14.06.2015 Views

20110802_Shailesh T. Prajapati et al, IJAPR.pdf - international ...

20110802_Shailesh T. Prajapati et al, IJAPR.pdf - international ...

20110802_Shailesh T. Prajapati et al, IJAPR.pdf - international ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

<strong>IJAPR</strong><br />

Available Online through<br />

www.ijapronline.org<br />

Review Article<br />

ISSN: 2230 – 7583<br />

PEGYLATION: AN APROACH FOR PROTEIN AND PEPTIDE DRUG DELIVERY SYSTEMS<br />

<strong>Shailesh</strong> T. <strong>Prajapati</strong>*, Amit N. Patel, Chhagan N. Patel<br />

Shri Sarvajanik Pharmacy College, Mehsana - 384 001, India.<br />

Received on 20 – 04 - 2011 Revised on 21 – 05- 2011 Accepted on 01 – 06 – 2011<br />

ABSTRACT<br />

A number of novel drug-delivery mechanisms have been developed to increase the utility of drugs having poor<br />

solubility, distribution and permeation, one of them is PEGylation. PEGylation defines the modification of a protein,<br />

peptide or non-peptide molecule by the linking of one or more poly<strong>et</strong>hylene glycol (PEG) chains. PEGylated<br />

products gener<strong>al</strong>ly have longer plasma h<strong>al</strong>f-lives and durations of bioactivity than their non PEGylated counterparts.<br />

Pegylation now play the important role in drug delivery and enhancing the potenti<strong>al</strong> of peptide and protein drugs.<br />

INTRODUCTION<br />

A number of novel drug-delivery mechanisms have<br />

been developed to increase the utility of drugs that<br />

are otherwise limited by suboptim<strong>al</strong> pharmacokin<strong>et</strong>ic<br />

properties, such as poor absorption, distribution, and<br />

elimination. These include continuous-release<br />

injectable and liposom<strong>al</strong> systems, which <strong>al</strong>ter the<br />

formulation of the drug, and PEGylation, which <strong>al</strong>ters<br />

the drug molecule. [1]<br />

PEGylation defines the modification of a protein,<br />

peptide or non-peptide molecule by the linking of one<br />

or more poly<strong>et</strong>hylene glycol (PEG) chains. This<br />

polymer is nontoxic, non-immunogenic, nonantigenic,<br />

highly soluble in water and FDA approved.<br />

The PEG-drug conjugates have sever<strong>al</strong> advantages: a<br />

prolonged residence in body, a decreased degradation<br />

by m<strong>et</strong>abolic enzymes and a reduction or elimination<br />

of\protein immunogenicity. Thanks to these favorable<br />

properties, PEGylation now plays an important role<br />

in drug delivery, enhancing the potenti<strong>al</strong>s of peptides<br />

and proteins as therapeutic agents. [2]<br />

Corresponding Author:<br />

Dr. <strong>Shailesh</strong> T. <strong>Prajapati</strong>,<br />

Shri Sarvajanik Pharmacy College,<br />

Mehsana- 384 001, India;<br />

Mob:+91 9924456583,<br />

Email: stprajapati@gmail.com<br />

PEGylation was first described in the 1970s by<br />

Davies and Abuchowsky and reported in two key<br />

papers on <strong>al</strong>bumin and cat<strong>al</strong>ase modification. This<br />

was an important milestone, because at that time it<br />

was not conceivable to modify an enzyme so<br />

extensively and still maintain its activity. Proteins<br />

were in fact considered very delicate entities and only<br />

few gentle modifications with low molecular-weight<br />

products were carried out, mainly to study SARs. [2]<br />

PEGylation is a new delivery technology that differs<br />

from tradition<strong>al</strong> formulation in a number of ways.<br />

Formulated products such as tabl<strong>et</strong>s, liquids and<br />

capsules, the formulation process is reversible, the<br />

drug becomes active after its release from the<br />

formulation and the API remains unchanged. In<br />

PEGylated products, on the other hand, the API is<br />

chemic<strong>al</strong>ly modified in a durable fashion, and the<br />

drug is not released from a formulation but has a<br />

permanent action and is in fact classed as a new API.<br />

Consequently, PEGylation has to be considered early<br />

in the drug development process. [2]<br />

The advantages conferred by PEGylation include an<br />

increased molecule weight and hydrodynamic<br />

volume and a masking of the surface of the molecule<br />

with highly mobile PEG chains. PEGylation <strong>al</strong>so<br />

reduces the rapid ren<strong>al</strong> clearance of sm<strong>al</strong>l proteins<br />

and makes it possible for liposomes to evade remov<strong>al</strong><br />

from the plasma by lipolytic enzymes and the<br />

r<strong>et</strong>iculoendotheli<strong>al</strong> system. As a result, pegylated<br />

agents gener<strong>al</strong>ly have longer plasma h<strong>al</strong>f-lives and<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 252


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

durations of bioactivity than their nonpegylated<br />

counterparts and benefits of pegylated product given<br />

in table 1. [1]<br />

PROPERTIES OF PEG<br />

Poly<strong>et</strong>hylene glycols are pH-neutr<strong>al</strong>, nontoxic watersoluble<br />

polymers that consist of repeating <strong>et</strong>hylene<br />

oxide subunits, each with a molecular weight of 44,<br />

and two termin<strong>al</strong> hydroxyl groups. They are either<br />

linear (5 to 30 kd) or branched (40 to 60 kd) chain<br />

structures.PEG has Poly-dispersity i.e. Molecular<br />

weight distribution is narrow (1.01 – 1.1). The<br />

pharmacokin<strong>et</strong>ic properties of PEGs vary according<br />

to their molecular weight and site of injection. The<br />

area under the time-concentration curve and the h<strong>al</strong>flife<br />

of PEGs increase with their molecular weight.<br />

For example, after intravenous administration in mice<br />

the h<strong>al</strong>f-life of 50kd PEG is substanti<strong>al</strong>ly longer than<br />

that of 6 kd PEG (987vs 17.6 minutes); 50kd PEG is<br />

<strong>al</strong>so r<strong>et</strong>ained longer at the injection site after<br />

subcutaneous or intramuscular injection than is 6 kd<br />

PEG. Poly<strong>et</strong>hylene glycols appear to undergo<br />

oxidation by the cytochrome P450 enzyme system,<br />

and lowmolecular- weight PEGs are excr<strong>et</strong>ed into the<br />

bile. [1]<br />

PEGylation – MECHANISM OF ACTION<br />

After administration, when PEG comes in contact of<br />

aqueous environment, <strong>et</strong>hylene glycol sub-unit g<strong>et</strong>s<br />

