throbber
Second Edition
`
`Pharmaceutical Formulation
`Development of Peptides
`and Proteins
`
`Edited by
`Lars Hovgoard
`Sven Frokjoer
`Marco van de Weert
`
`CRC Press
`Taylor & Francis Group
`Boca Raton London N ew York
`
`CRC Press Is an imp rint of the
`Taylor & Francis Group, an info rm a business
`
`Bausch Health Ireland Exhibit 2009, Page 1 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`CRCPress
`Taylor & Prnncls Group
`6000 Broken Sound Parkway NW, Suite 300
`Bo a Ralon, fL 33487-2742
`
`<O 2013 by Taylor& Erancis Group, LLC
`CRC Press Is an imprint ofTayior& Francis Group, an lnforma business
`
`No claim to origin•I U.S. Government works
`
`Printed in the United Slates of America on Cid-free paper
`Version Date: 2012 1003
`
`International Standa rd Book Number: 978-1 -4398-5388-7 (Hardback)
`
`This book contains information obtained from authentic an<I highly regarded sources. Reasonable efforts
`have been made to publish reliable data and i nformation, bul the author and publtsher cannot assume
`responsibilit y for the validity of all materials nr the consequences of theic use. The au thors and publishers
`have attemptrd to t race the copyright holders of all material reproduced iu this publication and apologiie to
`copyright holdcrs lfpermi<slon to publish in this form hos not been obtained. lfnnycopyright material has
`not been acknowledged please write and let us know so we may rectify In any l'uture reprint.
`
`Except as permitted unde.r U.S. Copyright Law, no part of thi, book may be reprinted, reproduced, transmit(cid:173)
`ted, or utilized in any form by any electronic, mechanical, or other means. now known or hereafter invented,
`Including photocopying, micro£ilmlng, and recording, or In nny information storage or retrieval system,
`without written permission from lhe publishers,
`
`For permission to photocopy or use materiul elecrronkally from lhis work, please access www.copyright.
`com (http://www.co pyright.com/) or contact the Copyright C learance CMter, Inc. (CCC), 222 Rosewood
`Drive, Danvers, MA 01923, 978-750-8100. CCC is a not-for-profit o r~anization that prnvid&s licenses and
`rcgistrntion for a variety of user,. For organizations that hove been granted a photocopy license by the CCC,
`a scp.trate S)'Sl~m of payment has bec-n arranged.
`
`Trademark Notice, Product occorporate names maybe trademarks or registered trademarks, and are used
`only for Identification and explanation withoul Intent to infringe.
`
`Library of Congress Cataloging-in-P ublicatio n Data
`
`Pharmaceutical formulation development of peptides and proteins / editors, Lars
`Hovgaard , Sven Frukjaer, Marco van de Weert. •- 2nd ed.
`p. ;cm.
`Includes bibliographical referencesand inde><.
`ISBN 978 -l -•1398-5388-7 (hardcover : alk, paper)
`I. Hovganrd, Lars, 1962- 11. Frllkjrer, Sven, 1947- UL Weert, Marco von de, 1973 -
`IDNLM : 1. Pq,tidc Blosynthesis. 2. Chemistr y, Pharmaceutical--melhods, 3. Protein
`Biosynthesis. QU 68}
`
`615.1'9 --dc23
`
`2012030490
`
`Visit the Taylor & Francis Web site •t
`http://www.taylorandfrancis.com
`
`and the CRC Press Web site at
`hllp;//www.crcpress.com
`
`Bausch Health Ireland Exhibit 2009, Page 2 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`Contents
`
`Preface ..................................................................................................................... , vii
`Ed it ors ...................... , ............. ···-· ........................................................................... ,. ix
`Contributors ............................................ , .................................... , ....................... , ..... xi
`
`Chapter 1 Peptide Synthesis .................................................................................. I
`
`K1111d J. Jensen
`
`Chapter 2 Protein Expression ...... ........................................................................ 17
`
`Nanni Din
`
`Ch11ptcr 3 Protein Purification ........................................................................... 35
`
`Lars Hovgaard. Lars Skriver, a,1d Sven Frokjaer
`
`Chapter 4 Characterization of Therapeutic Peptides and Proteins .................... .49
`
`Marco van de Weer/ and Tudor Arvittte
`
`Chapter 5 Chemical Pathways of Peptide aod Protein Degradation ................... 79
`
`Teru110 J. Siahuan. and Chris/fan Schoneich
`
`Chapter 6 Physical lnscabilily of Peptides a nd Proteins ................... .............. .. 107
`
`Marco van de Wee rt a,rd Theodore W Randolph
`
`Chapter 7 Peptide and Protein Derivatives . ., ..... r ................................ , .. , .... ., •• 131
`
`Kristian Str¢mgaard and Thomas H¢eg-Jensen
`
`Chapter 8 Peptides and Proteins as Parenteral Solutions .................. _ .............. 149
`
`Michael J. Akers and Michael R. De Felippis
`
`Chupter 9 Peptides and Proteins as Parenteral Suspensions: An Overview
`of Desiga, DevelopmenL. and Manufacturing Considerations ......... 193
`
`Michael R. DeFelippis and Michael J. Akers
`
`II
`
`Bausch Health Ireland Exhibit 2009, Page 3 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`vi
`
`Contents
`
`Chapter 10 Rational Design of Solid Protein Formulations ........................... u .. 239
`
`Bingq11a11 (Stuart) Wang and Michael J. Pikal
`
`Chapter 11 Peptide and Protein Drug Delivery Systems for Nonparenreral
`Routes of Administration . ................................................................ 269
`
`Ulrik Lyn Rahbek. Fra11tisek H11balek. and Simon Bjerregcwrd
`
`Chapter 12 lmmunogenicity of Therapeutic Proteins .......... , .............................. 297
`
`Grzegorz K,janka, WimJiskoor, Melody Suuerbom.
`Huub Schellekens, and Vera Brinks
`
`Chapter 13 Biosimulation of Peptides and Proteins ............................................ 323
`
`Tt1e S¢eborg, Chrisrian Hove Rasmussen, Erik Mosekilde. and
`Morren Coldi11g-J¢rgense11
`
`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 - IPR2022-01102
`
`

