D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug...

download D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery and diagnostic imaging

of 7

Transcript of D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug...

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    1/7

    Cellular Delivery of Therapeutic Macromolecules 61

    Dendrimers as multi-purpose nanodevices foroncology drug delivery and diagnostic imagingD.A. Tomalia*1, L.A. Reyna* and S. Svenson**Dendritic Nanotechnologies Inc., 2625 Denison Drive, Mt. Pleasant, MI 48858, U.S.A., and Department of Chemistry, Central Michigan University,

    Mt. Pleasant, MI 48859, U.S.A.

    AbstractDendrimers are routinely synthesized as tuneable nanostructures that may be designed and regulated

    as a function of their size, shape, surface chemistry and interior void space. They are obtained with

    structural control approaching that of traditional biomacromolecules such as DNA/RNA or proteins and are

    distinguished by their precise nanoscale scaffolding and nanocontainer properties. As such, these important

    properties are expected to play an important role in the emerging field of nanomedicine. This review will

    describe progress on the use of these features for both targeted diagnostic imaging and drug-delivery

    applications. Recent efforts have focused on the synthesis and pre-clinical evaluation of a multipurpose

    STARBURSTR

    PAMAM (polyamidoamine) dendrimer prototype that exhibits properties suitable for use as:

    (i) targeted, diagnostic MRI (magnetic resonance imaging)/NIR (near-IR) contrast agents, (ii) and/or forcontrolled delivery of cancer therapies. Special emphasis will be placed on the lead candidate, namely

    [core: 1,4-diaminobutane; G (generation)= 4.5], [dendri-PAMAM(CO2Na)64]. This dendritic nanostructure

    (i.e. 5.0 nm diameter) was selected on the basis of a very favourable biocompatibility profile [The

    Nanotechnology Characterization Laboratory (NCL), an affiliate of the National Cancer Institute (NCI), has

    completed extensive in vitro studies on the lead compound and have found it to be very benign, non-

    immunogenic and highly biocompatible], the expectation that it will exhibit desirable mammalian kidney

    excretion properties and demonstrated targeting features.

    IntroductionThe extraordinary level of structural control that is possible

    by either the Tomalia-type divergent synthesis or theFrechet-type convergent synthesis of dendrimers [13] rivals

    that observed for precise biological nanostructures such as

    DNA/RNA and proteins. In fact, owing to the widely

    recognizedmimicry of protein sizes, shapes and functionality,

    dendrimers are often referred to as artificial proteins [4,5].

    Several critical properties that differentiate dendrimers from

    proteins include: (i) their coreshell architecture, wherein the

    core is concentrically surrounded by annular shells [i.e. gen-

    erations (G)] of covalently linked branch cells that create an

    interior upon which terminal surface groups(Z) are presented

    and amplified according to the equation Z=NcNbG, whereNc is core multiplicity and Nb is branch cell multiplicity

    (see Figure 1); (ii) a wide range of robust non-amino-

    acid-derived dendrimer compositions; (iii) three-dimensional

    shape presentations of surface functionality defined by the

    shape/size of the core; (iv) a unique feature of developing

    interior void space capable of hosting high multiples of

    smaller guest molecules by surface-induced congestion; and

    (v) their lack of immunogenicity. These features create a wide

    variety of protein-like nanocontainer/scaffolding structures

    Key words: dendrimer, diagnostic imaging, nanocontainer, nanomedicine, nanoscaffolding,

    targeted delivery.

    Abbreviations used: EPR, enhanced permeability retention; MRI, magnetic resonance imaging;

    PAMAM, polyamidoamine; PEG, poly(ethylene glycol); P-gp, P-glycoprotein.1

    To whom correspondence should be addressed (email [email protected]).

    with demonstrated advantages compared with proteins in

    nanomedical applications such as drug delivery [6], MRI

    (magnetic resonance imaging) and DNA/RNA transfections[7]. This very active area has been reviewed recently [810].

