throbber
REVIEWS
`
`Effects of Glycosylation on the Stability of
`Protein Pharmaceuticals
`
`RICARDO J. SOLA´ , KAI GRIEBENOW
`
`Laboratory for Applied Biochemistry and Biotechnology, Department of Chemistry, University of Puerto Rico,
`Rı´o Piedras Campus, Facundo Bueso Bldg., Lab-215, PO Box 23346, San Juan 00931-3346, Puerto Rico
`
`Received 21 December 2007; revised 14 May 2008; accepted 19 June 2008
`
`Published online 25 July 2008 in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21504
`
`In recent decades, protein-based therapeutics have substantially expanded
`ABSTRACT:
`the field of molecular pharmacology due to their outstanding potential for the treatment
`of disease. Unfortunately, protein pharmaceuticals display a series of intrinsic physical
`and chemical instability problems during their production, purification, storage, and
`delivery that can adversely impact their final therapeutic efficacies. This has prompted
`an intense search for generalized strategies to engineer the long-term stability of
`proteins during their pharmaceutical employment. Due to the well known effect that
`glycans have in increasing the overall stability of glycoproteins, rational manipulation of
`the glycosylation parameters through glycoengineering could become a promising
`approach to improve both the in vitro and in vivo stability of protein pharmaceuticals.
`The intent of this review is therefore to further the field of protein glycoengineering by
`increasing the general understanding of the mechanisms by which glycosylation
`improves the molecular stability of protein pharmaceuticals. This is achieved by pre-
`senting a survey of the different instabilities displayed by protein pharmaceuticals,
`by addressing which of these instabilities can be improved by glycosylation, and by
`discussing the possible mechanisms by which glycans induce these stabilization
`effects. ß 2008 Wiley-Liss, Inc. and the American Pharmacists Association J Pharm Sci
`98:1223–1245, 2009
`Keywords: biopharmaceutics; biophysical models; chemical stability; glycosylation;
`molecular modeling; physical stability; physicochemical properties; proteins; stabiliza-
`tion; thermodynamics
`
`INTRODUCTION
`
`The employment of proteins as pharmaceutical
`agents has greatly expanded the field of molecular
`pharmacology as these generally display thera-
`peutically favorable properties, such as, higher
`target specificity and pharmacological potency
`
`Correspondence to: Ricardo J. Sola´ and Kai Griebenow
`(Telephone: 787-764-0000 ext 2391/4781; Fax: 787-756-8242;
`E-mail: rsola@bluebottle.com; kai.griebenow@gmail.com)
`
`Journal of Pharmaceutical Sciences, Vol. 98, 1223–1245 (2009)
`ß 2008 Wiley-Liss, Inc. and the American Pharmacists Association
`
`when compared to traditional small molecule
`drugs.1,2 Unfortunately, the structural instability
`issues generally displayed by this class of
`molecules still remain one of the biggest chal-
`lenges to their pharmaceutical employment, as
`these can negatively impact their final therapeu-
`tic efficacies (Tab. 1).2–50 In contrast to traditional
`small molecule drugs whose physicochemical
`properties and structural stabilities are often
`much simpler to predict and control, the struc-
`tural complexity and diversity arising due to the
`macromolecular nature of proteins has hampered
`the development of predictive methods and
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`1223
`
`Exhibit 2074
`Page 01 of 23
`
`

