throbber
Advanced Drug Delivery Reviews 54 (2002) 459–476
`
`www.elsevier.com / locate / drugdeliv
`
`Chemistry for peptide and protein PEGylation
`*
`M.J. Roberts , M.D. Bentley, J.M. Harris
`
`Shearwater Corporation,490 Discovery Drive, Huntsville, AL 35806, USA
`
`Received 17 December 2001; accepted 22 January 2002
`
`Abstract
`
`Poly(ethylene glycol) (PEG) is a highly investigated polymer for the covalent modification of biological macromolecules
`and surfaces for many pharmaceutical and biotechnical applications. In the modification of biological macromolecules,
`peptides and proteins are of extreme importance. Reasons for PEGylation (i.e. the covalent attachment of PEG) of peptides
`and proteins are numerous and include shielding of antigenic and immunogenic epitopes, shielding receptor-mediated uptake
`by the reticuloendothelial system (RES), and preventing recognition and degradation by proteolytic enzymes. PEG
`conjugation also increases the apparent size of the polypeptide, thus reducing the renal filtration and altering biodistribution.
`An important aspect of PEGylation is the incorporation of various PEG functional groups that are used to attach the PEG to
`the peptide or protein. In this paper, we review PEG chemistry and methods of preparation with a particular focus on new
`(second-generation) PEG derivatives, reversible conjugation and PEG structures.
`2002 Elsevier Science B.V. All rights
`reserved.
`
`Keywords: PEGylation; PEG-protein; PEG conjugation; PEG chemistry
`
`Contents
`
`1. Introduction ............................................................................................................................................................................
`2. Properties of PEG ...................................................................................................................................................................
`3. Chemistry of pegylation...........................................................................................................................................................
`3.1. First-generation PEG chemistry.........................................................................................................................................
`3.1.1. PEG chemistry for amine conjugation......................................................................................................................
`3.2. Second-generation PEGylation chemistry...........................................................................................................................
`3.2.1. PEG chemistry for amine conjugation......................................................................................................................
`3.2.2. PEG chemistry for cysteine modification .................................................................................................................
`3.2.3. PEG chemistry for oxidized carbohydrates or N-terminus .........................................................................................
`3.2.4. PEG chemistry for reversible PEGylation ................................................................................................................
`3.2.5. Heterobifunctional PEG chemistry ..........................................................................................................................
`3.3. PEG structures .................................................................................................................................................................
`4. Conclusions ............................................................................................................................................................................
`References ..................................................................................................................................................................................
`
`460
`460
`461
`462
`462
`464
`464
`466
`467
`467
`469
`473
`474
`474
`
`*Corresponding author. Tel.: 11-256-704-7524; fax: 11-256-533-4201.
`E-mail address: mroberts@shearwatercorp.com (M.J. Roberts).
`
`0169-409X / 02 / $ – see front matter
`P I I : S 0 1 6 9 - 4 0 9 X ( 0 2 ) 0 0 0 2 2 - 4
`
`2002 Elsevier Science B.V. All rights reserved.
`
`MPI EXHIBIT 1044 PAGE 1
`
`MPI EXHIBIT 1044 PAGE 1
`
`(cid:211)
`(cid:211)
`

