throbber
Exp,ert
`Opiíríon
`
`1. lntroduction
`2. General aspects of PEGYIation
`3. Polyethylene glycol (PEG), a
`biocor¡jugation polymer
`4. Chemistry of polyethylene
`glycol (PEG) conjugation
`5. PEc-prote¡n coqiugates
`6. PEcylation of drugs
`7. Expert opinion
`
`For reprint orders, please
`contact:
`re pr ¡ nts@ ashl ey - pu b. com
`
`Ashtey Publ¡cat¡ons
`www,ashley-pub.com
`
`Review
`Protein, peptide and non-pePtide
`drug PEGylation for theraPeutic
`application
`G Pasut, A Guiotto & FM Veroneser
`ÍDpartment of Pharmaceutical Sciencæ, [Jnivenily of Padua, vk E lVfarzo]o 5, 3513[-Padoua, Ialy
`
`For many years proteins have been investigated as therapeutic agents, but
`unfortunately the¡r potent¡al advantages could not be completely exploited.
`The main drawbacks are their intrinsic short life in the body, immunological
`adverse reaction and proteolytic digestion. Among all the approaches studied
`for overcoming these problems, PEGylation (the modificat¡on of molecules
`w¡th polyethylene glycol [PEG]) achieved the most interesting results, leading
`to a novel series of products that have already reached the market, and hope-
`fully other promising agents will soon be available. Since the first studies in this
`f¡eld, the cor.¡jugation of PEG to a prote¡n has shown the possibility of improv-
`ing the pharmacokinetic profile of a linked drug. ln the last few years this tech-
`nology, firstly developed for proteins, has been transferred to non-peptide
`drugs, opening a new area of investigation that is now receiving increasing
`¡nterest. This leads to new opportun¡t¡es for many therapeutic treatments as lt
`is possible to use molecules that could not before be exploited due to limita-
`tions such as inadequate water solubility, high nonspecific toxicity and poor
`pharmacokinetic profiles. ln th¡s review the most recent achievements in
`PEGylation of protein, peptide and non-peptide drugs are described concern-
`ing the binding chemistry, and many examples from the literature are
`reported, in the fields of both prote¡n therapeutics and non-peptide drugs.
`
`Keywords: poþethylene glycol (PEG), PEGdrugs, PEG protein, PEGylation, polyrrnr
`therapeutics
`
`Expert Opn. Ther. Patenx (2004) 14(6):855894
`
`t. Introduction
`
`Polymer bioconjugation is receiving inffeasing interest in applied biotechnology,
`and polyethylene glycol (PEG), m particular, has been extensively used in the modi-
`fication of different substrates of therapeutic interest'
`It is usually common to divide therapeutic polymer coniugates into two main cat-
`egoriesl conjugates with drugs having a peptide structure (so far the most studied
`area) and those with a non-peptide structure, in particular low molecular weight
`drugs such as anticancer drugs. The success of this technology is reflected not only
`by the many drug-polymer conjugates already available or under investigation but
`also by the growing number of publications and patents published each year.
`Polypeptides and many low molecular weight molecules usually have shortcom-
`ings that restrict or even prevent clinical use. common limits for many drugs are
`shãrt ft vivo :¡alf-life (due to enzyme degradation or rapid kidney clearance) and
`physicochemical drawbacks, for example low solubility or instability; polypeptides
`may present additional restrictions, such as immunogenicity and antigenicity [1-3ì.
`These problems are often addressed by covalent linking of the pharmaceutically
`active molecule to polymers (Box 1), PEG being the most successful among those
`used [¿-oì. Polymer conjugation, especially for small drugs, is also exploited to
`achieve an active or passive targeting, an improved biodistribution and a selected
`cellular uptake by endocytosis 1z-0¡.
`
`2004 o Ashley Publlcations Ltd ISSN 1 354-3776
`
`859
`
`MPI EXHIBIT 1041 PAGE 1
`
`MPI EXHIBIT 1041 PAGE 1
`
`

