive conformation. Finally, unfolded proteins are often more sus(cid:173

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