nano receptors

Upload: ramik

Post on 07-Jul-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/18/2019 Nano Receptors

    1/12

    Nanoparticle targeting of anti-cancer drugs that alter intracellular

    signaling or inuence the tumor microenvironment☆

    Mathumai Kanapathipillai a, Amy Brock b, Donald E. Ingber a,c,d,⁎a Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USAb Department of Biomedical Engineering, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX USAc Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USAd School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA

    a b s t r a c ta r t i c l e i n f o

    Available online 9 May 2014

    Keywords:

    Nanoparticle

    Tumor

    Nanotherapeutics

    Delivery

    Microenvironment

    Drug

    Nanoparticle-based therapeutics are poised to become a leading delivery strategy for cancer treatment because

    they potentially offerhigher selectivity,reduced toxicity, longerclearancetimes, and increased ef cacy compared

    to conventional systemic therapeutic approaches. This article reviews existing nanoparticle technologies and

    methods that areused to targetdrugsto treat cancerby altering signaltransductionor modulating thetumor mi-

    croenvironment. We also consider the implications of recent advances in the nanotherapeuticseldfor thefuture

    of cancer therapy.

    © 2014 Elsevier B.V. All rights reserved.

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107

    2. Targeting signaling networks in cancer cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1092.1. Surface-receptor mediated pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109

    2.1.1. Transmembrane delivery vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110

    2.2. Targeting intracellular signaling molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110

    2.2.1. Focused delivery of siRNAs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

    3. Targeting the tumor microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

    3.1. Vasculature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

    3.1.1. Angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

    3.1.2. Hypoxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

    3.2. Immune response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

    3.3. Extracellular matrix . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

    3.4. Multi-drug resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114

    4. Challenges for the future . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114

    Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116

    1. Introduction

    Current cancer chemotherapies often failto improve patient mor-

    tality and morbidity due to severe adverse effects on normal tissues.

    Targeted nanocarrier systems potentially offer a solution to this

    problem becausethey can be designed to deliver cargo preferentially

    to the tumor cells, sparing healthy tissue from dose-limiting side

    effects. Because their size and shape can be tailored as needed,

    nanotherapeutics also enhance bioavailability and plasma solubility,

    Advanced Drug Delivery Reviews 79–80 (2014) 107–118

    ☆   Thisreviewis partof the Advanced Drug Delivery Reviews themeissue on "Engineering

    of Tumor Microenvironments".

    ⁎   Corresponding author at: Wyss Institute for Biologically Inspired Engineering at

    Harvard University, Boston, MA, USA.

    E-mail address: [email protected] (D.E. Ingber).

    http://dx.doi.org/10.1016/j.addr.2014.05.005

    0169-409X/© 2014 Elsevier B.V. All rights reserved.

    Contents lists available at ScienceDirect

    Advanced Drug Delivery Reviews

     j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / a d d r

    http://dx.doi.org/10.1016/j.addr.2014.05.005http://dx.doi.org/10.1016/j.addr.2014.05.005http://dx.doi.org/10.1016/j.addr.2014.05.005http://dx.doi.org/10.1016/j.addr.2014.05.005http://dx.doi.org/10.1016/j.addr.2014.05.005mailto:[email protected]://dx.doi.org/10.1016/j.addr.2014.05.005http://www.sciencedirect.com/science/journal/0169409Xhttp://www.sciencedirect.com/science/journal/0169409Xhttp://dx.doi.org/10.1016/j.addr.2014.05.005mailto:[email protected]://dx.doi.org/10.1016/j.addr.2014.05.005http://crossmark.crossref.org/dialog/?doi=10.1016/j.addr.2014.05.005&domain=pdf

  • 8/18/2019 Nano Receptors

    2/12

    decrease systemic toxicity, increase patient compliance and improve

    pharmacokinetics of biologics and small molecules that otherwise

    have short half-lives in vivo.

    The   eld of nanotherapeutics has been made possible by the

    development of manufacturing methods that enable synthesis, self-

    assembly or fabrication of objects with dened size, shape and chemis-

    try on the nanometer scale and that are also biocompatible. These are

    either composed of multiple molecules or single molecules that are

    engineered to contain multiple functional cassettes. Examples of biocompatible nanocarriers include those composed of liposomes,

    polymers, micelles, engineered multi-functional antibodies, and nano-

    particles that can be composed of metals (e.g., gold, silver), polymer,

    quantum dots, dendrimers, fullerenes, ferritin, proteins, DNA, other bio-

    logical molecules or combinations of these materials (Fig. 1) [1–4].

    Nanomaterials also can be precisely designed to achieve desired

    physical and chemical properties necessary to target delivery of drugs

    to the dynamic tumor microenvironment with higher therapeutic

    ef cacy and lower toxicity. For example, surface to volume ratio, size,

    geometry, surface charge, shape and stimuli-responsive properties are

    examples of design features that can be controlled when formulating

    nanotherapeutics. Once the nanoparticles are created, they also can be

    functionalized with additional chemical moieties, such as antibodies

    or ligands for cell surface receptors, to target to selective sites, prevent

    clearance from blood, and provide other desired functional properties.

    To achieve effective targeting to cancerous tissues, systemically de-

    livered nanoparticles must   rst extravasate from the bloodstream,

    pass through the tumor extracellular matrix (ECM), bind to cells, and

    then cross the surface membrane to enter the target cells and reach ap-

    propriate intracellular sites (Fig. 2) [5–7]. Two targeting strategies, one

    active and the other passive, have been utilized to enhance nanoparticle

    delivery to tumors. In passive targeting, one capitalizes on the leaky na-

    ture of tumor microvessels, which results in enhanced permeability and

    retention (EPR) responses that facilitate greater accumulation of nano-

    particles compared to individual soluble molecules or drugs in the

    perivascular tumor microenvironment. The EPR effect varies depending

    on the size and surface properties of the nanocarriers, and hence, nano-

    particles must be designed to achieve maximum targeting and thera-

    peutic ef cacy. Nanocarriers in the size range of 20–200 nm can easily

    extravasate through the walls of poorly formed microvessels in the an-giogenic tumor niche, and poor lymphatic drainage further enhances

    their accumulation. Several nanoparticles that utilize this EPR-based

    passive targeting strategy are currently in human clinical trials  [8,9].

    Examples include the doxorubicin-liposome carriers Myocet and Doxil

    [10,11], which have increased half-lives in the blood due to the size of 

    the nanocarrier and in the case of Doxil, chemical coating of the lipo-

    some with poly-ethyleneglycol (PEG). Passive targeting polymeric

    micelles, nanoparticles, and polymer-drug conjugates are also in clinical

    trials for breast, lung, pancreatic and ovarian cancers [9,11].

    The creation of targeting antibodies in late1970s led to the develop-

    ment of the rst active or targeted nanoparticle delivery strategies  [12,

    13]. The concept was simple: by coating nanoparticles with antibodies,

    they could be selectively directed to sites expressing the target antigen.

    This active targeted delivery approach was later extended by coating

    nanoparticles with targeting ligands, lectin-carbohydrates, or antigen

    molecules that recognize specic sites or endogenous antibodies to fa-

    cilitate intracellular drug delivery [5,14]; these targeting ligandsinclude

    peptides, antibodies, aptamers, small molecules and proteins among

    others [5,14,15]. The density of these ligands on the surface of thenano-

    particles also can be ne-tuned to optimize binding and targeting using

    several physical and chemical immobilization techniques. Examples

    Fig. 1. Multi-valent, multi-functional and stimuli-responsive nanocarrier deliverysystems for tumortargeting. Examples of nanocarrierdelivery vehicles include (left to right) liposomes,

    polymers, micelles, engineered antibodies and nanoparticles decorated with stimuli-responsive cleavable stealth coatings (blue rectangles), targeting moieties (arrows), and anti-cancer

    drugs or imaging agents (stars and suns).

    108   M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

  • 8/18/2019 Nano Receptors

    3/12

    include technologies that enable precise control of size, shape, composi-

    tion and surface modication of nanoparticles [16,17] and a lipid inser-

    tion method that is used for targeted functionalization of erythrocyte-cloaked nanoparticles [18]. There are only a few active targeted cancer

    nanotherapeutics in clinical trials. The two that are farthest along are

    doxorubicin-containing immunoliposomes that are targeted to cancers

    using a human monoclonal antibody, and transferrin-targeted oxaliplatin

    liposomes; however, mostof the published resultson these nanotechnol-

    ogies refer to preclinical studies [11,19,20].

