14
381 REVIEW Immunotherapy (2011) 3(3), 381–394 ISSN 1750-743X 10.2217/IMT.11.5 © 2011 Future Medicine Ltd Antibody-targeted nanoparticles for cancer therapy Despite the advances in our understanding of the molecular and cellular basis of cancer, con- ventional front line cancer treatment remains centered on systemic chemotherapy and radio- therapy. These regimes are plagued by broad distribution, frequently resulting in suboptimal dosing of the tumor and/or induction of toxic side effects in normal tissues [1–3] . These issues have also inhibited the development of new drugs, where experimental agents have demon- strated exciting efficacies in vitro, but have failed in their ability to alleviate the disease in vivo. This can be due to inadequate dosing of the target area and drug accumulation in normal tissue [4–6] . The improved formulation and delivery of chemotherapies has the potential to address these current hurdles in the effective drug- ging of the diseased tissue. The application of nanoparticle (NP) delivery systems has been extensively studied for a range of drug molecules, with various perimeters such as size, entrap- ment efficiencies and release profiles examined in depth. Indeed, nanoparticulate formula- tions of daunorubicin (DaunoXome ® ; NeXstar Pharmaceuticals, CO, USA) and doxorubicin (Doxil ® /Caelyx ® , Janssen, UK and Myocet ® , Sopherion Therapeutics, Inc., NJ, USA) are used clinically to treat breast cancer and Kaposi’s sarcoma [7] . Although the use of NP formulations has improved drug bioavailability, targeting of these entities has the potential to further improve both the therapeutic effectiveness of drug compounds at the disease site whilst also reducing off-target effects. This targeting can be achieved through the coating of a broad range of molecules on the surface of the NP to achieve targeting and local- ization of the drug conjugate. In the following sections, the main classes of NP are briefly dis- cussed before a more in-depth review of the cur- rent progress with targeting strategies, focusing in particular on antibodies, are examined. Nanoparticle fabrication diversity Gold nanoparticles The therapeutic application of gold-based NPs has been documented in the treatment of rheumatoid arthritis since the 1930s and has been extensively investigated in diagnostic imag- ing and therapeutic applications. Gold NP-based diagnostic strategies have been developed, exploiting gold interaction with near infrared radiations, based on surface plasmon scatter- ing, or surface plasmon resonance, which can be used to detect NPs at concentrations as low as 10 –16 M [8] . For therapeutic purposes, gold NPs have been used as photosensitizers, emitting heat to bring about cell death when excited by minimal near infrared radiation [9] . Research into the use of gold-based NPs has examined different compositions and shapes, such as the conventional nanospheres, gold nanoshells (composed of a silica core enclosed in a thin layer of gold) [10] and gold nano- rods [11,12] . Each of these particles exploits the physiochemical properties of elemental gold [13] . Gold is resistant to oxidative corrosion and In recent years, nanoparticulate-mediated drug delivery research has examined a full spectrum of nanoparticles that can be used in diagnostic and therapeutic cancer applications. A key aspect of this technology is in the potential to specifically target the nanoparticles to diseased cells using a range of molecules, in particular antibodies. Antibody–nanoparticle conjugates have the potential to elicit effective targeting and release of therapeutic targets at the disease site, while minimizing off-target side effects caused by dosing of normal tissues. This article provides an overview of various antibody-conjugated nanoparticle strategies, focusing on the rationale of cell-surface receptors targeted and their potential clinical application. KEYWORDS: anticancer drug delivery immunotargeting liposome nanoparticle quantum dot receptor-mediated endocytosis Francois Fay 1 & Christopher J Sco †1 1 School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK Author for correspondence: Tel.: +44 289 097 2350 [email protected] For reprint orders, please contact: [email protected]

Antibody-targeted nanoparticles for cancer therapy

Embed Size (px)

Citation preview

Page 1: Antibody-targeted nanoparticles for cancer therapy

381

Review

Immunotherapy (2011) 3(3), 381–394 ISSN 1750-743X10.2217/IMT.11.5 © 2011 Future Medicine Ltd

Antibody-targeted nanoparticles for cancer therapy

Despite the advances in our understanding of the molecular and cellular basis of cancer, con-ventional front line cancer treatment remains centered on systemic chemotherapy and radio-therapy. These regimes are plagued by broad distribution, frequently resulting in suboptimal dosing of the tumor and/or induction of toxic side effects in normal tissues [1–3]. These issues have also inhibited the development of new drugs, where experimental agents have demon-strated exciting efficacies in vitro, but have failed in their ability to alleviate the disease in vivo. This can be due to inadequate dosing of the target area and drug accumulation in normal tissue [4–6].