`

`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 Structure ........................................................................ 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 Degradarioo ~ ..................................................................... I 14
`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 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 function of peptides and proteins is highly dependent on their three(cid:173)
`dimensional structuJe. Changes in that strucLUrc, which may arise due to chemical
`or physical processes, may alter or abolish th:n 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 tl1e 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 extcr.
`nal stress factors. The three.dimensional structure of proteins is mostly held log_ethcr
`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 intra- 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 - IPR2022-01102
`
`

`

`108
`
`Pharrnaceulica.1 Formulation Development of Peptides and Proteins
`
`In this chapter, we will discuss the noncovaJent interactions that result in the for(cid:173)
`mation of the specific three-dimensional fold of most proteins, the most important
`stress factors thut may cause changes in that protein fold, and the resulting physical
`instability of the protein. lt should be noted that we will 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 interdependent, 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 instability of proteins in pharmaceutical formulations. Several of
`these strategies will be discussed with more specific examples in other chapters.
`Throughout the chapter, a number of semantic issues will be discussed, which are of
`importance when reading the literatun:. The commonly used terminology within the
`field of protein structure, folding, and stability is not always strictly defined. and defini(cid:173)
`tions may differ over time and depending on the context. Unfortunately, the definitions
`used in a particula r scientific paper are often not explicit, which may lead to confus.ion
`when the reader is insufliciently aware of the different descriptions that are in use.
`
`6.2 PROTEIN STRUCTURE
`
`6.2.1
`
`PEPTIDES, POLYPEPTIDES, ANO PROTEINS
`
`All peptides, polypeptides, and proteins are considered condensation polymers of
`amioo 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 descriptions,
`rather than definitions, of the difference between peptide and protein alone. The cur(cid:173)
`rently most co1nmon 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 indudc several simple dipeptides (i.e ..
`two amino acids linked together), which can have a biological function. Finally, the
`absence or presence of a well-defined tertiary structure has been used to distinguish
`peptides from proteins. Also, this distinction is not without problems; there arc pro•
`teins that are referred to as "natively unfolded," so called because they do not have
`a specific tertiary structure. ln addition, some " peptides" can form fully reversible
`multimeric structures, such as glucagon (forming trimers) (Formisano et al., 1977),
`whiclJ involves the formation of a defined 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 acids a protein.
`
`6.2.2 PROTEIN STRUCTURE: PRIMARY, SECONDARY,
`TERTIARY, ANO QUATERNARY STRUCTURE
`
`The 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 - IPR2022-01102
`
`