tightly attached to the water molecule. This binding<br />

to water renders them high mobility and hydration.<br />

Hydration and rapid motion causes PEGylated<br />

protein to function, as it causes PEG to sweep out a<br />

large volume which acts like a shield to protect the<br />

attached drug from enzymatic degradation and<br />

interaction with cell surface proteins. This increased<br />

size <strong>al</strong>so helps to prevent rapid ren<strong>al</strong> filtration and<br />

clearance sustaining the drug bioavailability. The<br />

high steric hindrance of branched-chain PEGs<br />

gener<strong>al</strong>ly affords greater protection than do linear<br />

chain. [1]<br />

FACTORS AFFECTING PERFORMANCE OF<br />

PEGylated PRODUCT<br />

Molecular Weight<br />

Molecular weight less than 1000 Da of PEG broken<br />

down into sub-units, and have some toxicity, while<br />

Molecular weight greater than 1000 Da of PEG: does<br />

not demonstrate any toxicity in vivo.<br />

Molecular Weight upto 40,000 – 50,000 Da: used in<br />

clinic<strong>al</strong> and approved pharmaceutic<strong>al</strong> application.<br />

The molecular weight of PEG has a direct impact on<br />

the activity; Higher molecular weight PEGs tends to<br />

have higher in-vivo activity due to the improved<br />

pharmacokin<strong>et</strong>ic profile like increasing h<strong>al</strong>f life as<br />

earlier discussed. [1]<br />

Structure<br />

Branched structure has more size than same<br />

molecular weight linear structure so; it’s <strong>al</strong>so helps to<br />

prevent rapid ren<strong>al</strong> filtration and clearance sustaining<br />

the drug bioavailability. The high steric hindrance of<br />

branched-chain PEGs gener<strong>al</strong>ly affords greater<br />

protection than do linear chain. [1,4]<br />

Number of PEG chains<br />

Two or more lower molecular weight chains can be<br />

added to increase tot<strong>al</strong> molecular weight of PEG<br />

complex<br />

Specific location of PEG site of attachment to the<br />

molecule.<br />

Optim<strong>al</strong> PEGylation is product-specific, and can vary<br />

depending on the site of attachment, the chemistry<br />

used to create the conjugate, and the characteristics of<br />

the PEG used. Effective PEGylation of a drug may<br />

be achieved by attaching a single large PEG at a<br />

single site, a branched PEG at a single site, or sever<strong>al</strong><br />

sm<strong>al</strong>l PEG chains at sever<strong>al</strong> sites. [1]<br />

CHEMISTRY OF PEGYLATION<br />

To couple PEG to a molecule (i.e. polypeptides,<br />

polysaccharides, polynucleotide’s and sm<strong>al</strong>l organic<br />

molecules) as shown in Figure 1, it is necessary to<br />

activate the PEG by preparing a derivative of the<br />

PEG having a function<strong>al</strong> group at one or both<br />

termini. The function<strong>al</strong> group is chosen based on the<br />

type of available reactive group on the molecule that<br />

will be coupled to the PEG. For proteins, typic<strong>al</strong><br />

reactive amino acids include lysine, cysteine,<br />

histidine, arginine, aspartic acid, glutamic acid,<br />

serine, threonine, tyrosine, N-termin<strong>al</strong> amino group<br />

and the C-termin<strong>al</strong> carboxylic acid. In the case of<br />

glycoproteins, vicin<strong>al</strong> hydroxyl groups can be<br />

oxidized with periodate to form two reactive formyl<br />

moi<strong>et</strong>ies. [3]<br />

The most common route for PEG conjugation of<br />

proteins has been to activate the PEG with function<strong>al</strong><br />

groups suitable for reaction with lysine and N-<br />

termin<strong>al</strong> amino acid groups. Lysine is one of the most<br />

prev<strong>al</strong>ent amino acids in proteins and can be upwards<br />

of 10% of the over<strong>al</strong>l amino acid sequence. In<br />

reactions b<strong>et</strong>ween electrophilic<strong>al</strong>ly activated PEG<br />

and nucleophilic amino acids, it is typic<strong>al</strong> that sever<strong>al</strong><br />

amines are substituted. When multiple lysines have<br />

been modified, a h<strong>et</strong>erogeneous mixture is produced,<br />

which is composed of a population of sever<strong>al</strong><br />

poly<strong>et</strong>hylene glycol molecules attached per protein<br />

molecule (‘PEGmers’) ranging from zero to the<br />

number of ´-and a-amine groups in the protein. [3]<br />

PEGylation technology is classified into two types:<br />

1. Early PEGylation technology (First<br />

generation PEGylation)<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 253


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

2. Advanced PEGylation technology (Second<br />

generation PEGylation)<br />

1. First Generation PEGylation<br />

First generation PEGylation m<strong>et</strong>hods were fraught<br />

with difficulties.<br />

With first generation PEGylation, the PEG polymer<br />

was gener<strong>al</strong>ly attached to ε amino group of lysine,<br />

and gave mixtures of PEG isomers with different<br />

molecular masses. The existence of these isomers<br />

makes it difficult to reproduce drug batches, and can<br />

contribute to the antigenecity of the drug and poor<br />

clinic<strong>al</strong> outcomes. In addition, first generation<br />

m<strong>et</strong>hods mainly used linear PEG polymers of 12 kDa<br />

or less. Unstable bonds b<strong>et</strong>ween the drug and PEG<br />

were <strong>al</strong>so som<strong>et</strong>imes used, which leads to<br />

degradation of PEG-drug conjugate during<br />

manufacturing and injection Early PEGylation was<br />

performed with m<strong>et</strong>hoxy-PEG (m-PEG), which was<br />

contaminated with PEG diol and which resulted in<br />

the cross-linking of proteins to form inactive<br />

aggregates. Diol contamination can reach upto 10-15<br />

%. [1]<br />

Despite these limitations, sever<strong>al</strong> first generation<br />

PEGylated drugs receive regulatory approv<strong>al</strong>.<br />

Example: Still in use today are Pegademase<br />

(ADAGEN ® ), a PEGylated form of the enzyme<br />

adenosine de-aminase for the treatment of Severe<br />

Combined Immuno-Deficiency (SCID) and<br />

Pegaspargase (ONCASPAR ® ), a PEGylated form of<br />

enzyme asparginase for the treatment of Leukemia. [5]<br />

2. Second Generation PEGylation<br />

Second generation PEGylation strives to avoid<br />

pitf<strong>al</strong>ls associated with the first generation<br />

PEGylation.<br />

Over<strong>al</strong>l go<strong>al</strong> of this technology is to create larger<br />