    The most common anti-cancer nanotherapeutics in development

    target receptors on the surface membranes of tumor cells to enhance

    local drug concentrations, facilitate drug entry into the cell, and selec-

    tively perturb specic receptor-mediated signaling pathways [5]. How-

    ever, as our understanding of cancer biology has advanced, there has

    been increased recognition of the importance of intracellular signaling

    molecules and particular genes for cancer development, as well as

    new appreciation for the key role that the tumor microenvironment(e.g., capillary blood vessels, stroma, ECM composition and mechanics,

    immune cells, etc.) contributes to cancer control. Thus, nanocarriers

    are also being employed to selectively concentrate drugs at tumor

    sites based on the tumor microenvironment, as well as to target specic

    intracellular signaling molecules, and genes. Nanoparticles with multi-

    valency, multi-functionality, and triggered or stimuli-responsive release

    properties (Fig. 1) are all currently being explored for tumor targeting

    [6,21–23]. Clearly, there are still many challenges that need to be over-

    come before we can design fully effective, targeted, nanoscale, drug

    delivery systems that will overcome the complex interactions between

    the tumor cell's intracellular information processing network and the

    ever-changing tumor microenvironment that govern cancer formation

    and expansion. Thus, in this review, we focus on various nanoparticle-

    based drug delivery approaches that are being developed to either tar-get intracellular signaling pathways or the tumor microenvironment,

    as shown in the schematic in  Fig. 3. We also explore the implications

    of recent developments in this  eld for the future of targeted cancer

    therapy.

    2. Targeting signaling networks in cancer cells

    Cancers are heterogeneous in nature, and their growth and progres-

    sion depend on reciprocal interactions between tumor cells and their

    surrounding microenvironment [24–26]. Information processing net-

    works that govern cancer behavior include both chemical and genetic

    signaling pathways inside individual tumor cells, as well as inter-

    cellular signaling networks that involve soluble cues transferred back

    and forth between tumor cells and surrounding vascular endothelial

    cells, cancer-associated  broblasts, and cells of the immune system

    [24,27,28]. In this section, we examine nanoparticle-based delivery

    strategies that target drugs to these diverse information handling net-works that process soluble cues in the tumor.

     2.1. Surface-receptor mediated pathways

    Detailed molecular characterization of tumor cell signaling networks

    has identied changes in growth signaling receptors that characterize

    specic tumor types. Active targeting of nanoparticles to these signaling

    components couldpotentially increase therapeutic effectiveness and re-

    duce collateral damage caused by chemotherapeutic drugs on normal

    healthy tissues. Molecular recognition of tumor cells may exploit li-

    gand–receptor interactions or antigen–antibody binding to ef ciently

    target the nanoparticle drug delivery vehicle to the tumor. Humanized

    antibodies are attractive for targeting cell surface receptors due to

    their high specicity and low immune reactivity [29]. Active targetingvia antibody interactions can distinguish cancerous cells from normal

    tissue and also can target specic subpopulations of cancer cells [30].

    Folate is one of the most extensively utilized ligands for targeted

    cancer nanotherapeutics due to its high af nity bindingto the folate re-

    ceptor (K d ~10−9) and because it is frequently overexpressed in diverse

    tumor types including uterine sarcomas, ovarian carcinomas, osteosar-

    comas, and non-Hodgkin's lymphomas [30]. Folate recently was used

    to target delivery of tamoxifen with albumin and alginate-based nano-

    particles [31] and enhanced in vivo targeting of camptothecin to pancre-

    atic xenografts was accomplished through the use of folate-coated

    mesoporous silica nanoparticles loaded with this drug [32]. Signicant

    reduction in tumor volume and enhanced renal clearance were ob-

    served compared with uncoated particles in the camptothecin nanopar-

    ticle studies.Transferrin is an iron-binding plasma glycoprotein that facilitates

    iron uptake by cells through binding to its membrane receptor, TfR 

    [33], and this receptor is overexpressed by as much as 10-fold in

    many tumor cell types [34]. Ligand binding of TfR initiates endocytosis,

    which has been exploited for targeted delivery of transferrin-coated

    nanoparticles.  In vivo examples of this approach include the targeting

    of transferrin-conjugated paclitaxel-loaded nanoparticles to prostate

    [34], and delivery of transferrin-coated PLGA-nanoparticles to gliomas

    [35] in animal studies.

    Epidermal growth factor receptor (EGFR) is over-expressed in many

    cancers and can serve as a target for nanotherapeutics through binding

    of one of its two natural ligands: epidermal growth factor (EGF) or

    transforming growth factor-alpha (TGF-α). EGF-conjugation of stearoyl

    gemcitabinenanoparticles (GemC18-NPs) has been shown to result in a

    Fig. 2. A schematic depicting the mechanism by which nanoparticles are transported to the tumor and its surrounding microenvironment. Nanoparticles that escape reticuloendothelial

    system (RES)during circulation,reach tumorblood vessels and subsequentlyextravasate into the tumorinterstitium. After overcoming transport barriers and diffusion limitations, nano-

    particles reach the targeted tumor cells, where they deliver their cargo or imaging agents.

    109M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

    http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B2%80

  • 8/18/2019 Nano Receptors

    4/12

    2-fold improvement in its targeting to tumor cells that over-express

    EGFR  [36]. Human epidermal growth factor receptor 2 (HER2) is over-

    expressed in breast, ovarian, gastricand uterine cancers and it is the tar-

    get of the monoclonal antibody trastuzumab (marketed as Herceptin).

    Human serum albumin nanoparticles that were surface-coated with

    trastuzumab and loaded with the cytostatic drug doxorubicin displayed

    enhanced cell uptake and biological activity   [37]. Such targetedapproaches may be key to reducing clinical issues of cardiotoxicity

    that can be a major source of morbidity in patients receiving doxorubi-

    cin. The challenge of doxorubicin resistance also may be overcome, or

    at least reduced, using nanoparticles that are not recognized by

    P-glycoprotein, which is one of the main protein pumps that mediates

    the multidrug resistance response.

     2.1.1. Transmembrane delivery vehicles

    Several transmembrane nanoparticle-based delivery strategies have

    been reported to permit the delivery of small molecules, proteins, small

    interfering ribonucleic acids (siRNAs) and imaging agents to intracellu-

    lar compartments of tumor cells. Smallmolecule-mediated nanoparticle

    delivery of small molecules is commonly accomplished by utilizing

    mechanisms of endosomal escape and cytosolic release of nanoparticles.Delivery of doxorubicin by mesoporous silica nanoparticles coated with

    polyethylenimine (PEI) [38] and gold nanoparticles coated with doxo-

    rubicin conjugated to an acid cleavable PEG linker  [39] have both been

    shown to effectivelydeliver small molecules into cytosol. More recently,

    it was shown to be possible to target chemotherapeutic drugs to mito-

    chondria using poly(lactic-co glycolic acid) (PLGA)–PEG nanoparticles

    blended with triphenylphosphonium, a lipophilic cation known to

    cross mitochondrial membranes [40,41].

    For intracellular protein delivery, silica nanoparticles that target

    phospho-Akt (protein kinase B) to inhibit its kinase activity have been

    developed [42]. The nanoparticles enter cells through the endosomal

    pathway and subsequent escape into the cytosol, causing protein deliv-

    ery and eventually, cell death. Intracellular protein delivery also has

    been accomplished by targeting the ribosome inhibiting proteins (RIP)

    that suppress translation [43]. Several  “cell penetrating peptides,” such

    as the TAT (transactivated-transcription) sequence, have been utilized

    to mediate transfer of peptides and proteins across cell membranes and

    directly intothe cytosol [44]. These havebeen used to create nanocarriers

    that can enter the cytoplasm as well. Examples include TAT-conjugated

    PLGA nanoparticles to deliver K237 peptides that target the kinase insert

    domain receptor for brain drug delivery [45], and intracellular delivery of quantum dot-mediated protein delivery and imaging [46].

    The physical properties of nanoparticles also have been leveraged to

    enhance cellular internalization.For example, the specicity of targeting

    ligand-coated nanoparticles can be augmented by engineering their

    shape to exhibit distinct 3D geometries. PEG-hydrogel rods with a

    high aspect ratio of 150 × 450 nm exhibit higher cellular uptake com-

    pared with cylindrical particles with 200 × 200 nm dimensions  [47].