`

`Physical Instability of Peptides and Proteins
`
`109
`
`Intra- or interchain cross-links are also prevalent, usually through cystcine residues
`(forming a cystine or S- S bridges).
`T he 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:
`helical structures such as the alpha-helix, pleated structures sut:h as the beta-strand
`and beta-sheet, turn structures such as the beta-turns. and loop structures. The lattec
`are often referred to as "random" structures.
`The three-dimensional alignment of the secondary structural elements is known
`as the tertiary structure of a protein. Tbis 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-helix bundles (Orengo and Thornton, 2005).
`Some proteins exist under physiological conditions as specific muhimeric pro(cid:173)
`teins linked through noncovalent interactions. This multimerization is known as the
`quaternary structure of a protein. Examples of proteins with a quaternary structure
`include hemoglnhin, alpha-crystallin, and HIV-I protease. In general, the hiologi(cid:173)
`ca) function of such mullimeric proteins depends on this roullimerization, but some
`proteins may also form specific (and reversible) nmhirners that arc not biologically
`active. Perhaps the best known example of the lallcr is insulin; insulin forms dimers
`and hexamers 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 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(cid:173)
`gest there are multiple "native" structures. Furthermore, artificially created proteins
`(e.g., fusion proteins created by genetic engineering techniques) may assemble 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 the native structure. T he mechanism of protein folding is discussed
`in the following section.
`
`6.3 PROTEIN FO LDING: WHY DO PROTEINS FO LD?
`
`6.3.1 RO LE OF W ATER AND STABILIZING I NTERACTIONS
`
`The observation that most proteins are folded into a specific structure in simple
`aqueous solutions suggests that foldjng is a therruodynarnically favorable process.
`Many decades of research have been aimed at elucidating why, and how, proteins
`fold (Anfinsen, 1973). Although there are 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 before moving to the
`more complex siwation of a protein in solution.
`For a single protein molecule in the gas phase, there are four fundamental forces
`to take into account. The fi rst is the entropy of the amino acid chain, which tends
`
`Bausch Health Ireland Exhibit 2009, Page 7 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`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. ln
`contrast, hydrogen bonding and van der Waals forces favor folding. Blectrostatic
`interactions may either favor or d isfavor folding, depending on the sign of the charges.
`Experiments with peptides in the gas phas.e have shown that folding can be spontane(cid:173)
`ous (Chin er al., 2006); hence, at least in some circumstances rhe entropy loss upon
`folding 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 et al., 2006).
`Because proteins are typically fouod in an aqueous environment, these gas-phase
`experiments offer only limited insights to the understanding of protein folding under
`solmi.on conditions. The high dielectric constant of water means that the strength of
`electrostatic interactions is signiiicantly reduced, and therefore is a much less impor(cid:173)
`tant driving force for folding, if at all. Moreover, the peptide chain now has the ability
`to form hydrogen bonds with waler, 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 not immediately apparent driving forces for folding.
`And yet, proteins do fold in water, Ao important driving force of 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 el(change 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 (Dil l 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 d issolve. Thus, the negative energetic contribution by distorting
`the dynamic water network needs to be coumerbalanced by the positive contribution
`of 1he solute dissolving, which includes incccased er\tropy of the solute upon dis(cid:173)
`solution as well as hydrogen bonding and van der Waals interactions with the warer
`molecules.Due to the ability to form hydrogen bonds and significant van dcr Waals
`interactions, polar (hydrophilic) compounds dissolve to a much larger extent in water
`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 unfavorable.
`In contrast, the d issolution of the polar amino acids would be a favorable process.
`By folding of the amino acid chain such that the hydrophobic amino acids are hid(cid:173)
`den 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 resuH(cid:173)
`ing " hiding" of nonpolar amino acids io 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 - IPR2022-01102
`
`