`

`Protein, peptide and non-peptide drug PEGylation for therapeutic apPl¡cat¡on
`
`Box 1. General advantages of bioconjugat¡on ¡n
`therapeutic application.
`
`Stabilisation of labile drugs from chemical degradation
`Protection from proteolytic degradation
`Reduction of immunogenicity
`Decreased antibody ræognition
`lncreased body residence time
`Modif ication of biodistri bution
`Drug penetration by endocytosis
`New strategies lor drug targeting
`lncreased water solubi lity
`Reduced toxicity
`
`All protein and peptide drugs are candidates for alternative
`delivery methods. It is noteworthy that peptide drugs are
`worth > US$10 billion of the world pharmaceutical market
`and represent a rapidly growing area. As drug detivery is
`closely tied with pharmaceutical manufacture, it is anticipated
`that its market will be worth an estimated US$120 billion by
`2007 and bioconjugation appears to be one of the most prom-
`ising approaches to reach this goal.
`Although polymer conjugation to proteins originated in
`the 1950s and 1960s using polysaccharide polymers, the real
`boost in this field was represented by the use of PEG, thanks
`to the pioneering studies conducted in the late 1970s by Pro-
`fessor Frank Davis at Rutgers University [10]. Since then many
`excellent reviews have been dedicated to different aspects of
`this technique, known as PEGylation, whereas this review
`focuses on more recent pharmaceutical applications of PEG
`with a particular attention to the patent literature.
`
`z. General aspects of PEGylation
`
`PEG, approved by the FDA for human use, has a variety of
`interesting properties such as the absence of immunogenicity,
`antigenicity and toxicity, and high solubility in water and in
`many organic solvents. These properties can be transferred to
`the final conjugates by PEGylation, obtaining modification of
`the pharmacokinetic and pharmacodynamic profiles of native
`drugs [ll]. In general, PEGylated drugs, when compared with
`the parent molecules, show: i) reduced kidney excretion and
`altered biodistribution, mainly due to the increased molecular
`weight; ii) reduced degradation by proteolytic enzymes or
`hydrolytic media; iii) enhanced water solubility; iv) reduced
`reticuloendothelial (RES) clearance and v) reduced immuno-
`genicity and antigenicity U2,l3l.
`The evolution of protein PEGylation has been frequently
`described, and is divided into two generations:
`
`. The first generation of conjugates refers to PEGs with low
`molecular weights (< 12 kDa) and with a relevant percent-
`age of PEG Aiot impurities, a potential crosslinking
`agent originating from the synthesis of methoxy-PEG.
`
`Furthermore, the chemistry employed often presented
`side reactions or led to weak and reversible linkages. Despite
`these initial difficulties, important products were created
`and some reached the market, such as Enzon Pharmaceuti-
`cals PEG-adenosine deaminase (Adagen@, Enzon, Inc.)
`tl4,20lì for the treatment of severe combined immunodefi-
`ciency disease (SCID) and PEG-asparaginase (Oncaspar@,
`Enzon, Inc.) [15,202] for the treatment of leukaemia.
`. The second generation of conjugates was an improvement
`on the first, in particular as far as PEG purity is concerned;
`an important reduction in polydispersivity and in diol
`amounts was achieved in industrial production and
`improvements are also rmade in selectivity of protein mod-
`ification and in the range of available activated PEGs.
`Heterobifunctional PEGs were prepared in order to link a
`second molecule with a targeting role. Moreover, spacers
`between the polymer and the drug were studied to allow
`the release of bound drug under specific triggering condi-
`tions. Several products of this second generation reached
`the market, such as PEG-IFN-o¿Zb (PEG-Intron@, Scher-
`ing-Plough) [16,203], branched PEG (40 kDa)-IFN-o2a
`(Pegasys@, Roche Pharmaceuticals) t17,18,204Ì, PEG-growth
`hormone receptor antagonist (Pegvisomant, Somavert@,
`Pfizer) Itg,zosl and PEG-granulocyte-colony-stimulating
`factor (G-CSF) þegfilgrastim, Neulasta@, Amgen) ¡zo,zoo¡
`but many others are presently undergoing clinical trials and
`will hopefully be available in the near future.
`
`s. Polyethylene glycol (PEG), a bioconjugat¡on
`polymer
`
`PEG, as obtained by ethylene oxide polymerisation,
`presents one or two terminal hydroxyl groups (mPEG-OH
`or HO-PEG-OH) depending upon the initiator of polym-
`erisationl methanol or water, respectively. A branched form
`may also be obtained, as well as products with multiple reac-
`tive groups, at one or at both extremes (Figure 1). Whlle the
`monofunctional polymers, linear or branched, are used for
`protein modification, those with multiple groups are indi-
`cated to enhance the loading of low molecular weight drugs.
`The main characteristics of PEG, as compared to other poly-
`mers, are low polydispersivity (fu[*tA'[" spanning from 1.01 for
`PEG < 5 kDa molecular weight and up to I .1 for PEG as high as
`50kDa molecular weight), solubility in both aqueous and
`organic solvents and biocompatibility. The unique solvation
`properties of PEG are due to the ability to coordinate 2 - 3 water
`molecules per ethylene oxide tnit and to the higtrly flexible the
`backbone chain I2l. These characteristics give PEG an apparent
`molecular weight 5 - l0 times higher than that of a globular pro-
`tein of comparable mass, as may be verified by gel permeation
`chromatography (see tztl for a recent discussion) and this explains
`the protein-rejecting property, at the basis of the anti-immuno-
`genicity and antigenicity conveyed to a conjugate tz2l. This also
`explains the prolonged blood residence time and the decreased
`degradation by mammalian cells and enzf/mes [23].
`
`860
`
`Expert Opin. Ther. Patents (2004) 14(61
`
`MPI EXHIBIT 1041 PAGE 2
`
`MPI EXHIBIT 1041 PAGE 2
`
`

