1-s2.0-s0079670006000116-main

39
Polymer–drug conjugates: Progress in polymeric prodrugs Jayant Khandare a , Tamara Minko a,b,c, * a Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854-8020, USA b The Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA c New Jersey Center for Biomaterials, 610 Taylor Road, Piscataway, NJ 08854, USA Received 13 May 2005; received in revised form 30 August 2005; accepted 12 September 2005 Abstract Polymers are used as carriers for the delivery of drugs, proteins, targeting moieties, and imaging agents. Several polymers, poly(ethylene glycol) (PEG), N-(2-hydroxypropyl)methacrylamide (HPMA), and poly(lactide-co-glycolide) (PLGA) copolymers have been successfully utilized in clinical research. Recently, interest in polymer conjugation with biologically active components has increased remarkably as such conjugates are preferably accumulated in solid tumors and can reduce systemic toxicity. Based on the site and the mode of action, polymer conjugates possess either ‘tuned’ degradable or non-degradable bonds. In order to obtain such bonds, most of the strategies involve incorporation of amino acids, peptides or small chains as spacer molecules through multiple steps to include protections and deprotections. There is a need to design efficient synthetic methods to obtain polymeric conjugates with drugs and other bioactive components. Designs should aim to decrease the steric hindrance exhibited by polymers and the biocomponents. In addition, the reactivity of polymer and drug must be enhanced. This is especially true for the use of high molecular weight linear polymers and bulkier unstable drugs such as steroids and chemotherapeutic agents. Further, it is essential to elucidate the structure activity relationship (SAR) of a drug when it is conjugated with a polymer using different conjugation sites, as this can vary the efficacy and mechanism of action when compared with its free form. This review will discuss the current synthetic advances in polymer-conjugation with different bioactive components of clinical importance. In addition, the review will describe the strategies for reduction of steric hindrance and increase in reactivity of the polymers, drugs and bioactive agents and highlight the requisite structure activity relationship in polymer–drug bioconjugates. Finally, we will focus on passive and active targeting of polymeric drug delivery systems to specific site of drug action. q 2006 Elsevier Ltd. All rights reserved. Keywords: Polymer; Conjugation; Drug delivery; Prodrug; Enhanced permeability and retention; In vivo and in vitro Contents 1. Introduction .......................................................................... 360 2. Design and synthesis of polymeric prodrugs ................................................... 362 2.1. Polymeric drug delivery system (PDDS) ................................................ 364 2.2. n-hydroxysuccinimide (NHS) ester and coupling methods .................................... 365 2.3. Incorporation of spacers in prodrug conjugates ............................................ 366 Prog. Polym. Sci. 31 (2006) 359–397 www.elsevier.com/locate/ppolysci 0079-6700/$ - see front matter q 2006 Elsevier Ltd. All rights reserved. doi:10.1016/j.progpolymsci.2005.09.004 * Corresponding author. Address: Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854-8020, USA. Tel.: C1 732 445 3831x214; fax: C1 732 445 3134. E-mail address: [email protected] (T. Minko).

Upload: pollockpagu

Post on 24-Nov-2015

11 views

Category:

Documents


0 download

TRANSCRIPT

  • 2.3. Incorporation of spacers in prodrug conjugates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366

    Prog. Polym. Sci. 31 (2006) 359397

    www.elsevier.com/locate/ppolysci0079-6700/$ - see front matter q 2006 Elsevier Ltd. All rights reserved.Polymerdrug conjugates: Progress in polymeric prodrugs

    Jayant Khandare a, Tamara Minko a,b,c,*

    a Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey,

    160 Frelinghuysen Road, Piscataway, NJ 08854-8020, USAb The Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA

    c New Jersey Center for Biomaterials, 610 Taylor Road, Piscataway, NJ 08854, USA

    Received 13 May 2005; received in revised form 30 August 2005; accepted 12 September 2005

    Abstract

    Polymers are used as carriers for the delivery of drugs, proteins, targeting moieties, and imaging agents. Several polymers,

    poly(ethylene glycol) (PEG), N-(2-hydroxypropyl)methacrylamide (HPMA), and poly(lactide-co-glycolide) (PLGA) copolymers

    have been successfully utilized in clinical research. Recently, interest in polymer conjugation with biologically active components

    has increased remarkably as such conjugates are preferably accumulated in solid tumors and can reduce systemic toxicity. Based on

    the site and the mode of action, polymer conjugates possess either tuned degradable or non-degradable bonds. In order to obtain

    such bonds, most of the strategies involve incorporation of amino acids, peptides or small chains as spacer molecules through

    multiple steps to include protections and deprotections. There is a need to design efficient synthetic methods to obtain polymeric

    conjugates with drugs and other bioactive components. Designs should aim to decrease the steric hindrance exhibited by polymers

    and the biocomponents. In addition, the reactivity of polymer and drug must be enhanced. This is especially true for the use of high

    molecular weight linear polymers and bulkier unstable drugs such as steroids and chemotherapeutic agents. Further, it is essential

    to elucidate the structure activity relationship (SAR) of a drug when it is conjugated with a polymer using different conjugation

    sites, as this can vary the efficacy and mechanism of action when compared with its free form. This review will discuss the current

    synthetic advances in polymer-conjugation with different bioactive components of clinical importance. In addition, the review will

    describe the strategies for reduction of steric hindrance and increase in reactivity of the polymers, drugs and bioactive agents and

    highlight the requisite structure activity relationship in polymerdrug bioconjugates. Finally, we will focus on passive and active

    targeting of polymeric drug delivery systems to specific site of drug action.

    q 2006 Elsevier Ltd. All rights reserved.

    Keywords: Polymer; Conjugation; Drug delivery; Prodrug; Enhanced permeability and retention; In vivo and in vitro

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360

    2. Design and synthesis of polymeric prodrugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 362

    2.1. Polymeric drug delivery system (PDDS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364

    2.2. n-hydroxysuccinimide (NHS) ester and coupling methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365doi:10.1016/j.progpolymsci.2005.09.004

    * Corresponding author. Address: Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey,

    160 Frelinghuysen Road, Piscataway, NJ 08854-8020, USA. Tel.:C1 732 445 3831x214; fax: C1 732 445 3134.E-mail address: [email protected] (T. Minko).

  • cross-linkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 378

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380

    opolymer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388

    R) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388

    n of spacers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 391

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 393

    . . . .

    . . . .

    . . . .

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397360polymer forms so-called polymeric prodrug. Poly-

    meric conjugates of conventional drugs (polymeric

    prodrugs) have several advantages over their low

    molecular weight precursors. The main advantages

    include: (1) an increase in water solubility of low

    soluble or insoluble drugs, and therefore, enhancement

    of drug bioavailability; (2) protection of drug from

    deactivation and preservation of its activity during

    circulation, transport to targeted organ or tissue and

    intracellular trafficking; (3) an improvement in phar-

    macokinetics; (4) a reduction in antigenic activity of the

    drug leading to a less pronounced immunological body

    response; (5) the ability to provide passive or active

    targeting of the drug specifically to the site of its action;

    (6) the possibility to form an advanced complex drug

    delivery system, which, in addition to drug and polymer

    carrier, may include several other active components

    that enhance the specific activity of the main drug. Due

    to these advantages over to free form of a drug, the

    Various architectures of polymers have been used as

    vehicles to deliver drugs, along with relevant targeting

    agents [2,3]. Depending on the nature and site of action

    of a drug, either homopolymers, or graft or block

    Table 1

    Current status of polymeric bioconjugates in cancer therapy

    Polymer Drug/biomolecules

    in conjugate

    Current

    phase

    Name of the

    company

    PEGa Paclitaxel I Pharmacia

    P-HPMAb Doxorubicin (DOX)

    micelle and Platinate

    II/III Access Pharma

    P-HPMA Paclitaxel I Pharmacia

    P-HPMA Camptothecin I Pharmacia

    Poly

    (glutamate)

    Camptothecin III Cell

    Therapeutics

    PEG Camptothecin II Enzon

    PEG Aspartic acid I NCI

    P-HPMA DOX II Pharmacia

    P-HPMA DOX-Galactosamine II Pharmacia

    a Polyethylene glycol (PEG).2.4. Carbodiimide coupling reactions or zero lengths

    3. Design and synthesis of polymeric conjugates . . . . . .

    3.1. Dextran . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    3.2. Poly(ethylene glycol) (PEG) and conjugates . . .

    3.3. PEGHIV agent conjugates . . . . . . . . . . . . . . .

    3.4. PEGdrug and targeted delivery . . . . . . . . . . .

    4. Poly-amino acid conjugates . . . . . . . . . . . . . . . . . . . .

    4.1. N-(2-hydroxypropyl)methacrylamide (HPMA) c

    5. Dendrimer and its conjugates . . . . . . . . . . . . . . . . . . .

    6. Critical aspects of polymer conjugation . . . . . . . . . . .

    6.1. Structureactivity relationship of conjugates (SA

    6.2. Steric hindrance . . . . . . . . . . . . . . . . . . . . . . .

    6.3. Enhanced reactivity of polymers by incorporatio

    6.4. Targeting of polymeric drugs . . . . . . . . . . . . .

    6.4.1. Passive tumor targeting . . . . . . . . . . . . . . . . . .

    6.4.2. Active targeting . . . . . . . . . . . . . . . . . . . . . . .

    7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . .

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    1. Introduction

    A prodrug is a form of a drug that remains inactive

    during its delivery to the site of action and is activated

    by the specific conditions in the targeted site. In other

    words, a prodrug is an inactive precursor of a drug.

    Prodrug reconversion (i.e. its conversion into its active

    form) occurs in the body inside a specific organ, tissue

    or cell. In most cases, normal metabolic processes such

    as the cleavage of a bond between a polymer and a drug

    by specific cellular enzymes are utilized to achieve

    prodrug reconversion. A conjugation of a drug with apolymeric prodrug conjugates has lead into a new era of. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394

    polymeric drug delivery systems (PDDS). The task of

    obtaining a versatile polymer as an ideal candidate in

    drug delivery apparently seems to be intricate, as it has

    to undergo several vigorous clinical barriers. Therefore,

    many researchers rely on the polymers which have been

    approved and well established. For decades, the

    delivery of biomolecules using polymeric materials

    has attracted considerable attention from polymer

    chemists, chemical engineers and pharmaceutical

    scientists [1]. However, the design and synthesis of

    new polymeric candidates in view of their biological

    implications has just been initiated.b Poly N-(2-Hydroxypropyl) methacrylamide (P-HPMA).