    Dendrimer properties of importanceto nanomedicineThe objective of nanomedicine is to control and manipulate

    biomacromolecular constructs and supramolecular assem-

    blies that are critical to living cells in order to improve the

    quality of human health. By definition, these constructs and

    assemblies are nanoscale and include entities such as proteins,

    DNA/RNA, viruses, cellular lipid bilayers, cellular receptor

    sites and antibody variable regions critical for immunology

    and are involved in events of nanoscale proportions. Theemergence of such nanotherapeutics/diagnostics will allow

    a deeper understanding of human longevity and human ills

    that include cancer, cardiovascular disease, genetic disorders

    and trauma. A technology platform that provides a wide

    range of synthetic nanostructures that may be controlled

    as a function of size, shape and surface chemistry and scale

    to these nanobiological dimensions will be a critical first

    step in developing appropriate tools and a scientific basis

    for understanding nanomedicine. Many of the properties

    of these critical parameters are offered by dendrimers and

    include the following. (i) Monodisperse, nanoscale sizes

    that scale with important bio-building block dimensions

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    2/7

    62 Biochemical Society Transactions (2007) Volume 35, part 1

    Figure 1 Dendrimer topology

    (a) Illustration of general dendrimer architectural topology with the three architectural components: (i) core, (ii) interior,

    and (iii) terminal surface groups (Z). (b) Passive size-mediated targeting: dendrimer-based diagnostic imaging and

    therapy delivery nanodevices involving [A] imaging moieties, [B] small-molecule therapy components, and (Z) low-toxicity

    terminal surface groups. (c) Active receptor-mediated targeting: dendrimer-based diagnostic imaging and therapy delivery

    nanodevices involving [A] imaging moieties, [B] small-molecule therapy components, [C] receptor-mediated targeting

    groups, and (Z) low-toxicity surface groups.

    (i.e. protein, cellular lipid bilayer, virus and DNA/RNA),

    as a function of generation level. (ii) Mathematicallydefined numbers of terminal surface groups (Z) suitable for

    bioconjugation of drugs, signalling groups, targeting moieties

    or biocompatibility groups that amplify as a function of

    generation level according to the equation Z=NbNcG.

    (iii) Dendrimer surfaces may be functionally designed to

    enhance or resist trans-cellular, epithelial or vascular bioper-

    meability. (iv) Well-defined interior void space within the

    dendrimer suitable for encapsulation (i.e. interior isolation) of

    small-molecule drugs,metals or signalling groups[i.e. MRIor

    NIR (near-IR)]. Incarceration in this void space reduces drug

    toxicity and allows controlled release. The interior is defined

    by the size and nature of the core (i.e. hydrophobic compared

    with hydrophilic), as well as surface congestion, whichincreases with generation level. (v) Positive biocompatibility

    patterns are associated with lower generation [i.e. G= 15

    for PAMAM (polyamidoamine) dendrimers], anionic or

    neutral polar terminal surface groups compared with higher

    generation neutral apolar and cationic surface groups.

    (vi) Non/low-immunogenicity properties for most dendri-

    mers functionalized with small-molecule or PEGylated

    {PEG [poly(ethylene glycol)]-modified} surface groups.

    (vii) The ability to statistically modify and optimize the

    number/ratio of dendrimer surface groups that influence

    biodistribution, receptor-mediated targeting, therapy dosage

    or controlled release of drugs from the dendrimer interior.

    (viii) The ability to tune dendrimer mammalian excretion

    mode (i.e. urinary compared with bile) as a function ofnanoscale diameter (i.e. generation level).

    Critical parameters for designing multi-purpose dendrimer-based diagnosticimaging/therapy nanodevicesBased on the published literature and unpublished work from

    our group, it is widely recognized that dendrimers may be

    used in twomajormodalities forthe detectionand therapeutic

    treatment of cancer and other diseases, namely by (i) passive

    targeting nanodimension mediated via EPR (enhanced

    permeability retention) effect [11] involving primary tumour

    vascularization or organ-specific targeting [12], and (ii) activetargeting receptor-mediated cell-specific targeting involving

    receptor-specific targeting groups. The combinatorial design

    of these nanodevices may be visualized utilizing the four

    construction parameters, A, B, C and Z, as described in

    Figure 1. The scope, use and performance of these parameters

    will be selectively reviewed in the remainder of this account.