`

`mStabilizationismostlyachievedbykeepingtheproteinawayfromdenaturinginterfacesorbysimplyavoidingsuchinterfacesaltogether.39,42,46,76
`lPrecipitationpriortotheprocedureaffordedstabilization.
`kThemechanismofstabilizationisbelievedtobeacombinationofhydrogen-bondformingpropensityandincreaseintheglasstransitiontemperatureinthesolid.23
`jSuchexcipientsincludesugars,polyols,andaminoacidsthatstabilizeproteinstructurebyso-calledpreferentialexclusion.2,75
`iMilddetergentsatlowconcentrationcanpreventdetrimentalinteractionsofproteinswithhydrophobicsurfaces/interfaces.42
`hToremovemetalions.2
`gOtherprominentchemicalinstabilitiesareoxidationsanddisulfidescrambling.2
`fAprominentpathwaytoaggregationisbyso-calledsulfide–disulfideinterchange.11
`eThepotentiallymostharmfulsurfacesarehydrophobic,e.g.,Teflon.45
`dControloftemperaturecanbenontrivialwhenultrasonicationisbeingusedbecauseoflocalheatingevents.
`cContaminating(transition)metalionsandproteasescancatalyzefragmentations.22
`bThesoleFDAapprovedformulationthusfarconsistsintheencapsulationoftheproteininmicrospherescomprisedofpoly(lactic-co-glycolic)acid.
`aThereferencescitedincludemanyreviewstowhichtheinterestedreaderisreferredtofordetails.
` CovalentmodificationascountermeasuresareexcludedinthetablebecausetheyarediscussedinthepaperandinTable2forglycosylatedproteins.
`
`1224
`
`SOLA´ AND GRIEBENOW
`
`39–44,77
`
`31–38,74
`
`4,16–18,30
`
`4,18,23–29,48,73
`
`50,68–72
`
`interfacesm
`avoidanceofwater/organic
`andstabilizingexcipients,j
`
`mechanicalstress
`hydrophobicsurfaces,e
`
`Additionofsurfaceactivei
`
`Aggregation,finactivation
`
`Liquid–organicsolventinterface,
`
`precipitationl
`
`formulationsb
`
`Sustained-release
`
`Spray-freezedrying
`
`Similartolyophilization,
`
`Similartolyophilization
`
`Liquid–airinterface,dehydration
`
`Spray-drying,
`
`Similartolyophilization
`stabilizingexcipientsj,k
`surfaceactiveiand
`Colyophilizationwith
`
`stabilizingexcipientsj
`surfaceactiveiand
`antioxidants,additionof
`chelatingagents,h
`
`inactivation
`oxidation,deamidation,
`
`contacts,moisturef
`
`Aggregation,ffragmentation,
`
`Contaminations,cprotein–protein
`
`Solid-phasestorage
`
`Aggregation,finactivation
`
`inactivation
`adsorption,aggregation,f
`deamidation,gdenaturation,
`b-elimination,racemization,
`crosslinking,
`hydrolysis,oxidation,
`
`dehydration,phaseseparation
`Ice–waterinterface,pHchanges,
`
`Lyophilization
`
`interfaces,hydrophobicsurfacese
`freezethawing,amphipatic
`highproteinconcentrations,
`temperature,dchemicaldenaturants,
`
`2,5–12,19–22,47,49,
`
`ControlofpHandtemperature,
`
`Fragmentations,chemical
`
`Contaminations,cextremesofpH,
`
`Liquidstorage
`
`2,5,6,10,19–22,68–72
`
`andstabilizingexcipientsj
`additionofsurfaceactivei
`agents,hantioxidants,
`temperature,chelating
`controlofpHand
`Proteaseinhibitors,
`
`inactivation
`adsorption,aggregation,f
`deamidation,gdenaturation,
`crosslinking,oxidation,
`hydrolysis,fragmentations,
`
`surfacese
`amphipaticinterfaces,hydrophobic
`highsaltandproteinconcentrations,
`temperature,dchemicaldenaturants,
`extremesofpH,highpressures,
`
`Proteolyticandchemical
`
`Proteases,contaminations,c
`
`Refs.a
`
`TypicalCountermeasures
`
`MainDegradationPathways
`
`MainStressFactors
`
`Purification
`
`Process
`
`Table1.ChemicalandPhysicalInstabilitiesEncounteredbyProtein-BasedPharmaceuticalsandTypicalCountermeasures
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`DOI 10.1002/jps
`
`Exhibit 2074
`Page 02 of 23
`
`