  • polymers are being extensively used in bioconjugates.

    In general, a biologically active component undergoes

    numerous physio-chemical barriers, which can be

    further complicated by converting it into a polymeric

    prodrug. In addition, due to their higher molecular

    weight, polymers are known to dominate the physical

    properties of the bioconjugated moiety. It has been well

    established that polymers can enhance the aqueous

    solubility of drug molecules by conjugation. Moreover,

    polymers are being used for the preparation of various

    formulations such as liposomes, microparticles or

    nanoparticles [4]. Along with the polymer, the

    physico-chemical properties of the drug or biomolecule

    to be conjugated are equally important. The following

    properties of the drug molecules make it suitable as an

    ideal candidate to form the polymeric conjugate: (1)

    lower aqueous solubility, (2) instability at varied

    physiological pHs, (3) higher systemic toxicity, and

    (4) reduced cellular entry. Numerous polymeric

    prodrugs are in clinical phases (Table 1) and several

    others have been approved (Table 2).

    Despite progress, the delivery of active components

    using clinically approved polymers remains a challenge.

    Methods such as encapsulation, complexation or

    covalent conjugation are routinely used in drug delivery

    research [5]. But, the resulting complexes formed are

    often unstable in a physiological environment. Various

    bioconjugate methodologies that would form a stable

    bond are being reported. Covalent conjugation of

    biomolecules, e.g. protein drugs to synthetic polymers,

    particularly poly(ethylene glycol) (PEG) does increase

    their plasma residence, reduces protein immunogenicity

    and can increase therapeutic index [6]. Successful

    bioconjugation depends upon the chemical structure,

    molecular weight, steric hindrance and the reactivity of

    the biomolecule as well as the polymer. In order to

    synthesize a bioconjugate, both chemical entities need to

    posses a reactive or functional groups such as COOH,

    OH, SH or NH2. However, the presence of multiple

    reactive groups makes the task a bit complex. Therefore,

    the synthetic methodology to form a conjugate involves

    either protection or deprotection of the groups. There is

    Table 2

    Polymeric drug delivery systems (PDDS) that have received regulatory approval. [17]

    Polymer/Formulation Drug or Active agent Brand/Trade

    name/s

    Manufacturer Therapeutic Indication Approved

    Year/Country

    Liposomal Amphotericin B AmBisome Gilead, Fujisawa Fungal infection 1990 (Europe),

    1997

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 361PEG Adenosine deaminase Adagen

    Styrene maleic acid and

    neocarzinostatin

    copolymer in Ethiodol

    Zinostatin stimalamer SMANCS/

    Lipiodol

    Stealth (PEG-stabilized)

    liposomal

    Doxorubicin Doxil/Caelyx

    Liposomal Doxorubicin Myocet

    Liposomal cytosine Arabinoside DepoCyt

    PEG Interferon a-2b PEG-Intron

    Liposomal Verteporfin Visudyne

    PEG Granulocyte colony

    stimulating factor or

    pegfilgrastim

    Neulasta

    PEG L-asparaginase Oncaspar/

    pegaspargase

    PEG Adenosine deaminase. AdagenLeishmaniasis 2000

    Enzon Severe combined

    immunodeficiency

    1990

    Yamanouchi Hepatocellular carcinoma 1993 1996

    (Japan)

    Schering Plough,

    Alza

    Kaposis sarcoma 1995

    Refractory ovarian cancer 1999

    Refractory breast cancer 2003 (Europe,

    Canada)

    Elan Metastatic breast cancer in

    combination with

    cyclophosphamide

    2000 (Europe)

    SkyePharma Lymphomatous meningitis 1999

    Neoplastic meningitis Phase IV

    Enzon, Schering-

    Plough

    Hepatitis C 2001

    QLT, Novartis Wet macular degeneration

    in conjunction with laser

    treatment

    2000 2001

    (Japan)

    Amgen Reduction of febrile

    neutropenia associated with

    chemotherapy

    2002

    Enzon Antineoplastic 1997

    Enzon immunodeficiency disease 1993

  • type of prodrugthe drug delivery system (DDS).

    Such a system can be constructed not only to target a

    desired organ as a whole, its cells or specific organelles

    inside certain cells but also to release a specified

    amount of the drug at specific times. The polymeric

    prodrug conjugate can also increase aqueous solubility,

    enhance biodistribution and retain the inherent phar-

    macological properties of the drug intact [11]. Three

    major types of polymeric prodrugs are currently being

    used [12]. Prodrugs of the first type are broken down

    inside cells to form active substance or substances. The

    second type of prodrug is usually the combination of

    two or more substances. Under specific intracellular

    conditions, these substances react forming an active

    drug. The third type of prodrug, targeted drug delivery

    systems, usually includes three components: a targeting

    moiety, a carrier, and one or more active component(s).

    The targeting ability of the delivery system depends on

    the several variables including: receptor expression;

    ligands internalization; choice of antibody, antibody

    fragments or non-antibody ligands; and binding affinity

    of the ligand [13]. Therefore, the selection of a suitable

    polymer and a targeting moiety is vital to the

    effectiveness of prodrugs.

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397362further need to design simple and yet appropriate

    polymeric-conjugation methodology. Many of the

    most commonly used strategies involve use of both

    coupling agents such as dicyclohexyl carbodiimide

    (DCC) and 1-ethyl-3-(3-dimethylaminopropyl)carbo-

    diimide or use of N-hydroxysuccinimide esters.

    Chemical conjugation of drugs or other biomole-

    cules to polymers and its modifications can form stable

    bonds such as ester, amide, and disulphide. The

    resulting bond linkage should be relatively stable to

    prevent drug release during its transport before the

    cellular localization of the drug. Covalent bonds (e.g.

    ester or amide) are stable and could deliver the drug at

    the targeted site, but such bonds may not easily release

    targeting agents and peptides under the influence of

    acceptable environmental changes [7]. In the past, most

    of the polymeric prodrugs have been developed for the

    delivery of anticancer agents. High molecular weight

    prodrugs containing cytotoxic components have been

    developed to decrease peripheral side effects and to

    obtain a more specific administration of the drugs to the

    cancerous tissues [8,9]. Favorably, a macromolecular

    antitumor prodrug is expected to be stable in circulation

    and should degrade only after reaching the targeted

    cells or tissues. Polymerdrug conjugates can therefore

    be tailored for activation by extra- or intracellular

    enzymes releasing the parent drug in situ.

    In this review, our focus is on the most promising

    polymer candidates that are being used to form

    bioconjugates. There is a necessity to decrease the

    steric hindrance and increase the reactivity of polymers

    as well as biomolecules to be chemically conjugated. In

    this regard, we will discuss the current progress and

    advances with various methodologies to obtain poly-

    meric prodrugs.

    2. Design and synthesis of polymeric prodrugs

    Over the last decade, polymer chemists have been

    actively involved in designing polymeric materials for

    biomedical applications. One particular approach

    towards an improved use of drugs for therapeutic

    applications is to design polymeric prodrugs. A

    prodrug is a chemical entity of an active parent drug

    with altered physico-chemical properties [10]. In a

    prodrug, a drug precursor remains inactive during

    delivery to the site of action and is specifically activated

    at the target site. The utilization of prodrugs, to a certain

    extent, allows for the preservation of specific activity of

    a drug and targets its release to certain cells or their

    organelles. The most complete realization of theprodrug approach is possible by the use of an advancedFig. 1. Schematic presentation for (a) polymeric prodrug with

    targeting agent and (b) hyperbranched polymer conjugate withtargeting and imaging agent.

  • Fig. 2. Schemes of various polymer architectures showing steric hindrance and crowding effect for chemical conjugation of drug molecules

    (a) linear polymer, (b) copolymer, (c) branched polymer, and (d) hyperbranched polymer.

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 363

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397364Numerous polymeric conjugation methods have

    been attempted since 1950s. In 1955, Jatzkewitz

    reported peptaminpolyvinylpyrrolidone conjugates

    improve the efficacy of the drug [14]. Subsequential

    biological aspects of polymeric prodrugs were not

    previously taken into consideration. However, the

    conjugation methodology would be more applied in

    the forthcoming years. In 1975, a rational model for

    pharmacologically active polymers was proposed by

    Ringsdorfconsidered to be the pioneer of polymeric

    prodrug research [15]. In general, an ideal polymeric

    prodrug model consists mainly of a combination of one

    or more components: (a) a polymeric backbone as a

    vehicle, (b) one or more drugs of the biological active

    components, (c) spacer(s) for hydrolysis of the

    biomolecule and versatility for conjugation, (d) an

    imaging agent and (e) targeting moiety (Fig. 1a and b).

    The drug delivery carrier can be either biocompa-

    tible or an inert biodegradable polymer. The drug is

    coupled directly or via a spacer arm onto the polymer

    backbone. Selection of the spacer arm is critical as it

    opens the possibility of controlling the site and the rate

    of release of the active drug from the conjugates either

    by hydrolysis or by enzymatic degradation. The most

    challenging aspect of this protocol is the possibility of

    altering the body distribution and the cellular uptake by

    cell-specific or non-specific uptake enhancers.

    Due to current interdisciplinary research, molecular

    biologists and organic, polymer chemists can now

    design tailor-made polymeric carriers with different

    structural architecture. Soluble polymers as potential

    drug carriers have been reviewed in detail [16]. The

    polymers selected for preparing macromolecular pro-

    drugs can be categorized according to: (a) chemical

    nature (vinylic or acrylic polymers, polysaccharides,

    poly(a-amino acids), etc., (b) biodegradability, (c)origin (either natural polymers or synthetic polymers)

    and (d) molecular weight (oligomers, macromers and

    polymers). Below are the schematic polymeric archi-

    tectural structures in conjugated form with their

    bioactive components. Most of the polymers possess

    crowding effect for chemical conjugation (Fig. 2).

    Further synthetic strategies will need to be designed to

    reduce such effects that will improve the conjugation

    ratio and payload of component with the polymer.