    Uptake and internalization of trastuzumab-coated nanoparticles by

    breast cancer cells also were found to be higherfor rod-shapedparticles,

    followed by disks, and then spheres [48]. Interestingly, rod-shaped par-

    ticles were also found to provide improved specicity of endothelial

    targeting in lung and brain compared with other geometries  [49].

    Thus, the abilityto shape nanocarriersprovides an alternative way to af-

    fect drug delivery, independently of the drug being delivered or theirsurface coating.

     2.2. Targeting intracellular signaling molecules

    Intracellular signaling molecules control cell proliferation, differenti-

    ation, andsurvival, as well as myriadmetabolic functionsin cells.During

    cancer progression, the signaling networks undergo severe alteration

    and deregulation. Therapeutic targeting of specic molecules within

    these complex tumor signaling networks remains a challenge due to

    poor drug penetration into intracellular compartments. Nanocarriers

    offer a new way to overcome this challenge because they can be tuned

    to deliver the cargo by receptor-mediated internalization as well as

    organelle- or intracellular molecule-specic targeting. Below, we out-

    line some recent developments in this area.

    Fig. 3. Schematic of potential strategies forselectively targeting nanoparticles to various key structures and features of the tumormicroenvironment, including the vasculature (1), hypoxic

    regions (2), and intracellular signaling networks (3, a) and surface receptors (3, b) on tumor cells, extracellular matrix (4) and the immune response (5). Insert at left shows a multi-

    functional nanoparticle decorated with targeting moieties and loaded with drugs.

    110   M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

    http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B3%80

  • 8/18/2019 Nano Receptors

    5/12

     2.2.1. Focused delivery of siRNAs

    The recent development of siRNAs, which direct sequence-specic

    degradation of target mRNA transcripts, has led to the development of 

    an entirely new class of therapeutics for cancer. As the siRNA molecule

    itself is strongly negatively charged, delivery to cells is typically accom-

    plished by complexing it with a cationic carrier or lipid-based nanopar-

    ticle. In the rst-in-human trials studies, siRNA was targeted to the M2

    subunit of ribonucleotide reductase via cyclodextrin–PEG nanoparticles

    coated with human transferrin [50]. TfR receptor is expressed at highlevels on malignant cells as described above, and thus this enhanced

    accumulation of the nanoparticles at tumor sites in patients with refrac-

    tory melanoma.

    A new class of nanoparticle materials, termed  “lipidoids,” has been

    developed to facilitate siRNA delivery. Lipidoids are derived from an

    amine-containing backbone from which aliphatic chains extend; the

    amines appear to interact electrostatically with the nucleic acid phos-

    phate backbone and facilitate endosomal escape as the compartment

    becomes acidied. The ef cacy of lipidoid nanoparticle-siRNA formula-

    tions has been demonstrated in the silencing of tight junction protein

    claudin-3 in animals models of ovarian cancer [51] as well as Parp1 in

    Brca1-decient ovarian tumors [52] and glycolytic enzyme pyruvate ki-

    nase in several xenograft models [53]. Injection of lipidoid nanoparticles

    carrying siHoxA1 through the nipple and directly into the mammary

    duct prevented the progression of mammary tumors in the C3(1)

    SV40TAg transgenic mice [54].

    Although siRNA delivery by nanoparticles is able to delivery micro-

    grams of drug per kilogram of animal body weight, there remains

    room for additional improvement. For example, better understanding

    of the endocytotic pathways of particular siRNA–nanoparticle formula-

    tions may improve their ef ciency of delivery. After lipid nanoparticles

    enter the cell, a signicant amount of siRNA remains trapped in endocy-

    totic vesicles and does not reach its intracellular target. In parallel, vesicle

    recycling continues to rapidly clear the siRNA–nanoparticles from the cell

    before the payload is delivered. One recent study identied Niemann–

    Pick type C1 (NPC1) as a critical regulator of the major recycling path-

    ways of lipid nanoparticle-formulated siRNAs. In cells lacking NPC1,

    there was enhanced retention of nanoparticles inside the endosome

    and lysosomes of the recycling pathway, and the level of gene silencingachieved with RNAinterference was 10 to 15 times greaterthan that pro-

    duced in normal cells [55]. Strategies to target specic molecular compo-

    nents of endosome production, traf cking and recycling may be

    incorporated into the design of the next generation of nanocarriers for

    siRNA delivery. Another challenge is that immune stimulation through

    α /  β  interferons, RNA-dependent kinase effects, and Toll-like immunity

    couldlimit the safety proles of siRNAs. However, if this immune activat-

    ing activity of the siRNAs can be controlled or even exploited for use

    against the tumor, this sideeffectcould potentially improve treatment ef-

    cacy. The current status of nanoparticle-based RNAi therapies in clinical

    trials for oncology applications is shown in Table 1.

    3. Targeting the tumor microenvironment

    Over the past few decades, it has become recognized that tumor

    growth and progression are not solely the result of cumulative gene

    mutations, but are also signicantly inuenced by the surrounding

    tumor microenvironment. In addition to cancer cells, the dynamic

    tumor microenvironment consists of ECM, stromal cells, endothelial

    cells, and immune cells that are recruited from surrounding tissues as

    well as from the bone marrow  [25,56]. Interestingly, the chemistry,

    structure and mechanical properties of the tumor microenvironment

    all inuence the cellular information processing networks that drive

    phenotypic changes leading to tumor malignancy [24,56–59]. Hence, it

    is important to consider both cellular and non-cellular components of tumor microenvironment for cancer therapy as their complex interac-

    tions play a critical role in cancer progression. In this section we discuss

    nanoparticle-based delivery systems that have been reported to target

    various components of the tumor microenvironment.

     3.1. Vasculature

    In contrast to normal tissues, the vasculature of tumors is marked by

    a heterogeneous distribution of vessel sizes and shapes, generally larger

    vessel diameters, higher vascular density, and enhanced permeability.

    This inherently leaky vasculature promotes preferential accumulation

    of nanoparticles in the tumor interstitial space, and when coupled

    with impaired lymphatic drainage of macromolecules, this results in

    the EPR effect characterized by increased accumulation of nanoparticles

    that is observed in many solid tumors [60], as described above. The EPR 

    effect has been successfully exploited for passive targeting of drugs to

    solid tumors in animals that areencapsulatedin a variety of high molec-

    ular weight carrier systems, including polymeric drug conjugates, lipo-

    somes, polymeric NPs and micelles [61–64]. However, below we focus

    on strategies for active targeting of nanotherapeutics to the tumor

    vasculature.

     3.1.1. Angiogenesis

    One of the major angiogenic factors elaborated by tumors– vascular

    endothelial growth factor (VEGF)  –  both stimulates new capillary in-

    growth and increases the permeability of blood vessels  [65]. Targeting

    the VEGF receptor on endothelial cells is therefore an effective method

    to prevent cancer growth by blocking tumor angiogenesis. Targeting

    the VEGF receptor-2 (VEGFR-2) has been investigated as a way to en-hance delivery of variousnanoparticle systems to tumor vessels, includ-

    ing boronated dendrimers and radioisotype-labeled polymerized

    liposomes [66]. VEGF targeting also was accomplished in an animal

    model of hepatic carcinoma using dextran magnetic nanoparticles

    (DMNs) conjugated to radiolabeled 131I-anti-VEGF monoclonal anti-

    bodies [67].

    One of the  rst angiogenic inhibitors TNP-470 demonstrated broad

    spectrum activity in early trials, including ef cacy in metastatic disease

    in human patients, but then stalled in part due to neurotoxicities. A

    reformulation of this angiogenesis inhibitor composed of polymeric

    PEG–PLGA nanoparticles surrounding a TNP-470 core, called Lodamin,

    was shown to prevent the compound from crossing the blood–brain

    barrier, eliminating its neurotoxicity, while retaining its potency and

    broad-spectrum activity [68–70]. The new Lodamin formulation alsogreatly improved oral availability as the polymeric PEG and PLA protect

    the inner active compound from degradation by the acidic stomach

     Table 1

    Nanocarrier-mediated siRNA delivery systems in clinical trials.