`

`460
`
`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`1. Introduction
`
`The use of proteins and peptides as human thera-
`peutics has expanded in recent years due to: (1)
`discovery of novel peptides and proteins, (2) a better
`understanding of the mechanism of action in vivo,
`(3)
`improvements in expression or synthesis of
`proteins and peptides that closely resemble fully
`human proteins and peptides, and (4) improvements
`in formulation or molecule-altering technologies that
`have the ability to deliver polypeptides in vivo with
`improved pharmacokinetic and pharmacodynamic
`properties. It was estimated that in the year 2000, as
`many as 500 biopharmaceutical products were under-
`going clinical trials, and the estimated annual growth
`rates
`of
`protein
`products
`(glycoproteins,
`un-
`glycosylated proteins and antibodies) will range from
`10 to 35% [1].
`Although more biopharmaceuticals are in develop-
`ment than ever before, many of these have problems
`that are typical of polypeptide therapeutics, including
`short
`circulating
`half-life,
`immunogenicity,
`proteolytic degradation, and low solubility. Several
`strategies have emerged as ways to improve the
`pharmacokinetic and pharmacodynamic properties of
`biopharmaceuticals, including: (1) manipulation of
`amino acid sequence to decrease immunogenicity
`and proteolytic cleavage, (2) fusion or conjugation to
`immunoglobulins and serum proteins, such as al-
`bumin, (3) incorporation into drug delivery vehicles
`for protection and slow release, and (4) conjugating
`to natural or synthetic polymers [2–6].
`Those in the biomedical, biotechnical and pharma-
`ceutical communities have become quite familiar
`with the improved pharmacological and biological
`properties that are associated with the covalent
`attachment of poly(ethylene glycol) or PEG to
`therapeutically useful polypeptides. For
`instance,
`PEG conjugation can shield antigenic epitopes of the
`polypeptide, thus reducing reticuloendothelial (RES)
`clearance and recognition by the immune system and
`also reducing degradation by proteolytic enzymes.
`PEG conjugation also increases the apparent size of
`the polypeptide,
`thus reducing renal filtration and
`altering biodistribution. Contributing factors that
`affect the foregoing properties are: (1) the number of
`PEG chains attached to the polypeptide, (2) the
`molecular weight and structure of PEG chains at-
`
`tached to the polypeptide, (3) the location of the
`PEG sites on the polypeptide and (4) the chemistry
`used to attach the PEG to the polypeptide.
`The importance of chemistry and quality of PEG
`reagents for peptide and protein modification has
`only been realized in the last several years as more
`and more PEG-conjugates have reached late phase
`clinical trials. The first few PEG-protein products,
`(cid:210) (cid:210)
`now on the market (Adagen , Oncospar , and PEG-
`Intron ), were developed using first generation PEG
`chemistry. One characteristic of first generation PEG
`chemistry is the use of low molecular weight linear
`PEGs ( # 12 kDa) with chemistry that may result in
`side reactions or weak linkages upon conjugation
`with polypeptides.
`The next generation of PEG-protein therapeutics,
`which will come to market in the next several years,
`uses second-generation PEG chemistries. Second-
`generation PEGylation was designed to avoid the
`problems of first generation chemistry, notably
`diactivated PEG impurities, restriction to low molec-
`ular weight mPEG, unstable linkages and lack of
`selectivity in modification. Readers are referred to
`several detailed reviews on different aspects of
`PEGylation [7–11]. In this paper, we review chemis-
`tries of both first- and second-generation, with an
`emphasis on newer PEGylation technologies,
`in
`order to provide an introduction to those chemistries
`that will be used in the following reviews.
`
`2. Properties of PEG
`
`In its most common form poly(ethylene glycol),
`PEG, is a linear or branched polyether terminated
`with hydroxyl groups and having the general struc-
`ture:
`
`HO–(CH CH O) –CH CH –OH
`2
`2
`2
`2
`n
`
`PEG is synthesized by anionic ring opening
`polymerization of ethylene oxide initiated by nu-
`cleophilic attack of a hydroxide ion on the epoxide
`ring. Most useful for polypeptide modification is
`monomethoxy PEG, mPEG, having the general
`structure:
`
`CH O–(CH CH O) –CH CH –OH
`3
`2
`2
`2
`2
`n
`
`MPI EXHIBIT 1044 PAGE 2
`
`MPI EXHIBIT 1044 PAGE 2
`
`(cid:210)
`