    2.1. Polymeric drug delivery system (PDDS)

    During the last decade, polymer chemistry was

    dedicated to synthesis, derivatization, degradation,

    characterization, application, and evaluation, fornewer biocompatible and biodegradable polymers, allof which were used as carriers for polymeric drug

    delivery systems. These systems possess unique roles in

    enhanced physiological drug distribution, bioavailabil-

    ity, drug targeting, time-controlled release, sensor-

    responsive release, etc. The emergence of polymer

    therapeutics has instigated research interfacing polymer

    chemistry and the biomedical sciences including

    initiation of nanosized (5100 nm) polymer-based

    pharmaceuticals [6]. Polymer therapeutics include:

    rationally designed macromolecular drugs, polymer

    drug and polymerprotein conjugates, polymeric

    micelles containing covalently bound drug, and

    polyplexes for DNA delivery. The clinical applications

    of polymerprotein conjugates and their resulting

    outcomes seem to be promising, especially for

    polymeranticancer-drug therapeutics [6].

    Many of the pharmacological properties of conven-

    tional free drugs can be improved through the use of

    polymeric drug delivery systems (PDDS), which

    include particulate carriers composed primarily of

    lipids and/or polymers, and their associated thera-

    peutics [17]. Several PDDS have already reached the

    market (Table 2). The majority of the PDDS approved

    are mainly targeted for parenteral administration and

    include either liposomal or therapeutic molecules

    linked to poly(ethylene glycol) (PEG) polymers. The

    pegylation process is being used extensively in prodrug

    conjugates and the synthesis approach involves

    attachment of repeating units of poly(ethylene glycol)

    to a polypeptide drug. More than 30 years, scientists

    have developed various techniques to build PEG

    polymers and attach them to a drug of choice. Recently,

    pegylated drug forms in therapeutics of hepatitis C,

    acromegaly, rheumatoid arthritis, neutropenia, various

    cancers, wound healing, and other disorders either have

    been approved or are undergoing clinical trials [18].

    Proteins and peptides hold great promise as therapeutic

    agents; however, they are prone for degradation by

    proteolytic enzymes. These bioactives can be rapidly

    cleared by the kidneys, generate neutralizing anti-

    bodies, and have a short circulating half-life. Therefore,

    pegylation of such drugs can overcome these and other

    shortfalls. In addition, increased molecular mass of

    proteins and peptides shields them from proteolytic

    enzymes and enhances pharmacokinetics [18].

    Modification of a polymer to form a conjugate with a

    biomolecule depends upon two interrelated chemical

    reactions: (1) reactive functional groups present in the

    polymer and (2) functional groups present on the

    biological component. In general, most of the biomo-

    lecules such as ligands, peptides, proteins, carbo-hydrates, lipids, polymers, nucleic acid and

  • oligonucleotide possess combinations of these

    functional groups. Selection of a suitable method,

    process, and reagents are crucial for successful

    chemical conjugation.

    Polymers may undergo several structural changes

    with solvents, coupling agents, and reactants. Peptide

    and protein PEGylation problems and solutions have

    cross-linking which will lead to an unstabilized form

    and undesired products.

    Modification of the polymer or its bioconjugate can

    provide increased biocompatibility, reduced immune

    response, enhanced in vivo stability, and passive tumor

    targeting for anticancer DDS. In addition, modification

    can significantly increase the water solubility of the

    S lea

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 365been reviewed [19]. A detail of bioconjugation

    strategies involving PEG and other polymers with

    various biomolecules has been discussed briefly in

    Section 3.2. Most of the bioconjugation strategies

    involve coupling reactive nucleophiles with the

    following order of reactivity: thiol, a-amino groups,epsilone amino group, carboxyl and hydroxyl. The

    order of reactivity depends upon the pH in the reaction

    and presence of steric hindrance on the coupling

    moiety. Recent advances in bioconjugation use homo-

    bifunctional amine or heterobifunctional coupling

    reagents. A number of electrophilic groups are capable

    of reacting with amines and other nucleophiles, e.g.

    epoxides, vinylsulphones, and aziridines. In addition,

    sulphonyl chlorides also react with amines; however,

    they are generally water-insoluble and may get

    hydrolyzed. Homobifunctional compounds can

    form proteinprotein linkages such as N,N 00-ethylenei-minoyl-1-6-diaminohexane, bis-aziridin, divinyl sul-

    phone (DVS), nitrogen mustard and bis-sulphonyl

    chloride.

    On the other hand, heterobifunctional reagents are

    useful to couple amines with other functional groups.

    The most important amine reacting heterobifunctional

    compounds are used in protein chemistry; e.g.

    photoactivation, biotinylation and thiolation reactions.

    Reactive groups in proteincarboxyl functions offer

    alternating thiol reactions as a site for heterobifunc-

    tional coupling with amines. The linkages between

    these two functions are formed without incorporation of

    additional atoms by dehydration to an amide. A class of

    such reagents contain compounds with an amine at one

    end to allow a reaction with an activated carboxyl

    function and an amine reactive moiety at the other.

    However, such reactions may undergo cyclization or

    R-NH2 +

    = OR'

    Amine component NHS ester derivative

    Fig. 3. NHS esters compounds react with nucleophiles to release the NHto form COOH group, which can further form amide or ester bond with blysine residues involves use of a heterobifunctional

    reagent comprising of a N-hydroxysuccinimide

    functional group, together with a maleimide or

    = O

    R' NHR

    Amide bond

    +

    H

    NHS leaving group

    ving group and form an acetylated product. PEG can be succiniylatedinsoluble component. The following are common

    strategies adapted to obtain a polymeric drug delivery

    system as biologically active prodrug conjugates.

    2.2. N-hydroxysuccinimide (NHS) ester and coupling

    methods

    Due to their higher reactivity at physiological pH

    makes NHS a choice for amine coupling reactions in

    bioconjugation synthesis [20]. NHS ester compounds

    react with nucleophiles to release the NHS leaving

    group and form an acylated product (Fig. 3). NHS ester

    is the most common activation chemical agent used to

    form reactive acylating agent. Carboxyl groups acti-

    vated with NHS esters are highly reactive with amine

    nucleophiles. Polymers containing hydroxyl groups

    (e.g. PEG) can be modified to obtain anhydride

    compounds. PEG or mPEG can be acetylated with

    anhydrides to form an ester terminating to free

    carboxylate groups (Fig. 4). PEG and its succinimidyl

    succinate and succinimidyl glutarate derivative can be

    further used for conjugation with drugs or proteins.

    The differences in the mode of action of conjugates

    have created awareness of the importance in coupling

    protocols appropriate for different components. The

    successful application of conjugates for therapeutic

    implication requires conjugates of defined composition

    and of low molecular weight using heterobifuntional

    coupling reagents under controlled and optimized

    conditions [21]. Among the commonly used coupling

    procedures, a heterobifunctional reagent is used to

    couple modified lysine residues on one protein to

    sulphydryl groups on the second. The modification ofiomolecules.

  • 45% of scFv-c protein was conjugated as PEG(scFv-c)2using the smallest PEG(Mal)2 (2 kDa). However, no

    significant increase in scFv-c conjugation was observed

    spacer arm can enhance ligandprotein binding and has

    -NH2

    +

    EDC

    OOO

    H3Cn

    NH=O

    mPEG with amidebond=O

    R

    R-OH

    +

    or EDC.HClnic Solvent

    OOO

    H3Cn

    o=

    O

    mPEG with ester bond

    =OR

    sing a carbodiimide as coupling agent to form (a) amide bond and (b) ester

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397366protected sulphydryl group. The linkage formed is one

    of two basic types, a disulphide bridge or a thioether

    bond (Fig. 5); the difference depends on whether the

    introduced group is a sulphydryl or maleimide,

    respectively. The thiol group on the second protein

    may be an endogenous free sulphydryl or chemically

    introduced by modification of lysine residues. NHS is

    widely used as an acylating agent and is preferred for

    conjugation with amine terminal compounds.

    To optimize PEGylation, scFvs have been recombi-

    nantly developed in a vector that adds an unpaired

    cysteine (c) near the scFv carboxy terminus (scFv-c), to

    provide a specific site for thiol conjugation (Fig. 6).

    Random PEG conjugation (PEGylation) of small

    molecules via amine groups demonstrated variations in

    structural conformation and binding affinity [22]. The

    authors evaluated applicability of unpaired cysteine for

    PEGylation of scFv-c. Conjugation efficiency was

    determined for four different scFvs along with several

    PEG molecules having thiol reactive groups. ScFvs

    R

    OOO

    H3Cn

    OH=

    O

    Succinylated mPEG

    =O(a)

    DCCOrga

    OOO

    H3Cn

    OH=

    O

    Succinylated mPEG

    =O(b)

    Fig. 4. Succinylated mPEG coupled to amine terminated component u

    bond conjugate.produced as scFv-c and purified by anti-E-TAG affinity

    chromatography were conjugated using PEG molecules

    with maleimide (Mal) or o-pyridyl disulfide (OPSS).

    Conjugations were carried out at pH 7.0, with 2 M

    excess tris(2-carboxyethyl)phosphine hydrochloride

    (TCEP)/scFv and PEGMal or PEGOPSS, using 5:1

    (PEG:scFv). PEGMal conjugation efficiency was

    further evaluated with 1:5 (PEG:scFv). PEGylation

    efficiency was determined for each reaction by

    quantitation of the products on SDS-PAGE. ScFv-c

    conjugation with unifunctional maleimide PEGs

    resulted in PEG conjugates incorporating 3080% of

    the scFv-c with a consistent average above 50%. The

    efficiency of scFv-c conjugation to both functional

    groups of the bifunctional PEG(Mal)2 varied between

    the PEG and scFv-c molecules studied. A maximum ofapplication in prodrug conjugates and in biotechnology

    [24]. Ideal linkers possess the following characteristics:

    (1) stability in the physiological pH if the drug is to be

    O

    NH-C-X-by use of a greater than 5 M excess of PEG/scFv-c.

    Specific examples related to the coupling reactions have

    been described in Section 3.

    2.3. Incorporation of spacers in prodrug conjugates

    Various spacers have been incorporated along with

    the polymers and copolymers to decrease the crowding

    effect and steric hindrance [23]. The incorporation of aNO

    S

    O

    (a) Thioether linkage (x = Spacer)

    Toxin- NH-C-CH2-CH2-S-S-

    AntibodyEnzyme

    (b) Disulphide linkage

    O

    Fig. 5. Types of proteinprotein linkages used for synthesis of

    antibodyenzyme or antibodytoxin conjugates. The positions of the

    proteins with respect to the linkage may be varied (modified from

    Ref. [21]).

  • delivered to the tumor vasculature and (2) they release

    the bioactive agent at an appropriate site of action.