    Drug Disease Target Nanoparticle Company Stage Status

    FANG Ovarian tumors FURIN Liposome Gradalis, Inc. Phase II Active

    CALAA-01 Solid tumors RRM Cyclodextrin/PEG, transferrin Calando Pharm Phase I Ongoing

    ALN-VSP02 Solid tumors with liver involvement VEGF, KSP SNALP Alnylam Pharm. Phase I/II Completed

    Atu027 Advanced or metastatic solid tumors PKN3 Liposome Silence Therapeutics Phase I Completed

    siG12D LODER Pancreatic ductal adenocarcinoma KRASG12D Polymer matrix Silenseed Ltd. Phase I Ongoing

    TKM 080301 Solid tumors PLK1 SNALP Tekmira Pharma Phase I Recruiting

    siRNA–EphA2–DOPC Solid tumors EphA2 Liposome M.D. Anderson Cancer Center Phase I Recruiting

    111M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

  • 8/18/2019 Nano Receptors

    6/12

    environment. Lodamin is also under investigation as a therapeutic for

    ophthalmologic diseases of the vasculature including diabetic retinopa-

    thy and age-related macular degeneration.

    Vascular cell adhesion molecule-1 (VCAM-1) is an immunoglobulin-

    like transmembrane glycoprotein receptor whichmediates immune cell

    adhesion that is expressed at high levels on the surfaces of endothelial

    cells in tumor blood vessels. Liposome nanoparticles coated with anti-

    VCAM-1 antibodies selectively target the human tumor vasculature

    in vivo in xenograft studies [71]. Thus, VCAM-1-targeted nanocarrierscould provide a way to both enhance delivery of drugs to growing

    tumor vessels and alter endothelial function.

    Knowledge of the mechanismby which nanoparticles enter vascular

    endothelial cells also has given rise to modications that further en-

    hance targeting. Caveolae-mediated endocytosis is the most prominent

    transendothelial delivery pathway and has attracted interest because it

    has the capability to bypass lysosomes. Surface modications that

    enhance caveolae targeting, such as coating gold nanoparticles with

    an antibody to the caveolae-enriched protein aminopeptidase P, have

    been shown to be effective in concentrating the nanomedicine in lung

    tumor microvasculature [72].

     3.1.2. Hypoxia

    Hypoxia is a hallmark of cancer, which results from rapid tumor cell

    growth that overcomes the rate of vascular ingrowth. Hypoxic regions

    in tumors experience high interstitialpressurebuild up dueto compres-

    sion of blood vessels, poor lymphatic drainage and deterioration of 

    structural features of tissues that support diffusion. Hypoxia is also com-

    monly associated with poor circulation, tumor progression, malignancy

    and multi-drug resistance and hence, the delivery of drugs to hypoxic

    microenvironments remains a signicant clinical challenge in cancer

    therapy [73–75].

    Several nanotherapeutic strategies have been used to develop

    theranostics (combined therapeutics and diagnostics) for hypoxic envi-

    ronments within tumors. For example, a layer-by-layer assembled poly-

    electrolyte nanoparticle has been shown to selective target to hypoxic

    regions of tumors [76]. Targeting is based on the low pH in the hypoxic

    microenvironment, which triggers shedding of the outer layer of the

    nanoparticle, revealing a charged nanoparticle surface that is subse-quently taken up by cells. Hypoxia-activated, near-infrared (NIR)

    light-triggered chitosan nanoparticles also have been developed to

    deliver drugs to cancer cells [77]. This smart nanosystem consists of a

    hypoxic sensor, a photoactivatable trigger, and a caged drug; as it is

    highly specic to the hypoxic tumor microenvironment, its selectivity

    is greatly increased. Polystyrene nanoparticles doped with an oxygen-

    sensitive NIR-emissive palladium meso-tetraphenylporphyrin that se-

    lectively activate and emit optical signals when they experience hypoxic

    conditions also have been developed [78]. In vitro, the nanoparticles

    were easily phagocytosed by murinealveolar macrophages and exhibit-

    ed sensitivity to low oxygen concentrations resulting from induction of 

    hypoxia inducible factor-1α (HIF-1α). Experiments performed in vivo

    with tumor-bearing mice conrmed that these nanoparticles can be

    used as imaging probesfor tumor hypoxia; however, thesame approachpotentially could be leveraged to develop therapeutics.

     3.2. Immune response

    Renewed recognition of the critical role of the immune system in

    cancer has driven the development of immunotherapies that target

    existing tumors and enhance long-term immune surveillance. Nanopar-

    ticle carriers improve specicity of immune-cell targeting, permit

    simultaneous delivery of antigens and immune agents, and enhance

    sustained release.

    Human interleukin 12 (IL-12) induces a potent anti-tumor immune

    response, but systemic administration of IL-12 in early clinical trials

    resulted in serious dose-dependent toxicities, including death. In xeno-

    graft models, intratumoral injection of IL-12 loaded liposomes resulted

    in an enhanced memory T cell response [79]. Importantly, the drug

    did not enter the peripheral blood circulation, suggesting that this

    could help to improve its safety prole [79].

    Biodegradable nanoparticles are emerging as tools for targeting

    tumor antigens to dendritic cells, which is a critical step in eliciting cyto-

    toxic T cell activation for antitumor vaccine ef cacy [80]. Several studies

    have demonstrated the successful use of mannose-grafted nanocarriers

    and an increase in the uptake of these carriers by dendritic cells. For ex-

    ample, PLGA nanoparticles coated with mannan, a natural polymannoseisolated from the cell wall of  Saccharomyces cerevisae, have been shown

    to have a stronger binding af nity and produce increased CD4+ and

    CD8+ T cell responses compared to unmodied nanoparticle surfaces

    [81]. Other particulate vaccines have been successfully developed to spe-

    cically target the mannose receptor, such as mannose–PEG3–NH2-dec-

    orated PLGA mannose [82], mannosylated-gelatin nanoparticles [83] and

    liposomes containing mannosyl lipid derivatives [84]. Targeting antigens

    to C-type lectin receptors also has been shown to result in enhanced hu-

    moral and cellular responses which are suf cient to induce immunity

    against melanomas tumors in vivo [85]. Moreover, similar targeting can

    be accomplished using anti-lectin antibodies instead of lectin ligands

    [86].

    Another attractive property of nanoparticle carriers is thatthey offer

    the potential to deliver both antigenand adjuvant simultaneously to the

    same cell, thus improving the sustained immune response. Several Toll-

    like receptor (TLR) ligands have been tested as adjuvants of cancer vac-

    cines in clinical trials for various cancers, including melanoma, lympho-

    ma, glioblastoma, breast, prostate, ovarian and lung. As some TLR 

    ligands have severe systemic side effects, nanoparticle delivery to the

    desired site of action can greatly improve their safety prole and permit

    use of lower doses. In addition, nanocarriers can improve intracellular

    uptake, thus expanding the range of targeting options to intracellular

    receptor family members such as TLR3, 7, 8 and 9 [87].

    Another approach uses programmable macroporous 3D PLGA scaf-

    folds containing nanoparticles to induce endogenous dendritic cells to

    recognize tumor antigens placed within the scaffold, and to mount a

    systemic anti-cancer response in vivo [88]. The scaffolds contain tumor

    antigens from biopsy samples along with PLGA microspheres that

    release the chemokine GM-CSF to recruit the dendritic cells, andpolyethylenimine (PEI)–CpG cationic nanoparticles containing the im-

    mune adjuvant CpG to stimulate dendritic cell maturation. When im-

    planted in a mice tumor melanoma model, this nanoparticle-enabled

    vaccine resulted in survival of 90% of tumor-bearing animals  [88] and

    intracranial regression of glioma has been demonstrated in rats [89].

    These programmable scaffolds comprising micro and nanoparticles

    show potential in future cancer immune therapy, and human Phase I

    clinical trials were recently initiated with this technology.

    Onenew approach to stimulating the host immuneresponseis using

    light-activated nanoparticles: biodegradable polymer and zinc phthalo-

    cyanine (ZnPc) photosensitizer nanoparticles target the mitochondria

    of cancer cells and subsequent light activation elicits the production of 

    tumor antigen, triggering dendritic cell activation  [90]. These ex vivo

    studies suggest that secreted compounds from mitochondria-targetedand light-activated cancer cells also might be useful as cancer vaccines.