    Dendrimers as nanoscale scaffoldingDendrimer surfaces provide an excellent platform for

    the attachment and presentation of cell-specific targeting

    groups, solubility modifiers, stealth moieties that reduce

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    3/7

    Cellular Delivery of Therapeutic Macromolecules 63

    immunological interactions and imaging tags. The ability to

    attach any or all of these molecules in a well-defined and

    controllable manner on to a robust dendritic surface clearly

    differentiates dendrimers from other vectors such as micelles,

    liposomes, emulsion droplets or engineered particles. The use

    of dendrimers as targeting vectors for diagnostic imaging,

    drug delivery and gene transfection had been proposed in the

    patent literature nearly a decade ago [13]. These applications

    evolved from early observations by Tomalia et al. [14]. It

    was noted that bioactive agents could be encapsulated into

    the interior of the dendrimers [15], physically adsorbed or

    chemically attached [16] on to the dendrimer surface, with

    an option to tailor the properties of the vector to the specific

    needs of the active material and its therapeutic applications.

    Furthermore, the mathematically defined number density of

    surface groups allows rational attachment of desired ratios of

    drugs, targeting groups or functionality that may be required

    to obtain optimum solution, stealth, targeting or release

    properties with minimal dendrimer toxicity. Concurrently it

    was observed that appropriate surface-modified dendrimersthemselves may act as nanodrugs against viruses, bacteria

    or tumours. Based on their now recognized multivalency

    properties, a dendrimer-derived microbicide (i.e. VivaGelR

    ,

    Starpharma) for HIV or genital herpes is in its final stage

    of approval by the U.S. FDA (Food and Drug Admini-

    stration) (http://www.biotech-intelligence.com/html/html/

    070156594d92398317df5fd4e6324ccf.html).

    Historically, the first in vivo diagnostic imaging applic-

    ations using dendrimer-based MRI contrast agents were

    demonstrated in the early 1990s by Lauterbur and colleagues

    [18]. In contrast with the commercial small-molecule agent

    (Magnevist

    R

    , Schering, AG), the dendrimer-based reagentsexhibited blood pool properties and extraordinary relaxivity

    values when chelated gadolinium groups (Magnevist R

    ) were

    attached to PAMAM dendrimer surfaces. These generation-

    dependent, dramatic enhancements of MRI contrast prop-

    erties were some of the first examples of a dendritic

    effect. These architecturally driven properties were unique

    to dendrimers as a member of the fourth major polymer ar-

    chitecture class(i.e. dendritic), after (i) linear, (ii) cross-linked,

    and (iii) branched types [19], and have notbeen observed with

    any of the other three traditional polymer architecture types.

    Subsequently, the work by Kobayashi and Brechbiel [12]

    extended this concept by demonstrating the efficacy of size-

    mediated targeting, using the discrete dendrimer generationsizes for in vivo imaging of primary tumours via the EPR

    effect designing organ-specific diagnostic imaging modalit-

    ies and defining size-dependent mammalian excretion routes

    (i.e. urinary compared with bile pathways). Each of these

    passive targeting modalities was based on appropriate use

    of dendrimer scaffolding dimensions for presenting the MRI

    imaging moieties and is illustrated in Figure 2.

    One of the most effective cell-specific targeting agents

    presented by dendrimer scaffolding is folic acid. Membrane-

    associated high-affinity folate receptors are folate-binding

    proteins that are overexpressed on the surface of a variety

    of cancer cells (e.g. ovarian). They have been found to

    preferentially internalize dendrimers modified with folic acid

    into receptor-bearing cells via receptor-mediated endocytosis

    over normal cells. The first in vivo cell-specific active

    targeting work with dendrimers was reported by Wiener

    et al. [20] against breast cancer. Functionalizing dendritic

    imaging agents with folic acid allowed them to be specifically

    directed to overexpressed folic acid receptor sites found in

    these cancer types. Folatedendrimer conjugates have been

    shown to be well suited for targeted cancer-specific drug

    delivery of cytotoxic substances [2123]. In a very recent

    study, branched poly(L-glutamic acid) chains were organized

    around PAMAM and poly(ethyleneimine) dendrimer cores

    to create new biodegradable polymers with improved

    biodistribution and targeting ability [24]. These constructs

    were surface terminated with PEG chains to enhance their

    biocompatibility, and folic acid to introduce cell-specific

    targeting against the epidermal KB carcinoma cell line [24].