`

`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`461
`
`Monomethoxy PEG is synthesized by anionic ring
`opening polymerization initiated with methoxide
`ions. Commercially available mPEG contains a
`considerable amount of diol PEG due to the presence
`of trace amounts of water during polymerization.
`This diol PEG is also of relatively high molecular
`weight due to polymerization at both ends of the
`polymer. The amount of diol PEG can exceed 15%
`of the composition of mPEG. A solution to the
`problem of diol contamination has been developed in
`our laboratories [12]. In this work, a crude benzylox-
`y-PEG, containing diol impurity, is methylated and
`then hydrogenated to remove the benzyl group. Thus
`diol is converted to the inert dimethyl ether, which
`can be subsequently removed after activation and
`polypeptide attachment.
`BzO–PEG–OH 1 HO–PEG–OH fi fi
`OCH 1 CH O–PEG–OCH
`3
`3
`
`HO–PEG–
`
`3
`
`is to
`Another common route to remove diol
`convert the PEGs to PEG-carboxylic acids that can
`then be purified by ion-exchange chromatography
`[13]. PEG with various end groups can be prepared
`by use of suitable initiator and / or termination re-
`agents. Numerous functionalities can be introduced
`as end groups on PEG in this manner,
`including
`heterobifunctional products. For instance, Kataoka et
`al. synthesized a heterobifunctional PEG derivative
`containing aldehyde and thiol end groups [14].
`Polymerization was initiated with 3,3-diethoxy-1-
`propanol, which forms a propionaldehyde after acid
`hydrolysis, and the polymerization was terminated
`with methansulfonyl chloride with successive con-
`version to ethyldithiocarbonate and a free thiol.
`Compared with other polymers, PEG has a rela-
`tively narrow polydispersity (M /M ) in the range of
`w
`n
`1.01 for low molecular weight PEGs (,5 kDa) to
`1.1 for high molecular weight PEGs (.50 kDa).
`The unique ability of PEG to be soluble in both
`aqueous solutions and organic solvents makes it
`suitable for end group derivatization and chemical
`conjugation to biological molecules under mild
`physiological conditions. Studies of PEG in solution
`have shown that PEG typically binds 2–3 water
`molecules per ethylene oxide unit. Due to both the
`high flexibility of
`the backbone chain and the
`binding of water molecules, the PEG molecule acts
`
`as if it were five to 10 times as large as a soluble
`protein of comparable molecular weight. These
`factors have been suggested as the reason that PEG
`exhibits the ability to precipitate proteins [15],
`exclude proteins and cells from surfaces [16], reduce
`immunogenicity and antigenicity [17] and prevent
`degradation by mammalian cells and enzymes [18].
`Low molecular weight oligomers of PEG (,400
`Da) have been shown to be degraded in vivo by
`alcohol dehydrogenase to toxic metabolites, however
`the lack of toxicity of PEGs with a molecular weight
`above 1000 Da has been revealed over many years of
`use in foods, cosmetics and pharmaceuticals [18].
`PEG is rapidly cleared in vivo without structural
`change and clearance is dependent on molecular
`weight. Below a molecular weight of about 20 kDa
`the molecule is cleared in the urine, and higher
`molecular weight PEGs are cleared more slowly in
`the urine and feces. PEG is only weakly immuno-
`genic even at high molecular weights. Antibodies to
`PEG have been generated when attached to a highly
`immunogenic molecule under
`an immunization
`protocol with Freund’s adjuvant [19–21]. There are
`no known situations in which anti-PEG antibodies
`have been generated under ‘normal’ clinical adminis-
`tration of a PEG-modified protein.
`
`3. Chemistry of pegylation
`
`To couple PEG to a molecule (i.e. polypeptides,
`polysaccharides, polynucleotides and small organic
`molecules) it is necessary to activate the PEG by
`preparing a derivative of the PEG having a func-
`tional group at one or both termini. The functional
`group is chosen based on the type of available
`reactive group on the molecule that will be coupled
`to the PEG. For proteins,
`typical reactive amino
`acids include lysine, cysteine, histidine, arginine,
`aspartic
`acid, glutamic
`acid,
`serine,
`threonine,
`tyrosine, N-terminal amino group and the C-terminal
`carboxylic acid. In the case of glycoproteins, vicinal
`hydroxyl groups can be oxidized with periodate to
`form two reactive formyl moieties.
`The most common route for PEG conjugation of
`proteins has been to activate the PEG with functional
`groups suitable for reaction with lysine and N-termi-
`nal amino acid groups. Lysine is one of the most
`
`MPI EXHIBIT 1044 PAGE 3
`
`MPI EXHIBIT 1044 PAGE 3
`
`