     3.3. Extracellular matrix

    Tumors have a dense ECM consistingof highly interconnected cross-

    linked collagen bers and associated large glycoproteins and proteogly-

    cans. Changes in the structure and mechanical properties of the ECM

    can alter cellular mechanical signaling pathways and thereby inuence

    subsequent tumor progression and metastasis [57,91,92]. For example,

    during tumor progression, overexpression of the ECM-modifying en-

    zyme lysyl oxidase (LOX) results increased collagen cross-linking,

    thereby increasing ECM rigidity and tissue tension  [93,94]. Moreover,

    experimental inhibition of LOX enzyme activity suppressestumor growth

    and prevents cancer progression, and this is mediated by decreasing

    112   M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

  • 8/18/2019 Nano Receptors

    7/12

    collagen cross-linking and restoring more normal-like tissue compliance

    [91,95]. Importantly, when blocking LOX antibodies were delivered on

    biodegradable PLGA-based polymer nanoparticles (~200 nm diameter)

    created using self-assembly, they produced more effective tumor inhibi-

    tion in animals using lower doses, and they had a higher therapeutic

    index (Fig. 4) [96]. Compared to systemic injection of free LOX antibody,

    this LOX antibody-coated nanotherapeutic exhibited signicant

    tumor inhibition with only ~ 1/30th dose normally used for systemic

    administration.Proteoglycans are also a major contributor to ECM mechanics and in

    particular, studies have focused on the roles of hyaluronic acid (HA)and

    the proteoglycan versican in tumor ECM mechanics. Interaction

    between versican with HA and its cell surface receptor, CD44, serves

    to modulate the ECM mechanical environment, thereby altering cancer

    cell proliferation and migration [97,98]. Several studies have utilized

    HA-conjugated nanocarriers to target CD44 over-expressing cancer

    cells and to deliver anti-cancer drug to the tumor site. For example,

    intelligent core-corona HA–PEG–PCL nanoparticles have been used

    to deliver doxorubicin in Ehrlich ascites tumor-bearing mice, and

    HA-decorated PLGA nanoparticles enhance doxorubicin uptake by cul-

    tured human breast adenocarcinoma cells   [99,100]. Ef cient CD44

    targeting also has been accomplished using HA-modied magnetic

    nanocrystals for MRI imaging of breast cancer cells  [101]. Importantly,

    HA is also a viable target for cancer delivery (independently of CD44)

    because it is often over-expressed during tumor progression. Hyaluron-

    idase liposomes disrupt the HA matrix when injected intravenously in a

    human osteosarcoma solid tumor model [102] and silica nanoparticles

    coated with hyaluronidase enzyme have been used to deliver adjuvantdrugs to treat skin cancer [103].

    Apart from activation of chemical signaling, the tumor stroma phys-

    ically limits the penetration of nanotherapeutics and molecular drugs

    into the tumor. For this reason, co-delivery of proteases, such as collage-

    nase, gelatinase and hyaluronidase, signicantly aids in breakdown of 

    the dense ECM barrier and thereby, increases transport of nanoparticles

    into the tumor and enhances targeted delivery [1,104,105]. Other en-

    zymes (e.g., transglutaminase) and proteins (e.g., the proteoglycans

    lumican, decorin and biglycan) also may play a role in modifying tumor

    ECM properties and inuencing cancer development [106–111].  For

    Self-assembly in

    aqueous solutionPLGA-b-PEG-COOH

    A

    LOXAb

    EDC/NHS

    PLGA-b-PEG-COOHNanoparticle (NP)

    O OH

    O

    r

    OO H

    O

    r

    LOXAbNP

    LOX

    Untreated

    AbNP

    O

    OOr N

    O

    OO

    OOr N

    O

    O

    Or

    O

    LOX AbNH

    O O H

    O

    rPLGA

    500 nm

    1

    2

    0

       T  u  m  o  r   S   i  z  e   (  x   1   0   3   m  m

       3   )

    Days

    5 8 122

      Untreated Tumor

    LOXAbNP treated Tumor

    0 100 10000

    10

    20

    30

       I  n   t  e  n  s   i   t  y   % 

    Size (nm)

    B

    C

    Fig. 4. A) Schematic of the process by which lysyl oxidase-antibody (LOX-ab) coated PLGA nanoparticles are formed by solvent displacement, aqueous self-assembly and surface conju-

    gation. B) Size characterization of the LOX-ab nanoparticles using transmission electron microscopy (TEM) (left) and dynamic light scattering (DLS) (right) con rming that their size is

    ~200nm indiameter andthatthey aresphericalin form.C) Thetherapeuticpotential ofthese nanoparticleswas demonstratedby injectingthe LOX-abcoated nanoparticlesintravenously

    in a mammary tumor xenograft model; this resulted in signicant inhibition of tumor growth when analyzed over time (left) or by visual inspection at the end of the experiment after

    removal of the tumors (right) (adapted with permission from Kanapathipillai et al.  [96]).

    113M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

    http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80http://localhost/var/www/apps/conversion/tmp/scratch_5/image%20of%20Fig.%E0%B4%80

  • 8/18/2019 Nano Receptors

    8/12

    example, macromolecular drugs (e.g., FITC-dextran, FITC-albumin) pene-

    trate more deeply into tumor tissue after degradation of collagen and

    decorin in the ECM [112]. Thus, nanoparticle-based strategies to modu-

    late these ECM enzymes and protein functions, will likely improve cancer

    targeting specicity, drug diffusion and accumulation, as well as drug re-

    tention at the tumor site.

    The ECM is a dynamic structure as it is maintained through ongoing

    coupling between synthetic and degradative mechanisms; however,

    ECM remodeling becomes deregulated during tumor progression [57]and this is mediated by elevated expression and activity of various pro-

    teolytic enzymes, including matrix metalloproteinases (MMPs) and

    urokinase-based plasminogen activator systems [113,114]. Interesting-

    ly, several nanoparticle-based theranostics have been designed to both

    monitor MMP expression and inhibit their activity in the tumor micro-

    environment. For example, liposomes modied with the GPLPLR pep-

    tide sequence to target MT1-MMP showed higher binding ef ciency

    and higher therapeutic ef ciency in tumors than uncoated liposomes

    [115]. Nanoparticle-based prodrug strategies also have been developed

    by taking advantage of MMP-sensitive activation mechanisms which

    are selectively triggered in the tumor microenvironment. An albumin-

    binding doxorubicin prodrug that is sensitive to MMP2 has been

    shown to be effective in malignant melanoma [116]. Gold nanoparticles

    conjugated to MMP-sensitive peptides also have been used in tumor

    imaging and drug delivery   [117,118]. Recently, MMP-2-sensitive

    denuding of gold nanoparticles in human breast adenocarcinoma cells

    has been shown to improve their uptake and hence, enhance targeted

    delivery to tumor cells [119]. Tumor-triggered release of doxorubicin

    from MMP-sensitive gold nanoparticles also has been demonstrated in

    a SSC-7 subcutaneous tumor model [120].

    The serine protease inhibitor, urokinase (uPA), plays a major role in

    tumor cell invasion and metastasis and uPA-targeting nanoparticles

    have been developed for tumor targeting and imaging. Magnetic nano-

    particles coated with uPA receptor (uPAR)-targeting peptides have

    higher rates of uptake in uPAR positive cancer cells compared with un-

    coated particles [121], and uPA-targeting iron oxide nanoparticles have

    been used for in vivo imaging of breastcancer [122]. Thesenanoparticles

    were coated with a recombinant peptide of the amino terminal frag-

    ment of uPA, which targets the uPAR that is overly expressed in thebreast cancer tissue microenvironment.

    Nanoparticles or small molecule drug carriers also have been de-

    veloped to target tumor support structures. The cyclic decapeptide,

    CGLIIQKNEC, which is known to bind  bronectin–brin complexes

    in the tumor ECM, has been conjugated to gadolinium-containing

    dendrimers for targeting breast tumor xenografts [123]. The tumor

    stromaalso containsan abnormal meshwork of clotted plasma proteins,

    such as brin. The plasma clot-binding peptide, CREKA, which was se-

    lected using phage display [124] has been used for targeted delivery

    to the brin mesh in tumor stroma [125]. Nanoparticles and liposomes

    were conjugated to the CREKA peptide and shown to both target to the

    tumor stroma and also to induce self-triggered clotting.

     3.4. Multi-drug resistance

    Nanoparticles also have the potential to overcome multi-drug resis-

    tance (MDR) that can severely restrict the effectiveness of conventional

    chemotherapeutic drugs in the tumor microenvironment [126]. Due to

    non-specic and ligand receptor-mediated targeted uptake, nano-

    particles are able to bypass ef ux by ABC transporter proteins and

    accumulate intracellularly; this is in contrast to free drugs which

    can be highly susceptible to ABC transporter P-glycoprotein (P-gp)

    capture and ef ux  [127,128]. For instance, P-gp ef ux and multi-

    drug resistance could be overcome in cultured P388 leukemia cells

    using doxorubicin-loaded poly(alkyl-cyanoacrylate) nanoparticles

    [129,130]. In murine cancer models, these nanoparticles circulated

    longer and displayed increased accumulation in tumor tissue when

    coated with PEG-modied drug.