    For example, amino acid spacers such as glycine,

    alanine, and small peptides are preferred due to their

    chemical versatility for covalent conjugation and

    biodegradability. Heterobifunctional coupling agents

    containing succinimidyl have also been used exten-

    sively as spacers (Fig. 7). Therapeutic potential of a

    carboxypeptidase monoclonal antibody conjugate were

    reported using N-succinimidyl anhydrides [2529].

    The higher conjugation ratio of an antibody with a

    drug can result in a decrease in the ability of the

    antibody to bind to its specific receptor. This could be

    overcome by introducing a polymer spacer between the

    targeting antibody and the drug. The use of an

    intermediate polymer with drug molecules carried in

    Fig. 6. PEGmaleimide (PEGMal) structures and the maleimide

    (Mal) thiol conjugation reaction with scFv-c (scFvSH): (a) methoxy-

    PEGMal; (b) PEG(Mal)2; (c) branched methoxy-PEGMal; (d)

    thioether bond formation between maleimide and cysteine of scFv-c

    (reproduced from Ref. [22]).

    Fig. 7. Structures of commonly used heterobifunctional coupling

    agents with spacer lengths N-succinimidyl-4-(N-maleimidomethyl)-

    cyclohexane-l-carboxylate (SMCC), N-succinimidyl-4-(p-maleimi-

    dophenyl)-butyric acid (SMPB), N-maleimidobenzoyl-N-hydroxy-

    succinimide (MBS) (reproduced from Ref. [29]).

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 367Fig. 8. Structure of (a) acid-sensitive bifunctional reagents used for

    coupling of anthracycline drugs with polymers or antibodies and (b)

    antibodydrug and antibodypolymerdrug conjugates (reproducedfrom Ref. [28]).

  • its side chains increases the potential number of drug

    molecules able to attach to that antibody by modifi-

    cation of only a minimum amount of existing amino

    acid residues (Fig. 8a and b) [29].

    In most of the bioconjugates, the NHS ester

    anhydride is reacted with primary NH2 of the peptide

    at slightly higher pH (7.5) to form an amide bond which

    links the maleimide group to the protein and releases

    NHS (Figs. 9 and 10). N-hydroxysuccinimide released

    2.4. Carbodiimide coupling reactions or zero lengths

    cross-linkers

    Coupling and condensation reactions are unique to

    obtain chemical conjugates involving drugs or other

    biocomponents with polymers. The smallest possible

    reagents for bioconjugate synthesis are called zero

    length cross-linkers [30]. Modification of these con-

    jugates depends upon the following two interrelated

    Fig. 9. Scheme for protein coupling using N-hydroxysuccinimide ester/maleimide heterobifunctional agents. X represents the spacer groups of

    varying chain lengths (reproduced from Ref. [29]).

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397368from the protein can be easily removed either by

    dialysis or by gel filtration using Sephadex columns

    such as G10 or G25. Thereafter, the maleimide group

    can be further reacted with the thiol containing moieties

    or proteins to form a thioether bond in the presence of a

    slightly acidic or neutral pH [29]. The applications of

    spacers to form a polymer conjugate are discussed in

    the polymer conjugate text.Fig. 10. Schemes for two N-hydroxysuccinimide-based reagents commonl

    Ref. [29]).chemical reactions; the reactive functional groups on

    the different cross-linking or deriavatizing reagents and

    the functional groups present on the target biomolecule

    to be modified. Coupling agents mediate the conju-

    gation of the two molecules by forming a bond with no

    additional spacer atom. Therefore, one atom of the

    molecule is covalently linked to an atom of the second

    molecule with no additional linker or spacer needed.y used for insertion of thiol residues into proteins (reproduced from

  • Carbodiimides (Figs. 11 and 12a and b) are most

    commonly used as coupling reagents to obtain amide

    linkage between a carboxylate and an amine or

    phosphoramidate linkage between a phosphate and an

    amine. They are unique due to their efficiency and

    versatility to form a conjugate between two polymers,

    between protein molecules, between a peptide and a

    drug molecule, or between a peptide and a protein plus

    any combination of these small molecules.

    The carbodiimides are either water-soluble or water-

    insoluble. The water-soluble carbodiimides are used for

    biomacromolecular conjugation with variations of

    buffer solutions. Water-insoluble carbodiimides are

    used in presence of organic solvents for conjugation of

    polymers with drugs and imaging agents, polymers and

    peptides, and even polymerpolymer conjugates.

    Usually, a byproduct is formed; which is mostly

    insoluble in the solvent medium and facilitates easier

    purification (Fig. 12).

    Biomacromolecules containing phosphate groups

    such as the 50 phosphate of oligonucleotide can beconjugated to amine containing molecules by using a

    carbodiimide mediated reaction (e.g. EDC). Carbodii-

    mide activates the phosphate to an intermediate phos-

    phate ester, identical to its reaction with carboxylates.

    Further, in the presence of an amine on a polymer

    containing NH2 terminal groups, carbodiimide can be

    conjugated to form a stable phosphoramidate bond

    (Fig. 13). The application of carbodiimide as a coupling

    agent to form a polymeric bioconjugate, with examples,

    has been discussed in the subsequent polymeric text.

    3. Design and synthesis of polymeric conjugates

    3.1. Dextran

    Several natural and synthetic polymers have

    attracted the attention as prodrugs in biomedical

    CH3

    NC

    N

    NH

    Cl

    CH3

    +

    CH3

    Fig. 11. Structure of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide

    hydrochloride (EDC) coupling agent.

    ON

    CCl-

    CH3O

    R

    o-acylisourea active intermediate

    HCl

    NH

    +

    (a)

    H

    -acyli

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 369CH3

    NC

    N

    CH3 N

    Amide bond formation

    EDC.

    R-COOH+

    R'-NH2

    R-COO-NHR'

    NH-C-N

    O

    R-COOH+

    o(b)CN

    DCCR'-NH2

    R-COO-NHR'Amide bond formation

    Fig. 12. Formation of amide or ester bond. EDC$HCl (a) and DCC (b) activaform amide or ester bond, respectively.Cl-CH3

    CH3

    CH3

    Esterbondformation

    NH

    + R-COOH+

    R'-OH

    R-COO-R'

    NR-COOH

    +

    R'-OH

    R-COO-R'

    sourea intermediate

    Esterbondformation

    tes carboxylic acid and further reacts with amine or hydroxyl group to

  • lysosomal fraction, but not in the control samples

    lacking lysosomes. The hydrolysis rate constants for

    DMP conjugate over to MP and MPS in the lysosomal

    fraction were not significantly different from those in

    the control samples. The authors also delineated the

    applications of dextrans for targeted and sustained

    delivery of therapeutic and imaging agents [35]. The

    dextran was chemically conjugated with the amine

    containing drugs or proteins using the periodate method

    (Fig. 16).

    The method to obtain b-(4-hydroxy-3,5-di-tert-butylphenyl) propionic acid (PA)dextran conjugates

    + R'-NH2O

    O

    O-

    POH EDC

    O

    O

    O-

    PNH

    R'

    Phosphoramidate bond5' phosphate oligonucleotideR'~

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397370applications. In particular, natural biopolymers, e.g.

    dextran and chitosan have been used extensively for

    prodrug conjugation research. Dextran is a natural

    polysaccharide containing monomers of simple sugar

    glucose (Fig. 14). This polyglucose biopolymer is

    characterized by a-1,6 linkages, with hydroxylatedcyclohexyl units and generally produced by enzymes

    from certain strains of Leuconostoc or Streptococcus.

    However, dextran has more compact structure than any

    other polymers. The chemical properties of dextran can

    be modified chemically for the specific applications.

    Dextran possesses multiple primary and secondary

    hydroxyl groups and therefore can be easily conjugated

    with drugs and proteins with reactive groups either by

    direct conjugation or by incorporation of a spacer arm.

    After oral administration, the polymer is not signifi-

    cantly absorbed. Therefore, most of the effective

    applications of dextran as polymeric carriers are

    through injections. A few studies have reported the

    potential of dextran in colon-specific delivery of drugs

    via the oral route [31]. On the other hand by systemic

    administration, the pharmacokinetics of the dextran

    conjugates along with therapeutic agents are signifi-

    cantly affected by the kinetics of the dextran. Animal

    and human studies have shown that both the distri-

    bution and elimination of dextrans are dependent on

    molecular weight and the net charge.

    Dextran has been extensively evaluated as a

    polymeric vehicle for delivery of anticancer drugs to

    the tumor tissue through a passive accumulation of the

    dextrananticancer conjugate [32]. Conjugates of

    dextrans with corticosteroids have been evaluated

    Fig. 13. Phosphate oligonucleotide-polymer conjugation using EDC

    as couplng agent.previously for the local delivery of steroids in colon

    as anti-inflammatory agents [33]. A macromolecular

    O

    O

    CH2

    OH

    OHOHO

    O

    OH

    OHHOO

    Fig. 14. Structure of dextran.prodrug of methylprednisolone (MP) was synthesized

    by conjugating MP with dextran using succinic acid (S)

    as a spacer [34]. MP-succinate (MPS) conjugate was

    prepared using 1,1 0-carbonyldidmidazole as a couplingagent with succinic acid as a spacer (Fig. 15). The

    hydrolysis of dextran MP (DMP) conjugate in rat blood

    was achieved with a half-life ofw25 h. The hydrolysisof MPS to MP was proved to occur in the liver

    OCH2

    O

    OHO

    OH

    OGlu

    CO

    H2C

    H2C C

    OO

    C=OOH

    O

    HO

    CH3Methylprednisolone

    Succinate linker

    Glu

    Dextran

    H2C

    Fig. 15. Dextranmethylprednisolone conjugate. MP-succinate

    (MPS) conjugate was prepared using 1,1 0-carbonyldidmidazole as acoupling agent with succinic acid as a spacer (reproduced from

    Ref. [34]).(PADC) was reported [36]. The reaction was carried

    out using dicyclohexyl carbodiimide (DCC) as a

    coupling agent (Fig. 17). In most of the coupling

    reactions, either pyridine or p-dimethylaminopyridine

    (DMAP) was used as a catalyst. Dicyclohexylurea was

    formed as a byproduct in the form of precipitate during

    the reaction, easily removed by filtration.