PEG polymers to improve the Pharmacokin<strong>et</strong>ics and<br />

Pharmacodynamic effects seen with lower molecular<br />

mass PEGs.<br />

Newer pegylation m<strong>et</strong>hods create conjugates with<br />

strong linkages that are resistant to side-reactions and<br />

are able to withstand purification to remove dio1<br />

contaminants, thereby making it possible to use high<br />

molecular- weight PEGs . These m<strong>et</strong>hods attach an<br />

activated PEG to the drug molecule by incorporating<br />

part of the activating group as a link b<strong>et</strong>ween the two<br />

entities. For example, PEGFILGRASTIM ® is formed<br />

by cov<strong>al</strong>ently attaching a 20 kd PEG chain through a<br />

stable secondary amine bond directly to the termin<strong>al</strong><br />

amino group of the filgrastim molecule. In this way,<br />

the nitrogen atom to which the PEG chain is attached<br />

r<strong>et</strong>ains its surface charge, a factor that has been<br />

shown to be cruci<strong>al</strong> in conserving the bioactivity of<br />

some molecules. [1]<br />

Amine PEGylation and N-termin<strong>al</strong> PEGylation<br />

Since most applications of PEG conjugation involve<br />

labile molecules, the coupling reaction gener<strong>al</strong>ly<br />

requires mild chemic<strong>al</strong> conditions. In case of<br />

polypeptides, the most common reactive groups<br />

involved in coupling are nucleophiles with the<br />

following decreasing rank order of reactivity: thiol,<br />

<strong>al</strong>pha amino group, epsilon amino group,<br />

carboxylate, hydroxylate. However, this order is not<br />

absolute, since it depends <strong>al</strong>so on the reaction pH,<br />

furthermore other residues may react in speci<strong>al</strong><br />

conditions, as the imidazole group of histidine. The<br />

thiol group is rarely present in proteins, furthermore<br />

it is often involved in active sites. The carboxylic<br />

groups cannot be easily activated without having.<br />

reaction with the protein amino groups, to yield intra<br />

or inter molecular cross linking. Therefore, amino<br />

groups, namely the <strong>al</strong>pha amino or the epsilon amino<br />

of lysine, are the usu<strong>al</strong> sites of PEG linking. [4]<br />

PEGylating Agents used for amino PEGylation<br />

shown in Table 2.<br />

Carboxyl PEGylation<br />

PEG reagents react with carboxylic acid in the<br />

presence of coupling agents such as<br />

DCC (N,N'-dicyclohexylcarbodiimide) and EDIC (N-<br />

(3-dim<strong>et</strong>hylaminopropyl)-N' <strong>et</strong>hylcarbodiimide, HCl<br />

s<strong>al</strong>t). However, the procedure is successful only when<br />

amines are not present in the compound, as for<br />

instance in the case of non-peptide drugs. In peptides<br />

and proteins the risk of cross-linking is difficult to<br />

avoid. [3] PEGylating Agents used for Carboxyl<br />

PEGylation are shown in Table 3.<br />

PEGylation at the –SH (thiol) groups of Cysteine of<br />

polypeptides<br />

PEGylation of free cysteine residues in proteins is the<br />

main approach for site-specific modification because<br />

reagents that specific<strong>al</strong>ly react with cysteines have<br />

been synthesized, and the number of free cysteines on<br />

the surface of a protein is much less than that of<br />

lysine residues. In the absence of a free cysteine in a<br />

native protein, one or more free cysteines can be<br />

added by gen<strong>et</strong>ic engineering. PEGylating site<br />

specific<strong>al</strong>ly can minimize the loss of biologic<strong>al</strong><br />

activity and reduce immunogenecity. [3] PEGylating<br />

Agents used for thiol PEGylation are shown in Table<br />

3.<br />

Hydroxyl PEGylation<br />

PEG-isocyanate is useful for hydroxyl group<br />

conjugation yielding a stable ur<strong>et</strong>hane linkage.<br />

However, its reactivity may be best exploited for<br />

non-peptide moi<strong>et</strong>ies such as drugs or hydroxylcontaining<br />

matrices to yield biocompatible surfaces.<br />

PEG-isocyanate is in fact highly reactive with amines<br />

<strong>al</strong>so. [3] PEGylating Agents used for Hydroxyl<br />

PEGylation are shown in Table 3.<br />

H<strong>et</strong>ero-bifunction<strong>al</strong> PEGs<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 254


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

As applications of PEG chemistry have become more<br />

sophisticated, there has been an increasing need for<br />

h<strong>et</strong>erobifunction<strong>al</strong> PEGs, which are PEGs bearing<br />

dissimilar termin<strong>al</strong> groups. Such h<strong>et</strong>erobifunction<strong>al</strong><br />

PEGs bearing appropriate function<strong>al</strong> groups may be<br />

used to link two entities where a hydrophilic, flexible,<br />

and biocompatible spacer is needed.<br />

H<strong>et</strong>erobifunction<strong>al</strong> PEG can be used in a vari<strong>et</strong>y of<br />

ways that includes linking macromolecules to<br />

surfaces (for immunoassays, biosensors or various<br />

probe applications), targ<strong>et</strong>ing of drugs, liposomes and<br />

viruses to specific tissues, liquid phase peptide<br />

synthesis and many others. Preferred end groups for<br />

h<strong>et</strong>ero-bifunction<strong>al</strong> PEGs are NHS esters, m<strong>al</strong>eimide,<br />

vinyl sulfone, pyridyl disulfide, amine, and<br />

carboxylic acids. [3]<br />

Branched structures<br />

Second generation PEGylation uses branched<br />

structures of PEG, in contrast to the solely linear<br />

structures found in first generation PEGs. Branched<br />

PEGs of greatly increased molecular masses – upto<br />

60 kDa or more, as compared with the 12 kDa or less<br />

found in the first generation PEGs – have been<br />

prepared. A branched PEG ‘acts’ as if it were much<br />

larger than a corresponding linear PEG of the same<br />

molecular mass. Branched PEGs are <strong>al</strong>so b<strong>et</strong>ter at<br />