The improved formulation and delivery of chemotherapies has the potential to address these current hurdles in the effective drug-ging of the diseased tissue. The application of nanoparticle (NP) delivery systems has been extensively studied for a range of drug molecules, with various perimeters such as size, entrap-ment efficiencies and release profiles examined in depth. Indeed, nanoparticulate formula-tions of daunorubicin (DaunoXome®; NeXstar Pharmaceuticals, CO, USA) and doxorubicin (Doxil®/Caelyx®, Janssen, UK and Myocet®, Sopherion Therapeutics, Inc., NJ, USA) are used clinically to treat breast cancer and Kaposi’s sarcoma [7].

Although the use of NP formulations has improved drug bioavailability, targeting of these entities has the potential to further improve both the therapeutic effectiveness of drug compounds

at the disease site whilst also reducing off-target effects. This targeting can be achieved through the coating of a broad range of molecules on the surface of the NP to achieve targeting and local-ization of the drug conjugate. In the following sections, the main classes of NP are briefly dis-cussed before a more in-depth review of the cur-rent progress with targeting strategies, focusing in particular on antibodies, are examined.

Nanoparticle fabrication diversity � Gold nanoparticles

The therapeutic application of gold-based NPs has been documented in the treatment of rheuma toid arthritis since the 1930s and has been extensively investigated in diagnostic imag-ing and therapeutic applications. Gold NP-based diagnostic strategies have been developed, exploiting gold interaction with near infrared radiations, based on surface plasmon scatter-ing, or surface plasmon resonance, which can be used to detect NPs at concentrations as low as 10–16 M [8]. For therapeutic purposes, gold NPs have been used as photosensitizers, emitting heat to bring about cell death when excited by minimal near infrared radiation [9].

Research into the use of gold-based NPs has examined different compositions and shapes, such as the conventional nanospheres, gold nanoshells (composed of a silica core enclosed in a thin layer of gold) [10] and gold nano-rods [11,12]. Each of these particles exploits the physiochemical proper ties of elemental gold [13]. Gold is resistant to oxidative corrosion and

In recent years, nanoparticulate-mediated drug delivery research has examined a full spectrum of nanoparticles that can be used in diagnostic and therapeutic cancer applications. A key aspect of this technology is in the potential to specifically target the nanoparticles to diseased cells using a range of molecules, in particular antibodies. Antibody–nanoparticle conjugates have the potential to elicit effective targeting and release of therapeutic targets at the disease site, while minimizing off-target side effects caused by dosing of normal tissues. This article provides an overview of various antibody-conjugated nanoparticle strategies, focusing on the rationale of cell-surface receptors targeted and their potential clinical application.

KEYWORDS: anticancer � drug delivery � immunotargeting � liposome � nanoparticle � quantum dot � receptor-mediated endocytosis

Francois Fay1 & Christopher J Scott†1

1School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, UK†Author for correspondence:Tel.: +44 289 097 2350 [email protected]

For reprint orders, please contact: [email protected]

Page 2: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)382 future science group

Review Fay & Scott

is therefore considered stable to the various environments encountered within the human body [13]. However, although some studies have shown that gold would appear safe in vivo [14], others have demonstrated a significant reduction in cell viability when exposed to high concen-trations of gold-based NPs [15–18]. Nonetheless, gold-based NPs have now entered clinical examination. For example, polyethylene glycol (PEG)-coated NPs, conjugated to recombinant TNF-a, have been examined in a Phase I clinical study. In this study, patients with non responsive advanced and/or metastatic solid organ cancers were treated with single or several injections of various doses (from 50 to 600 mg/m2) of the NPs, without encountering dose limiting toxicity or immunogenicity [19].

� Single-wall carbon nanotubesSingle-wall carbon nanotubes consist of single-atom thick cylindrical structures [20]. Carbon nanotubes have been shown to absorb light of near infrared light wavelengths, making them useful for imaging [21], biosensing [22–24] and therapeutic strategies. Single-wall carbon nano-tube surfaces can be functionalized by poly-mers or biomolecules, and various therapeutic applications have been developed, including anti microbial [25,26] and photothermal tumor therapy [27,28]. In these strategies, similar to gold NPs, the carbon nanotubes are used as photosensitizers to kill targeted cells.