    Carbohydrates constitute another important class of bio-

    logical recognition moieties, displaying a wide variety of

    spatial structures because of their branching possibilities andanomericity. To achieve sufficiently high binding affinities

    between simple monosaccharide/oligosaccharide ligands and

    cell membrane receptors, these ligands have to be presented

    to the receptors in a multivalent or cluster fashion [25,26].

    The highly functionalized surface of dendrimers provides

    an excellent platform for such presentations. The design,

    synthesis and biomedical use of glycodendrimers, as well as

    their application in diagnostics andfor vaccinationshave been

    reviewed thoroughly [2729].

    As early as 1990, Tomalia, Roberts and colleagues demon-

    strated conjugations of antibodies to PAMAM dendrimers

    [16]. More recently, antibodies specific to CD14 and PSMA(prostate-specific membrane antigen) were conjugated to a

    G= 5 PAMAM dendrimer bearing a fluorescein imaging tag.

    Cell binding and internalization of the antibody-conjugated

    dendrimersto antigen-expressing cellswere evaluated by flow

    cytometry and confocal microscopy. It was found that the

    conjugates bound specifically to the antigen-expressing cells

    in a time- and dose-dependent fashion with an affinity similar

    to that of the free antibody. Confocal microscopy indicated

    that cellular internalization of the dendrimer conjugate did

    occur [30].

    Recently, it has been demonstrated that PAMAM dendri-

    mers modified with VEGF (vascular endothelial growth

    factor) could be targeted to overexpressed tumour neovas-culatures in breast carcinoma [31].

    Dendrimers as nanoscale containersTomalia and colleagues first demonstrated that certain

    carboxylated PAMAM dendrimers possess topologies and

    dimensional features reminiscent of regular classical micelles

    [14,15]. Subsequent studies clearly described the ability to hy-

    drophobically modify PAMAM dendrimers (i.e. with hydro-

    phobic groups) to mimic inverse micelles and demonstrated

    the unlimited guest-host properties [8,32] that were possible

    for these two types of so-called covalently fixed dendritic

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    4/7

    64 Biochemical Society Transactions (2007) Volume 35, part 1

    Figure 2 Dendrimer scaffolding dimensions (a) for presenting MRI imaging (b)

    (a) Scaled spheroids illustrating the relative sizes (nm) for G= 18 PAMAM dendrimer series, wherein the (core:

    1,2-diaminoethane; G= 18); [dendri-PAMAM(NH2)NcNbG] generational series is categorized into the observed periodic

    properties of (i) flexible scaffolding (G= 13), (ii) nanocontainer properties (G= 46), and (iii) rigid surface scaffolding

    (G= 7 and greater), due to enhanced surface congestion as a function of generation. Note, reversible entry and departure

    of most guest molecules is possible for G= 46; however, the surface is too congested for entry into G= 7 and

    greater. (b) MRI images of mice using MagnevistR

    -modified PAMAM dendrimers, i.e. (core:1,2-diaminoethane; G= 18);

    [dendri-PAMAM(NH2)NcNb G]; wherein, MagnevistR and G= 34 are excreted completely through the kidney, G= 5 is

    excreted through both kidney and liver, and G= 69 are excreted exclusively through the liver. Note: G= 39 are excellent

    blood pool agents relative to MagnevistR

    (i.e. diethylenetriaminepenta-acetic acid) and G= 9 is very organ-specific for the

    liver, presumably due to its large nanosize [12].

    micelles. Theseresultingnanoscale container propertieswerereferred to as unimolecular encapsulations [33], and have

    been utilized to encapsulate a wide variety of bioactive drugs,

    vitamins, metal salts and diagnostic imaging moieties. Par-

    ticular interest has been focused on tailoring encapsulation,

    release and biopermeabilty features to utilize the passive and

    active targeting properties of dendrimers for either (i) size-

    mediated targeting (EPR effect), or (ii) receptor-mediated

    targeting to desired disease sites (organs) or cells. Many of

    these issues have been reviewed recently [6,9,10,34].