cloaking the attached polypeptide drug from the<br />

immune system and proteolytic enzymes, thereby<br />

reducing its antigenecity and likelihood of<br />

destruction. [1,3]<br />

Specific PEGylation by enzymes or by reversible<br />

protection<br />

The specific conjugation of PEG to the amide group<br />

of glutamines or to the hydroxyl group of serines and<br />

threonines is only possible under mild conditions<br />

using enzymes. Sato discovered that glutamine in<br />

proteins can be the substrate of the transglutaminase<br />

enzymes, if an amino PEG is used as the nucleophilic<br />

donor. Through a transglutamination reaction the<br />

enzyme links PEG to the protein at the level of the<br />

glutamine residue as shown in Figure 2. [2] Now a<br />

days, PEG conjugates with different enzyme like<br />

arginines n histaminase are <strong>al</strong>so available.<br />

LIMITATIONS IN TH USE OF PEG<br />

PEG is obtained by chemic<strong>al</strong> synthesis and, like <strong>al</strong>l<br />

synth<strong>et</strong>ic polymers, it is polydisperse, which means<br />

that the polymer’s batch is composed of molecules<br />

having different number of monomers, yielding a<br />

Gaussian distribution of the molecular weights. This<br />

leads to a population of drug conjugates, which<br />

might have different biologic<strong>al</strong> properties, mainly in<br />

body-residence time and immunogenicity.<br />

Polydispersivity problem must be still taken into<br />

consideration, especi<strong>al</strong>ly when de<strong>al</strong>ing with low<br />

molecular weight drugs, either peptide or nonpeptide<br />

drugs, where the mass of linked PEG is more<br />

relevant for conveying the conjugate’s<br />

characteristics, mainly those related to the molecular<br />

size. A second problem for the use of this polymer<br />

relates to the excr<strong>et</strong>ion from the body. As for other<br />

polymers, PEGs are usu<strong>al</strong>ly excr<strong>et</strong>ed in urine or<br />

feces but at high molecular weights they can<br />

accumulate in the liver, leading to macromolecular<br />

syndrome. [2]<br />

APPLICATION OF PEGYLATION<br />

PEG as Diagnostic Carrier<br />

In vivo non invasive diagnosis is done by using<br />

tracers d<strong>et</strong>ected through magn<strong>et</strong>ic resonance or<br />

radioactivity. Usu<strong>al</strong>ly they are administered in a<br />

chelated form using compounds that can give<br />

specific biodistribution, stability or targ<strong>et</strong>ing.<br />

PEGylation increases the body-residence time of<br />

paramagn<strong>et</strong>ic chelates that will be cleared more<br />

slowly than the unmodified molecules through the<br />

kidney or liver, thus <strong>al</strong>lowing more d<strong>et</strong>ailed images<br />

by magn<strong>et</strong>ic resonance. C225 is a monoclon<strong>al</strong><br />

antibody directed against the epiderm<strong>al</strong> growth factor<br />

receptor, which was conjugated to a<br />

h<strong>et</strong>erobifunction<strong>al</strong> PEG bearing a radiom<strong>et</strong><strong>al</strong> chelator<br />

(di<strong>et</strong>hylen<strong>et</strong>riaminepentaac<strong>et</strong>ic acid, DTPA) at one<br />

terminus. The conjugate DTPA–PEG–C225 r<strong>et</strong>ained<br />

66% of binding affinity, and, more importantly,<br />

when labeled with Indium-111 (111In) it showed<br />

narrower steady-state distribution than the non-<br />

PEGylated 111In–DTPA–C225, because of reduced<br />

nonspecific binding. Therefore, in case of protein<br />

targ<strong>et</strong>ed diagnostic, PEG could help to collect b<strong>et</strong>ter<br />

defined images by limiting the background noise due<br />

to nonspecific protein–protein interaction. [2]<br />

PEG oligonucleotides<br />

Mainly antisense oligonucleotides and are now under<br />

active investigation as new potenti<strong>al</strong> drugs because<br />

of their extremely high selectivity in targ<strong>et</strong><br />

recognition. All of them, however, share the<br />

problems of short h<strong>al</strong>f-life in vivo because of either<br />

low stability towards the eso- and endo-nucleases<br />

(present in plasma and inside the cells) or their rapid<br />

excr<strong>et</strong>ion caused by their sm<strong>al</strong>l size. Furthermore,<br />

their negative charge prevents an easy pen<strong>et</strong>ration<br />

into the cells. A PEG molecule, bound to the<br />

hydroxyl group of a nucleic acid (directly or through<br />

a spacer link), was found to increase the stability<br />

towards enzyme degradation,prolong the plasma<br />

permanence and enhance the pen<strong>et</strong>ration into cells by<br />

masking the negative charges of oligonucleotides. A<br />

PEGylated aptamer, the 28mer oligomeraptanib, has<br />

<strong>al</strong>ready been approved by FDA for the treatment of<br />

age-related macular degeneration of r<strong>et</strong>ina. In this<br />

product, a branched PEG of 40 kDa was attached to<br />

the oligonucleotides through a pentamino linker. [2]<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 255


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

PEGylated conjugate as Anticancer agent [7]<br />

PEG conjugates with low molecular weight<br />

anticancer drugs<br />

PEG has been successful for protein modification but<br />

in the case of low molecular weight drugs it presents<br />

a cruci<strong>al</strong> limit, the low drug payload accompanying<br />

the available m<strong>et</strong>hoxy or diol forms of this polymer.<br />

This intrinsic limitation had for many years<br />

prevented the development of a sm<strong>al</strong>l drug-PEG<br />

conjugate. A few studies have been conducted to<br />

overcome the low PEG loading by either branching<br />

the end chain groups or coupling on them sm<strong>al</strong>l<br />

Dendron structures.<br />

Pegamotecan ® (Enzon Pharmaceutic<strong>al</strong>s, Inc.) is a<br />

prodrug obtained by coupling two molecules of<br />

camptothecin to a diol PEG of 40 kDa. The drug is<br />

linked through an ester bond involving the C-20<br />

hydroxyl group and a carboxylic group of PEG. The<br />

aim of this approach was double, to increase the drug<br />

h<strong>al</strong>f-life in blood by PEGylation and to stabilize by<br />

acylation the active lactone configuration of<br />

camptothecin.<br />

PEG-irinotecan: The architecture of new multi-arm<br />

PEGs was <strong>al</strong>so exploited for the preparation of PEGirinotecan<br />