`

`EFFECTS OF GLYCOSYLATION ON PROTEIN STABILITY
`
`1225
`
`generalized strategies concerning their chemical
`as well as their physical stabilizations.51,52 While
`the protein primary structure is subject to the
`same chemical instability issues as traditional
`small molecule therapeutics (e.g., acid-base and
`redox chemistry, chemical fragmentation, etc.),
`the higher levels of protein structure (e.g.,
`secondary, tertiary) often necessary for therapeu-
`tic efficacy can also result in additional physical
`instability issues (e.g.,
`irreversible conforma-
`tional changes, local and global unfolding) due
`to their noncovalent nature.2,15,53–55 The innate
`propensity of proteins to undergo structural
`changes coupled with the fact that there is only
`a marginal difference in thermodynamic stability
`between their folded and unfolded states provides
`a significant hurdle for the long-term stabilization
`of protein pharmaceuticals. This is due to the fact
`that a thermodynamically stabilized protein could
`still inactivate kinetically even at the relatively
`low temperatures used during storage.2,53,55–59
`Additionally, as a result of their colloidal nature,
`proteins are prone to pH, temperature, and
`concentration dependant precipitation, surface
`adsorption, and nonnative supramolecular aggre-
`gation.11,14,20,47,60–65 These instability issues are
`further compounded by the fact that the various
`levels of protein structure can become perturbed
`differently depending on the physicochemical
`environment to which the protein is exposed.2
`This is of special relevance in a pharmaceutical
`production setting where proteins can be simul-
`taneously exposed to several destabilizing envir-
`onments during their production, purification,
`storage, and delivery (Tab. 1).
`Due to these stability problems much emphasis
`has been given to the development of strategies for
`the effective long-term stabilization of protein
`pharmaceuticals.2,4,11,61,66–77 These include exter-
`nal stabilization by influencing the properties of
`the surrounding solvent through the use of
`stabilizing excipients (e.g., amino acids, sugars,
`polyols) and internal stabilization by altering the
`structural characteristics of the protein through
`chemical modifications (e.g., mutations, glycosy-
`lation, pegylation).2,53,58 While many protein
`pharmaceuticals have been successfully formu-
`lated by employing stabilizing mutations, excipi-
`ents, and pegylation, their use can sometimes be
`problematic due to limitations, such as, predicting
`the stabilizing nature of amino acid substitutions,
`the occurrence of protein and excipient dependant
`nongeneralized stabilization effects, protein/
`excipient phase separation upon freezing, cross-
`
`reactions between some excipients and the multi-
`ple chemical functionalities present in proteins,
`acceleration of certain chemical (e.g., aspartate
`isomerization) and physical (e.g., aggregation)
`instabilities by some excipients (e.g., sorbitol,
`glycerol, sucrose), detection interferences caused
`by some sugar excipients during various protein
`analysis methods, and safety concerns regarding
`the long-term use of pegylated proteins in vivo
`due to possible PEG induced immunogenecity
`and chronic accumulation toxicity resulting
`from its reduced degradation and clearance
`rates.2,4,33,48,66,78–95
`Due to these limitations, there is still a need
`for further development of additional strategies of
`protein stabilization.2 Amongst
`the chemical
`modification methods, glycosylation represents
`one of the most promising approaches as it is
`generally perceived that through manipulation of
`key glycosylation parameters (e.g., glycosylation
`degree, glycan size and glycan structural compo-
`sition) the protein’s molecular stability could be
`engineered as desired.2,66,96–105 In this context, it
`is important to highlight the fact that glycosyla-
`tion has been reported to simultaneously stabilize
`a variety of proteins against almost all of the
`major physicochemical instabilities encountered
`during their pharmaceutical employment (Tab. 2),
`suggesting the generality of these effects.
`Even though a vast amount of studies have
`evidenced the fact that glycosylation can lead to
`enhanced molecular stabilities and therapeutic
`efficacies for protein pharmaceuticals (Tab. 3), an
`encompassing perspective on this subject is still
`missing due to the lack of a comprehensive review
`of the literature. The intent of this article is
`therefore to further the field of protein glycoengi-
`neering by increasing the general understanding of
`the mechanisms by which glycosylation improves
`the molecular stability of protein pharmaceuticals.
`This is achieved by presenting a survey of the
`different instabilities displayed by protein phar-
`maceuticals, by addressing which of these instabil-
`ities can be improved by glycosylation, and by
`discussing the possible mechanisms by which
`glycans induce these stabilization effects.
`
`PROTEIN GLYCOSYLATION
`
`Protein glycosylation is one of the most common
`structural modifications employed by biological
`systems to expand proteome diversity.106–108
`Evolutionarily,
`glycosylation
`is widespread
`found to occur in proteins through the main
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`Exhibit 2074
`Page 03 of 23
`
`