`

`462
`
`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`prevalent amino acids in proteins and can be up-
`wards of 10% of the overall amino acid sequence. In
`reactions between electrophilically activated PEG
`and nucleophilic amino acids,
`it
`is typical
`that
`several amines are substituted. When multiple lysines
`have been modified, a heterogeneous mixture is
`produced that is composed of a population of several
`polyethylene glycol molecules attached per protein
`molecule (‘PEGmers’) ranging from zero to the
`number of «- and a-amine groups in the protein. For
`a protein molecule that has a single PEG attached by
`this nonspecific modification method,
`the poly-
`ethylene glycol moiety may be attached at a number
`of different amine sites. Therefore there is the
`potential for a large number of positional isomers (P)
`as the degree of modification increases:
`
`N!
`P 5 (N 2 k)! 3 k!
`]]]]
`
`where N is the number of possible sites and k is the
`number of sites modified. The extent of modification
`is important
`in determining the pharmacological
`properties of the bioconjugate. Typically, a higher
`degree of modification will extend the circulation
`half-life and reduce the likelihood of antigenicity
`[22]. Each positional isomer of the heterogeneous
`mixture is likely to have an influence on whether the
`conjugate is active or whether an antibody will bind
`an antigenic epitope. The heterogeneity in lysine
`substitution and in PEG molecular weights is of
`some concern for PEG-protein pharmaceuticals, and
`it
`is generally necessary to demonstrate that
`the
`pattern for a particular pharmaceutical can be mea-
`sured and is reproducible. Many of the important
`benefits of PEGylation can be controlled by proper
`conjugation of various molecular weight PEGs to the
`protein at specific locations on the protein’s surface.
`The monofunctionality of methoxyPEG makes it
`particularly suitable for protein and peptide modi-
`fication because it yields reactive PEGs that do not
`produce crosslinked polypeptides, as long as diol
`PEG has been removed. As we will see in the
`discussion of second generation PEGylation,
`it
`is
`also possible in some instances to reduce or elimi-
`nate heterogeneity in the position of substitution.
`
`3.1. First-generation PEG chemistry
`
`3.1.1. PEG chemistry for amine conjugation
`Since most applications of PEG conjugation in-
`volve labile molecules, the coupling reactions require
`mild chemical conditions. In the case of polypep-
`tides, the most common reactive groups involved in
`coupling are the alpha or epsilon amino groups of
`lysine. In Fig. 1 is listed a wide range of first
`generation PEG derivatives used for protein PEGyla-
`tion of either the alpha or epsilon amino groups.
`First-generation chemistries are generally plagued by
`PEG impurities, restriction to low molecular weights,
`unstable linkages, and lack of selectivity in modi-
`fication. Examples of first-generation PEG deriva-
`tives include: (a) PEG dichlorotriazine, (b) PEG
`tresylate, (c) PEG succinimidyl carbonate, (d) PEG
`benzotriazole carbonate, (e) PEG p-nitrophenyl car-
`bonate, (f) PEG trichlorophenyl carbonate, (g) PEG
`carbonylimidazole and (h) PEG succinimidyl succi-
`nate.
`The initial work of Davis et al. used cyanuric
`chloride to prepare activated PEG for attachment to
`proteins [6,17]. The PEG dichlorotriazine (Fig. 1a)
`derivative can react with multiple nucleophilic func-
`tional groups such as lysine, serine, tyrosine, cys-
`teine, and histidine, which results in displacement of
`one of the chlorides and produces a conjugate with
`retained charge in the form of a secondary amine
`linkage [23]. The remaining chloride is less suscep-
`tible to reactions with nucleophilic residues. Un-
`fortunately,
`the reactivity is sufficient
`to allow
`crosslinking of protein molecules containing addi-
`tional nucleophilic residues. To solve this problem,
`Inada et al. synthesized 2,4-bis(methoxypolyethylene
`glycol)-6-chloro-s-triazine
`(mPEG -chlorotriazine)
`2
`as shown in Fig. 2 [24]. The lower reactivity of the
`remaining chlorine translates into a more selective
`modification of lysine and cysteine residues without
`further side reactions.
`Another alkylating reagent used to nonspecifically
`modify multiple amino groups to form secondary
`amine linkages to proteins, viruses and liposomes is
`PEG tresylate (Fig. 1b) [25]. Although more specific
`to amino groups than PEG dichlorotriazine,
`the
`chemistry of conjugation and the conjugation prod-
`ucts are not unique and well defined. For example,
`
`MPI EXHIBIT 1044 PAGE 4
`
`MPI EXHIBIT 1044 PAGE 4
`
`