(NKTR-102) by Nektar Therapeutics. The<br />

drug has been cov<strong>al</strong>ently bound to a four arms PEG.<br />

In preclinic<strong>al</strong> studies NKTR-102 plasma h<strong>al</strong>f-life<br />

was ev<strong>al</strong>uated in a mouse model taking into<br />

consideration the active m<strong>et</strong>abolite SN-38, released<br />

from irinotecan. The conjugate showed prolonged<br />

pharmacokin<strong>et</strong>ic profiles with a h<strong>al</strong>f-life of 15 days<br />

when compared to 4 h with free irinotecan.<br />

PEG-doc<strong>et</strong>axel : PEGylated doc<strong>et</strong>axel (NKTR-105)<br />

has been prepared with the same multi-arm PEG<br />

technology. The derivative has shown good<br />

preclinic<strong>al</strong> activity in colon and lung cancer<br />

xenograft models.This product has just entered phase<br />

1 clinic<strong>al</strong> studies enrolling approximately 30 patients<br />

with refractory solid tumours who have failed <strong>al</strong>l<br />

prior available therapies.<br />

PEG-Protein conjugates<br />

In PEGylation of protein conjugate two different<br />

approaches can be identified based on the type of<br />

protein studied:<br />

H<strong>et</strong>erologous protein, Usu<strong>al</strong>ly the main limit of these<br />

proteins is the immunogenicity rather than a short<br />

pharmacokin<strong>et</strong>ic. Therefore, both PEG molecular<br />

weight and coupling chemistry should ensure a wide<br />

shielding of the protein surface or, at least, the<br />

immunogenic sites. Basic<strong>al</strong>ly, in these cases low<br />

molecular weight PEGs (5– 10 kDa) and random<br />

amine coupling are used. It is important to note that<br />

<strong>al</strong>l the enzymes studied possess sm<strong>al</strong>l substrates;<br />

these can cross the PEG layer, around the protein,<br />

and easily reach the active site. Conversely, active<br />

site approach of large and hindered substrates would<br />

be prevented this compromising the enzyme activity.<br />

This would suggest that PEGylation may be not a<br />

suitable approach for immunogenic enzymes having<br />

big substrates.<br />

Endogenous protein, For these biopharmaceutic<strong>al</strong><br />

drugs the prolongation of body circulation h<strong>al</strong>f-life is<br />

the driving force in seeking for a polymer conjugate.<br />

Most of the endogenous proteins act through a<br />

receptor-mediated activity. This dictates the strategy<br />

for an optimum PEGylation approach, namely a site<br />

specific conjugation to generate monoPEGylated<br />

isomers. In particular the site of polymer attachment<br />

must be far from the receptor recognition area. In this<br />

case, it is mandatory the use of high molecular<br />

weight polymers to reach the PEG mass for the<br />

desired h<strong>al</strong>f-life prolongation.<br />

PEG-antibody fragment angiogenesis inhibitor<br />

(CDP791)<br />

Vascular endotheli<strong>al</strong> growth factor receptor-2<br />

(VEGFR-2) is involved in the formation of new<br />

blood vessels in tumours (angiogenesis), <strong>al</strong>lowing<br />

cancer cells to receive nutrients and to maintain<br />

growth. Therefore, a molecule able to block VEGFR-<br />

2 can interfere with the development of tumour<br />

vasculature. CDP791 is a PEGylated diFab antibody<br />

that binds the VEGFR-2, with a Kd of 49pM,<br />

preventing the activation by VEGF ligands. The<br />

unconjugated CDP791 antibody fragment is affected<br />

by a too fast in vivo clearance, because it has a<br />

reduced mass due to the absence of Fc region. This<br />

problem was overcome by PEGylation of the<br />

cysteine present at the C-terminus.<br />

PEG-interferon-<strong>al</strong>pha conjugates<br />

Sever<strong>al</strong> clinic<strong>al</strong> studies are ev<strong>al</strong>uating the<br />

effectiveness of PEGinterferon-α2b (PEG-<br />

INTRON®), presently used for the treatment of<br />

hepatitis B and C, as adjuvant therapy in certain<br />

anticancer protocols. The native interferon-α2b is one<br />

of the most studied agents for adjuvant therapy in<br />

stage IIb and stage III melanoma. Improvements in<br />

the recurrence-free surviv<strong>al</strong> have been shown when<br />

interferon- α2b therapy was prolonged for 12–15<br />

months. This long therapy, consisting in a daily drug<br />

administration, can particularly compromise the<br />

patient compliance. This can be highly improved<br />

using the PEGylated form of interferon-α2b, a<br />

monoPEGylated derivative obtained by conjugating<br />

the protein with a linear 12 kDa amino reactive<br />

PEGylating agent. The conjugate maintains the<br />

therapeutic level of interferon-α2b by a weekly, self<br />

administered, dose schedule and its saf<strong>et</strong>y has been<br />

studied in sever<strong>al</strong> cancers.<br />

PEG-Interferon-α2a, a conjugate obtained by linking<br />

a branched PEG 40 kDa to the protein and mark<strong>et</strong>ed<br />

as PEGASYS®, is used in clinic to treat hepatitis as<br />

PEG-INTRON®. The higher polymer molecular<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 256


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

weight of PEGASYS® (40 kDa versus 12 kDa of<br />

PEG-INTRON®) and the higher stability of the<br />

PEG-protein linkages (i.e. His residues are involved<br />

in this case) <strong>al</strong>lowed for the prolonging of the in vivo<br />

h<strong>al</strong>f-life to 65 h with respect to the 27–37 h of PEG-<br />

INTRON®.<br />

PEGylation: the in vitro activity is reduced to about<br />

7% of that of native interferon, this being the<br />

weakness of stable polymer conjugation, but this<br />

limitation is more than counterb<strong>al</strong>anced by the<br />

enhanced in vivo h<strong>al</strong>f-life of the conjugate.<br />

PEG-granulocyte colony stimulating factor<br />

Granulocyte colony stimulating factor (G-CSF) is<br />

used as adjuvant therapy to treat granulocytes<br />

depl<strong>et</strong>ion during chemotherapy. The fast blood<br />

clearance of the free drug was addressed by<br />

PEGylation. Different PEG coupling approaches<br />

were conducted but the most successful one consisted<br />

of a reductive <strong>al</strong>kylation with PEG <strong>al</strong>dehyde<br />