`

`./o
`
`OH
`
`OH
`
`Pasut, Gu¡otto & Veronese
`
`U
`
`OH
`
`a mPEG-OH
`
`b HO-PEG-OH
`
`c mPEG,-COOH
`
`o
`
`o
`
`X
`
`d X=Reactivegroup
`
`"{--^-"']:---"
`
`X
`
`Figure 1. Different PEG structures. a) Linear monometho)ry PEG; b) Linear diol PEG; c) Branched PEG; and d) Multifunctional PEGs.
`PEG: Polyethylene glycol.
`
`In uivo, PEG undergoes limited chemical degradation and
`its clearance depends on its molecular weight, < 20 kDa.
`PEG is easily secreted into urine, whereas for higher molecu-
`lar weights it is eliminated more slowly into urine and faeces,
`up to the threshold of 40 - 60 kDa (a hydrodynamic radius of
`- 45 Ä tzal), which represents the albumin excretion limit.
`Above this limit the polymer remains in circulation and is
`mainly accumulated in the liver. Alcohol dehydrogenase can
`degrade low molecular weight PEGs and chain cleavage can
`also be catalysed by cytochrome P450 microsomal enzymes
`t25ì. Many years of the use of PEG as an excipient in foods,
`cosmetics and pharmaceuticals without toxic reactions are a
`clear proof of its safety 1231.
`Tì"ends for new PEGs are directed towards: i) monodisperse
`polymer batches, not only in the case of low molecular weight
`PEG (< 600 Da), which have recently reached the market, but
`also for an extended range of higher molecular weights, ii)
`PEG polymers with specific functions tailored to provide con-
`jugation with reactive groups on target molecules, also in view
`of a controlled releasei and iii) high loading PEGs to increase
`the payload of active molecules conjugated to the chain,
`through the construction of dendrimeric structures or multi-
`arm forms at the polymer extremes 126,27,2071.
`
`4. Chemistry of polyethylene glycol(PEG)
`coqiugat¡on
`
`The first generation PEGs were mainly designed for amine
`modification, as amines are a widely represented functional
`group in proteins. Among these, of particular not are: i)
`PEG succinimidyl succinate (SS-PEG); iÐ PEG succinimidyl
`carbonate (SC-PEG); iii) PEG ¡rnitrophenyl carbonate
`(pNPC-PEG); iv) PEG benzotriazoþl carbonate (BTC-PEG);
`v) PEG trichlorophenyl carbonate (TCP-PEG);vi) PEG carb-
`onylimidazole (CDI-PEG); vii) PEG tresylate; and viii)
`PEG dichlorotriazine (Figure 2).
`Carbonate PEGs, such as pNPC-PEG, CDI-PEG and
`TCP-PEG, show slower reactivity than other PEG derivatives,
`
`thus allowing a selective conjugation on the basis of reactivity
`of the different amino groups. It is noteworthy that
`PEG dichlorotriazine, PEG tresylate and PEG aldehyde (after
`sodium borohydride reduction) dlow the same total charge of
`the native protein to be maintained because the conjugation,
`based on alkylation, leads to a secondary amine, whereas the
`coupling performed with the other PEGs, based on acylation,
`yields neutral acidic amide or carbamate linkages. Although,
`the primary amino group is the more reactive in proteins,
`PEGs such as SC-PEG, BTC-PEG and PEG dichlorotriazine
`can slowly react with hydroxyl groups (Ser, Tyr) and the histi-
`dine imidazole side chain to give hydrolytically unstable link-
`ages. For example, one histidine residue of a-IFN was
`conjugated to SC-PEG or BTC-PEG under slightly acidic
`conditions t2081. PEG was also linked to hydroxyl groups of
`serine and tyrosine in the decapeptide antide and in the epider-
`mal growth factor (EGF) [28,20eì, respectively.
`The improvement of polymer synthesis and conjugation
`chemistry is now yielding a second generation of PEGs,
`mainly characterised by lower percentages of diol contami-
`nant in polymer batches. This was achieved by the isolation of
`the monocarboxylic acid intermediate of PEG from the bicar-
`boxylic intermediate, coming from the diol by ionic exchange
`chromatography t2r0ì. Higher molecular weight polymers,
`with an improved pharmacokinetic profile and stability for
`non-peptide drugs, were also obtained. Among the new PEGs
`are reportedi
`
`' PEG-propionaldehyde, also in the form of the more stable
`acetal [211]: the reaction with the amino group leads to a
`Schiff base that is reduced by sodium borohydride, giving a
`derivative that maintains the same total ionic charge of the
`parent drug.
`. PEG-succinimidyl derivatives: higtily reactive towards
`amine group. The reaction rate of these derivatives may
`significantly change depending upon the extension and the
`composition of the alkyl chain between PEG and
`succinimidyl moiety Í29,2121.
`
`Expert Op¡n. Ther. Patents(2004\ 14(6)
`
`861
`
`MPI EXHIBIT 1041 PAGE 3
`
`MPI EXHIBIT 1041 PAGE 3
`
`