    A macromolecular prodrug of Tacrolimus (FK506),

    FK506dextran conjugate, was developed and its

    physico-chemical, biological and pharmacokinetic

  • drug

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 371characteristics were studied [37]. The conjugate was

    estimated to have 0.45% of Tacrolimus (FK506) agent

    and the coupling molar ratio was approximately 1:1

    (dextran to FK506) (Fig. 18). Low molecular weight

    radioactive compound(s) which eluted in the same

    fractions as [3H]FK506 were released from

    [3H]FK506dextran conjugate by chemical hydrolysis,

    with a half-life of 150 h in phosphate buffer. The

    dextranFK506 conjugate was synthesized using a

    solution of carboxy-n-pentyl-dextran (C6D-ED) in

    phosphate buffer with activated ester of FK506 in di-

    oxane.

    Fig. 16. Dextran was chemically conjugated with the amine containingPotent anticancer agents, such as camptothecin have

    been conjugated to dextran to form prodrugs (Fig. 19).

    Carrier and dose effects on the pharmacokinetics of

    T-0128, a camptothecin analogue-carboxymethyl dex-

    tran conjugate, were reported in control and tumor

    bearing rats [38].

    Conjugation of drugs with dextrans has exhibited

    prolonged effect, reduced toxicity, and immunogeni-

    city. Most of the studies have been carried out in

    animals, however, with only a few experiments being

    performed on humans [35]. The multiple OH groups

    Fig. 17. Synthetic scheme for sterically hindered phenoon the polymeric dextran backbone provides possible

    functional sites for drug conjugation. Dextran of Mw70,000 Da was conjugated to doxorubicin via an acid-

    labile linkage for intratumoral delivery [39]. Dextran is

    considered as a polymeric carrier because of its

    biocompatibility and biodegradability [40].

    An example of tumor-targeted dextran-based drug

    delivery system has been described by Chau et al. [41].

    A dextranpeptidemethotrexate conjugate for tumor-

    targeted delivery of chemotherapeutics contains Pro-

    Val-Gly-Leu-Ile-Gly peptide linker cleavable by matrix

    metalloproteinase II (MMP-2) and IX (MMP-9). Both

    s or proteins using the periodate method (reproduced from Ref. [35]).enzymes are over expressed in tumors. The linker

    chemistry and the backbone charge were optimized to

    allow high sensitivity of the conjugates toward the

    targeted enzymes. In the presence of the targeted

    enzymes, the peptide linker was cleaved and peptidyl

    methotrexate (anticancer drug) was released. Satisfac-

    tory stability of the new conjugates was demonstrated

    in serum containing conditions, suggesting that the

    conjugates can remain intact in systemic circulation.

    Apart from natural polymers such as dextran, other

    synthetic polymers such as PEG conjugates have

    ldextran conjugate (reproduced from Ref. [36]).

  • intracellular targets. Covalent conjugation of synthetic

    Fig. 18. Scheme for FK506dextran conjugate. The dextranFK506 conjugate was synthesized using a solution of carboxy-n-pentyl-dextran (C6D-

    ED) in phosphate buffer with activated ester of FK506 in di-oxane (reproduced from Ref. [37]).

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397372polymersparticularly poly(ethylene glycol) (PEG)

    to bioactive components increases plasma residencegained interest for numerous therapeutic applications

    and will be discussed in Section 3.2.

    3.2. Poly(ethylene glycol) (PEG) and conjugates

    PEG conjugates are classical prodrugs with an

    enhanced permeability and retention (EPR) effect, and

    which can accumulate significantly into tumor mass

    and cross the cell membranes by endocytosis to reachFig. 19. Synthesis of FITC-labeled CPT (T-2513)dtime and the therapeutic index yet protein immuno-

    genicity is reduced. Several PEGylated enzymes

    (adenosine deaminase, L-asparaginase) and cytokines

    (including interferon a and G-CSF) have now enteredroutine clinical use. PEG-modified adenosine de-

    aminase (ADAGENw) and PEG-L-asparaginase

    (ONCASPARw) were the first PEG modified enzymes

    being used in early 1990s.

    Most commonly used, monomethoxy poly(ethylene

    glycol) (mPEGOH), can be functionalized and

    conjugated with drug and other biological components.

    Having only one or two terminal functional groups atextran conjugate (reproduced from Ref. [38]).

  • the end of polymer chain, PEG has a limitation with a

    poor loading capacity. PEG has a linear or branched

    polyether terminated with hydroxyl groups and is

    synthesized by an anionic ring opening polymerization

    of ethylene oxide initiated by a nucleophilic attack of a

    hydroxide ion on the epoxide ring. Most used PEGs for

    prodrug modification are either monomethoxy PEG

    (Fig. 20a) or di-hydroxyl PEG (Fig. 20b). High aqueous

    solubility makes PEG polymer a versatile candidate for

    the prodrug conjugation. PEG is also considered to be

    somewhat hydrophobic due to its solubility in many

    organic solvents.

    O OHO

    n

    H3C(a)

    HOO

    OHn

    PEG

    (b)

    Fig. 20. Structures for (a) monomethoxy-poly(ethylene glycol) and

    (b) di hydroxyl terminated poly(ethylene glycol).

    X C

    O

    H2C

    nC

    O

    O SumPEG

    3 x =O,n=24 x =O, n =25 x = NH, n = 2

    mPEG O C

    O

    O Su

    6

    mPEG-OH

    mPEG O CO

    AA

    mPEG O C

    O

    O SumPEG O C

    O

    O Su

    mPEG O 2HCOF3

    O N

    NN

    Cl

    R

    1 R = Cl2 R = mPEG-O

    O Su

    7 R = Imidazole8 R = O-TCP9 R = O-pNP10 R = O-Su

    11 AA=Gly,Ala etc13

    SO O

    12

    mPEG

    (a)

    mPEG O N

    NN

    NH

    R

    R = Clor mPEG-O

    Protein

    1 or 2

    3 -11H2N

    12 13 and NaCN BH3

    HN Protein

    Protein

    mPEG-

    (b)

    Fig. 21. (a) mPEG-based protein-modifying methods. Protein modification w

    amides, derived from active esters 36 and 11, or carbamates, derived from 7

    secondary amine conjugation with amino-containing residues. As represented

    in reductive alkylation reactions. The numbering (113) represent to the orde

    Ref. [42]).

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 373X C

    O

    mPEG HN Protein

    x = O2C (CH2)n , OCH2,O, or an amino acid residue

    ith all of these agents results in acylated amine-containing linkages:

    to 10. Alkylating reagents (12 and 13) both react with proteins forming

    in (b) tresylate (12) alkylates directly, while acetaldehyde (13) is used

    r in which these activated polymers were introduced (reproduced from

  • T) co

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397374Coupling reactions between NH2 groups of

    proteins and mPEG with an electrophilic functional

    group have been used in most cases for preparation of

    PEGprotein conjugates [42]. Such reactions usually

    result in formation of conjugates composed of a

    globular protein in its core to which several polymer

    chains are covalently linked. The composition of such a

    graft copolymeric system is dependent on the number

    of available attachment sites (NH2 and other nucleo-

    philic groups) on the reactivity of the mPEG, and

    reaction conditions. Fig. 21a and b illustrates the most

    commonly used methods of mPEG-based protein

    modifying reagents. Figure represents multiple com-

    ponents of mPEGOH polymer conjugates. Derivatives

    1 and 2 contain a reactive aryl chloride residue, which is

    displaced by a nucleophilic amino group by a reaction

    with peptides or proteins, as shown in Fig. 21b.

    Derivatives (1 and 2) are acylating reagents, whereas

    derivatives (311) contain reactive acyl groups refer-

    enced as acylating agents. Protein modification with all

    of these agents results in acylated amine-containing

    linkages: amidesderived from active esters (36 and

    Fig. 22. PEG-carbamate-paclitaxel (PC11)or carbamatesderived from (710). Alkylating

    reagents (12 and 13) both react with proteins forming

    secondary amine conjugation with amino-containing

    residues. As represented in Fig. 21b, tresylate (12)

    alkylates directly, while acetaldehyde (13) is used in

    reductive alkylation reactions. Numbers (113) rep-

    resent the order in which these activated polymers were

    introduced [42].

    It has been established that PEGylation greatly

    enhances water solubility and decreases immuno-

    genicity [43]. However, bioactive compounds con-

    jugated with PEG polymers must be chemically

    stable in its conjugated form until released.

    Successful applications of covalently bonded PEG

    with proteins and anticancer agents have beenreported [44]. The authors designed and evaluated

    water-soluble mPEG5000 paclitaxel-7-carbamates

    conjugates (Fig. 22).

    PEG has severe limited conjugation capacity since

    only two terminal functional groups exist at the end of

    the polymer chain (or just one in the case of the most

    used monomethoxy poly(ethylene glycol) (mPEG

    OH)), which can be functionalized and conjugated to

    the biocomponents. Recently, this limitation of PEG

    was overcome by coupling amino acids, such as

    bicarboxylic amino acid and aspartic acid, to the PEG

    [45,46]. Such derivatization doubled the number of

    active groups of the original molecule of PEG. Using

    the same method with recursive derivatization, den-

    drimeric structures were achieved at each PEGs

    extremity. However, the authors encountered some

    problems in this study, namely the lower reactivity of

    the bicarboxylic acids groups towards Ara-C binding. It

    was inferred that low reactivity is a result of steric

    hindrance between two molecules of Ara-C when they

    are conjugated to neighboring carboxylic moieties. It

    was suggested that this effect might be overcome by

    njugate (reproduced from Ref. [44]).incorporating the dendrimer arms with an amino

    alcohol (H2N[CH2CH2O]2H).

    PEG polymers with hydroxyl terminals can be

    modified easily using small amino acids or other

    aliphatic chains molecules. For example, linear or

    branched PEG of varying molecular weight PEGAra-

    C conjugates for controlled release were reported [47].