performed at acidic pH. Under this condition, a<br />

monoPEGylated conjugate was preferenti<strong>al</strong>ly<br />

obtained in which the polymer was linked to the<br />

protein N-termin<strong>al</strong> α amino group. The PEG 20 kDa<br />

conjugate showed an improved pharmacokin<strong>et</strong>ic<br />

profile as consequence to the reduced kidney<br />

excr<strong>et</strong>ion.<br />

The PEG-G-CSF conjugate (Pegfilgastrim,<br />

Neulasta®) was approved for human use in 2002 for<br />

the first and subsequent cycleadministration against<br />

febrile neutropenia in patients with nonmyeloid<br />

m<strong>al</strong>ignancies receiving myelosuppressive<br />

chemotherapy associated with a 30%–40% risk of<br />

febrile neutropenia.<br />

PEG conjugates with enzymes<br />

PEG conjugated Asparaginase<br />

sever<strong>al</strong> leukemic lymphoblasts cells rely on the<br />

serum supply of asparagine, for their growth,<br />

because they lack the enzyme asparagine synth<strong>et</strong>ase.<br />

Asparaginase, the enzyme that converts asparagine<br />

into aspartate and ammonia, has therefore been<br />

proposed as a therapeutic agent for acute<br />

lymphoblastic leukaemia (ALL). FDA approv<strong>al</strong> for<br />

PEG-asparaginase (Rhone-Poulenc Rorer as<br />

Oncaspar®) was granted in 1994 for treatment of<br />

patients with ALL who are hypersensitive to the two<br />

native isoforms of the enzyme. PEGylated<br />

asparaginase has been used in combination with<br />

sever<strong>al</strong> tradition<strong>al</strong> anticancer molecules, often in a<br />

multiagent regimen, including for example one or<br />

more of the following drugs: cyclophosphamide,<br />

daunorubicin, vincristine, cytarabine, prednisone,<br />

<strong>et</strong>c. In these studies the PEGylated enzyme was well<br />

tolerated, showing hyperbilirubinaemia and<br />

hyperglycaemia as the most common adverse effects.<br />

Arginine deiminase and Arginase<br />

In literature two types of arginine degrading enzymes<br />

are reported, and both have been suggested as<br />

antitumour agents: i) arginine deiminase (ADI),<br />

which degrades arginine in citrulline and<br />

ammonia, ii) arginase (ARG) that cat<strong>al</strong>yses the<br />

conversion of arginine in ornithine and urea. This<br />

enzyme was shown to be even more powerful than<br />

asparaginase in killing human leukaemia cells.<br />

arginine depl<strong>et</strong>ing enzymes can be useful in treating<br />

these tumours. Indeed, arginine deficiency inhibits<br />

tumour growth, angiogenesis and nitric oxide<br />

synthesis.<br />

Chitosan–PEG nanocapsules as new carriers for<br />

or<strong>al</strong> peptide delivery<br />

Chitosan–PEG nanocapsules and the control PEGcoated<br />

nanoemulsions were obtained by the solvent<br />

displacement technique. Their size was in the range<br />

of 160–250 nm. Their z<strong>et</strong>a potenti<strong>al</strong> was greatly<br />

affected by the nature of the coating, being positive<br />

for chitosan–PEG nanocapsules and negative in the<br />

case of PEG-coated nanoemulsions. The presence of<br />

PEG, wh<strong>et</strong>her <strong>al</strong>one or grafted to chitosan, improved<br />

the stability of the nanocapsules in the<br />

gastrointestin<strong>al</strong> fluids. Using the Caco-2 model cell<br />

line it was observed that the pegylation of chitosan<br />

reduced the cytotoxicity of the nanocapsules. Fin<strong>al</strong>ly,<br />

the results of the in vivo studies showed the capacity<br />

of chitosan–PEG nanocapsules to enhance and<br />

prolong the intestin<strong>al</strong> absorption of s<strong>al</strong>mon<br />

c<strong>al</strong>citonin. Addition<strong>al</strong>ly, they indicated that the<br />

pegylation degree affected the in vivo performance of<br />

the nanocapsules. Therefore, by modulating the<br />

pegylation degree of chitosan, it was possible to<br />

obtain nanocapsules with a good stability, a low<br />

cytotoxicity and with absorption enhancing<br />

properties. [8]<br />

Gene Delivery<br />

Poly<strong>et</strong>hyleneglycol modified poly<strong>et</strong>hylenimine for<br />

improved CNS gene transfer<br />

One problem of using polycation DNA complexes,<br />

especi<strong>al</strong>ly in an in vivo study, is their poor solubility.<br />

They may immediately precipitate out of a solution<br />

when prepared at a higher concentration.<br />

Poly<strong>et</strong>hylene glycol (PEG) modification<br />

(PEGylation) often can improve the solubility of<br />

macromolecules, minimize aggregation of<br />

particulates and reduce their interaction with proteins<br />

in the physiologic<strong>al</strong> fluid. PEGylation of PEI reduced<br />

surface charge of PEI/DNA particles, increased their<br />

dispersion ability at high concentrations, decreased<br />

plasma protein binding and erythrocyte aggregation,<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 257


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

prolonged blood circulation and reduced systemic<br />

toxicity & increased invivo transgene expression of<br />

PEI. The study provides the in vivo evidence that an<br />

appropriate degree of PEG modification is decisive in<br />

improving gene transfer mediated by PEGylated<br />

polymers. [9]<br />

Sm<strong>al</strong>l interfering RNA (siRNA) delivery<br />

Sm<strong>al</strong>l interfering RNA was conjugated with<br />

poly(<strong>et</strong>hylene glycol) (PEG) at four different termin<strong>al</strong><br />