`

`1226
`
`SOLA´ AND GRIEBENOW
`
`Table 2. Protein Instabilities Improved by Glycosylation
`
`Instability
`
`Proteolytic degradation
`Oxidation
`Chemical crosslinking
`pH denaturation
`Chemical denaturation
`Heating denaturation
`
`Freezing denaturation
`Precipitation
`Kinetic inactivation
`Aggregation
`
`Refs.
`
`96,121–141
`145
`97,146,149
`124,137,171–178
`136,164,171,172,181–185,187,188
`98,101–103,119,124,128,129,146,149,159,170,171,181,182,
`188–195,202,204,205
`201
`159–165
`101,103,136,146,186,212–218
`97,101,103,130,218,222
`
`eubacteria, and
`(archaea,
`life
`of
`domains
`eukarya).109,110 The prevalence of glycosylation
`is such that it has been estimated that 50% of all
`proteins are glycosylated.111 Functionally, glyco-
`sylation has been shown to influence a variety of
`critical biological processes at both the cellular
`(e.g., intracellular targeting) and protein levels
`(e.g., protein–protein binding, protein molecular
`stability).103 It should therefore not come as a
`surprise that a substantial fraction of the cur-
`rently approved protein pharmaceuticals need to
`be properly glycosylated to exhibit optimal ther-
`apeutic efficacy.100,112
`Structurally, glycosylation is highly complex
`due to the fact that there can be heterogeneity
`with respect to the site of glycan attachment
`(macroheterogeneity) and with respect to the
`glycan’s structure (microheterogeneity). Although
`many protein residues have been found to be
`glycosylated with a variety of glycans (for a
`detailed discussion see review by Sears and
`Wong), in humans the most prevalent glycosyla-
`tion sites occur at asparagine residues (N-linked
`glycosylation through Asn-X-Thr/Ser recognition
`sequence) and at serine or threonine residues
`(O-linked glycosylation) with the following mono-
`saccharides: fucose, galactose, mannose (Man),
`N-acetylglucosamine (GlcNAc), N-acetylgalacto-
`samine, and sialic acid (N-acetylneuraminic
`acid).109,113–115 Since all of the potential glycosy-
`lation sites are not simultaneously occupied this
`leads to the formation of glycoforms with differ-
`ences in the number of attached glycans. Further
`structural complexity can occur due to variability
`in
`the
`glycan’s monosaccharide
`sequence
`order, branching pattern, and length. In humans
`N-linked glycan structures are classified in three
`principal categories according to their monosac-
`
`charide content and structure: high mannose
`type (Man2-6Man3GlcNAc2), mixed type (GlcNAc2-
`Man3GlcNAc2), and hybrid type (Man3GlcNAc-
`Man3GlcNAc2).113 The terminal ends of these
`glycans are often further functionalized with
`chemically charged groups (e.g., phosphates,
`sulfates, carboxylic acids) in human glycopro-
`teins, leading to even greater structural diversity.
`These charged glycans most probably impact to
`some degree the overall stability of glycoproteins
`since they can alter their
`isoelectric point
`(pI).116,117 Some of these charged terminal glycans
`(e.g., sialic acid) have also been found to be critical
`in regulating the circulatory half-life of glycopro-
`teins. This has led to the development of
`glycosylation as a novel strategy to improve the
`therapeutic efficacies of protein pharmaceuticals
`by engineering their pharmacokinetic profiles (for
`a detailed discussion see the recent review by
`Sinclair and Elliot).100
`Due to the high degree of structural variability
`arising from physiological (natural) glycosylation,
`novel strategies are currently being pursued to
`create structurally homogeneous pharmaceutical
`glycoproteins with humanized glycosylation pat-
`terns.118 These include engineered glycoprotein
`expression systems (e.g., yeast, plant, and mam-
`malian cells) as well as enzymatic, chemical, and
`chemo-enzymatic in vitro glycosylation remodel-
`ing methods. Alternatively, to understand the
`mechanisms by which glycosylation influences
`protein physicochemical properties researchers
`have employed comparatively simpler glycosyla-
`tion strategies. These include enzymatic deglyco-
`sylation of natural glycoproteins,
`chemical
`glycosylation via the use of structurally simple
`chemically activated glycans, and glycation of the
`lysine residues with reducing sugars via the
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`DOI 10.1002/jps
`
`Exhibit 2074
`Page 04 of 23
`
`