    The antitumor agent 1-b-D-arabinofuranosilcytosyne(Ara-C) was covalently linked to varying molecular

    weight OH terminal PEGs through an amino acid

    spacer in order to improve the in vivo stability and

    blood residence time. Conjugation was carried out with

    one or two available hydroxyl groups at the polymers

    terminals. Furthermore, to increase the drug loading of

    the polymer, the hydroxyl of PEG was functionalized

  • HO-PEG-OH + Cl C O

    O

    NO2

    CH2Cl2MW 10000 Da

    PEG-O CO

    O

    NO2

    13

    H2O/CH2CN 1) Amino adipic acid

    2

    PEG-O CHN

    O

    CH

    14

    COOH

    CH2-CH2-CH2-COOH 2

    CH2Cl2

    EDC / NHSPEG- O C

    HN

    O

    CH

    15

    CONHS

    CH2-CH2-CH2-CONHS

    2Ara CPyridine

    PEG-AD2-Ara-C7

    2) Et3N

    Et3N

    AD= Amino adipic acid

    (a)

    H2O/CH2CNPEG-O C

    HN

    O

    CH

    16

    CONH

    CH2-CH2-CH2-

    2

    PEG -O CHN

    O

    CH

    15

    CONHS

    CH2-CH2-CH2-CONHS

    2

    Et3NAmino adipic acid CH

    COOH

    CH2-CH2-CH2-COOH

    CONH

    CHCOOH

    CH2-CH2-CH2-COOHEDC / NHS

    CH2Cl2

    PEG-O CHN

    O

    CH

    17

    CONH

    CH2-CH2-CH2-

    2

    CHCONHS

    CH2-CH2-CH2-CONHS

    CONH

    CHCONHS

    CH2-CH2-CH2-CONHSAra-C

    Pyridine

    PEG-A2-AD4-Ara-C8

    AD= Amino adapic acid

    8

    (b)

    Fig. 23. Synthetic schemes for PEG10000AD2Ara-C4 (7) (a) and PEG10000AD2AD4Ara-C8 (8) conjugates (b). The antitumour agent 1-b-D-

    arabinofuranosilcytosyne (Ara-C) was covalently linked to varying molecular weight OH terminal PEGs through an amino acid spacer in order to

    improve the in vivo stability and blood residence time (reproduced from Ref. [47]).

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 375

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397376PEG

    (a)

    -O CHN

    O

    CH

    CONH CHCO-Ara-C

    CH2-CH2-CH2-COOHwith a bicarboxylic amino acid to form a tetrafunctional

    derivative. Finally, the conjugates with four or eight

    Ara-C molecules for each PEG chain were prepared

    (Fig. 23). The authors investigated steric hindrance in

    (b)

    CH2-CH2-CH-

    HC COOH

    CH2-CH2-CH2-CO Ara- C

    CONH

    PEG-O CHN

    O

    CH

    CONH

    -H2C

    CHCO-Ara-C

    CH2-COOH

    HC COOH

    CH2-CO - Ara -C

    CONH

    Fig. 24. (a) Most stable molecular structure of PEG(aminoadipic

    acid)(Ara-C)2 conjugate. The two carboxylic acid functions are

    emphasized by a space filling representation, and the distance (A)

    between them is reported. (b) Most stable conformation of PEG

    (aspartic acid)(Ara-C)2 conjugate. The two carboxylic acid functions

    are emphasized by space filling, and the distance (A) between them is

    reported (reproduced from Ref. [47]).PEGAra-C conjugates using molecular modeling to

    investigate the most suitable bicarboxylic amino acid

    with the least steric hindrance (Fig. 24a and b).

    Computer aided design suggested that aminoadipic

    acid was most suitable, because the carboxylic groups

    are sufficiently separated to accommodate Ara-C

    without the necessity to incorporate spacer arms. The

    theoretical findings were confirmed and supported by

    the experimental conjugation results. PEG conjugates

    with Ara-C were prepared through an amino acid spacer

    (e.g. non-leucine or lysine). Hydroxyl groups of PEG

    were activated by p-nitrophenyl chloroformate to form

    a stable carbamate linkage between PEG and the amino

    acid. The degree of PEG hydroxyl group activation

    with p-nitrophenyl chloroformate was determined by

    UV analysis of the p-nitrophenol released from PEGp-

    nitrophenyl carbonate after alkaline hydrolysis. Acti-

    vated PEG was further coupled with amino acid and the

    intermediate PEGamino acid was linked to Ara-C by

    EDC/NHS activation.

    Design and synthesis of non-targeted or antibody-

    targeted biodegradable PEG multi-block coupled with

    N2,N5-diglutamyllysine tripeptide with doxorubicin

    (Dox) attached through acid-sensitive hydrazone bond

    was reported [4851]. PEG activated with phosgene

    and N-hydroxysuccinimide was reacted with NH2groups of triethyl ester of tripeptide N2,N6-diglutamyl-

    lysine to obtain a degradable multi-block polymer. The

    polymer was then converted to the corresponding

    polyhydrazide by hydrazinolysis of the ethyl ester

    with hydrazine hydrate. The non-targeted conjugate

    was prepared by direct coupling of Dox with the

    hydrazide PEG multi-block polymer (Fig. 25). Whereas

    the antibody-targeted conjugates, a part of the polymer-

    bound hydrazide group was modified with succinimidyl

    3-(2-pyridyldisulfanyl) propanoate to introduce a

    pyridyldisulfanyl group for subsequent conjugation

    with a modified antibody. Dox was coupled to the

    remaining hydrazide groups using acid-labile hydra-

    zone bonds to obtain a polymer precursor.

    In addition, human immunoglobulin IgG modified

    with 2-iminothiolane was conjugated to the polymer by

    substitution of the 2-pyridylsulfanyl groups of the

    polymer with SH groups of the antibody. It was

    demonstrated that Dox was rapidly released from the

    conjugates when incubated in phosphate buffer at

    lysosomal pH 5 and 7.4 (blood).

    PEG has been used to modify a number of

    therapeutically interesting proteins [52]. Conjugates

    of adriamycin with PEGpoly(aspartamide) block

    copolymers forms micelles [53]. Adriamycin (ADR),an anthracycline anticancer drug, was bound to the

  • ne) w

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 377poly(aspartic acid) chain of poly(ethylene glycol)

    poly(aspartic acid) block copolymer by amide bond

    formation between an amino group of adriamycin and

    the carboxyl groups of the poly(aspartic acid) chain

    (Fig. 26). The hydrophilic PEG chains form the outer

    shell and the hydrophobic poly(aspartic acid)doxor-

    ubicin components form the inner core. It was

    Fig. 25. Scheme for multi-block PEGDox (hydrazodemonstrated that these systems have very high

    in vivo antitumor activity and show a reduced non-

    specific accumulation in the heart, lungs and liver.

    Water-soluble PEG polymerdrug conjugates are

    considered to be most promising with unique delivery

    systems [5459]; especially PEG polymer which is the

    most utilized carrier to deliver drugs in cancer therapy.

    In tumor tissue, spacers are necessary to be incorpor-

    tated between the drug and its carrier in order to enable

    the release drug from that carrier either in slightly

    acidic extracellular fluids or, after endocytosis, in

    endosomes or lysosomes of cancer cells [60]. Acid-

    sensitive hydrazone bond was formed between the C13

    carbonyl group of anthracyclines (Dox, daunomycin

    (Dau) and polymer hydrazides or amide bond of a cis-

    aconityl residue containing spacer. Structure of Dox

    bound to the polymer via pH-sensitive trityl spacer is

    shown in Fig. 27.

    It is worthwhile to note that even the larger proteins

    or peptides have been successfully conjugated with

    linear polymers. Various amounts of a releasable PEG

    linker (rPEG) were conjugated to the protein lysozyme

    (Fig. 28). rPEGlysozyme conjugate is relatively stablein pH 7.4 buffer for over 24 h. However, regeneration

    of native protein from the rPEG conjugates occurred, as

    expected, in a higher pH buffer of rat plasma [61].

    Lysozyme is released more rapidly from the mono-

    substituted conjugate than from the di-substituted

    conjugate, suggesting possibility of steric hindrance

    for the enzyme cleavage. PEG, being a linear polymer,

    ith antibody conjugate (reproduced from Ref. [50]).is first activated at the OH terminal of either diol or

    mono methoxy PEG. Modification of a branched PEG

    Fig. 26. Adriamycin-conjugated poly(ethylene glycol)poly(aspartic

    acid) block copolymer. Adriamycin (ADR), an anthracycline antic-

    ancer drug, was bound to the poly(aspartic acid) chain of

    poly(ethylene glycol)poly(aspartic acid) block copolymer by

    amide bond formation between an amino group of adriamycin and

    the carboxyl groups of the poly(aspartic acid) chain (reproduced from

    Ref. [53]).

  • aqueous solubility of drugs [6466]. Recently, HIV

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397378prodrug conjugates were designed, synthesized, and

    evaluated to overcome several biopharmaceutical

    challenges associated with HIV-1 protease inhibitorchain can provide an umbrella-like covering (U-PEG,

    PEG 2) and has demonstrated applications in protein or

    peptide conjugation [62,63].

    3.3. PEGHIV agent conjugates

    PEGylation significantly enhances biopharmaceuti-

    cal characteristics such as circulation half-life and

    Fig. 27. Structure of Doxpolymer conjugate with a pH-sensitive

    trityl spacer (reproduced from Ref. [60]).(PIs) and prodrugs [67]. Various PEG-based prodrug

    conjugates of the HIV-1 (PI) saquinavir (SQV) were

    synthesized using different chemical groups having the

    capability to modify pharmacokinetic properties. The

    prodrug conjugates included SQVcysteinePEG3400,

    SQVcysteinePEG3400biotin, SQVcysteine

    Fig. 28. Scheme for PEGylation of lysozyme with PEG-BE linker

    (reproduced from Ref. [61]).(R.I.CK-Tat9) [a cationic retro-inverso-cysteine-

    lysine-Tat nonapeptide]PEG3400, and SQVcysteine

    (R.I.CK(stearate)-Tat9)PEG3400. In all the conju-

    gates, SQV was linked with cysteine to form a

    degradable SQVcysteine ester bond. In addition, the

    amino group of the cysteine moiety provided an

    attachment site for degradation of the amide bond

    with N-hydroxysuccinimide-activated forms of PEG

    and PEGbiotin (Fig. 29).

    3.4. PEGdrug and targeted delivery

    Targeting of the cancer drug and other anti-

    neoplastic agents is critical to the specific sites as it

    can maximize cell-death effect during the tumor

    growth phase during which majority of the cells remain

    sensitive to pharmacotherapy. Also, healthy cells are

    protected from exposure to the cytotoxic agent [68].

    Recently, a two-tier approach was demonstrated using

    the drug, camptothecin (CPT), and two different

    targeting agentsluteinizing hormone-releasing hor-

    mone (LHRH) and BCL2 homology 3 (BH3) peptide.