ends (sense 3′, sense 5′, antisense 3′, and antisense 5′)<br />

via cleavable disulfide and noncleavable thio<strong>et</strong>her for<br />

gene silencing efficiencies. The PEGylation site at<br />

the four siRNA termini and PEG molecular weight<br />

were not critic<strong>al</strong> factors to significantly affect gene<br />

silencing activities. Cleavable siRNA-PEG<br />

conjugates showed comparable gene silencing<br />

activities to naked siRNA, and exhibited sequencespecific<br />

degradation of a targ<strong>et</strong> mRNA. Interestingly,<br />

noncleavable siRNA-PEG conjugates were processed<br />

by Dicer, enabling to exert RNAi effect without<br />

showing a targ<strong>et</strong> sequence-specific manner.<br />

However, only cleavable siRNA-PEG conjugates<br />

significantly reduced the extent of INF-α release as<br />

compared to noncleavable siRNA-PEG conjugates,<br />

suggesting that they can be potenti<strong>al</strong>ly used for<br />

therapeutic siRNA applications. [10]<br />

Dendrimer<br />

in drug delivery mostly in antivir<strong>al</strong> and cancer<br />

therapy. [11]<br />

Insulin PEGylation<br />

Despite the robust structure of<br />

polyamidoamine(PAMAM)dendrimers, they are not<br />

stable when complexed with surfactants.<br />

Modification of PAMAM dendrimers by grafting<br />

PEG chains on the surface of PAMAM substanti<strong>al</strong>ly<br />

improves its colloid<strong>al</strong> stability in the presence of<br />

sodiumdodecylsulfate(SDS). Michael addition<br />

reaction was employed to synthesize PEGylated-<br />

PAMAM by activating MPEG with 4-<br />

nitrophenylchloroformate.The PEGylated-PAMAM<br />

dendrimers did not aggregate in the presence of upto<br />

100mM SDS as the complexes were steric<strong>al</strong>ly<br />

stabilized by PEGchains. ITC and z<strong>et</strong>apotenti<strong>al</strong><br />

measurements reve<strong>al</strong>ed that the binding mechanism<br />

of SDS and PEGylated-PAMAM was induced by<br />

electrostatic interaction and polymer-induced<br />

micellization of SDS on PEG chains. The interaction<br />

of PEGylated-PAMAM and amphiphilic molecules,<br />

such as SDS was elucidated, and this provided a<br />

useful basis for the application PEGylated-PAMAM<br />

A novel long-acting insulin based on the following<br />

properties: (i) action as a prodrug to preclude supra<br />

physiologic<strong>al</strong> concentrations shortly after injection;<br />

(ii) maintenance of low-circulating level of<br />

biologic<strong>al</strong>ly active insulin for prolonged period; and<br />

(iii) high solubility in aqueous solution. A<br />

spontaneously hydrolyzable prodrug was thus<br />

designed and prepared by conjugating insulin through<br />

its amino side chains to a 40 kDa poly<strong>et</strong>hylene glycol<br />

containing sulfhydryl moi<strong>et</strong>y (PEG40-SH),<br />

employing recently developed h<strong>et</strong>ero-bifunction<strong>al</strong><br />

spacer<br />

9-hydroxym<strong>et</strong>hyl-7(amino-3-<br />

m<strong>al</strong>eimidopropionate)-fluorene-Nhydroxysucinimide<br />

(MAL-Fmoc-0Su). A conjugate<br />

trapped in the circulatory system and capable of<br />

releasing insulin by spontaneous chemic<strong>al</strong> hydrolysis<br />

has been created. PEG40-Fmoc-insulin is a watersoluble,<br />

reactivatable prodrug with low biologic<strong>al</strong><br />

activity. Upon incubation at physiologic<strong>al</strong> conditions,<br />

the cov<strong>al</strong>ently linked insulin undergoes spontaneous<br />

hydrolysis at a slow rate and in a linear fashion,<br />

releasing the nonmodified immunologic<strong>al</strong>ly and<br />

biologic<strong>al</strong>ly active insulin with a t1/2 v<strong>al</strong>ue of 30h. A<br />

single subcutaneous administration of PEG40-Fmocinsulin<br />

to he<strong>al</strong>thy and diab<strong>et</strong>ic rodents facilitates<br />

prolonged glucose-lowering effects 4- to 7-fold<br />

greater than similar doses of the native hormone. The<br />

benefici<strong>al</strong> pharmacologic<strong>al</strong> features endowed by<br />

PEGylation are thus preserved. In contrast,<br />

nonreversible, ‘‘convention<strong>al</strong>” pegylation of insulin<br />

led to inactivation of the hormone. [12]<br />

PEGylated derivatives of rosin (PD) used as<br />

sustained release film forming materi<strong>al</strong>s for<br />

controlled release formulation. The mechanism of<br />

drug release from these coated systems however<br />

followed class II transport (n>1.0). [13]<br />

Recently approved pegylated products shown in<br />

Table 4.<br />

CONCLUSION<br />

PEGylation improves the biopharmaceutic<strong>al</strong><br />

properties of drugs that increase stability, resistant to<br />

proteolytic inactivation, decrease to nonexistent<br />

immunogenicity, increase circulatory lives and low<br />

toxicity. These type of <strong>al</strong>ter properties improve the<br />

efficacy of protein and peptide drug delivery.<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 258


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

Table 1. Potenti<strong>al</strong> benefits of PEGylated products<br />

Greater biologic activity<br />

Greater passive tumour targ<strong>et</strong>ing of liposomes<br />

Longer circulating h<strong>al</strong>f-life<br />

Lower peak plasma concentrations<br />

Sm<strong>al</strong>ler fluctuations in plasma concentrations<br />

Less enzymatic degradation<br />

Less immunogenicity and antigenicity<br />

Greater solubility<br />

Less-frequent administration<br />

Greater patient adherence and improved qu<strong>al</strong>ity of life<br />

PEG reagents<br />

PEG-NHS<br />

PEG-<strong>al</strong>dehyde<br />

PEG-isocyanate<br />

PEG epoxide<br />

PEG-isothiocyanate<br />

PEG-COOH<br />

PEG-NPC<br />

PEG-acrylate<br />

Table 2. PEGylating agent for Amino PEGylation [6]<br />

PEGylation<br />

The N-hydroxysuccinimide (NHS) activated ester of PEG carboxylic<br />

acid can react with the amino group of lysine. The coupling requires<br />

only mild conditions, pH 7-9, low temperature (5-25ºC) for short<br />

period of time. The formed amide bond is physiologic<strong>al</strong>ly stable.<br />

Reductive amination with primary amines to produce secondary<br />

amines, in the presence of reducing agents such as sodium<br />

borohydride and sodium cyanoborohydride. pH is important for<br />

reductive amination.<br />

Reaction with amine to produce a stable ur<strong>et</strong>hane linkage.<br />

Nucleophilic addition<br />

React with amine to produce a stable thiourea linkage.<br />

Usu<strong>al</strong>ly the acid needs to be activated, such as NHS ester.<br />

Amine reacts with NPC function<strong>al</strong>ized PEG under proper conditions.<br />

Michael addition b<strong>et</strong>ween amine and acrylate ester<br />

Table 3. PEGylating agent for Carboxyl, Thiol and Hydroxyl PEGylation [6]<br />

PEG reagents for Carboxyl PEGylation<br />

PEG-amine<br />

PEG-hydrazide<br />

Amide formation under DCC or EDIC coupling conditions<br />

After activated by EDIC at mild acidic pH, the carboxyl group of proteins<br />

readily react with PEG-hydrazide, while the amino groups present in <strong>al</strong>l<br />

reagents remain inactive in this particular conditions.<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 259