`

`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`463
`
`Fig. 1. First-generation amine reactive PEG derivatives.
`
`Fig. 2. Branched PEG (PEG2) based on PEG-triazine.
`
`Gais et al. have shown that PEG-tresylate conjuga-
`tion to small molecule amines can produce a product
`that contains a degradable sulfamate linkage [26].
`Therefore, a heterogeneous mixture that results from
`
`attaching PEG-tresylate to proteins may contain a
`population of conjugates with degradable linkages.
`Most first-generation PEG chemistries are those
`that produce conjugates through acylation. Two
`widely used first-generation activated mPEGs are
`succinimidyl carbonate (SC-PEG in Fig. 1c) [27,28]
`and benzotriazole carbonate (BTC-PEG in Fig. 1d)
`[29]. SC-PEG and BTC-PEG react preferentially
`with lysine residues to form a carbamate linkage, but
`are also known to react with histidine and tyrosine
`residues. SC-PEG is slightly more stable to hy-
`
`MPI EXHIBIT 1044 PAGE 5
`
`MPI EXHIBIT 1044 PAGE 5
`
`

`

`464
`
`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`drolysis than BTC-PEG with a half-life of 20.4 min
`at pH 8 and 25 8C compared to the 13.5 min
`hydrolysis half-life of BTC-PEG under the same
`conditions [30]. It has recently been observed that
`SC-PEG and BTC-PEG couple to histidine residues
`of a-interferon at slightly acidic conditions to form a
`hydrolytically unstable imidazolecarbamate linkage
`[31]. The weak linkage could be used to advantage
`in preparation of controlled-release formulation, or it
`could be a disadvantage if conjugate instability were
`not desired.
`Other PEG acylating reagents which produce
`urethane linked proteins include p-nitrophenyl car-
`bonate (pNPC-PEG in Fig. 1e), trichorophenyl car-
`bonate (TCP-PEG in Fig. 1f) and carbonylimidazole
`(CDI-PEG in Fig. 1g) [32,33]. These reagents are
`prepared
`by
`reacting
`chloroformates
`or
`car-
`bonylimidazole with the terminal hydroxyl group on
`mPEG, and these have much lower reactivity than
`either the SC-PEG or BTC-PEG. Generally,
`the
`slower the reaction the more specific the reagent is to
`certain amino acid groups of the protein. In this way,
`some selectivity is achieved. The extent and rate of
`modification can easily be followed in the case of
`pNPC-PEG and TCP-PEG by monitoring the
`phenolate-ion leaving-group by colorimetric analysis.
`The remaining first-generation PEG reagent
`is
`succinimidyl succinate (SS-PEG in Fig. 1h) [34].
`SS-PEG is prepared by reaction of mPEG with
`succinic anhydride, followed by activation of the
`carboxylic acid to the succinimidyl ester. The poly-
`mer backbone contains a second ester linkage that
`remains after the conjugation reaction with a protein.
`This linkage is highly susceptible to hydrolysis after
`the polymer has been attached to the protein. Not
`only does this hydrolysis lead to loss of the benefits
`of PEG attachment, but the succinate tag that re-
`mains on the protein after hydrolysis can act as a
`hapten and lead to immunogenicity of the remaining
`protein [35].
`Techniques used to form first generation PEG
`derivatives are generally straightforward and involve
`reacting the PEG polymer with a group that
`is
`reactive with hydroxyl groups, typically anhydrides,
`chlorides, chloroformates and carbonates. With the
`exception of
`the work by Bentley et al.,
`these
`techniques lack the ability to produce pure mono-
`functional PEG derivatives of high molecular weight
`
`[12]. Since the diol content of high molecular weight
`PEGs can reach 15%, high-molecular-weight, first-
`generation PEG chemistry is inefficient for protein
`conjugation. The ability to generate an intermediate
`that can be purified from diactivated species renders
`second-generation chemistry a valuable tool
`for
`protein modification.
`
`3.2. Second-generation PEGylation chemistry
`
`3.2.1. PEG chemistry for amine conjugation
`Second-generation PEGylation chemistry has been
`designed to avoid the above noted problems of diol
`contamination, restriction to low molecular weight
`mPEG, unstable linkages, side reactions and lack of
`selectivity in substitution. One of the first examples
`of second-generation chemistry is mPEG-propional-
`dehyde [36]. mPEG-propionaldehyde is easier to
`prepare and use than PEG-acetaldehyde because the
`acetaldehyde is very susceptible to dimerization via
`aldol condensation. A key property of mPEG-
`priopionaldehyde, as disclosed by Kinstler et al. in
`work on PEGylation of G-CSF, sTNF-RI, and con-
`sensus IFN, is that under acidic conditions (approxi-
`mately pH 5), aldehyde is largely selective for the
`N-terminal a-amine because of the lower pK of the
`a
`a-amine compared to other nucleophiles [37–39].
`The conjugation of electrophilic PEGs to amino acid
`residues on proteins is highly dependent on the
`nucleophilicity of each amino acid residue. Nu-
`cleophilic attack will only take place when the pH of
`the protein solution is near or above the residue’s
`pK . Therefore the reactivity of each residue also
`a
`depends on neighboring amino acid residues. Al-
`though complete selectivity is not observed,
`the
`extensive heterogeneity frequently seen with lysine
`chemistry is greatly reduced. Coupling of aldehydes
`to primary amines proceeds through a Schiff base,
`which is reduced in situ to give a stable secondary
`amine linkage as shown in Fig. 3.
`An alternative approach to using PEG-aldehyde is
`to use the acetal derivative of PEG-propionaldehyde
`or PEG-acetaldehyde [40]. The aldehyde hydrate of
`the acetal derivatives can be generated in situ by acid
`hydrolysis (Fig. 4). The pH of the solution can then
`be adjusted to values sufficient for protein modi-
`fication with the same mechanism as the free alde-
`hyde derivative in Fig. 3. The benefit of using the
`
`MPI EXHIBIT 1044 PAGE 6
`
`MPI EXHIBIT 1044 PAGE 6
`
`