`

`EFFECTS OF GLYCOSYLATION ON PROTEIN STABILITY
`
`1227
`
`(Continued)
`
`132
`
`149,159,160
`
`97
`
`142,194,195
`
`145,171,216,221
`
`127
`
`191
`
`101–103,188
`
`126
`
`181
`
`193
`
`161
`
`4
`
`3
`
`2
`
`b
`
`1
`
`2
`
`10
`
`b
`
`11
`
`3
`
`6
`
`3
`
`proteolyticdegradation
`
`Protectsagainst
`
`aggregation
`thermaldenaturation,and
`crosslinking,precipitation,
`
`Protectsagainstdisulfide
`
`crosslinkingandaggregation
`
`Protectsagainstnondisulfide
`
`thermaldenaturation
`
`Protectsagainstproteolysisand
`
`andaggregation
`inactivation,
`denaturation,kinetic
`thermal,chemical,andpH
`
`Protectsagainstoxidation,
`
`degradation
`
`Protectsagainstproteolytic
`
`denaturation
`
`Protectsagainstthermal
`
`denaturationandaggregation
`chemical,andkinetic
`
`Protectsagainstthermal,
`
`degradation
`
`Protectsagainstproteolytic
`andthermaldenaturation
`
`Protectsagainstchemical
`
`denaturation
`
`Protectsagainstthermal
`
`andprecipitation
`
`Protectsagainstaggregation
`
`Refs.
`
`Glycan(#)
`
`EffectsofGlycosylation
`
`granulomatousdisease
`
`Actimmune1(Intermune)Treatmentofchronic
`
`Interferongamma-1b
`
`Treatmentofmultiplesclerosis
`
`(Pfizer/EMDSerono)
`
`Avonex1(Biogen),Rebif1
`
`Interferonbeta-1a(rHuInf-b1)
`
`Treatmentofdiabetes
`
`Multipleindications
`
`a
`
`a
`
`Insulin
`
`IgG-likeantibodies
`
`withchronicrenalfailure(CRF)
`
`Treatmentofanemiaassociated
`
`Biotech)
`
`Epogen1(Amgen),
`
`Procrit1(Ortho
`
`Epoetinalfa
`ProteinC)
`
`Treatmentofseveresepsis
`
`Xigris1(EliLilly)
`
`Drotrecoginalfa(CF-XIV,
`
`Gonal-F1(EMDSerono)Treatmentofinfertility
`
`Corifollitropinalfa(FSH)
`
`myeloma
`
`Nutraceuticals)
`
`Adjuncttherapyformultiple
`
`insufficiency
`relatedtoexocrinepancreatic
`
`Treatmentoflipidmalabsorption
`
`deficiencywithemphysema
`
`Treatmentofcongenitala1-AT
`
`WobeMugos1(Marlyn
`
`Therapeutics)
`
`Merispase1(Meristem
`
`Biotherapeutics)
`
`Prolastin1(Talecris
`
`Chymotrypsin
`
`Bucelipasealfa(cholesterol
`
`esterase)
`
`Alpha1-antitrypsin(a1-AT)
`
`TreatmentofPompedisease
`
`Myozyme1(Shire)
`
`Alglucosidasealfa(a-glucosidase)
`
`TreatmentofFabrydisease
`
`Replagal1(Shire)
`
`Agalsidasealfa(galactosidase)
`
`Indication
`
`BrandName(Company)
`
`INN
`
`Table3.PartialListofApprovedProtein-BasedPharmaceuticalProductsStabilizedbyGlycosylation
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`Exhibit 2074
`Page 05 of 23
`
`

`

`1228
`
`SOLA´ AND GRIEBENOW
`
`131,192
`
`130
`
`124,125,146,170
`
`128,129,189,190
`
`b
`
`8
`
`3
`
`2
`
`124,125,146,170
`
`1
`
`denaturation
`degradationandthermal
`Protectsagainstproteolytic
`
`degradationandaggregation
`
`Protectsagainstproteolytic
`
`andkineticinactivation
`thermalandpHdenaturation,
`degradation,
`crosslinking,proteolytic
`Protectsagainstdisulfide
`
`denaturation
`degradationandthermal
`Protectsagainstproteolytic
`
`kineticinactivation
`pHdenaturation,and
`degradation,thermaland
`crosslinking,proteolytic
`Protectsagainstdisulfide
`
`bCommerciallyavailableproteinisnotglycosylated.
`aMultipleapprovedproducts.Furtherinformationavailableatwww.fda.govandwww.biopharma.com.
`INN,Internationalnonproprietaryname.
`InformationwasobtainedfromthePrescribingInformation(PI)foreachproduct.
`
`pulmonaryemboli
`
`Therapeutics)
`
`Treatmentofacutemassive
`
`andhypothyroidism
`
`Abbokinase1(ImaRx
`
`Urokinasealfa
`
`Thyrogen1(Genzyme)Detectionofthyroidcancer
`
`Thyrotropinalfa(TSH)
`
`myelogenusleukemia
`chemotherapywithacute
`Leukin1(BayerHealthcare)Treatmentafterinduction
`
`Sargramostin(G-CSF)
`
`mesothelioma
`
`Onconase1(AlfacellCorp.)Treatmentofmalignant
`
`Ranpirnase(RNAse)
`
`inducedneutropenia
`
`(ChugaiPharma)
`
`Treatmentofchemotherapy
`
`Granocyte1
`
`Lenograstim(G-CSF)
`
`Refs.
`
`Glycan(#)
`
`EffectsofGlycosylation
`
`Indication
`
`BrandName(Company)
`
`INN
`
`Table3.(Continued)
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`DOI 10.1002/jps
`
`Exhibit 2074
`Page 06 of 23
`
`