� Quantum dotsQuantum dots (QDs) are semiconductor crystal nanostructures. QDs have fluorescent properties that can be modified as a result of changing the diameter or composition of the particle [29]. These properties make them ideal for biological applications, as in the case of bio-imaging [30–32]. Furthermore, the conjugation of the QD to other molecules, such as targeting proteins, does not affect their spectral properties. However, as the core of the QD contains toxic metal, leakage from this has raised some toxicity concerns [33–35].

� LiposomesLiposomes are spherical vesicles composed of one or more aqueous cores, which are enclosed in a single or multilamellar lipid bilayer (Figure 1). The development of liposomal vesicular carri-ers has been examined, attempting to mimic natural lipid bilayers since the first description of an endosome/lysosome in the early 1960s. Liposomal drug formulations can provide longer

drug half lives as well as tailored drug-release pro-files, reducing high peak plasma concentrations. Moreover, it has been shown that liposomes can accumulate within the tumor tissue as a result of the enhanced permeability and retention (EPR) effect; a consequence of leaky vasculature asso-ciated with the developing cancer [36,37]. A fore-most example of the application of liposomes in drug delivery is in the application of doxorubicin. Doxorubicin can induce off-target myocardial toxicity, which is significantly reduced when the drug is encapsulated within a liposomal prepa-ration [38,39]. Consequently, liposomal prepara-tions of doxorubicin (Doxil/Caelyx and Myocet) and daunorubicin (DaunoXome), have been clinically approved to date [40–43].

Liposomal lipid bilayers are usually composed of naturally derived phospholipids such as egg phosphatidylethanolamine, hydrogenated soy phosphatidylcholine and cholesterol. Cationic surfactants, such as diol-eolyl-phosphatidyl-etha-nol-amine, or ionizable cationic lipid, such as dio-leoyl-trimethyl ammonium propane (DODAP), have also been used to produce positively charged structures that promote interaction with nega-tively charged molecules such as DNA and gene-silencing RNA (siRNA) [44–49]. Liposomes may also have potential in the co encapsulation of different types of hydrophilic biologics, such as siRNA in combination with imatinib, to treat chronic myeloid leukemia [50].

� NiosomesNiosomes, or nonionic surfactant vesicles, have a similar structure to liposomes but their membranes are only composed by nonionic surfactants, such as polyglyceryl-alkyl ethers or N-palmitoylglucosamine [51]. Niosomes comprise a wide spectrum of vesicles, from small, unila-mellar to large, multilamellar vectors and con-stitute an alternative to liposomes with improved chemical stability in vivo [52]. Various studies have examined niosome formulations for oral and sys-temic delivery of molecules, such as doxorubi-cin, camptothecin and insulin, demonstrating an increase in drug bioavailability [53].

� DendrimersDendrimers essentially encompass a range of branched polymer complexes. Generally, they consist of an initiator core, surrounded by a fur-ther layer of selected polymer, which is grafted to this core, forming a tree-like branched macro-molecular complex. Dendrimer synthesis is per-formed through controlled repetitive chemical polymerization steps. These repetitions permit

Page 3: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 383future science group

Antibody-targeted nanoparticles for cancer therapy Review

tight regulation and control of features, such as shape, size and surface functionality [54]. Dendrimers, produced using polymers such as poly(amidoamine) or poly(l-lysine), have excel-lent water-solubility, biocompatibility and are considered sufficiently nonimmunogenic [55]. Dendrimers have been used in various therapeu-tic and diagnostic applications for the delivery of a broad range of molecules including DNA [56,57], RNA [58,59], bioimaging contrast agents [54] and chemotherapies [60].

� Polymeric micellesPolymeric micelles are composed by aggregates of amphiphilic polymers constituted into hydropho-bic cores, surrounded by a corona of hydrophilic polymeric chains exposed to the aqueous envi-ronment [61]. Polymers used in recent studies are mainly heterobifunctional copolymers, composed of a hydrophilic block of PEG, poly(vinyl pyrrol-idone) and hydrophobic poly(l-lactide) or poly(l-lysine) that will form the particle core [62,63]. Encapsulation of anti cancer drugs, such as doxo-rubicin [64,65] and camptothecin [66,67] has been developed through conjugation to the hydropho-bic core. Furthermore, as with liposomes, formu-lations of cationic micelles have been developed to carry DNA or RNA molecules [61,68].