`

`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`465
`
`Fig. 3. Reductive amination using PEG-propionaldehyde.
`
`Fig. 4. In situ generation of PEG-aldehyde hydrate for use in reductive amination.
`
`acetal derivative over the free propionaldehyde or
`acetaldehyde is longer storage stability and higher
`purity.
`Active esters of PEG carboxylic acids are the most
`used acylating agents for protein modification. Ac-
`tive esters react with primary amines near physiolog-
`ical conditions to form stable amides as shown in
`Fig. 5. Generating the carboxylic acid intermediate
`allows the PEG to be purified from unsubstituted or
`disubstituted impurities by ion-exchange chromatog-
`raphy [41]. Purities of greater than 97% are routinely
`obtainable by this method. Activation of PEG-car-
`boxylic acids to the succinimidyl active esters is
`accomplished by reacting the PEG-carboxylic acid
`with N-hydroxysuccinimide (NHS or HOSu) and a
`carbodiimide.
`The first carboxylic acid derivative of PEG not
`containing a degradable linkage to the PEG back-
`bone, as in SS-PEG, was carboxymethylated PEG
`
`this
`[42]. The succinimidyl ester of
`(CM-PEG)
`compound (SCM-PEG) is extremely reactive (hy-
`drolysis t
`of 0.75 min at pH 8 and 25 8C) and is
`1 / 2
`therefore difficult to use. To take advantage of the
`intermediate purification step and have an active
`ester that had more favorable kinetics for protein
`modification, Harris et al. prepared propionic acid
`(PEG–O–CH CH –COOH)
`and
`butanoic
`acid
`2
`2
`(PEG–O–CH CH CH –COOH) derivatives of PEG
`2
`2
`2
`(Fig. 5(1)) [13]. Changing the distance between the
`active ester and the PEG backbone by the addition of
`methylene units had a profound influence on the
`reactivity towards amines and water. For example,
`SBA-PEG, which has two additional methylene
`groups, has a longer hydrolysis half-life of 23 min at
`pH 8 and 25 8C. SPA-PEG, which has one additional
`methylene group, has a hydrolysis half-life of 16.5
`min at pH 8 and 25 8C.
`Reactivity of PEG active esters towards amines
`
`MPI EXHIBIT 1044 PAGE 7
`
`MPI EXHIBIT 1044 PAGE 7
`
`