`

`Physical Instability of Peptides and Protelns
`
`111
`
`salt bridge formation, and van der Waals interactions between lightly packed resi(cid:173)
`dues (Rose and Wolfenden, 1993). Finally, the ultimate fold may be stabilized by the
`formation of cystines.
`The importance 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. Thal is, even though the overall amino acid sequence may be
`significantly different 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 Shakhnovich, 2001), and mutations iu these amino acids are more
`likely to yield an inactive protein (Guo et al., 2004). As a result, even proteins with a
`mere 30- 40% similarity in amino acid sequence cnn 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 hydrophohic amino acids. In foct, it is likely
`the absence of a significant amount of hydrophobic amino acids, along with many
`charged residues, that allows these proteins to have little tertiary fold (Uversky and
`Dunker, 2010). However, tbey often do have a specific secondary structure, which
`suggests that for amino acid chains, the intrachaio 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 fold in aqueous solution. That means
`that the change in Gibbs free energy upon folding is negative, that is, 6G, < 0, and
`thus the change in Gibbs free energy of unfolding is positive (6G0 > 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) and in a folded state (N) (Scheme 6.L):
`
`(Scheme 6.1)
`
`The change in Gibbs free energy for this folding process can be approximated
`using a modified form of the Gibbs-Helmholtz equation (Equation 6.1), in which the
`temperature dependence of l:i.H and l:i.S are approximated by a constant <lifference
`in heat capacity between :.he native and unfolded stated of the protein, l:i.CP. In this
`equation, T,. is a temperature where l:i.G is zero; l:i.Hf is the enthalpy change upon
`folding at this temperaLure, and t.Cp.r is the change in heal capacity upon folding.
`
`(6.1)
`
`Data on Tm, l:i.Cr, and ti.Hr can he obtained. for example, u.~ing differential scan(cid:173)
`ning calorimetry (DSC)*. Plotting this data using 8quatlon 6.1 will yield a parabola
`
`• Note lhnt in a typical DSC cxp,rimenl the protein is folded111 the start of thecxpcrimcnl. Thus. the ti.fl
`ond <!IC, obtnincd are those for the unfolding process.
`
`Bausch Health Ireland Exhibit 2009, Page 9 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`112
`
`Pharmaceutical Formulation Development of Peptides and Proteins
`
`40
`
`i 20
`~ 0
`J
`<I - 20
`
`240
`
`280
`T (I<)
`
`320
`
`360
`
`FIGURE 6.1 Graphical representation of the thcrmodyn~mic s1ability of two model pro(cid:173)
`teim as a function of temperature as derived (rom the modified Gibbs-Helmholtz equation
`(Equation 6.1). Figure created using numerical data from Anjum el aL (2000), with T,.:
`340.4 K, t:.Hr = -343 kJ/moJ, and t:.C.,,: -1 1.45 kJ/mol for myoglobin in a pH 6.1 buffer:
`7~ = 335.7 K, AH,"' -372 kJ/mol, and t.C9 _., = -6.52 kJ/mol for lysozyme in a pH 4.8 buffer.
`(Data from Anjum el al., Biochim. Biophy.,. Acla 1476, 2000.)
`
`(Figure 6.1), which is known as the protein stability curve. It has been observed lha1
`many proteins have their highest tbermodynamk stability around 283 K, indepen(cid:173)
`dent of their melliog temperatures (Rees and Robertson, 2001). This is significantly
`below physiological temperatures for many organisms, probably because some struc(cid:173)
`tural flexibility is required for activity.
`Figure 6 .1 shows there are two crossings where AG= 0, suggesting that p roteins
`can unfold due to both increased as well as decreased temperatures. The latter, cold
`denaturation (Privalov, 1990), usua.lly occurs at temperatures below 270 K and thus
`is less likely to be observed io standard aoalylical techniques due Lo ice formation.
`Furthermore, the kinetics of unfolding slows down with decreased temperature,
`which may result in kinetic trapping of the protein in its folded structure. Finally, it
`is important 10 note that under physiological conditions, the magnitude of AGr .• .,, is
`relatively small, typically ca. l0-50 kJ/mol. This is a rather weak stabilizing interac(cid:173)
`tion, considering that a typical hydrogen bond conlributes about 5-30 kJ/moL
`The pathway from unfolded to fold\:d stare is. for mnny proteins, likely not as sin,(cid:173)
`pte as suggested by Scheme 6.1. Unfolded proteins may assume an enormous number
`of conform ational states; indeed, a simple calculation shows that in a typical sample
`of unfolded protein molecules, each molecule is likely to be found in a different con(cid:173)
`formational state.* Yet, proteins can spontaneously fold to Lheir native conformalion
`within a second. This suggests that each protein molecule must necessarily follow a
`slightly different pathway to the folded stale. This complex folding process can be
`conceptualized in terms of a biased random walk, wherein proteins fold via a large
`number of small conformational changes, with the likelihood of any conformational
`change occurring being biased toward those that lower the overall free energy of the
`protein (Bryngelson et al., 1995 ). The collection of all possible conformational trajec(cid:173)
`tories and associated free energies forms an "e.nergy landscape." To better visualize
`
`• Take.for example, n protein of 100 amino acids. and allow each amino aoid only two differenl coofor•
`mations. This already yields 2''° = IO'° different potenliat conforma1ions. This exceeds 1hc nurnbe.r of
`molec.ulcs of a sped fie protein o n Ear1h.
`
`Bausch Health Ireland Exhibit 2009, Page 10 of 27
`Mylan v. Bausch Health Ireland - IPR2022-01102
`
`