`

`Protein, pept¡de and nm-peptide drug PEGylation for therapeutic apPlicat¡on
`
`/R
`
`NH
`
`/o
`
`PEG
`

`
`PEG'
`
`o
`
`o\\
`
`l,,-Å-.P + HrN-R +>
`
`/o
`
`PEG
`
`a
`
`Y"o
`
`b
`
`+
`
`H2N-R
`
`,ro'oyo$^o,
`o-
`
`+
`
`H2N-R
`
`'oY
`
`o\
`
`PEG
`
`o
`
`d
`
`+ H2N-R
`
`PEG
`
`H
`
`Y-o
`
`R
`
`/o
`
`PEG
`
`cl
`
`+
`
`H2N-R
`
`e
`
`ct
`
`o-*AI
`\zJ
`
`-oY
`
`PEG
`
`o
`
`r
`
`+ HrN-R
`
`*o-l>..(fi
`
`-F
`
`+ H2N-R
`
`---+
`
`H
`PEG,N- R
`
`s i
`
`h
`
`/o
`
`PEG
`
`ct
`
`-(N1
`
`cr
`
`+ H2N-R +
`
`-o
`PEG'
`
`ct
`
`rJY)N1
`
`-R
`
`il
`
`Figure 2. Elamples of act¡vated PEG molecules reactive tourards amino groups. a) PEG succinimidyl succinate; b) PEG succinimidyl
`carbonate; c) PEG p-nitrophenyl carbonate; d) PEG benzotriazol carbonate; e) PEG trichlorophenyl carbonate; 0 PEG ærbonylimidazole;
`gf PEG dichlorotriazine; and h) PEG tresylate.
`PEG: Polyethylene glycol.
`
`862
`
`Expett Opin. Ther. Patents(2004) 14(6)
`
`MPI EXHIBIT 1041 PAGE 4
`
`MPI EXHIBIT 1041 PAGE 4
`
`