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

PEG reagents for Thiol (-SH) PEGylation<br />

PEG-M<strong>al</strong>eimide<br />

PEG-OPSS<br />

PEG-vinylsulfone<br />

Michael addition, thiols react with the C=C bond in the m<strong>al</strong>eimic ring to<br />

form a physiologic<strong>al</strong> stable linkage. The best reaction condition is at pH 8.<br />

Disulfide S-S bond formation, which can be reversed by reducing agents<br />

such as sodium borohydride and thio<strong>et</strong>hanolamine.<br />

Michael addition, thiols react with the C=C bond to form<br />

a physiologic<strong>al</strong> stable linkage.<br />

PEG-thiol<br />

Oxidative disulfide S-S bond formation.<br />

PEG reagents for Hydroxyl PEGylation<br />

PEG-isocyanate<br />

PEG-NPC<br />

Hydroxyl groups react with PEG-NCO, however speci<strong>al</strong> considerations are<br />

required.<br />

Hydroxyl groups react with NPC to from a carbonate linkage.<br />

PEG-epoxide PEG-epoxide reacts with hydroxyls best at pH 8.5-9.5.<br />

Table 4. Approved PEGylated Products [5]<br />

Brand name Product Company Indication<br />

PEGasys<br />

PEG-INF α-2a<br />

(interferon)<br />

Hoffmann-La<br />

Roche<br />

Hepatitis<br />

PEG-Intron<br />

PEG-INF α-2b<br />

Enzon<br />

Hepatitis<br />

(interferon)<br />

Neulasta<br />

PEG- filgrastim(granulocyte<br />

colony stimulating factor)<br />

Amgen<br />

Neutropenia<br />

Adagen PEG-adenosine deaminase Enzon Immunodeficiency<br />

Oncaspar PEG-asparginase Enzon Cancer<br />

Somavert PEG-visomant Pfizer Acromeg<strong>al</strong>y<br />

PEG-hirudin PEG-recombinant hirudin Abbot Thrombosis(phase III)<br />

PEG- monoclon<strong>al</strong><br />

antibody<br />

PEG-CDP 870 Pfizer Rheumatoid arthritis<br />

(phase III)<br />

PEG-Axokine PEG-cilliary nurotrophic<br />

factor<br />

Regeneron<br />

Obesity (phase III)<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 260


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

Figure 1. PEGylation process in gener<strong>al</strong><br />

Figure 2. Specific PEGylation of Glutamine by transglutaminase enzyme.<br />

REFERENCES<br />

1. Molineux G, Pegylation: engineering<br />

improved pharmaceutic<strong>al</strong>s for enhanced<br />

therapy. Cancer treatment reviews 2002,<br />

28(A), 13-16.<br />

2. Veronese FM, Pasut G, PEGylation,successful<br />

approach to drug delivery. DDT(Drug<br />

Discovery Today), November 2005, 10(21).<br />

3. Roberts MJ, Bentley MD, Harris JM.<br />

Chemistry for peptide and protein PEGylation.<br />

Advanced Drug Delivery Reviews 2002, 54,<br />

459–476.<br />

4. Veronese FM. Review Peptide and protein<br />

PEGylation: a review of problems and<br />

solutions. Biomateri<strong>al</strong>s 2001, 22, 405-417.<br />

5. Leenders F,Kraehmer R. PEGylation<br />

Technology and PEGylated Drugs. Celares<br />

GmbH, Robert-Roessle-Str. 10, 13125 Berlin,<br />

Germany.<br />

6. PEGylating agent information available at<br />

http://www.creativepegworks.com/pegylation<br />

technology and chemistry.mht updated on 20<br />

April 2011.<br />

7. Pasut G, Veronese FM. PEG conjugates in<br />

clinic<strong>al</strong> development or use as anticancer<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 261


<strong>Shailesh</strong> T. <strong>Prajapati</strong>., <strong>et</strong> <strong>al</strong>. / Internation<strong>al</strong> Journ<strong>al</strong> of Advances in Pharmaceutic<strong>al</strong> Research<br />

agents: An overview. Advanced Drug Delivery<br />

Reviews 2009, 61, 1177–1188.<br />

8. Prego C, Torres D, Fernandez-Megia E,<br />

Novoa-Carb<strong>al</strong>l<strong>al</strong> R, Quiñoá E, Alonso MJ.<br />

Chitosan–PEG nanocapsules as new carriers<br />

for or<strong>al</strong> peptide delivery Effect of chitosan<br />

pegylation degree. Journ<strong>al</strong> of Controlled<br />

Release 2006, 111, 299–308.<br />

9. Tanga GP, Zenga JM, Gaoa SJ, Maa YX, Shia<br />

L, Toob HP, Wanga S. Poly<strong>et</strong>hylene glycol<br />

modified poly<strong>et</strong>hylenimine for improved CNS<br />

gene transfer: effects of PEGylation extent.<br />

Biomateri<strong>al</strong>s 2003, 24, 2351–2362.<br />

10. Jung S, Lee SH, Mok H, Chung HJ, Park TG.<br />

Gene silencing efficiency of siRNA-PEG<br />

conjugates: Effect of PEGylation site and PEG<br />

molecular weight. Journ<strong>al</strong> of Controlled<br />

Release 2010, 144, 306–313.<br />

11. Lima AH, Tamb K. Stabilization of<br />

polyamidoamine<br />

(PAMAM)<br />

dendrimers/sodium dodecyl sulfate complexes<br />

via PEGylation. Colloids and Surfaces<br />

A:Physicochem.Eng 2011.<br />

12. Shechter Y, Mironchik M, Rubinraut S,<br />

Tsubery H, Sasson K, Marcus Y, Fridkin M.<br />

Reversible pegylation of insulin facilitates its<br />

prolonged action in vivo. European Journ<strong>al</strong> of<br />

Pharmaceutics and Biopharmaceutics 70<br />

(2008) 19–28.<br />

13. Nande VS, Barabde UV, Morkhade DM, Joshi<br />

SB, Patil AT. Investigation of PEGylated<br />

Derivatives of Rosin as Sustained Release<br />

Film Formers. AAPS PharmSciTech March<br />

2008, 9(1).<br />

<strong>IJAPR</strong> / June 2011/ Vol. 2 / Issue. 6 / 252 - 262 262

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!