`

`466
`
`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`Fig. 5. PEG NHS esters. (1) PEG NHS esters based on propionic and butanoic acids and (2) a-branched PEG NHS esters based on
`propionic and butanoic acids.
`
`and water can be further decreased by introducing an
`a-branching moiety to the carboxylic acid as shown
`in Fig. 5(2). An a-methyl branched PA-PEG deriva-
`tive has a hydrolysis half-life of 33 min at pH 8 and
`25 8C.
`
`3.2.2. PEG chemistry for cysteine modification
`PEGylation of free cysteine residues in proteins is
`the main approach for site-specific modification
`because reagents that specifically react with cysteines
`have been synthesized, and the number of
`free
`cysteines on the surface of a protein is much less
`than that of lysine residues. In the absence of a free
`cysteine in a native protein, one or more free
`cysteines can be added by genetic engineering [43].
`The advantage of this approach is that
`it makes
`possible site-specific PEGylation at areas on the
`protein that will minimize a loss in biological
`activity but decrease immunogenicity. This strategy
`is not without its shortcomings. The addition of free
`cysteines by genetic engineering increases the possi-
`bility of incorrect disulfide formation and protein
`dimerization.
`PEG derivatives such as PEG-maleimide (Fig.
`6(1)), vinylsulfone (Fig. 6(2)), iodoacetamide (Fig.
`6(3)), and orthopyridyl disulfide (Fig. 6(4)) have
`been developed for PEGylation of cysteine residues,
`with each derivative having its own advantages and
`disadvantages [43–46]. PEG-vinylsulfone (PEG-VS)
`reacts slowly with thiols to form a stable thioether
`linkage to the protein at slightly basic conditions (pH
`7–8) but will proceed faster if the pH is increased.
`
`Although PEG-VS is stable in aqueous solutions, it
`may react with lysine residues at elevated pH. Unlike
`PEG-VS, PEG-maleimide (PEG-MAL) is more reac-
`tive to thiols even under acidic conditions (pH 6–7),
`but it is not stable in water and can undergo ring
`opening or addition of water across the double bond.
`PEG-iodoacetamide (PEG-IA)
`reacts slowly with
`free thiols by nucleophilic substitution, creating a
`stable thioether linkage. The reaction should be done
`in slight molar excess of PEG-IA in a dark container
`to limit the generation of free iodine that may react
`with other amino acids. The thioether linkage be-
`tween the PEG-MAL and protein is stable, but slow
`cleavage of one of the amide linkages can occur by
`hydrolysis. Orthopyridyl disulfide-PEG (PEG-OPSS)
`reacts specifically with sulfhydryl groups under both
`acidic and basic conditions (pH 3–10) to form a
`disulfide bond with the protein. Disulfide linkages
`are also stable, except in a reducing environment
`when the linkage is converted to thiols.
`Scientists in our laboratories recently prepared a
`highly active, long circulating and stable conjugate
`of IFN-b using a two-step method with PEG-OPSS
`[47]. The tertiary structure of IFN-b was determined
`by Karpusas et al. who showed that the free cysteine
`residue at position 17 was proximal to the surface
`but hidden [48]. In this case, the available thiol was
`not accessible to high molecular weight PEG that
`would be needed for improved pharmacokinetics.
`The approach that was ultimately adopted was to
`couple a low molecular weight di-OPSS PEG (Mw
`2000) to the interferon and then couple a PEG thiol
`
`MPI EXHIBIT 1044 PAGE 8
`
`MPI EXHIBIT 1044 PAGE 8
`
`