`


`
`Linear PEG
`
`Branched PEG
`

`
`a
`
`Ho'
`
`b
`
`OH
`
`Pasut, Guiotto & Veronese
`
`P%'
`
`o'
`
`Pq%1.^1" Y}" -f
`,auto
`tll
`
`PEGto
`
`,rcto
`
`OH
`
`OH
`
`X
`
`= Bnanching moiety
`X = Active group
`
`Figure 3. Structure of linear and branched PEG on the
`protein surface. The 'umbrella-like' structure of branched PEG
`explains its higher capacity for rgecting approaching molecules or
`cells compared to linear PEG.
`PEC: Polyethylene glycol.
`
`Figure 5. Different strategies to achieve multifunctional
`high loading PEGs. a) Multiarm PEGs. b) Dendronìsed PEGs, the
`branching moiety may be a bicarboxylic amino acid, lysine or
`other bifunctional molecule.
`PEG: Polyethylene glycol.
`
`Branched PEG
`
`jÉ
`
`,,,
`
`Active site cleft
`
`Linear PEG
`
`Figure 4. Effect of PEG hindrance on the en4me active site'
`The high steric hindrance of branched PEG may be advocated to
`explain the lower inactivation of enrymes compared to linear PEG
`of the same size.
`PEG: Polyethylene glycol.
`
`. 'Y'-shaped branched PEG l¡o,zts,zlal (see Figure lc): for its
`increased surface shielding (Figure 3), this PEG reagent is
`more effective in protecting the coqjugated protein from
`degradative enzymes and antibodies. Moreover, enzymes
`modified with this PEG retain more activity with respect to
`the same en4/me modified by linear PEGs. Tfris effect is
`probably due to the hindrance of the branching polymer
`that prevents the entrance of PEG inside the en4rme active
`site cleft (Figure 4).
`
`. PEGs reactive toward thiol groups: PEG-maleimide
`(MAL-PEG) Í^st, PEG-vinylsulfone (VS-PEG) and
`PEG-orthopyridyl-disulfide (OPSS-PEG). Even if the
`thiol addition rate to the first two derivatives is very rapid,
`some addition to the amino group (mainly present in pro-
`teins) may also take place, especially at basic pHs. On the
`other hand, the reaction with OPSS-PEG is very specific
`for thiol groups but the conjugates may be reversed in the
`presence of thiols as reducing agents.
`. Heterobifunctional PEGs [31,32,216]: these derivatives
`present two different functional groups, one for each
`extreme, which simplify the linking of different molecules
`to the same PEG chain. Therefore, it is easier to obtain
`conjugates that carry both a drug and a targeting molecule.
`Among the proposed and commercially available heterobi-
`functional PEGs, H2N-PEG-COOH, HO-PEG-COOH
`and H2N-PEG-OH are the most used.
`. PEG with linkers designed for a controlled release of the con-
`jugated drug: one of the most exploited linkers is a peptide
`sequence, designed to be recognised and cleaved by lysosomal
`enzymes when the conjugates reach the intracellular compart-
`ment. Examples of such peptide linkers are H-Gly-Phe-Leu-
`G1y-OH or H-Gly-Leu-Phe-Gly-OH 133,341. Altematively, a
`linker may respond to pH changes or release the drug by a
`1,6-elimination reaction or by a cyclisation reaction [35].
`Moreover, the linker and the polymer together can form a
`double prodrug system, where the drug released is obtained
`after polymer hydroþis (frst prodrug) that triggers the linker
`(second prodrug), as reported for he drug delivery system
`based on trimethyl lock (TML) lactonisation [36,217].
`. Multiarm or 'dendronised' PEGs (Figure 5): the former are
`compounds prepared by attaching linear PEG to a
`
`Expert Op¡n. Ther. Patenß(2004) 14(6)
`
`863
`
`MPI EXHIBIT 1041 PAGE 5
`
`MPI EXHIBIT 1041 PAGE 5
`
`

`

`Protein, pept¡de and non-peptide drug PEGylation for therapeutic application
`
`,ao'oyt"!^-otu+H2N-protein+pEG-o
`o
`
`H
`\1M"!x-N-Prot"in
`il
`
`BrCN+
`
`- Protein
`
`X = Nle or Ê-Ala
`
`Figure 6. Use of PECrlVlet.Nle-OSu or PEG-tVlet-p-Ala-OSu to introduce a reporter amino acid at the PEGylation site'
`PEG con-¡ugation is followed by polymer moiety release by BrCN to leave Nle or B-Ala that can be identified on the protein by amino acid
`sequence analysis.
`PEG: Polyethylene qlycol.
`
`+ NH¡
`
`HN
`
`Io^]t-"
`H
`
`o
`
`"-*H
`
`Adduct
`
`[*]t-,,,
`
`H
`
`l"
`
`+ R_NH,
`
`ca2*
`
`TGase
`
`o
`
`NH,
`
`Protein
`