    Cisplatin is effective in treating major cancers such as

    ovarian, head, neck and lung cancers, as well as melanomas,

    lymphomas, osteosarcomas, bladder, cervical, bronchogenic

    and oropharyngeal carcinomas. Unfortunately, cisplatinhas many adverse side effects, the most important being

    nephrotoxicity and cytotoxicity to non-cancerous tissue, due

    to the non-selective interaction between cisplatin and DNA

    [35]. Furthermore, thetherapeutic effect of cisplatin is limited

    by its poor water solubility (1 mg/ml), low lipophilicity

    and the development of resistance to cisplatin drugs.

    Encapsulation of cisplatin within PAMAM dendrimers gives

    complexes that exhibit lower toxicity, higher accumulation

    in solid tumours and slower release compared with free

    cisplatin[3639]. Furthermore, preliminary evidence suggests

    that dendrimer complexes are able to bypass the P-gp (P-

    glycoprotein) efflux transporter [40] which may contribute

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    5/7

    Cellular Delivery of Therapeutic Macromolecules 65

    Figure 3 Generalized patterns for in vitro toxicity, in vivo toxicity, biopermeability and immunogenic properties as a function of

    dendrimer surface functionality

    Note: anionic and non-charged polar surfaces in this series exhibit the most biocompatible least toxic functionality,

    wherein () indicates less of a specific property, whereas non-charged apolar and cationic surfaces exhibit higher

    toxicity/immunogenic properties designated by (+). From [10]. Reproduced by permission of The Royal Society of Chemistry.

    to drug resistance. Cisplatin is an anti-tumour drug that

    exerts its effects by forming stable DNAcisplatin complexes

    through intrastrand cross-links, resulting in an alteration of

    the DNA structure that prevents replication and initiates

    apoptosis. Largely pioneered by Duncan and colleagues

    [3639], the tumour activity of the cisplatindendrimer

    complexes was studied using B16F10 cells. These cells were

    injected into C57 mice subcutaneously to provide a solid

    tumour model. Compared with cisplatin alone, the cisplatindendrimer complex was found to accumulate preferentially

    in the tumour site relatively quickly after the injection. This

    presumably occurs due to the EPR effect. The tumour AUC

    (area under the curve) for the complex was 5-fold higher than

    that of free cisplatin, while that in the kidney only increased

    2.4 times, and accumulation in the liver was reduced [3639].

    Another recent study revealed a remarkable stability

    of cisplatindendrimer complexes, with a 20% release of

    cisplatin over the first 8 h, and an additional 60% release

    within 150 h (G. Krippner and S. Svenson, unpublished

    work). In vivo animal efficacy of the platinate was demon-

    strated using B16F10 tumour cells that were implanted

    subcutaneously into mice. The tumour was allowed to growfor 7 days before treatment with two doses of drug, on day 7

    and day 14, providing equal capsulation (5 mg/kg) doses in

    both the dendrimer capsulation complex and free cisplatin. A

    tumour mass reduction of40% compared with that for the

    free drug was found in this study.

    The anticancer drugs adriamycin and methotrexate were

    encapsulated into PAMAM dendrimers (i.e. G= 3 and 4)

    which had been modified with PEG monomethyl ether

    chains (i.e. 550 and 2000 Da respectively) attached to their

    surfaces. The encapsulation efficiency was dependent on the

    PEG chain length and the generation (size) of the dendrimer,

    with the highest encapsulation efficiencies (on average, 6.5

    adriamycin molecules and 26 methotrexate molecules per

    dendrimer) found for the G= 4 PAMAM terminated with

    PEG2000 chains. The drug release from this dendrimer was

    sustained at low ionic strength, again reflecting PEG chain

    length and dendrimer size, but fast in the isotonic solution

    [42]. In a related study, it was reported that the surface cover-

    age of PAMAM dendrimers with PEG2000 chains had little

    influence on the encapsulation efficiency of methotrexate, but

    affected the release rate [43]. A similar construct involvingPEG chains and PAMAM dendrimers was used to deliver