� Polymeric nanoparticlesThese nanoparticulate structures encom-pass both nanospheres and nanocapsules. Nanospheres are amongst the simplest NP for-mulations, consisting simply of a solid matrix of polymer, whist the nanocapsule contains an aqueous core (Figure 1). The formulation used is essentially dependent on the solubility of the drug substance; poorly water-soluble drugs are more readily encapsulated within the non-aqueous environment in nanospheres, while water-soluble and labile drug substances, such as DNA and proteins/peptides, are more easily encapsulated within nanocapsules.

Various polymers have been exam-ined for use in these formulations, includ-ing poly (acr y lamides ) , poly (e ster s ) , poly(alkylcyanoacrylates), poly(lactic acids), poly(glycolic acids), poly(lactic-coglycolic acid) (PLGA), chitosan (copolymer of d-glucosamine and N-acetyl-glucosamine) and alginates (copolymer of guluronic acid and mannuronic acid) [69–74]. PLGA is considered by some to be particularly attractive owing to its proven biocompatibility. Indeed, various PLGA-based clinical products such as bone fixations [75], con-trolled-release drug implants or micro particles including Nutropin™ Depot (Genentech, CA,

Nanospheres Nanocapsules Liposomes Dendrimers

Polymeric micelles

Polymeric matrix

Lipid core

Hydrophilic drug

Hydrophobic drug

Phospholipid

Polymer

Amphiphilic polymer

Polar head group

Hydrophobic tail

Figure 1. Diversity of nanoparticles used in drug delivery strategies. Nanoparticulate formulations can be tailored based on the required drug to be encapsulated/carried. While hydrophobic molecules can be incorporated inside the core of the nanoparticles, hydrophilic molecules can be carried more readily within an aqueous core protected by a polymeric or lipidic shell.

Page 4: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)384 future science group

Review Fay & Scott

USA), Trelsta™ Depot (Pfizer, CT, USA), Sandostatin™ LAR Depot (Novartis, MO, USA) are already available on the market.

Nanoparticle drug delivery: targeting strategiesAlmost as intense an area of research as NP formulation is NP targeting. The targeting of nanoparticulate formulations focuses on both the development of new diagnostic tools and improving the efficacies of therapeutic agents. Targeting approaches can be broadly classified into two areas; passive and active targeting.

� Passive targetingPassive targeting exploits the normal biodistri-bution that unadorned NPs will take within the body. Upon intravenous delivery, plain NPs are rapidly removed from circulation by opsoniza-tion and macrophage engulfment [76] or accumu-late in the liver and spleen [77]. Therefore, this clearance can be exploited to treat hepatic disorders such as leishmaniasis, a parasitic dis-ease [78], or for the targeting of accumulated macrophages in atherosclerosis [79].

Despite these opportunities to exploit opson-ization, the treatment of most diseases, in par-ticular tumors, requires the circumvention of this clearance mechanism. To avoid opsoniza-tion and subsequent phagocytosis, NPs coated with PEG have been widely investigated and shown to promote significant improvements in bioavailability of the particles [80,81]. By pre-senting a corona of hydrophilic polymer, the absorption of protein on the NP surface (which triggers opsonization) is prevented [82]. The form ation of a PEG shield can be achieved by using PEG during particle formulation [83–86] or by adsorption of PEG polymer onto preformed particles [87–89].

It has also been established that nontargeted NPs can accumulate in developed tumors. Studies have suggested that particles up to 400 nm can be passively targeted to tumors [90]. This is due to the ability of these particles to leach into the diseased tissue through the leaky vasculature network that is commonly associ-ated with tumorigenesis; a phenomenon called the EPR effect [91]. The increased permeability of the vasculature in tumors is due to incom-plete or disordered endothelial cell junctions that are frequently found in rapidly grow-ing tumors. Consequently, tumor vessels are more permeable to nanoparticulate formula-tions than the well-defined vasculature found in normal differentiated tissue. Furthermore,

tumors tend to have poor lymphatic drainage, leading to further accumulation of the NP at the diseased site. Indeed, the clinical use-fulness of Doxil has been attributed, at least in part, to the EPR effect with a significant decrease of doxorubicin side effects, such as cardiomyo pathy and myelo suppression; a result of decreased exposure of these normal tissues and cells to the drug [92].