`

`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`467
`
`Fig. 6. Thiol reactive PEGs. (1) PEG maleimide, (2) PEG vinyl sulfone, (3) PEG iodoacetamide, and (4) PEG orthopyridyl disulfide.
`
`to the remaining terminal OPSS group. The disulfide
`linkage between PEG and the protein was found to
`be stable in plasma circulation (unpublished data).
`
`3.2.3. PEG chemistry for oxidized carbohydrates
`or N-terminus
`Oxidation of carbohydrate residues or N-terminal
`serine or threonine is an alternative method for
`site-directed PEGylation of proteins. Carbohydrates
`can be oxidized with enzymes, such as glucose
`oxidase, or chemically with sodium periodate. Oxi-
`dation of the carbohydrate residues generates multi-
`ple reactive aldehyde groups, which can be reacted
`with either PEG-hydrazide to produce a hydrazone
`linkage or with PEG-amine to produce a reversible
`Schiff base (Fig. 7) [49]. The hydrazone linkage may
`be reduced with sodium cyanoborohydride to a more
`stable alkyl hydrazide and the Schiff’s base may be
`reduced to form a secondary amine. Reductive
`alkylation with PEG-amine is problematic because
`the amino groups of a protein possess similar
`reactivity to PEG-amines and thus may form cross-
`
`linked aggregates. PEG-hydrazides are more useful
`in these situations. Under acidic conditions (approx.
`pH 5), amino groups of the protein are predominant-
`ly protonated, but because the PEG-hydrazide is a
`weaker base (pK approx. 3) than primary amines
`a
`(pK approx. 10),
`the reaction is selective to the
`a
`PEG-hydrazone formation. Multiple attachment sites
`are generated using this method, but the modification
`site is specific to the carbohydrate.
`Another approach to site-specific conjugation is to
`take advantage of the presence of a N-terminal serine
`or threonine, which can be converted by periodate
`oxidation to a glyoxylyl derivative. Gaertner et al.
`oxidized the N-terminal serine of IL-8 to form a
`glyoxylyl derivative, which they conjugated to
`aminooxy and hydrazide PEG derivatives [50].
`
`3.2.4. PEG chemistry for reversible PEGylation
`Most PEGylation chemistry is designed to create a
`conjugate that contains a stable linkage to the
`protein. In most cases having a stable linkage to the
`protein is beneficial because of the suitability for
`
`MPI EXHIBIT 1044 PAGE 9
`
`MPI EXHIBIT 1044 PAGE 9
`
`

`

`468
`
`M.J. Roberts et al. / Advanced Drug Delivery Reviews 54(2002)459–476
`
`Fig. 7. Attachment of poly(ethylene glycol) to oxidized carbohydrates of glycoproteins.
`
`long-term storage, easier purification and availability
`of prefilled syringes. It is also generally observed
`that stable linkages to a protein can reduce the
`activity, possibly due to the presence of the PEG
`chain at the active or binding site of the protein or
`steric crowding at the active or binding site. Also the
`PEG molecular weight has a direct impact on the
`activity; higher molecular weight PEG conjugates
`tend to have lower in vitro activity but have higher in
`vivo activity due to the improved pharmacokinetic
`profile [51]. The objective of most PEG conjugation
`techniques is to increase the circulation half-life
`without altering activity. In the development of PEG-
`Intron , Enzon used a degradable linkage between
`the PEG and protein to improve the pharmacokinetic
`half-life but minimize loss of activity by releasing
`native
`interferon alpha-2b [52]. PEG-Intron is
`formed by conjugation of PEG-SC to interferon
`alpha-2b at low pH (around pH 5). The conjugation
`leads to a population of PEG conjugates coupled to
`34His
`(Fig. 8). In this case, the PEG is coupled to the
`d1N position of the imidazole ring in histidine to
`form a carbamate linkage and the PEG was found to
`be released from the protein over time. Note should
`be taken when comparing PEG-Intron to the branch-
`ed
`PEG
`-interferon
`alpha-2a
`conjugate
`40 kDa
`(Pegasys ) that the PEG-Intro

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