`

`EFFECTS OF GLYCOSYLATION ON PROTEIN STABILITY
`
`1229
`
`Maillard reaction. Although some of these glyco-
`sylation methods (e.g., glycation) may be unde-
`sired for use in protein pharmaceuticals their
`fundamental scientific value for the understand-
`ing the effects of glycosylation on protein stability
`cannot be ignored.119 This is due to the fact
`that independently of the method by which the
`structurally different glycans are attached to the
`protein surface (e.g., enzymatic and chemical
`glycosylation, or reductive glycation) they all
`seem to induce similar stabilization effects.103
`In the next sections, we thus focus on discussing
`which pharmaceutically relevant chemical and
`physical protein instabilities have been reported
`to be ameliorated by glycosylation and discuss
`possible mechanisms by which glycans achieve
`such effects.
`
`CHEMICAL INSTABILITIES PREVENTED BY
`GLYCOSYLATION
`
`The presence of multiple reactive chemical
`functionalities in the amino acids side chains of
`proteins makes them particularly sensitive to
`several chemical degradation processes. These
`can include: glutamine (Gln) and asparagine
`(Asn) deamidation; histidine (His), methionine
`(Met), cysteine (Cys),
`tryptophan (Trp), and
`tyrosine (Tyr) oxidation; serine (Ser), threonine
`(Thr), phenylalanine (Phe), lysine (Lys), and Cys
`b-elimination; disulfide fragmentation, exchange,
`and crosslinking; backbone peptide hydrolysis
`caused either by proteases or by pH sensitive
`backbone sequences (e.g., aspartic acid-proline
`(Asp-X));
`transamidation;
`racemization; and
`chemically triggered nonspecific
`crosslinking
`(Tab. 1).2,6,8,15,54,55,112 For further detailed dis-
`cussions on the general mechanisms which
`trigger these chemical instabilities the reader is
`referred to several excellent reviews on the
`subject.2,6,8,9,12,55 In the next section, we focus
`on those chemical instabilities which have been
`reported to be improved by glycosylation (e.g.,
`proteolytic degradation, oxidation, and chemical
`crosslinking) (Tab. 2).
`
`Proteolytic Degradation
`
`Protein pharmaceuticals are typically adminis-
`tered intravenously and not via the oral route due
`to their chemical degradation by the proteases of
`
`the digestive system.120 However, the systemic
`expression of proteases also makes proteins
`administered by other routes highly susceptibly
`to proteolytic degradation.120 Therefore,
`the
`in vivo molecular stability and therapeutic
`efficacy of protein pharmaceuticals is intimately
`related to their stability towards proteolytic
`degradation.2,6,100,120 In general, glycosylation
`has been found to protect proteins against
`proteolytic degradation.96,121–123 Some examples
`include granulocyte colony stimulating factor
`(GRANOCYTE1, Chugai Pharma,
`(G-CSF)
`(MERISPASE1;
`Tokyo, Japan),124,125
`lipase
`Meristem Therapeutics,
`Clermont-Ferrand,
`France),126 protein C (XIGRIS1; Eli Lilly,
`(ONCONASE1; Alfacell
`IN),127
`ribonuclease
`Corp., NJ),128,129 thyroid-stimulating hormone
`(THYROGEN1; Genzyme, MA),130 urokinase
`(ABBOKINASE1; ImaRx Therapeutics, AZ),131
`interferon-g (ACTIMMUNE1; Intermune, CA),132
`streptokinase,133 cellulose,134 ovomucoid,135 amy-
`lase,136,137 lysosomal integral membrane proteins
`Lamp-1 and Lamp-2,138 peroxidase,139 and cata-
`lase.140 There is also evidence that this proteolytic
`stability can be engineered into proteins as was
`described by Holcenberg et al.141 upon chemical
`glycosylation of asparaginase and by Raju and
`Scallon142 upon enzymatic glycosylation of IgG-
`like antibodies. Particularly, in this last study it
`was found that altering the end-terminal glycan
`structures (e.g., N-acetylglucosamine, galactose,
`and sialic acid) led to increasingly greater in vitro
`proteolytic stability when subjected to papain
`digestion.142 Mechanistically,
`it has been pro-
`posed that this proteolytic stability arises due to
`the fact that the glycan’s presence provides a
`steric hindrance around the peptide backbone of
`the amino acids adjacent to the glycosylation
`site.114,115,143 This prevents the contact between
`the glycoprotein’s surface and the cleaving pro-
`tease’s active site.
`
`Oxidation
`
`Protein pharmaceuticals can potentially lose their
`bioactivity during their manufacture and storage
`due to the oxidation of several of their amino
`acid side chains (His, Met, Cys, Trp, and
`Tyr).2,6,9,22,55,144 These oxidation events have
`been mainly attributed to the production of active
`oxygen-based radicals in protein formulations due
`to the combination of trace amounts of transition
`metals, atmospheric oxygen, and exposure to
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`Exhibit 2074
`Page 07 of 23
`
`