`Scheme 1. Reaction between a glutam¡ne residue in a protein and an alkyl amine, catalysed by TGase.
`TGase: Iransglutam¡nase.
`
`multimeric compound 12181, whereas the latter are linear
`PEGs with a dentritic structure at one or both chain
`extremes 126,37,2071. The aim of both derivatives is to
`increase the drug:polymer molar ratio, overcoming prob-
`lems of high viscosity that may occur with monofunctional
`drug conjugate solutions, in particular for therapeutic treat-
`ments that require quantity of drug.
`
`PEG and any polymer conjugate, especially when multifunc-
`tional drugs are involved, face great problems regarding repro-
`ducible synthesis, scaling-up, characterisation and methods of
`analysis, all issues that have been discussed in detail [38,39ì.
`One additional problem in protein conjugation is the localisa-
`tion of the PEGylation site into the primary amino acid
`sequence. So far the classical approach involves proteolytic
`digestion followed by Edman degradation, as reported for
`PEGylated IFN-2cx 1401, where the PEGylated sites are deter-
`mined by the missing amino acids. Beyond the lengthy proce-
`dure, the polymer may interfere v/ith the analysis as cleavage
`by proteolytic enrymes may be incomplete due to steric hin-
`drance. A more direct approach involves the use of two tailor
`made PEGs, PEG-MeI-N1e-COOH or PEG-Met-BAla-
`COOH tzrgl, which possess a labile bond in the peptide
`spacer arm that can be cleaved by treatment with BrCN
`(Figure6). After cleavage at the methionine level, tfre PEG
`chains are removed and the remaining nor-leucine or
`p-alanine tags, linked to the protein, are identified with stand-
`ard sequence investigation methods. The tag can be revealed
`by amino acid anaþis or mass spectrometry [411.
`A PEGylation procedure that differs from all others was
`recently developed by Sato and involves the use of the enzyme
`transglutaminase (TGase) 142,2201. In vivo TGase catalyses
`crosslinking between a protein containing an acceptor
`
`glutamine and another protein with an amino donor group,
`such as lysine t43,44ì. This enzyme catalyses the attachment of
`PEG-alkylamine, which behaves as a new substrate of TGase to
`the glutamine residues of the protein (S.h"-" l). This system
`provides new opportunities in polymer derivatisationi in fact,
`beyond the high homogeneity of the Tcase-catalysed conju-
`gates, recombinant DNA techniques, it may be used in com-
`bination to introduce amino acids sequences that are specific
`and more favourable TGase substrates. Amongst the several
`TGases isolated from natural sources, the guinea-pig liver
`TGase (G-TGase) and the microbial TGase (M-TGase) were
`reported for PEGylation procedures. The former possesses
`more stringent requirements on neighbouring glutamine
`amino acìds for enzyme activity and transglutamination site
`flexibility, although there is no unique amino acid recogni-
`tion sequence in proteins, G-TGase has been used for the
`preparation of site-specific PEGylated conjugates t45,461. With
`TGase, the suggested strategy consists of the introduction of
`a specific sequence, comprising a glutamine residue, at the
`N terminus of proteins, in order to maintain the peptide
`backbone flexibility. Instead, M-TGase does not require high
`specificity in the substrate sequence, although an optimal
`accessibility to the glutamine site is required either through
`peptide flexibility or high solvation of the sequence t¿zl. On
`the other hand, the amino donors (i.e., PEG-NH) do not
`need strict requirements to allow TGase conjugation, and
`high affinity PEG-NHZ has been designed to link the amino
`group to the PEG with a spacer chain of six carbon atoms
`(PEG-(CHr6-NH, l+at). In the case of IL-Z, the TGase-cata-
`lysed PEGylated form preserved a high degree of activity
`because the N terminus is, in this protein like many others, a
`flexible region and so is less involved in peptide recognition.
`The enzyme-catalysed coupling seems to yield more
`
`864
`
`Expert Op¡n. Ther. Patents (2004) 14(6)
`
`MPI EXHIBIT 1041 PAGE 6
`
`MPI EXHIBIT 1041 PAGE 6
`
`