� Active targetingActive targeting involves the modification of the NP with a targeting moiety. This modi-fication is usually on the corona of the par-ticle, introducing a ligand, which facilitates the homing, binding and internalization of the formulation to the targeted cells (Table 1). Although this approach has been used to target normal cells in vaccine formulations (e.g., tar-geting of gastro intestinal tract epithelial cells in oral delivery strategies [84,93]), most research has focused on the specific targeting of cells expressing disease-associated biomarkers, as in the case of cancer. Various moieties have been examined as targeting agents, including vita-mins [87,94], carbo hydrates [95], aptamers [85,96], peptides (e.g., Arg-Gly-Asp, allatostatin, trans- activating transcriptional activator) [84,97–99] and proteins (e.g., lectins, and transfer-rin) [98,100–103]. However, the majority of research to date has focused on antibodies, which will now be discussed in more depth.

Antibody-targeted nanoparticlesAntibodies are considered by many as ideal anticancer therapeutic agents and have been an area of intense research since customized monoclonal antibody production was reported in the mid-1970s. Indeed, antibody produc-tion display and screening innovations, such as phage display, mean that antibodies can be derived or engineered to bind with exceptional specificity to a wide range of target antigens. Importantly however, although antibodies are used very successfully as therapeutic agents in their own right, they also have the ability to be exploited as targeting agents [104,105].

The application of antibodies to deliver con-jugated agents to disease sites can be utilized for imaging or diagnostic purposes as in the example of fluorophores [106] or radioisotopes for lymphomas [10]. Alternatively, they can be used for the delivery of active therapeutics such as cytokines [107], prodrug activation enzymes (e.g., b-lactamase and carboxypepti-dase) [108,109] and chemotherapy toxins [110–112].

Page 5: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 385future science group

Antibody-targeted nanoparticles for cancer therapy Review

Clinically, immunoconjugates have been used in cancer treatment. For example, gem-tuzumab (Mylotarg®; Wyeth, CT, USA) con-sists of a CD-33 specific monoclonal antibody conjugated to a calicheamicin chemotherapy, and was used for the treatment of acute myeloid leukemia [113]. Furthermore, conjugation of radioisotopes with targeting antibodies has been developed for both imaging (immuno-scintigraphy) and radioimmunotherapy strat-egies. For example, anti-CD20 ibritumomab-tiuxetan (Zevalin®; Spectrum Pharmaceuticals, CA, USA) has been applied incorporating either 90Y metal isotope for use as a clinical therapy [114,115] or, alternatively, g-emitter 111In for in vivo imaging strategies in hematological malignan-cies [10]. Antibody–NP conjugates have poten-tial benefits over these current approaches as they have the ability to circumvent some of the issues associated with direct conjugates, such as the possible inactivation of the drug entity and the necessary release of the drug once internal-ized into endosomal/lysosomal vesicles through pH labile or reducible linkers. Furthermore, whereas stoichiometric ratios of drug to anti-body are only possible with direct conjugates, the potential of much higher drug to antibody ratios are possible with antibody–NP com-plexes; thereby maximizing the concentration of drug that can be targeted to the disease site.

This current background has led to focused interest in the development of antibody-coated nanoparticulates; both for lipid-based [116,117] and non-lipid-based NPs [118]. The majority of nanoparticulate antibody-targeting research has focused on antitumor strategies, using the antibody to target cell-surface markers of disease that are frequently upregulated or expressed specifically on either the tumor or tumor-associated cells (Table 2). In the following sections, the main targets examined for anti-body–NP conjugates to date are discussed in more depth.

� EGFR/HER1EGF receptor (EGFR/HER1) is a member of the human epidermal receptor family. This recep-tor binds various, closely related ligands, such as EGF or TGF-a [119]. When activated by ligand binding, it has been shown to trigger down-stream signaling to promote proliferation, migra-tion and angiogenesis [119]. This receptor is over-expressed or dysregulated in many solid tumors, such as breast or esophageal cancers [120–122] and its expression levels have been linked with poor prognosis [122,123]. Consequently, numerous antibody-based strategies have been developed to block EGFR activation and are used clini-cally, as in the example of cetuximab (colorec-tal and head/neck carcinomas) [124,125], panitu-mumab (colo rectal cancer) [125] or nimotuzumab (head/neck cancer) [126].