`

`1230
`
`SOLA´ AND GRIEBENOW
`
`ultraviolet light.2,6 Thus far, erythropoietin (EPO-
`GEN1, PROCIT1; Amgen, CA, Ortho Biotech,
`NJ) is the sole reported case of a protein whose
`bioactivity can be impacted by oxidation and
`where glycosylation has been found to ameliorate
`this chemical instability.145 The loss of bioactivity
`for this protein was found to correlate with the
`levels of tryptophan oxidation when exposed to
`oxidizing conditions.145 Comparison of the oxida-
`tive susceptibility for the naturally glycosylated
`erythropoietin with that of its deglycosylated form
`revealed that glycosylation diminished the tryp-
`tophan oxidation rates and the inactivation of
`this protein.145 These results suggest that glycosy-
`lation can protect the protein structure from
`damage by active oxygen radicals although more
`studies are still needed to shed some light on the
`mechanisms of this stabilization and to determine
`the extent to which engineered glycosylation could
`prevent this type of instability. Also, whether this
`stabilizing effect is specific to when the glycans
`are chemically attached to the protein surface or
`nonspecific having to do more with the radical
`scavenging capabilities of the glycans remains to
`be established.70
`
`Chemical Crosslinking
`
`Protein therapeutics can form covalent dimers
`and oligomers due to polymerization triggered by
`both disulfide and nondisulfide crosslinking path-
`ways.2,6 Preventing the formation of these cova-
`lently linked species in protein pharmaceuticals is
`important as these frequently lead to loss of
`bioactivity.2,6 Additionally, for many proteins it
`has been found that this type of instability, in
`addition to protein unfolding, could trigger the
`formation of larger soluble and insoluble protein
`aggregates.2,6,11 There are several reports in the
`literature were it has been found that glycosyla-
`tion prevents the formation of these crosslinked
`species. For example, Oh-eda et al.146 reported
`that the presence of the single glycan in human
`granulocyte colony-stimulating factor (G-CSF)
`(GRANOCYTE1; Chugai Pharma) prevented
`the polymerization-induced inactivation of the
`protein. The mechanism by which G-CSF poly-
`merizes was studied by Krishnan et al. and Raso
`et al. and found to be due to disulfide cross-
`linking.147,148 Interferon beta (REBIF1, Pfizer,
`NY/Serono, Geneva, Switzerland; AVONEX1,
`Biogen, MA) is another example of a therapeu-
`tically relevant protein where glycosylation
`
`prevents its inactivation due to disulfide cross-
`linking.149 Glycosylation has been also reported to
`prevent nondisulfide protein crosslinking. For
`example, Baudys et al.97 reported that engineered
`chemical glycosylation of insulin, especially at the
`PheB-1 amino group, suppressed the self-associa-
`tion of the protein into dimers and oligomeric
`species. The formation of these crosslinked insulin
`species occurs due to a transamidation reaction
`between AsnA-21 and PheB-1.2 This finding is
`highly significant since it demonstrates that this
`type of stabilization can also be engineered into
`proteins via rationally designed glycosylation.
`These results additionally suggest
`that
`the
`mechanism by which this type of instability is
`prevented is due to increased intermolecular
`steric repulsion between the crosslinking-prone
`protein species due to the glycan’s presence at the
`protein surface.
`
`PHYSICAL INSTABILITIES PREVENTED
`BY GLYCOSYLATION
`
`The functional efficacy of proteins critically
`depends on the conformational stability of their
`natively folded state.2 Most proteins adopt a
`tertiary structure by folding as to minimize the
`exposure of their hydrophobic residues in aqueous
`solution.56,150–152 This creates a compact native
`state with a hydrophobic core that is additionally
`energetically stabilized by the presence of several
`types
`of
`atomic
`interactions within
`the
`protein core (e.g., electrostatic and charge–charge
`interactions, hydrogen bonds, Van der Waals
`interactions).151–154 Unfortunately, the resulting
`thermodynamic and kinetic stability of this state
`tends to be intrinsically low due to the noncova-
`lent nature of these forces.2,53 Therefore, any
`physical or chemical phenomena which can
`disrupt these forces will trigger either small or
`large scale protein structural changes. These
`conformationally altered species are more prone
`to interact either with themselves or with the
`hydrophobic surfaces and interfaces present
`during protein manufacturing and storage lead-
`ing to additionally physical instabilities, such as,
`adsorption, aggregation, and precipitation.2,42
`Examples of pharmaceutically relevant phenom-
`ena that can lead to protein physical instability
`include exposure to extremes of temperature and
`pH; exposure to amphipatic interfaces (e.g.,
`aqueous/organic solvent, aqueous/air), hydropho-
`bic surfaces, and chemical denaturants; and
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 4, APRIL 2009
`
`DOI 10.1002/jps
`
`Exhibit 2074
`Page 08 of 23
`
`