`

`Pasut, Guiotto & Veronese
`
`H
`
`----------->
`
`mPEG
`
`Yl
`
`_lå1
`
`Nq
`
`+
`
`o
`
`mPEG/
`
`SC-PEG
`
`His34 of IFN
`
`His34 of PEG-IFN
`
`Scheme 2. Add$t formation at the level of His34 using SC-PEG as PEcylating agerìt. (Reproduced with permission from tszl)
`PEG: Polyethylene glycol; SC: Succinimidyl carbonate.
`
`mPEG-O
`
`mPEG-
`
`- IFN
`
`NH
`
`U>-o
`
`mPEG
`
`PEGr-lFN
`
`o
`
`o
`
`-t\ Y
`
`50mM
`sodium borate
`pH9
`
`IFN-NH2
`---+>
`
`mPEG
`
`PEG2-NHS
`
`Scheme 3. PEcylat¡on of IFN-q-2a by branched nPEG2-COOH (a0 kDa). (Reproduced with permission from tlzl),
`NHS: N-Hydroxysuccinimide; PEG: Polyethylene glycol,
`homogenous derivatives than the chemical PEGylation that
`isknowntooftenleadtoamixtureof isomers.
`
`5. Polyethylene glycol (PEG)-prote¡n conju gates
`
`Many proteins and peptides have short half-lifes when
`administered in vivo, due to other proteolytic enzyme activ-
`ity, fast kidney ultrafiltration and activation of immune sys-
`tem response. PEGylation may overcome all of these
`drawbacks thanks to the increased hydrodynamic volume of
`conjugates and the shielding effect of PEG towards enzymes
`and antibodies. Due to protein multivalence, the PEGs used
`for protein conjugation should have only one reactive group
`to avoid crosslinking reactions; for this reason, batches com-
`pletely devoid or with minimal traces of PEG aiol must be
`employed. Furthermore, to avoid protein denaturation, the
`conjugation chemistry should be gentle, yield stable bonds,
`avoid unmasking of epitopes or sites of proteolysis and,
`where possible, it should lead to homogeneous products or,
`at least, reproducible positional isomers. Far from addressing
`all and each of the above-mentioned requirements, several
`coûugates have been engineered showing interesting
`improvements over the starting materials, and some conju-
`gates have already hit the market.
`
`5.1 lnterferons
`Type 1 interferons (IFN-c, -Þ, -r, -t and -ol) are structurally
`and functionally related proteins belonging to a cytokìne fam-
`ily. Thet activity is mediated by lFN-inducible gene activa-
`tion t49l that occurs when the IFN blnds to a multimeric cell
`surface receptor. The activities of these proteins include a
`wide range of effects, mainly antiviral, antitumour and
`
`immunomodulatory properties tsol. IFN-ct' was first approved
`for hairy-cell leukaemia therapy, then for the treatment of
`hepatitis B and C and later for various dermatological pathol-
`ogies; INF-p was approved for the treatment of multiple scle-
`rosis. The low molecular weight of interferons (- 20 kDa)
`reflects the relatively short serum half-life of these proteins,
`which was improved by PEG conjugation.
`The first study in this direction was the modification of
`IFN-cr-2a with linear succinimidyl carbonate PEG (SC-PEG;
`5 kDa) via a urea linkage. The coupling, performed at equi-
`molar ratio of protein and polymer, mainly led to mono-
`PEGylated isomers and, in small amounts, di-PEGylated
`conjugates as well as free IFN [221ì. Characterisation of the
`conjugates indicated that lysine residues were the site of
`PEGylation 1401. Although a once a week schedule was possi-
`ble, while the unconjugated protein was usually given three
`times a week, the desired pharmacokinetic/activity profile was
`not yet achieved. After conjugation of PEG to IFN-cr-Zb vla a
`carbamate linkage, Enzon researchers proposed a method
`that, by acidification of the conjugate aqueous solution to
`pH 3 or lower, allowed the removal of the few PEG chains
`that were attached to INF's amino acid residues (i.e., His)
`through unstable carbamate bonds [222ì.
`Further investigations demonstrated that derivatives with
`better pharmacokinetic profiles and higher activity could be
`obtained by conjugation of IFN-u-Zb with SC-PEG (12 tDa)
`in sodium phosphate buffer solution at pH 6.5. This reaction
`gave an unexpected conjugate at His34, representing - 47o/o of
`the total PEGylated species (Scheme 2) tsr,zosì. The stability of
`this species was studied by [lH]-nuclear magnetic resonance
`(NMR) analysis following the ppm shift of Het of Hls34
`during its conversion from the PEGylatea to the
`
`Expert Op¡n. Ther. Patents(2004\ 14(6\
`
`865
`
`MPI EXHIBIT 1041 PAGE 7
`
`MPI EXHIBIT 1041 PAGE 7
`
`

`

`Protein, pept¡de and non-pePt¡de drug PEGylation for therapeutic application
`
`kDa
`
`200
`
`1 16.3
`97.4
`
`66.3
`55.4
`
`36.5
`31
`
`21.5
`14.4
`6
`
`kDa
`
`!-
`