This proven effectiveness of antibody thera-pies towards EGFR, has led to much interest in EGFR as a target for immunoconjugates includ-ing antibody conjugated liposomes [64,92,127] and NPs [128–131]. EGFR-targeted antibody–NP tar-geting strategies have also been developed for diagnostic purposes, using gold NPs as the con-trast agent in optoacoustic tomography. Using this approach, it has been possible to differenti-ate between cells preincubated with either EGFR antibody-labeled gold NPs, or nonspecific gold NPs [130]. More recently, it has been shown that cetuximab-conjugated gold NPs could be used in conjugation with radiotherapy to specifically target and kill cells expressing high levels of EGFR in mixed cultures [132].

The effectiveness of targeting chemotherapy-loaded NPs with EGFR antibodies has also been demonstrated. This successful targeting was exploited to deliver rapamycin (a poorly water soluble, antiproliferative drug), inducing a significant increase of cytotoxicity compared with native rapamycin and nontargeted loaded rapamycin NPs towards breast adenocarcinoma MCF7 cells [120].

Table 1. Main nanoparticle intracellular uptake mechanisms.

Endocytosis pathway

Particles size limit

Proteins involved Stimuli Ref.

Phagocytosis Up to 20 µm

Actin, dynamin Antibody Fc region, complement, vitronectin, phosphatidylserine

[175,176]

Macropinocitosis ~1 µm Actin Growth factor, antigen binding [177,178]

Caveolae-mediated endocytosis

~80 nm Caveolae, actin, dynamin

Cholera toxin, tetanus toxin, folic acid

[179,180]

Clathrin-mediated endocytosis

~200 nm Clathrin, actin, dynamin

Transferrin, low-density lipoprotein, EGF

[181–183]

Endocytosis pathways can be subdivided into four main categories: phagocytosis, macropinocitosis, caveolae-mediated and clathrin-mediated. These pathways are principally initiated through receptor activation on the cell membrane by various molecules.

Page 6: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)386 future science group

Review Fay & Scott

In addition to these in vitro investigations, EGFR targeting strategies have also now been examined in vivo. Administration of anti-EGFR monoclonal antibody-labeled dendrim-ers conjugated with nonradioactive boron-10 to treat F98 EGFR glioma-bearing rats, fol-lowed by boron neutron capture therapy, demonstrated an increase of lifespan of 107% when compared with untreated controls [129]. This work highlights that the successful tar-geting observed in vitro can be preserved and exploited in vivo, paving the way for clinical applications in the future.

� Her2/neuHuman EGF receptor 2 (Her2/neu, ErbB-2, CD340) is a membrane tyrosine kinase recep-tor from the same epidermal growth factor receptor family as EGFR. This receptor has been linked with the signal transduction of cell growth and differentiation pathways [133,134]. Moreover, it is a prominent cancer target, overexpressed in a signif icant number of breast cancer cases (20–30%) and linked with poorer prognosis and higher risk of disease recurrence [135–137].

Tra s tu zumab (Hercept in®; Roche Pharmaceuticals, CA, USA), is a humanized, monoclonal antibody raised against the extra-cellular domain of HER2/neu that is currently used clinically to treat HER2 positive breast cancer [138]. However, its precise in vitro and in vivo antitumor mechanisms remain con-troversial. Using experimental models, diverse molecular and cellular mechanisms have been proposed including inhibition of HER2 extra-cellular cleavage, inhibition of intra cellular signaling pathways and in vivo antibody-dependent, cell-mediated cytotoxicity [139,140]. Nonetheless, trastuzumab has been granted by various national agencies to treat early-stage HER2-positive breast cancer in combination with chemotherapy [141,142].