`

`EFFECTS OF GLYCOSYLATION ON PROTEIN STABILITY
`
`1231
`
`formulation at extreme protein concentrations
`(Tab. 1). For further detailed discussions on the
`general mechanisms which trigger these physical
`instabilities the reader is again referred to
`a series of excellent
`reviews on the sub-
`ject.2,6,8,9,11,12,14,42,61,63,64 In the next section, we
`focus on those physical protein instabilities which
`have been reported to be improved by glycosyla-
`tion (e.g., precipitation; pH, chemical, and ther-
`mal denaturation; and aggregation) (Tab. 2).
`
`increased linearly as the glycosylation degree was
`increased (Fig. 2A).103,166 The linear dependence
`of these results are agreement with the solubility
`findings of Tams et al.164 These results therefore
`suggest the mechanism by which glycosylation
`increases protein solubility is due to an increase in
`the number of possible interactions between the
`glycoprotein surface and the surrounding solvent
`molecules due to an overall greater molecular
`SASA caused by the presence of the glycans.
`
`Precipitation
`
`One of the most fundamental challenges when
`designing a protein-based formulation involves
`achieving the desired therapeutic protein concen-
`tration in solution.2,63 This is due to the fact that
`protein solubility is not only inversely propor-
`tional
`to the protein concentration but also
`dependant on the solution’s pH, temperature,
`ionic
`strength,
`and
`excipient
`concentra-
`tion.2,52,63,155,156 Therefore, as the target concen-
`tration of the formulation is increased (e.g.,
`100 mg/mL) protein precipitation becomes a
`more critical problem.63 Glycosylation has been
`shown to increase the solubility of many pro-
`teins,99,157 although the generality of this effect
`has been questioned.158 Some examples include
`interferon beta (REBIF1, Pfizer/Serono; AVO-
`NEX1, Biogen),159,160 alpha-galactosidase A
`(REPLAGAL1, Shire, England, UK),161 glucose
`oxidase,162 and invertase.163 While studying the
`effects of glycosylation on peroxidase, Tams
`et al.164 determined that the solubility of the
`protein showed a linear dependence with the
`glycosylation degree. Although one could logically
`consider that this increased solubility is due to a
`greater hydration potential since the glycans have
`a higher affinity for the aqueous solvent than the
`polypeptide chain, Bagger et al.165 recently
`showed that this is not the case. From this study
`it was concluded that it is unlikely that strength-
`ened interactions with the aqueous solvent are the
`mechanism for increased protein solubility due to
`glycosylation.165 An alternative explanation can
`be provided from a comparative in silico struc-
`tural and energetic analysis recently performed
`by Sola´ and Griebenow on a series of chemically
`glycosylated a-chymotrypsin conjugates with
`increasing levels of glycosylation (Fig. 1).103,166
`From these computer simulations it was found
`that the overall molecular solvent accessible
`surface area (SASA) for the whole glycoprotein
`
`pH Denaturation
`
`Exposure of proteins to extremes of pH can result
`in loss of structure by disruption of both internal
`electrostatic forces and charge–charge inter-
`actions.2 At extreme pH values, far from the
`isoelectric point (pI), the unfolding propensity
`of proteins increases as a result of electrostatic
`repulsions
`between
`similarly
`charged
`atoms.2,151,167,168 Additionally, the diminished
`capability of salt bridge formation b

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