    the anticancer drug 5-fluorouracil. Encapsulation of 5-

    fluorouracil into G= 4 PAMAM dendrimers modified with

    carboxymethyl PEG5000 surface chains revealed reasonable

    drug loading, a reduced release rate and reduced haemolytic

    toxicity compared with the non-PEGylated dendrimer

    [44].

    Routes of applicationDendrimeric vectors are most commonly used as parenteral

    injections, either directly into the tumour tissue or intra-

    venously for systemic delivery. However, oral drug deliverystudies using the human colon adenocarcinoma cell line,

    Caco-2, have indicated that low-generation PAMAM

    dendrimers cross cell membranes, presumably through a

    combination of two processes, i.e. paracellular transport and

    adsorptive endocytosis [45]. Remarkably, the P-gp efflux

    transporter does not appear to affect dendrimers, therefore

    drugdendrimer complexes are able to bypass the efflux

    transporter [40].

    Biocompatibility of dendrimersThe most important features required for the widespread

    use of dendrimers in nanomedicine are that they exhibit

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    6/7

    66 Biochemical Society Transactions (2007) Volume 35, part 1

    low toxicity, acceptable excretionary pathways and are non-

    immunogenic. To date, the cytotoxicity of dendrimers has

    been studied primarily in vitro, although a few in vivo

    mammalian studies have been performed on mice or rats [46].

    However, advanced human clinicals are currently in progress

    involving topical uses of an anionic surface functionalized

    polylysine dendrimer (i.e.VivaGelR

    ) which is used for the

    prevention of HIV and genital herpes [47,48]. In general,

    the dendrimer surface functionality in combination with the

    generation level (size) are the two most significant para-

    meters affecting toxicity either in vitro or in vivo. The most

    comprehensive reviewof these issuesmay be found in a recent

    paper by Boas, Christensen and Heegaard [10], wherein, they

    have attempted to categorize in vitro and in vivo toxicities

    of dendrimers according to four general types of dendrimer

    surface groups as shown in Figure 3.

    ConclusionsThe extraordinarily high level of synthetic control over the

    size, shape, surface functionality and interior void space

    makes these nanostructures ideal vectors for both passive and

    activedrugdelivery/diagnosticimagingapplications.Thebio-

    active agents may be encapsulated into the interior of the

    dendrimers, physically adsorbed or chemically attached on to

    the dendrimer surface, with the option to tailor the properties

    of the vector to the specific needs of the active material and

    its therapeutic applications. Furthermore, the ability to select

    nanoscale-sized vectors with mathematically determined

    numbers of surface groups and well-defined interior void

    space allows systematic size adjustments to determine excre-

    tionary pathways while producing optimal ratios of target-ing moieties, therapy and surface groups required in

    combination with desired solution behaviour, excretionary

    pathway and acceptable toxicity margins. Finally, certain

    anionic surface-modified dendrimers are proving to function

    as safe andeffective topicalnanodrugs against HIVand genital

    herpes. These dendrimer-based nanopharmaceutics are in the

    final stages of human clinical testing in the U.S. FDA ap-

    proval process ([47,48], and http://www.biotech-intelligence.

    com/html/html/070156594d92398317df5fd4e6324ccf.html).

    This brief review of dendrimer-based nanomedical applic-

    ations clearly illustrates the potential of this new fourth

    architectural class of polymers [19,49] and substantiates ahigh optimism for the future role of dendrimers in the

    emerging field of nanomedicine [50].

    We thank all contributors to this fascinating field of nanomedical

    research, as well as the funding agencies that have supported this

    work over the years. This includes the NCI (National Cancer Institute),

    NIH (National Institutes of Health) and DARPA (Defense Advanced

    Research Projects Agency). In particular, D.N.T. also acknowledges

    funding by the U.S. ARL (Army Research Laboratory) (Contract #

    W911NF-04-2-0030). We express sincere appreciation to Ms L.S.