`

`Physical Instability of Peptides and Proteins
`
`11 3
`
`the high-dimensional space represented by this enormous collection of conformational
`states and energies. the energy landscape is often conceptualized as a "Colding fun(cid:173)
`oel," wherein the vertica I position on the fonnel is representative of the free energy
`of a given conformation. and the circumference of the funnel is representative of the
`number of states having a given free energy (Bryngelson et al., l995). Thus, the large
`number of unfolded states would be found at the top of the funnel, and the ~ingular
`native state conformation would be found at the funnel bottom (Figure 6.2a).
`
`Entropy
`
`(a)
`
`Native state(~)
`
`Amorphous
`Rggregate~
`
`Fibril~
`(b)
`
`FIGURE 6.2 Energy landscape of a protein. (a) (See color insert.) An idealiicd folding fun
`ncl for a single protein molecule. At tbe high Gibbs free energy end (lop of' picture), the protein
`molecule can adopt many different confonnations: the width of the funnel can be viewed as a
`measure of the conformational entropy. At the bonom of the funnel a singular folded state with
`very limilod conformational entropy is present. (b) A more realistic two-dimensional representa(cid:173)
`tion of the energy landscape cf a protein. On the left-hand side. the folding of a single protein is
`shown; as depicted, there may be several fold~ with almost the same low Gibbs free energy. Also,
`there may be a folding intermcdiate(s) and misfoldcd species with higher energy which cnn be
`significantly populated due tokinetie barriers. In the middle and on Lhe right-hand side possible
`energy states are shown for ensembles of protein molecules, resulting in various aggregated
`species (oligomers, fibrils, and amorphous aggregates). These may have lower Gibbs free energy
`than the native protein, but 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 - IPR2022-01102
`
`

`

`114
`
`Pharmaceutical Formulation Development of Peptides and Proteins
`
`The energy landscape of proteins can probably be bener conceptualized as a
`jagged funnel (Figure 6.2b), where the protein fold, or a subpopulation of protein
`molecules, may be kinetically trapped in local minima, rather than in the thermo(cid:173)
`dynamically lowest energy state. Note that the aggregated states , in particular the
`librillar state, are shown in Figure 6.2b as being equally 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 fibrillar state (Baldwin et al., 20ll). Jn
`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. ln a pharmaceutical formulation, there are no processes
`that remove misfolded protein, meaning that aggregation and fibrillation can occur
`for proteins 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
`SecLion 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
`to the global minimum, possibly resulting in a new global minimum. This may also
`occur upon binding of a protein to a ligand or to its receptor, if this involves signifi(cid:173)
`cant changes in protein structure. The energy barriers between the variou. states will
`likely also change, and may either increase or decrease. This win affect the kinelics
`of the physical degradation prm:esses 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 bioactive protein that docs
`not involve formation or breakage of chemical bonds and is sometimes also referred
`to as denaturation. It 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 pro1ein into a specific three(cid:173)
`dimensional structure was di cussed. Here, we look al the reverse process: the
`spontaneous 1111/olding 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 intramolecu lar in teractions within the protein, as well as the inter(cid:173)
`actions between protein and water. Thus, one may expect a change in the protein
`folding stability upon changing U1e 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, (partial) unfolding is
`commoo.ly 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 - IPR2022-01102
`
`

`

`Physical Instability of Peptides and Proteins
`
`115
`
`(par tial) unfolding followed by subsequent refolding m ay trap the protein in a non(cid:173)
`native and thus inactive conformation. Finally, unfolded proteins are often more sus(cid:173)
`ceptible to chemical degradation. Protein unfoldi

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