    Another strategy to overcome MDR is to exploit the cell-surface re-

    ceptor binding strategies of nanoparticles. Studies using transferrin-

    conjugated nanoparticles or liposomes-containing paclitaxel and

    oxaliplatin, as well as pH-stimuli-responsive polymeric micelles contain-

    ing doxorubicin, have all shown less development of drug-resistance in

    tumor mouse models compared to free drug [20,34,131]. Other recent

    studies on MDR-modifying nanoparticles with increased therapeutic ef-

    cacy include doxorubicin-loaded lipid nanoparticles in human breast

    and ovarian cancer cells [132], anti-P-glycoprotein-conjugated PEG–

    PLGA, and poloxamer-modied PLGA nanoparticles for the targeted de-

    livery drugs to cervical carcinoma cells over-expressing P-gp [133].

    4. Challenges for the future

    While there have many great advances in the area of nanocarrier-

    based drug delivery over the past few decades, many challenges remain

    for the design of effective nanotherapeutics for clinical cancer therapy.

    Nanomaterials that target specically to the cancer cell or the tumor

    microenvironment, but do not exhibit long-term toxicity and can be

    effectively cleared from the circulation and removed from the body,

    without compromising circulation time or bioavailability must be de-

    veloped [134]. A deeper understanding of the complexities involved in

    cancer cell physiology and the tumor microenvironment, as well as

    drug and particle pharmacokinetics are key for the development of 

    successful new cancer therapeutics. It will be crucial to develop smart

    nanomaterials that improve stability, extend circulation time, enhance

    targeting and improve retention at the tumor site, while minimizing

    short- and long-term systemic toxicities. Emerging nanoparticle

    technologies for the delivery of siRNA, anti-metastatic agents, and

    multi-resistant drugs [8,135,136] show promise, and demonstrate

    the ability to overcome many of the roadblocks that have limited

    the effectiveness of conventional drug delivery approaches. More re-

    cent identication of CD47 as a  “marker of self ” whichhelps to evade

    phagocytosis uptake of nanoparticles by macrophages, and is also

    overexpressed in several solid tumors [137,138], raises the possibil-

    ity of further increasing the effectiveness of nanocarrier drug deliv-

    ery systems. In fact, nanobead circulation times and therapeutic

    ef cacy were shown to be signicantly enhanced in lung carcinoma-bearing mice when the nanoparticles were decorated with CD47 or

    anti-CD47 antibodies [138].

    Another emerging technology in nanotherapeutics is the develop-

    ment of DNA-based nanostructures that form through programmable

    self-assembly [139,140]. The ability to create virtually any 3D shape

    using this approach provides a potentially powerful new way to

    tailor nanoparticle design to optimize its function. For example, self-

    programmable DNA nanorobots that are capable of selectively deliver-

    ing anti-cancer payloads (ligands for apoptosis-inducing receptors)

    only when they bind to specic types of cancer cells (Fig. 5) were re-

    cently developed [141]. Exploration of the value of this approach for

    treatment of human cancer will require demonstration of biocompati-

    bility and stability of these materials when injected  in vivo. However,

    the range of design control and the programmability of these nanoscalerobots are huge, and hence, this area will be interesting to watch over

    the next decade.

    Further, in vitro tumor engineering and tumor organ on a chip tech-

    nologies could serve in optimal design and screening of nanoparticles

    prior to preclinical studies. For example, the potential of using tumor

    spheroids for drugscreening and delivery to pathological tissues has re-

    cently been reported [142]. Spheroids resemble the 3D architecture of 

    tumor tissues, mimicking cell–cell interactions and diffusional limita-

    tions of mass anduid transport, and therefore represent a moresuitable

    platform for testing drugs and nanotherapeutics than conventional pla-

    nar culture substrates [142,143]. A microuidic tumor culture device or

    “tumor-on-a-chip”model, alsohas beendeveloped to study nanoparticle

    transport in tissues  [144]. Tumor-like spheroids were incorporated

    into the microuidic channels and accumulation of nanoparticles was

    114   M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

  • 8/18/2019 Nano Receptors

    9/12

    measured in real-time at physiological blood ow rates. In a further ad-

    vancement of this organ-on-a-chip approach, the in vivo microenviron-ment of a breathing human lung alveolar-capillary interface was

    recreated in a microuidic chip [145], and this platform was shown to

    be effective at detecting both physiological nanoparticle absorption and

    relevant toxicities  in vitro [146]. Hence, these organ-on-chip devices

    might be used in the future as a replacement for animal studies for

    rapid testing nanotherapeutic ef cacy as well as toxicity. In this manner,

    emerging tumor engineering technologies may synergize with new ad-

    vances in nanobiotechnology to accelerate the development of more ef-

    fective nanocarriers while reducing both the time and cost of 

    investigations required to meet these goals.

    However, to develop truly effective cancer nanotherapeutics, we

    have to focus beyond the design of the nanoparticle itself and instead,

    learn more about the process of cancer formation and progression as

    well as how we might more selectively interfere with, or prevent,these critical processes. For example, as it is becoming increasingly

    clear that cancer is not merely a disease of uncontrolled cell growth,

    and instead that it is a disease of normal tissue development  [26],

    then we might think to explore ways to harness knowledge about de-

    velopment control to create new cancer therapeutics. In this context,

    it is interesting that developmental biology studies have shown that

    when ECM isolated from embryonic tissue is combined with adult epi-

    thelial cancers, it is capable of normalizing the cancer by suppressing

    growth and stimulating tissue-specic differentiation [147,148]. Thus,

    development of biomimetic nanomaterials that mimic the inductive

    cues found within embryonic ECM could lead to the development of 

    novel nanotherapeutics that might normalize the cancer tissue micro-

    environment rather than simply kill all growing cancer cells [26]. Inter-

    estingly, molecules within embryonic ECM that are responsible for this

    type of cancer-normalizing activity (e.g., biglycan) are beginning to be

    identied [111]. Given the key role of ECM mechanics in cancer devel-opment, it also might be possible to design nanotherapeutics that pre-

    vent tumor growth or normalize the cancer phenotype by selectively

    modulating the mechanical or structural properties of the tumor micro-

    environment [26]. This could be facilitated by development of smart

    nanotechnologies that have programmable mechanical properties, or

    that can be triggered to alter their physical properties by microenviron-

    mentalcues, as is possible,for example, with self assemblingDNA nano-

    structures [149].

    In conclusion, it is clear that simultaneously advancements in geno-

    mic, proteomic and nanotechnology elds have enabled many types of 

    new nanotherapeutics to be developed that offer the potential to trans-

    form cancer therapy. The newest wave of nanomaterials are multi-

    functional, programmable, and even self-assembling; they also offer

    ways to provide longer circulation times, higher selectivity, and moreeffective targeting to tumor sites. The recent clinical approval of the

    liposomal-doxorubicin nanoparticles, Doxil and Abraxane [1,11], has

    validated the value of nanotechnology for cancer treatment. However,

    the future is even brighter, and the continuing exciting advances in de-

    velopment nanoparticle-based therapeutics will hopefully attract many

    new investigators in various disciplines into this exciting interdisciplin-

    ary eld.

     Acknowledgments

    We thank K. Johnson for help with the graphics. This work was sup-

    ported by the Wyss Institute for Biologically Inspired Engineering at

    Harvard University, and grants from the Department of Defense

    (W81XWH-08-1-0659 and W81XWH-10-1-0565).

    Fig. 5. A) Schematic depicting self-programmable DNAnanorobots loadedwith antibody payload, in lockedposition(left) andthe ligandlocking mechanism(middle) andopen position of 

    nanorobot afterligand–receptor interactionexposingthe payload. B) Eightdifferentnanorobots were loadedwithuorescentlylabeled anti-HLA-A/B/C antibody fragments andtheir bind-

    ing specicity was tested in 6 different cancercell types expressing various combinations of apoptosis-inducing receptors. The histograms show count of cells versus uorescence due to

    anti-HLA-A/BC labeling (reproduced with permission from Ref.  [141]).

    115M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

  • 8/18/2019 Nano Receptors

    10/12

    References

    [1]   T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Drug targeting to tumors: princi-ples, pitfalls and (pre-) clinical progress, J. Control. Release 161 (2012) 175–187.