    LHRH peptide was targeted to extra-cellular LHRH

    receptors overexpressed in several cancer cells in order

    to: increase the cancer specificity of the drug, reduce

    adverse drug side effects, and enhance drug uptake by

    cancer cells. BH3 peptide was targeted to intracellular

    controlling mechanisms of apoptosis used to suppress

    the cellular anti-apoptotic defense to enhance drug

    anticancer activity [69]. CPT was first coupled to an

    amino acid via a biodegradable ester bond to the

    hydroxyl group at position 20, using Boc-Cys (Trt)

    amino acid. Diisopropylcarbodiimide, was used as a

    coupling agent and protecting groups were removed

    using trifluoroacetic acid in methylene chloride. The

    prodrug conjugate, CPTcysteine ester, had two

    potential, orthogonal conjugation sitesthe amino

    group and the thiol group (Fig. 30a).

    CPT-Gly ester was first reacted with bifunctional

    reagent, NHSPEGVinyl sulphone (VS), where the

    amino group formed an amide bond with the active ester

    (N-hydroxysuccinimide ester of PEG (Fig. 30b). An

    analog of BH3 (Ac-Met-Gly-Gln-Val-Gly-Arg-Gln-

    Leu-Ala-Ile-Ile-Gly-Asp-Asp-Ile-Asn-Arg-Arg-Tyr-

    CysNH2), containing an extra residue of cysteine at the

    C-terminus, was synthesized by the solid phase peptide

    method. The previous reaction mixture was coupled to

    the thiol group of BH3, which formed a thioether bond

    with the VS group on the PEG. The LHRH analog-

    LHRHLys6des-Gly10Pro9-ethylamide (Gln-His-

    Trp-Ser-Tyr-DLys-Leu-Arg-Pro-NH-Et), which had areactive amino group only on the side chain of the lysine

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 379at position 6, was first reacted with one equivalent of

    NHSPEGVS in DMF. CPT-Cys was then added to

    obtain the thioether bond formation between the VS

    group and the thiol group. The efficacy of the targeted

    CPTPEGBH3 and CPTPEGLHRH conjugates was

    higher than the non-targeted PEGCPT conjugate.

    The feasibility of a two tier targeting of CPT

    PEG conjugates to LHRH receptors and cellular anti-

    apoptotic defense using LHRH and BH3 peptides,

    respectively, was investigated [69]. In this study,

    human ovarian carcinoma cells were incubated with

    free CPT, CPTPEG, CPTPEGBH3 or CPTPEG

    LHRH conjugates and the mixture of CPTPEG

    BH3 or CPTPEGLHRH conjugates. Two

    Fig. 29. Scheme to prepare saquinavir (SQV) conjugates. SQV was linked wi

    the amino group of the cysteine moiety provided an attachment site for stea

    forms of PEG and PEGbiotin (reproduced from Ref. [67]).approaches were used to assess the induction of

    apoptosis: (1) measurement of the enrichment of cell

    cytoplasm by histone-associated DNA fragments

    (mono- and oligonucleosomes) and (2) the detection

    of single- and double-stranded DNA breaks (nicks)

    using terminal deoxynucleotidyl transferase mediated

    dUTP-fluorescein nick end labeling (TUNEL)

    method. The results obtained in these experiments

    demonstrated that conjugation of CPT to PEG

    increased its proapoptotic activity (Fig. 31). Further

    enhancement was achieved by using BH3 peptide in

    a CPTPEGBH3 and LHRH peptide in a CPT

    PEGLHRH conjugates and their mixture. These

    results were consistent with the cytotoxicity and

    th cysteine to form a degradable SQVcysteine ester bond. In addition,

    dy sate degrading amide bond with N-hydroxysuccinimide-activated

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397380gene expression analysis results and showed that

    simultaneous targeting and suppression of cellular

    anti-apoptotic defense substantially increased antic-

    ancer activity of camptothecin.

    Recent development has lead to the use of higher

    molecular weight PEG (O20,000 Da); especially the useof PEG 40,000 Da which is estimated to have a plasma

    circulating half-life of approximately 89 h in a mouse.

    Conjugation of small organic molecules with high

    molecular weight PEG conjugates to form a prodrug

    has proven to be a successful polymeric candidate [70].

    4. Poly-amino acid conjugates

    Due to demand of new biomaterials for a variety of

    biomedical application, new potential candidates are

    being designed and synthesized. Various amino acid

    based polymers and their complex forming nature are

    Fig. 30. Synthetic schemes for (a) CPTPEGBH3 conjugate. CPT was first c

    group at position 20, using Boc-Cys (Trt) amino acid and (b) LHRHPEGbeing explored to deliver drugs to genetic materials. Since

    poly(amino acid)s are structurally related to natural

    proteins, the synthesis of amino acid-based polymers is

    explored as a potential source of new biomaterials [71].

    In the past, various reviews have been published

    predicting poly(amino acids) as promising polymeric

    candidates [7274]. However, only few polymers such

    as poly(g-substituted glutamates) and copolymers havebeen screened as promising materials for biomedical

    applications. Cationic polymers such as poly(L-lysine)

    and their complexes with DNA have increased attention

    as synthetic vectors for the delivery of genes. Synthetic

    poly(a-amino acids) like poly(L-lysine), poly(L-gluta-mic acid), and poly((N-hydroxyalkyl) glutamines) can

    be synthesized by ring-opening polymerization of the

    N-carboxyanhydride monomers. These polymers have

    functionalities in their side groups (amine, hydroxyl,

    and carboxyl) that allow covalent coupling with drug

    oupled to an amino acid via a biodegradable ester bond to the hydroxyl

    CPT conjugate (reproduced from Ref. [69]).

  • A2780 human ovarian carcinoma cells after exposure to CPT, CPTPEG,

    3 and CPTPEGLHRH. Cells were incubated 48 h with equivalent CPT

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 381molecules (Fig. 32a and b). In general, poly(L-amino

    acids) are biocompatible and biodegradable.

    Poly-N-(2-hydroxyethyl)-L-glutamine (PHEG) pro-

    drugs of the antitumor antibiotic mitomycin C (MMC)

    were synthesized using peptidyl spacers along with tri and

    tetrapeptides to link drugs to a macromolecular carrier

    (Fig. 33) [75]. Conjugates having a terminal glycine in the

    spacer are less stable to hydrolysis than those with a

    terminal hydrophobic amino acid both in buffer and in

    serum. The Gly-Phe-Ala-Leu conjugate released MMC

    Fig. 31. Typical fluorescence microscopy images of TUNEL labeled

    CPTPEGBH3, CPTPEGLHRH and the mixture of CPTPEGBH

    concentration of 3 nM (modified from Ref. [69]).very rapidly in the presence of both lysosomal enzymes

    and collagenase IV. Biological experiments indicate that

    PHEGMMC conjugates act as prodrugs of MMC.

    Cytotoxicity was observed after hydrolytic release of

    the active compound in vitro. MMC was coupled to the

    Fmoc-protected oligopeptide pentafluorophenyl ester.

    After deprotection, the amine-containing spacer-MMC

    derivatives and tyrosinamide were coupled with 4-nitro-

    phenyl carbonate containing PHEG (Fig. 33).

    Delivery of genetic materials is a long-term goal for

    which researchers continually strive. Vectors for gene

    delivery formed by self-assembly of DNA with poly

    (L-lysine) grafted with hydrophilic polymers was reported

    [76]. Poly(L-lysine) (PLL) grafted with range of

    hydrophilic polymer blocks, including poly(ethylene

    glycol) (PEG), dextran and poly[N-(2-hydroxypropyl)-

    methacrylamide] (PHPMA) showed efficient binding to

    DNA. PEG-containing complexes increased transfection

    activity against cells in vitro. Complexes formed with all

    polymer conjugates exhibit greater aqueous solubility

    than simple PLL/DNA complexes, particularly at chargeneutrality. a-Methoxy-u-carboxypoly(ethylene oxide)(PEGCOOH) was prepared by the reaction of PEGNH2with succinic anhydride (Fig. 34). Conversely, PLL-g-

    PEG copolymers were synthesized by partially coupling

    amino functions of PLL with a-methoxy-u-carboxy-poly(ethyleneoxide) using EDC as a coupling agent.

    Further, dextran-grafted PLL was prepared by reductive

    coupling of NH2 groups of PLL with the terminal

    aldehyde groups of dextran using NaCNBH3 for selective

    reduction of the Schiff base. In addition, a-n

    (a)

    -NH-CH-Cn

    O=C-NHCH2CH2OH

    (CH2)2

    [ ]

    O(b)

    Fig. 32. Structures of (a) polylysine and (b) poly-N-(2-hydroxyethyl-

    L-glutamine).

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397382(carboxyethylenethio)poly(HPMA) (PHPMACOOH)

    was prepared by radical solution polymerization in the

    presence of 3-mercaptopropionic acid (MPA) as a chain

    transfer agent. The PLL and PHPMACOOH were

    dissolved in water and adjusted to pH 5.0 by dropwise

    addition of HCl (0.5%). 1-Ethyl-3-[(dimethylamino)pro-

    pyl]carbodiimide hydrochloride (EDC) was added as

    coupling agent and the pH was maintained at 5.

    Despite the advances in the field of therapeutics, the

    delivery of nucleic acids and other genetic materials

    remains a challenge [77,78]. Due to their intrinsic

    ability, viral vectors introduce exogenous DNA into

    host cells and have had limited success in delivery of

    therapeutic genes due to immunogenicity. In this

    regard, synthetic cationic polymers are a promising

    alternative to viral vectors, e.g. polycations [79].

    Whereas certain polycations can transfect mammalian

    cells, these vectors can be cytotoxic and are much less

    efficient than their viral counterparts [80].

    A polycation gene delivery vector, polylysine-graft-

    imidazoleacetic acid (Fig. 35), has recently been shown

    to deliver genes in vitro with low cytotoxicity [81]. The

    polycation imidazole groups were conjugated to the

    Fig. 33. Mitomycin-C (MMC) poly-N-(2-hydroxyethyl)-L-glutamine)

    (PHEG) conjugate. MMC was coupled to the Fmoc-protected

    oligopeptide pentafluorophenyl ester. After deprotections the amine-

    containing spacer-MMC derivatives and tyrosinamide were coupled

    with 4-nitrophenyl carbonate containing PHEG (reproduced from

    Ref. [75]).3-amines of Mw 34,300 Da poly-L-lysine in threedifferent molar ratios. It was observed that the reporter

    gene expression increased non-linearly with the

    increasing imidazole content in polycations, providing

    an example of the structureactivity relationships of

    these polymers. Amidation of the 3-amine of poly-L-lysine with 4-imidazoleacetic acid was achieved using a

    EDAC/NHS coupling conjugation method.

    Fig. 34. Structures of the grafted copolymers (a) PLL-g-PEG, (b)

    PLL-g-dextran and (c) PLL-g-PHPMA (reproduced from Ref. [76]).