`

`
`r-
`

`
`4
`
`iD
`
`b
`
`;
`
`ri=r
`
`120
`
`73.4
`
`41.6
`
`22.8
`
`!¡
`
`-
`
`J
`
`4
`
`Figure 7. SDS-PAGE analysis of the PEGylated IFN-a-2a
`m¡xture. a) Gel specifìcally stained for proteln with Coomassie
`blue, Lane 1 : molecular weight marker protein; Lane 2: PEGylation
`reaction m¡xture; Lane 3: purified PEG,-|FN; Lane 4: IFN-cr-2a.
`b) Gel specifically stained for PEG with iodine, Lane 1: molecular
`weight marker PEGs; Lane 2: PEGylation reaction mixture; Lane 3:
`purified PEGr-|FN; Lane 4: IFN-o-2a. Note that the IFN-a-2a is not
`stained by iodine, (Reproduced with permission from tr ul),
`PEG: Poìyethylene glycol; SDS PAGE: Sodium dodecyl sulfate polyacrylamide gel
`eleLtr ophores¡s.
`
`Table 1. Pharmacokinetic propert¡es of IFN-o-2a and its
`PEcylated form in rats [18¡.
`Half-life (h) Plasma res¡dence
`t¡me (h)
`'l 0
`20.0
`
`Prote¡n
`
`IFN-o-2a
`
`PEG2 (40 kDa)-lFN-a-2a
`
`PEG: Polyethylene glycol.
`
`866
`
`nonPEGylated form t5zl. The higher activity ol this IFN
`preparation was related to the ability to release free and fully
`active IFN by slow hydrolysis of the His-PEG bond t2081.
`Further studies were also carried out to find adequate for-
`mulations to prevent loss of activity and denaturation of
`IFN-PEG during the conjugation and the lyophilisation
`procedure t2231. These investigations allowed marketing
`PEG-Intron in 2000. Although the in vitro potency of this
`IFN-PEG is only one-quarter of the free interferon form, its
`serum life is approximately six times longer, allowing for a
`less frequent administration schedule while maintaining an
`efficacy comparable with unmodified IFN Ilo,ssl.
`A different approach to IFN-PEGylation exploited the
`special properties of branched PEGs. A high molecular
`weight branched PEG (PEG2, 40 kDa) was chosen on the
`basis of several preliminary studies disclosing that: i) the
`protein surface protection at the conjugation site with a sin-
`gle, long chain PEG is better than several small PEG chains
`at different sites [4], ii) branched PEGs have lower distribu-
`tion volumes than linear PEGs of identical molecular weight
`and the delivery to organs such as liver and spleen is faster
`[b4] and iii) proteins modified with branched PEG possess
`greater stability towards enzymes and pH degradation [30].
`The 40 kDa branched succinimidyl PEG (PEG2-NHS) was
`linked to IFN a-2a using a 3:1 PEG:protein molar ratio in
`50 mM sodium borate buffer pH 9 (Scheme 3) tl7l.
`PEGylation under these conditions led to a mixture con-
`taining 45 50% monosubstituted protein, 5 - 10%
`polysubstituted (essentially dimer) and 40 - 50o/o unmodi-
`fied IFN (FigureT). Identification of the major positional
`isomer within the mono-PEGylated fraction was carried out
`by combination of high performance cation exchange chro-
`matography, peptide mapping, amino acid sequencing and
`mass spectroscopy analysis, which showed that PEG was
`attached mainly to either Lys31, Lys121, Lysl31 or Lysl34
`117,2041. Even though lhe in vitro anlivlral activity for
`PEG2-IFN was greatly reduced (only 7o/o of residual activ-
`ity was found) , Itre in vivo activity, measured as the ability to
`reduce the size of various human tumours, was higher than
`that of free IFN. The positive result could be related to the
`extended blood residence time of the conjugated form as
`shown in Täble l. These studies brought into the market a
`long lasting blood IFN conjugate, Pegasys, that is effective
`in eradicating hepatic and extrahepatic hepatitis C virus
`(HCV) infection Ir8l.
`A different cytokine, IFN-P, is approved for the treatment
`of multiple sclerosis in the US, like other cytokines, it suffers
`lrom a short blood residence time, again suggesting that a
`PEGylation strategy might be a solution to the problem. An
`exhaustive study conducted by Pepinsky and colleagues [55]
`resulted in a PEG modification of IFN-B-1a that exploited a
`reductive alkylation of an amine residue in phosphate buffer
`at pH 6 with an excess of 20 kDa PEG aldehyde and sodium
`cyanoborohydride to reduce the intermediate Schiff base. The
`conjugate, after extensive purification by gel-filtration and ion
`
`2.1
`150
`
`Expert Op¡n. Ther. Patents (2004) 14(6)
`
`MPI EXHIBIT 1041 PAGE 8
`
`MPI EXHIBIT 1041 PAGE 8
`
`

`

`Pasut, Guiotto & Veronese
`
`while pharmacokinetic experiments showed a fivefold increase
`in serum halflife. A 20 kDa PEG was chosen, as the 5 kDa
`PEG led to a fully active conjugate but with no improvement
`in the pharmacokinetic profile, while the 40 kDa PEG deriva-
`tive was completely inactive.
`A different approach was followed by Shearwater and
`Serono researchers, taking advantage of a site-specific
`PEGylation at CyslT of IFN-p t2241 using a thiol-reactive
`PEGylating agent, OPSS-PEG. Conjugation could be spe-
`cifically directed to CyslT since the other two cysteines are,
`in the native form of INF-P, involved in a disulfide bridge.
`Derivatisation was carried out with PEGs of different molec-
`ular weights but with a special and original strategy, well
`described in the patent. Low molecular weight PEGs are, in
`general, more reactive than those with a high molecular
`weight and in this special case the low molecular weight
`PEGs can overcome the steric hindrance around the thiol
`moiety at Cys17 IFN-P, yielding polymer-IFN conjugates.
`On the basis of this observation, a modification with high
`molecular weight PEGs was obtained via a two-step proce-
`dure: in the first step the protein was modified with a low
`molecular weight, heterobifunctional PEG oligomer; conju-
`gation with a higher molecular weight PEG possessing spe-
`cific reactivity towards the free terminal end of the first
`oligomer completed the procedure (Figureg). This strategy
`implied the use of a special heterobifunctional PEG oli-
`gomer with a group reactive towards the free cysteine
`(OPSS) at one extreme and a hydrazine group at the other.
`Hydraz

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