    [2]   Y. Ding, S. Li, G. Nie, Nanotechnological strategies for therapeutic targeting of tumor vasculature, Nanomedicine (London) 8 (2013) 1209–1222.

    [3]   S. Nie, Y. Xing, G.J. Kim, J.W. Simons, Nanotechnology applications in cancer, Annu.Rev. Biomed. Eng. 9 (2007) 257–288.

    [4]   F. Alexis, E.M. Pridgen, R. Langer, O.C. Farokhzad, Nanoparticle technologies forcancer therapy, Handb. Exp. Pharmacol. (2010) 55–86.

    [5]   T.M. Allen, Ligand-targeted therapeutics in anticancer therapy, Nat. Rev. Cancer 2(2002) 750–763.[6]   A. Ozcelikkale, S. Ghosh, B. Han, Multifaceted transport characteristics of 

    nanomedicine: needs for characterization i n dynamic environment, Mol. Pharm.10 (2013) 2111–2126.

    [7]  L.Y. Rizzo, B. Theek, G. Storm, F. Kiessling, T. Lammers, Recent progress innanomedicine: therapeutic, diagnostic and theranostic applications, Curr. Opin.Biotechnol. 24 (2013) 1159–1166.

    [8]   Y. Matsumura, K. Kataoka, Preclinical and clinical studies of anticancer agent-incorporating polymer micelles, Cancer Sci. 100 (2009) 572–579.

    [9]   W.J. Gradishar, S. Tjulandin, N. Davidson, H. Shaw, N. Desai, P. Bhar, M. Hawkins, J.O'Shaughnessy, Phase III trial of nanoparticle albumin-bound paclitaxel comparedwith polyethylated castoroil-based paclitaxel in women with breast cancer, J. Clin.Oncol. 23 (2005) 7794–7803.

    [10] R.D. Hofheinz,S.U. Gnad-Vogt, U. Beyer, A. Hochhaus,Liposomalencapsulatedanti-cancer drugs, Anticancer Drugs 16 (2005) 691–707.

    [11]   F. Danhier, O. Feron, V. Preat, To exploit the tumor microenvironment: passive andactive tumor targeting of nanocarriers for anti-cancer drug delivery, J. Control. Re-lease 148 (2010) 135–146.

    [12]   P. Decuzzi, S. Lee,B. Bhushan, M. Ferrari, A theoretical model forthe margination of particles within blood vessels, Ann. Biomed. Eng. 33 (2005) 179–190.

    [13]   M. Tiwari, Nano cancer therapy strategies, J. Cancer Res. Ther. 8 (2012) 19–22.[14]   T.M. Allen, P. Sapra, E. Moase, J. Moreira, D. Iden, Adventures in targeting, J. Lipo-

    some Res. 12 (2002) 5–12.[15]   F. Alexis, J.W. Rhee, J.P. Richie,A.F. Radovic-Moreno, R. Langer,O.C. Farokhzad, New

    frontiers in nanotechnology for cancer treatment, Urol. Oncol. 26 (2008) 74–85.[16]   J.L. Perry, K.P. Herlihy, M.E. Napier, J.M. Desimone, PRINT: a novel platform toward

    shape and size specic nanoparticle theranostics, Acc. Chem. Res. 44 (2011)990–998.

    [17]   P. Kolhar, A.C. Anselmo, V. Gupta, K. Pant, B. Prabhakarpandian, E. Ruoslahti, S.Mitragotri, Using shape effects to target antibody-coated nanoparticles to lungand brain endothelium, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 10753–10758.

    [18]   R.H. Fang, C.M. Hu, K.N. Chen, B.T.Luk, C.W. Carpenter, W. Gao, S. Li, D.E. Zhang,W.Lu, L. Zhang, Lipid-insertion enables targeting functionalization of erythrocytemembrane-cloaked nanoparticles, Nanoscale 5 (2013) 8884–8888.

    [19]   Y. Matsumura, M. Gotoh, K. Muro, Y. Yamada, K. Shirao, Y. Shimada, M. Okuwa, S.Matsumoto, Y. Miyata, H. Ohkura, K. Chin, S. Baba, T. Yamao, A. Kannami, Y.Takamatsu, K. Ito, K. Takahashi, Phase I and pharmacokinetic study of MCC-465,a doxorubicin(DXR) encapsulated in PEG immunoliposome, in patients with met-astatic stomach cancer, Ann. Oncol. 15 (2004) 517–525.

    [20]   R. Suzuki,T. Takizawa, Y. Kuwata, M. Mutoh,N. Ishiguro, N. Utoguchi, A. Shinohara,M. Eriguchi, H. Yanagie, K. Maruyama, Effective anti-tumor activity of oxaliplatinencapsulated in transferrin-PEG-liposome, Int. J. Pharm. 346 (2008) 143–150.

    [21]   Y.C. Chen, C.L. Lo, G.H. Hsiue, Multifunctional nanomicellar systems for deliveringanticancer drugs, J. Biomed. Mater. Res. A 102 (2013) 2024–2038.

    [22]   A.A. Alexander-Bryant, W.S. Vanden Berg-Foels, X. Wen, Bioengineering strategiesfor designing targeted cancer therapies, Adv. Cancer Res. 118 (2013) 1–59.

    [23]   R. Cheng, F. Meng, C. Deng, H.A. Klok, Z. Zhong, Dual and multi-stimuli responsivepolymeric nanoparticles for programmed site-specic drug delivery, Biomaterials34 (2013) 3647–3657.

    [24]   D.E. Ingber, Tensegrity II. How structural networks inuence cellular informationprocessing networks, J. Cell Sci. 116 (2003) 1397–1408.

    [25]   H. Fang, Y.A. Declerck, Targeting the tu mor microenvironment: from understand-ing pathways to effective clinical trials, Cancer Res. 73 (2013) 4965–4977.

    [26]   D.E. Ingber, Can cancer be reversed by engineer ing the tumor microenvironment?Semin. Cancer Biol. 18 (2008) 356–364.

    [27]   M. Allinen, R. Beroukhim, L. Cai, C. Brennan, J. Lahti-Domenici, H. Huang, D. Porter,M. Hu,L. Chin,A. Richardson,S. Schnitt,W.R. Sellers,K. Polyak, Molecular character-ization of the tumor microenvironment in breast cancer, Cancer Cell 6 (2004)17–32.

    [28]   M.M. Mueller, N.E. Fusenig, Friends or foes  —  bipolar effects of the tumour stromain cancer, Nat. Rev. Cancer 4 (2004) 839–849.

    [29]   M.A. Phillips, M.L. Gran, N.A. Peppas, Targeted nanodelivery of drugs and diagnos-tics, Nano Today 5 (2010) 143–159.

    [30]   F.X. Gu, R. Karnik, A.Z. Wang, F. Alexis, E. Levy-Nissenbaum, S. Hong, R.S .Langer, O.C. Farokhzad, Targeted nanoparticles for cancer therapy, NanoToday 2 (2007) 14–21.

    [31]   A. Martinez, R. Olmo, I. Iglesias, J.M. Teijon, M.D. Blanco, Folate-targeted nanoparticlesbased on albumin and albumin/alginate mixtures as controlled release systems of ta-moxifen: synthesis and in vitro characterization, Pharm. Res. 31 (2013) 182–193.

    [32]   J. Lu, Z. Li, J.I. Zink, F. Tamanoi, In vivo tumor suppression ef cacy of mesoporoussilica nanoparticles-based drug-delivery system: enhanced ef cacy by folate mod-ication, Nanomedicine 8 (2012) 212–220.

    [33]  P. Ponka, C.N. Lok, The transferrin receptor: role in health and disease, Int. J.Biochem. Cell Biol. 31 (1999) 1111–1137.

    [34]   S.K. Sahoo, W. Ma, V. Labhasetwar, Ef cacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer, Int. J. Cancer 112(2004) 335–340.

    [35]   J. Chang, A. Paillard, C. Passirani, M. Morille, J.P. Benoit, D. Betbeder, E. Garcion,Transferrin adsorption onto PLGA nanoparticles governs their interaction with bi-ological systemsfrom blood circulationto brain cancercells,Pharm. Res.29 (2012)1495–1505.

    [36]   M.A. Sandoval, B.R. Sloat, P.D. Lansakara, A. Kumar, B.L. Rodriguez, K. Kiguchi, J.Digiovanni, Z. Cui, EGFR-targeted stearoyl gemcitabine nanoparticles show en-hanced anti-tumor activity, J. Control. Release 157 (2012) 287–296.