    Nixon for manuscript preparation.

    References1 Fr echet, J.M.J. and Tomalia, D.A. (2001) Dendrimers and Other Dendritic

    Polymers, Wiley2 Tomalia, D.A. (2004) Aldrichimica Acta 37, 39573 Tomalia, D.A. (2005) Prog. Polym. Sci. 30, 2943244 Tomalia, D.A., Huang, B., Swanson, D.R., Brothers, II, H.M. and Klimash,

    J.W. (2003) Tetrahedron 59, 379938135 Tomalia, D.A., Brothers, II, H.M., Piehler, L.T., Durst, H.D. and Swanson,

    D.R. (2002) Proc. Natl. Acad. Sci. U.S.A. 99, 508150876 Svenson, S. and Tomalia, D.A. (2005) Adv. Drug Deliv. Rev. 57,

    210621297 Kubasiak, L.A. and Tomalia, D.A. (2005) in Polymeric Gene Delivery

    Principles and Applications (Amiji, M.M., ed.), pp. 133157, CRC Press,Boca Raton

    8 Tomalia, D.A., Mardel, K., Henderson, S.A., Holan, G. and Esfand, R.(2003) in Handbook of Nanoscience, Engineering and Technology(Goddard, III, W.A., Brenner, D.W., Lyshevski, S.E. and Iafrate, G.J., eds.),pp. 134, CRC Press, Boca Raton

    9 Lee, C.A., MacKay, J.A., Frechet, J.M.J. and Szoka, F.C. (2005)Nat. Biotechnol. 23, 15171526

    10 Boas, U., Christensen, J.B. and Heegaard, P.M.H. (2006) Dendrimers inMedicine and Biotechnology, The Royal Society of Chemistry, Cambridge

    11 Maeda, H. and Matsumura, Y. (1986) Cancer Res. 33, 436312 Kobayashi, H. and Brechbiel, M.W. (2003) Mol. Imaging 2, 11013 Tomalia, D.A., Wilson, L.R., Hedstrand, D.M., Tomlinson, I.A., Fazio, M.J.,

    Kruper, Jr, W.J., Kaplan, D.A., Cheng, R., Edwards, D.S. and Jung, C.W.(1996) Dense Star Conjugates. U.S. Pat. 5, 527,524

    14 Tomalia, D.A., Berry, V., Hall, M. and Hedstrand, D.M. (1987)Macromolecules 20, 11641167

    15 Naylor, A.M., Goddard, III, W.A., Keifer, G.E. and Tomalia, D.A. (1989)J. Am. Chem. Soc. 111, 23392341

    16 Roberts, J.C., Adams, Y.E., Tomalia, D., Mercer-Smith, J.A. and Lavallee,D.K. (1990) Bioconjug. Chem. 2, 305308

    17 Reference deleted18 Wiener, E.C., Brechbiel, M.W., Brothers, II, H.M., Magin, R.L., Gansow,

    O.A., Tomalia, D.A. and Lauterbur, P.C. (1994) Magn. Reson. Med. 31,18

    19 Tomalia, D.A. and Fr echet, J.M.J. (2002) J. Polym. Sci. Part APolym. Chem. 40, 27192728

    20 Wiener, E.C., Konda, S., Shadron, A., Brechbiel, M. and Gansow, O. (1997)Invest. Radiol. 32, 748754

    21 Kono, K., Liu, M. and Frechet, J.M. (1999) Bioconjug. Chem. 10,1115112122 Baker, Jr, J.R., Quintana, L., Piehler, L., Banazak-Holl, M., Tomalia, D. and

    Raczka, E. (2001) Biomed. Microdevices 3, 616923 Kukowska-Latallo, J.F., Candido, K.A., Cao, Z., Nigavekar, S.S., Majoros, I.J.,

    Thomas, T.P., Balogh, L.P., Khan, M.K. and Baker, Jr, J.R. (2005)Cancer Res. 65, 53175324