    Fig. 35. Polylysine-graft-imidazoleacetic acid conjugates (reproduced

    from Ref. [81]).

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 383C

    CH3

    CO

    HN

    CH2

    CHHO

    CH3

    C

    CH3

    CO

    NH

    CH2

    C=O

    H2C

    H2C

    CH2-CH-CONH-CH-CONH-CH2-CO

    NH CH2

    CH

    CH3H3C

    HOH2CC O

    O

    O

    O

    CH3

    OHNH

    OCH3OH

    HO

    HO

    O

    Fig. 36. Structure of PHMPA copolymeradriamycin conjugate4.1. N-(2-hydroxypropyl)methacrylamide (HPMA)

    copolymer

    Since 1973, HPMA is the most investigated and

    advanced polymer used in therapeutics due to its

    versatility as a vehicle. Other clinically established

    polymers in terms of biocompatibility and biodegrada-

    tion kinetics are linear poly(ethylene glycol) and PLGA

    copolymers. HPMA homopolymer was designed and

    synthesized in Czechoslovakia as a plasma expander

    [82]. HPMA being hydrophilic, increases water

    solubility of the drugs and has proven to be non-toxic

    in rats. Currently, antitumor agent HPMADox

    conjugate is under clinical trials.

    An HPMA copolymer with adriamycin conjugated

    with the peptidyl linker Gly-Phe-Leu-Gly (PK1)

    (Fig. 36), has been developed [83]. It was demonstrated

    that HPMAadriamycin conjugates are less toxic than

    the free drug and can accumulate inside solid tumor

    models. HPMA drug conjugates are known to induce

    endocytosis for the delivery of drugs into the cells.

    PHPMA hydrazides were synthesized by modifying it

    with N-succinimidyl 3-(2-pyridyldisulfanyl) propano-

    ate (SPDP) to introduce the pyridyldisulfanyl groups

    (reproduced from Ref. [83]).for subsequent conjugation with a modified antibody

    (Fig. 37). Dox was bound to the remaining hydrazide

    groups via an acid-labile hydrazone bond [84,85].

    Human immunoglobulin IgG was modified with

    2-iminothiolane by conjugating to the HPMA polymer

    by substitution of the 2-pyridylsulfanyl groups of the

    polymer with SH groups of the antibody. Another type

    of the conjugate used a hydrazone linkage formed by

    direct coupling of the periodate-oxidized antibody with

    hydrazide groups remaining in the PHPMA-hydrazide

    polymer after Dox attachment.

    Anticancer drug, Paclitaxel, due to its low water

    solubility, has been an ideal candidate for preparing

    water-soluble prodrugs. It has been established that the

    2 0- or 7-hydroxy group of Paclitaxel is suitable forstructure modification. Many attempts have been made

    to couple low-molecular-weight solubilizing moieties

    at the C2 0 or C7 position. These prodrugs are mainlyester derivatives including succinate, sulfonic acid, and

    amino acid/phosphate derivatives [86,87].

    Polymerdrug conjugates have demonstrated high

    potential for targeting drugs to a cancerous tumor [88].

    Apart from the selection of the ideal polymer to form a

    prodrug, there are several other important aspects that

    govern the success of polymeric conjugation. Most of

    the carriers selected for covalent conjugation of drugs

    with polymers must be water-soluble, non-toxic, and

    possess a degree of compatability for chemical

    conjugation. Advanced study of the acid-sensitive

    DoxHPMA copolymer conjugates was reported

    [8991]. PolymerDox conjugates containing side

    chains of hydrazone-bound Dox moieties were attached

    via single-amino-acid or the longer oligopeptide

    spacers. Enzymatically degradable Gly-Phe-Leu-Gly

    or non-degradable Gly, Gly-Gly, b-Ala, 6-aminohex-anoyl (AH) or 4-aminobenzoyl (AB) spacers were used.

    Also HPMA-based conjugates with Dox attached

    through Gly-Phe-Leu-Gly, Gly-Gly, and AH

    spacerscontaining cis-aconityl residue at the spacer

    endwere synthesized and studied (Fig. 38). It was

    shown that the rate of Dox released from all the

    conjugates under study was pH-dependent, with highest

    rates obtained at pH 5.

    A detailed investigation showed that mechanisms of

    anticancer action of HPMA-copolymer bound antic-

    ancer drug, Doxorubicin, differ substantially from those

    of free drugs (Fig. 39) [9296]. It was shown that high

    molecular weight HPMA-copolymer bound drugs

    accumulated preferentially in solid tumors, with only

    a trace amount of drugs detected in healthy organs. In

    contrast, significant amount of free drugs were alsofound in the liver, heart, lungs, spleen and kidneys.

  • J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397384C

    CH3

    CO

    HN

    CH2

    CHHO

    CH3

    C

    CH3

    CO

    X

    NH

    N

    H2C

    CCH2OHO

    OOCH3 OH

    OH

    OH

    C

    CH3

    CO

    X

    NH

    H2C

    C=O

    (H2C

    CH2

    CH2Therefore, conjugation of the anticancer drug to HPMA

    copolymer substantially limited adverse side effects to

    healthy organs imposed upon by free drugs.

    In addition, the distribution of polymeric drugs

    within the tumor was considerably more uniform when

    compared with that of free drugs, which accumulated

    mainly in the tumor regions with maximal permeability

    of the vascular endothelium. HPMA-copolymer bound

    anticancer drugs overcame existing drug efflux pumps

    located in the plasma membrane of cancer cells and

    prevented de novo development of multi-drug resist-

    ance during repeated chemotherapy. In contrast, free

    low molecular weight drugs activated existing multi-

    drug resistance and initiated its de novo development

    after prolonged treatment. The polymeric drugs

    internalized into the cancer cells by endocytosis were

    transported through the cellular cytoplasm in

    O

    O

    H3C

    HONH2 . HCl

    NH

    S

    S

    N

    +

    S

    NH

    CHN (CH2)3 SH

    Fig. 37. Scheme of the synthesis of HPMA copolymerDox-antibody con

    N-succinimidyl 3-(2-pyridyldisulfanyl) propanoate (SPDP) to introduce the

    antibody (reproduced from Ref. [84]).C

    CH3

    CO

    HN

    CH2

    CHHO

    CH3

    C

    CH3

    CO

    X

    NH

    N

    H2C

    CCH2OHO

    OOCH3 OH

    OH

    OH

    C

    CH3

    CO

    X

    NH

    H2C(H2C

    NH

    C=O

    CH2membrane limited organelles. This protected the

    drugs from cellular detoxification enzymes and pre-

    served their high anticancer activity. Moreover,

    HPMA-copolymer bound drugs suppressed the activity

    of cellular detoxification enzymes. In contrast, free

    drugsin most casesactivated cellular detoxification

    mechanisms, antioxidant defense systems, and other

    non-specific cellular defensive mechanisms. Finally,

    the polymeric drugs activated caspase-dependent cell

    death signaling pathways as they induced both

    apoptosis and necrosis in tumor cells. Simultaneously,

    HPMA-polymer bound drugs suppressed cell death

    dependent mechanisms. In contrast, free low molecular

    weight anticancer drugs activated cellular cell death

    defensive mechanisms. As a result, cell death induction

    by polymeric drugs was more pronounced when

    compared with the free drugs. These data justify that

    O

    O

    H3C

    HONH2 .HCl

    CH2

    S

    NH

    CHN (CH2)3 S

    jugates. PHPMA hydrazides were synthesized by modifying it with

    pyridyldisulfanyl groups for subsequent conjugation with a modified

  • HO NH2 . HCl

    J. Khandare, T. Minko / Prog. Polym. Sci. 31 (2006) 359397 385C

    CH3

    CO

    (a)

    HNCH2CHHOCH3

    C

    CH3

    CO

    X

    NH

    N

    H2C

    CCH2OHO

    O

    O

    O

    H3C

    OCH3 OH

    OH

    OH

    C

    CH3

    CO

    X

    NH

    H2C

    NH2

    (H2Cconjugation of low molecular weight drugs to HPMA

    copolymer carrier substantially increased its antitumor

    activity.

    5. Dendrimer and its conjugates

    Dendritic polymers are emerging as potentially ideal

    drug delivery vehicles because they are easily

    manipulated and they provide a large density of

    functional groups [2,9799]. Dendrimers possess

    unique properties and are considered to be most

    promising candidates for delivery of drug and

    bioactives [100104]. The following characteristics of

    dendrimers makes them ideal candidates for conju-

    gation with biomolecules. Dendrimers are (1) mono-

    disperse (w1.0), (2) nanosize (w20 nm), (3) havemultiple functional groups at the terminal (generation

    28), and (4) possess end group tailorability for

    conjugation. Due to these unique properties, dendri-

    mers can be modified to obtain a high payload of

    Fig. 38. Structure of HPMA copolymerDox conjugates: (a) hyrazone bond-

    Dox conjugates containing side chains of hydrazone-bound Dox moieties

    (reproduced from Ref. [89]).(b)H2CC

    CH3

    CO

    HNCH2CHHOCH3

    C

    CH3

    CO

    X

    NH

    (CH2)2

    H2C

    NH

    C

    CH3

    CO

    X

    NH

    NH

    (H2C

    (CH2)2

    NH2

    O CH3

    OH

    C=O

    CH2

    HC COOH

    C=O

    NHbioactive components. So far they have been primarily

    functionalized to deliver anticancer agents along with

    targeting moieties. Recently, polyether dendritic

    compounds with folate residues on their surface were

    prepared as model drug carriers with potential tumor

    cell specificity [2]. Although the dendrimers have

    capability to conjugate high amounts of drug mol-

    ecules, most of the reports imply conjugation of only 4

    5 molecules.

    In general, the low conjugation ratio could be a net

    effect of all of the following: (1) lower reactivity, (2)

    higher steric hindrance exhibited by the biomolecule as

    well as the dendrimer, (3) small radius of gyration (Rh)

    for chemical conjugation, and (4) crowding effect of the

    functional groups at the terminals. Recently, nanoscale

    dendritic drug delivery was targeted to the tumor cells

    by using the folate receptor [102]. The nanodevice was

    ethylenediamine core polyamidoamine (PAMAM)

    dendrimer of generation 5. Folic acid, fluorescein, and

    methotrexate were covalently attached to the surface to

    HOH2COCO

    OO

    OCH3OH

    OH

    HO

    contain