    [37]   M.G. Anhorn, S. Wagner, J. Kreuter, K. Langer, H. von Briesen, Specic targeting of 

    HER2 overexpressing breast cancer cells with doxorubicin-loaded trastuzumab-modied human serum albumin nanoparticles, Bioconjug. Chem. 19 (2008)2321–2331.

    [38]   H. Meng, M. Liong, T. Xia, Z. Li, Z. Ji, J.I. Zink, A.E. Nel, Engineereddesign of mesopo-rous silica nanoparticles to deliver doxorubicin and P-glycoprotein siRNA to over-come drug resistance in a cancer cell line, ACS Nano 4 (2010) 4539–4550.

    [39]   F. Wang, Y.C. Wang, S. Dou, M.H. Xiong, T.M. Sun, J. Wang, Doxorubicin-tetheredresponsive gold nanoparticles facilitate intracellular drug delivery for overcomingmultidrug resistance in cancer cells, ACS Nano 5 (2011) 3679–3692.

    [40]  S. Marrache, S. Dhar, Engineering of blended nanoparticle platform for delivery of mitochondria-acting therapeutics, Proc. Natl. Acad. Sci. U. S. A. 109 (2012)16288–16293.

    [41]  R.A. Smith,C.M. Porteous, A.M. Gane, M.P. Murphy, Delivery of bioactive moleculesto mitochondria in vivo, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 5407–5412.

    [42]   S.S. Bale, S.J. Kwon, D.A. Shah, A. Banerjee, J.S. Dordick, R.S. Kane, Nanoparticle-mediated cytoplasmic delivery of proteins to target cellular machinery, ACSNano 4 (2010) 1493–1500.

    [43]   P.S. Lai, C.L. Pai, C.L. Peng, M.J. Shieh, K. Berg, P.J. Lou, Enhanced cytotoxicity of saporin by polyamidoamine dendrimer conjugation and photochemical internali-

    zation, J. Biomed. Mater. Res. A 87 (2008) 147–155.[44]   F. Heitz,M.C. Morris, G. Divita,Twenty years of cell-penetrating peptides: from mo-

    lecular mechanisms to therapeutics, Br. J. Pharmacol. 157 (2009) 195–206.[45]   D.H. Yu, Q. Lu, J. Xie, C. Fang, H.Z. Chen, Peptide-conjugated biodegradable nano-

    particles as a carrier to target paclitaxel to tumor neovasculature, Biomaterials 31(2010) 2278–2292.

    [46]   B.Y. Kim, W. Jiang, J. Oreopoulos, C.M. Yip, J.T. Rutka, W.C. Chan, Biodegradablequantum dot nanocomposites enable live cell labeling and imaging of cytoplasmictargets, Nano Lett. 8 (2008) 3887–3892.

    [47]  S.E. Gratton, P.A. Ropp, P.D. Pohlhaus, J.C. Luft, V.J. Madden, M.E. Napier, J.M.DeSimone, The effect of particle design on cellular internalization pathways,Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 11613–11618.

    [48]   S. Barua, J.W. Yoo, P. Kolhar, A. Wakankar, Y.R. Gokarn, S. Mitragotri, Particle shapeenhances specicityof antibody-displaying nanoparticles, Proc. Natl. Acad. Sci.U. S.A. 110 (2013) 3270–3275.

    [49]   R. Agarwal, V. Singh, P. Jurney, L. Shi, S.V. Sreenivasan, K. Roy, Mammalian cellspreferentially internalize hydrogel nanodiscs over nanorods and use shape-specic uptakemechanisms, Proc. Natl. Acad. Sci.U. S. A. 110(2013) 17247–17252.

    [50]   M.E. Davis, J.E. Zuckerman, C.H. Choi, D. Seligson, A. Tolcher, C.A. Alabi, Y. Yen, J.D.Heidel, A. Ribas, Evidence of RNAi in humans from systemically administeredsiRNA via targeted nanoparticles, Nature 464 (2010) 1067–1070.

    [51]   Y.H. Huang, Y. Bao, W. Peng, M. Goldberg, K. Love, D.A. Bumcrot, G. Cole, R. Langer,D.G. Anderson, J.A. Sawicki, Claudin-3 gene silencing with siRNA suppresses ovariantumor growth and metastasis, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 3426–3430.

    [52]   M.S. Goldberg, D. Xing, Y. Ren, S. Orsulic, S.N. Bhatia, P.A. Sharp, Nanoparticle-mediated delivery of siRNA targeting Parp1 extends survival of mice bearing tu-mors derived from Brca1-decient ovarian cancer cells, Proc. Natl. Acad. Sci. U. S.A. 108 (2011) 745–750.

    [53]   M.S. Goldberg, P.A. Sharp, Pyruvate kinase M2-specic siRNA induces apoptosisand tumor regression, J. Exp. Med. 209 (2012) 217–224.

    [54]   A. Brock, S. Krause, H. Li, M. Kowalski, M.S. Goldberg, J.J.Collins, D.E. Ingber, Silenc-ing HoxA1 by intraductal injection of siRNA lipidoid nanoparticles prevents mam-mary tumor progression in mice, Sci. Transl. Med. 6 (2014) 217ra212.

    [55]   G. Sahay, W. Querbes, C. Alabi, A. Eltoukhy, S. Sarkar, C. Zurenko, E. Karagiannis, K.Love, D. Chen, R. Zoncu, Y. Buganim, A. Schroeder, R. Langer, D.G. Anderson, Ef -ciency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling,Nat. Biotechnol. 31 (2013) 653–658.

    [56]   N.E. Sounni, A. Noel, Targeting the tumor microenvironment for cancer therapy,Clin. Chem. 59 (2013) 85–93.

    [57]   D.E. Ingber, J.A.Madri,J.D. Jamieson, Role of basallamina in neoplastic disorganiza-tion of tissue architecture, Proc. Natl. Acad. Sci. U. S. A. 78 (1981) 3901–3905.

    [58]   P. Lu, V.M. Weaver, Z. Werb, The extracellular matrix: a dynamic niche in cancerprogression, J. Cell Biol. 196 (2012) 395–406.

    [59]   J. Zhang, J. Liu, Tumor stroma as targets for cancer therapy, Pharmacol. Ther. 137(2013) 200–215.

    [60]   H. Maeda, The enhanced permeability andretention(EPR) effect in tumor vascula-ture: the key role of tumor-selective macromolecular drug targeting, Adv. Enzym.Regul. 41 (2001) 189–207.

    [61]   V.P. Torchilin, Recent advances with liposomes as pharmaceutical carriers, Nat.Rev. Drug Discov. 4 (2005) 145–160.

    [62]   V.P. Torchilin, Targeted pharmaceutical nanocarriers for cancer therapy and imag-ing, AAPS J. 9 (2007) E128–E147.

    [63]   J.K. Vasir, V. Labhasetwar, Targeted drug delivery in cancer therapy, Technol.Cancer Res. Treat. 4 (2005) 363–374.

    [64]   S. Parveen, S.K. Sahoo, Polymeric nanoparticles for cancer therapy, J. Drug Target.16 (2008) 108–123.

    116   M. Kanapathipillai et al. / Advanced Drug Delivery Reviews 79–80 (2014) 107 –118

    http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0005http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0005http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0005http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0005http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0010http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0010http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0010http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0010http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0015http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0015http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0015http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0015http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0020http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0020http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0020http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0020http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0025http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0025http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0025http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0025http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0030http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0030http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0030http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0030http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0030http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0730http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0730http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0730http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0730http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0730http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0040http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0040http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0040http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0040http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0045http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0050http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0050http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0050http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0050http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0055http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0055http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0055http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0055http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0055http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0060http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0060http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0060http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0060http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0065http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0065http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0065http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0070http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0070http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0070http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0070http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0075http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0075http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0075http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0075http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0080http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0085http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0085http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0085http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0085http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0085http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0090http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0090http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0090http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0090http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0090http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0095http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0100http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0100http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0100http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0100http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0100http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0735http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0735http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0735http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0735http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0110http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0110http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0110http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0110http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0115http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0120http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0125http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0125http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0125http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0125http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0130http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0130http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0130http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0130http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0135http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0140http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0145http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0145http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0145http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0145http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0150http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0150http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0150http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0150http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0150http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0740http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0740http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0740http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0740http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0740http://refhub.elsevier.com/S0169-409X(14)00099-4/rf0160http://refhub.elsevier.com/S016