    24 Tansey, W., Ke, S., Cao, X.Y., Pasuelo, M.J., Wallace, S. and Li, C. (2004)J. Controlled Release 94, 3951

    25 Lundquist, J.J. and Toone, E.J. (2002) Chem. Rev. 102, 55557826 Zanini, D. and Roy, R. (1997) J. Am. Chem. Soc. 119, 2088209527 Veprek, P. and Jezek, J. (1999) J. Pept. Sci. 5, 20322028 Roy, R. (1996) Curr. Opin. Struct. Biol. 6, 69270229 R ockendorf, N. and Lindhorst, T.K. (2001) Top. Curr. Chem. 217,

    20123830 Thomas, T.P., Patri, A.K., Myc, A., Myaing, M.T., Ye, J.Y., Norris, T.D. and

    Baker, Jr, J.R. (2004) Biomacromolecules 5, 22692274

    31 Backer, M.V., Gaynutdinov, T.I., Patel, V., Bandyopadhyaya, A.,Thirmumamagal, B.T.S., Tjarks, W., Barth, R.F., Claffey, K. and Backer, J.M.(2005) Mol. Cancer Ther. 4, 14231429

    32 Tomalia, D.A. (2005) in Supramolecular Polymers (Ciferri, A., ed.),pp. 187256, CRC Press, Boca Raton

    33 Tomalia, D.A., Naylor, A.M. and Goddard, III, W.A. (1990) Angew. Chem.Int. Ed. 29, 138175

    34 Esfand, R. and Tomalia, D.A. (2001) Drug Discov. Today 6, 42743635 Wang, D. and Lippard, S.J. (2005) Nat. Rev. 4, 30732036 Malik, N., Evagorou, E.G. and Duncan, R. (1999) Anti-Cancer Drugs 10,

    76777637 Malik, N. and Duncan, R. (2003) Dendritic-plantinate drug delivery

    system. U.S. Pat. 6, 585,95638 Malik, N. and Duncan, R. (2004) Method of treating cancerous tumors

    with a dendritic-plantinate drug delivery system. U.S. Pat. 6, 790,43739 Malik, N., Duncan, R., Tomalia, D.A. and Esfand, R. (2006)

    Dendritic-antineoplastic drug delivery system. U.S. Pat. 7, 005,124

    C2007 Biochemical Society

  • 8/3/2019 D.A. Tomalia, L.A. Reyna and S. Svenson- Dendrimers as multi-purpose nanodevices for oncology drug delivery an

    7/7

    Cellular Delivery of Therapeutic Macromolecules 67

    40 DEmanuele, A., Jevprasesphant, R., Penny, J. and Attwood, D. (2004)J. Controlled Release 95, 447453

    41 Reference deleted42 Kojima, C., Kono, K., Maruyama, K. and Takagishi, T. (2000)

    Bioconjug. Chem. 11, 91091743 Pan, G.F., Lemmouchi, Y., Akala, E.O. and Bakare, O. (2005) J. Bioactive

    Compat. Polym. 20, 11312844 Bhadra, D., Bhadra, S., Jain, S.K. and Jain, N.K. (2003) Int. J. Pharm. 257,

    111124

    45 El-Sayed, M., Rhodes, C.A., Ginski, M. and Ghandehari, H. (2003)Int. J. Pharm. 265, 151157

    46 Boas, U. and Heegaard, P.M.H. (2004) Chem. Soc. Rev. 33, 4363

    47 Jiang, Y.H., Emau, P., Cairns, J.S., Flanary, L., Morton, W.R.,McCarthy, T.D. and Tsai, C.C. (2005) AIDS Res. Hum. Retroviruses 21,207213

    48 McCarthy, T.D., Karellas, P., Henderson, S.A., Giannis, M., OKeefe, D.F.,Heery, G., Paull, J.R.A., Matthews, B.R. and Holan, G. (2005)Mol. Pharmacol. 2, 312318

    49 Tomalia, D.A. and Fr echet, J.M. (2005) Prog. Polym. Sci. 30,217219

    50 Tomalia, D.A. (2005) Chem. Today 23, 5255

    Received 22 August 2006

    C2007 Biochemical Society