In addition to its clinical application as a direct therapeutic target, HER2 has also been examined as a target for drug delivery. Cirstoiu-Hapca and coworkers observed that anti-Her2 labeled NPs were able to bind to, and inter-nalize into, SKOV-3 (HER2+) cells [143]. In agreement with these findings, another study observed similar internalization effects using NP conjugated to trastuzumab. This study also revealed that upon NP uptake, a 40% decrease in surface receptor density was observed. Furthermore, through incubation of a range of silver and gold NP formulations with SK-BR-3 cells overexpressing HER2, this study demon-strated that while nude particles had little effect, trastuzumab-conjugated NPs induced caspase-9 and caspase-3 activation, leading to a twofold enhancement in cytotoxicity compared with trastuzumab treatment alone [144]. This indi-cated that the density of the antibody paratopes on the surface of the NP could cross-link and activate the receptor. Moreover, by modifying the diameter, they were also able to demonstrate that internalization was dependent on NP size, with the most efficient uptake occurring within the 25–50-nm diameter range. Interestingly, although larger particles were less readily taken up, as was anticipated, so too were particles smaller than this optimum range. The authors argue that although these small particles can target and bind to the receptors on the surface, once the particles get to a critical size, the dis-played antibodies induce multivalent recep-tor crosslinking; stimulating ligand–complex internalization. This research suggests that the most efficient particle size is likely to be a com-promise between being large enough to induce receptor cross-linking, while small enough to facilitate efficient membrane wrapping. This multivalent receptor cross-linking has also been observed with anti-Her2 antibody-conjugated liposomes [145], as well as for receptors such as IgE [146], transferrin [147] and Fas [148].

Table 2. Main tumor-associated antigens used as targets for nanoparticle-based tumor therapy.

Antigen Antigen function Cell types targeted Ref.

Her2/neu EGF receptor Breast, colorectal and ovary cancers [55,104,140,143,144,184,185]

EGFR EGF receptor Brain tumor glioma, oral and pharyngeal, liver, head and neck cancers

[64,92,120,127–129, 129–132]

VEGF VEGF receptor Lymphoma breast, colorectal and lung cancers

[151–154]

PSMA Prostate-specific membrane antigen

Prostate cancers [164,165]

Tfr Transferrin receptor Brain tumor glioma, pancreatic, colorectal, lung and breast cancers

[102]

Page 7: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 387future science group

Antibody-targeted nanoparticles for cancer therapy Review

The application of anti-HER2-conjugated NPs has also been examined in vivo. Established control or HER2-expressing xenograft tumors, treated with anti-HER2 grafted dendrimers, revealed significant NP localization and inter-nalization into the HER2-expressing tumors in comparison with control tumors after 15 h; thus highlighting that antibody target-ing could be maintained in vivo [55]. Indeed, another study demonstrated that the efficacy of paclitaxel-loaded PLGA NPs could be improved in SKOV-3 xenografts with trastu-zumab targeting [104]. The authors argue that the effectiveness of the conjugates is a combi-nation of both passive targeting by EPR and receptor-mediated internalization.

� Vascular endothelial growth factor receptorsThe VEGF receptors (VEGFRs) are tyrosine kinase receptors, which, when activated by their ligand, promote angiogenesis and vasculogen-esis [149]. VEGF and VEGFR are upregulated in angiogenic tumors [150] and, as such, are of interest in both the development of diagnostics and therapeutics – particularly in development of antagonistic antibodies to block activation of VEGFR. Soman and Giorgio developed anti-VEGF labeled QDs as an early diagnos-tic strategy and demonstrated the possibility to measure the concentration of VEGF in solu-tion within a sensitivity limit of 50 × 10–15 M by flow cyto metry [151]. In another diagnostic approach, superparamagnetic iron oxide NPs, conjugated with anti-VEGF-R2 antibody, were used as probes to detect VEGF-R2 positive glioma- bearing mice to monitor progression of the disease using MRI [152].

The application of anti-VEGFR conjugated gold NPs has been examined as an ex vivo therapeutic approach for the treatment of B-cell chronic lymphocytic leukemia. It was demonstrated that the diseased cells extracted from patients, were able to take up the parti-cles regardless of the presence of antibody into early/late endosomes and in multivesicular bod-ies. However, the anti-VEGFR NPs were able to significantly increase apoptosis compared with naked, gold NPs and antibodies alone [153].

The potential of using VEGFR antibody conjugates to deliver radiotherapy has also been pursued. VEGFR-targeted dextran magnetic NPs containing the radioisotope iodine-131 were applied towards human liver HepG2 xeno-grafts, in combination with a magnetic field applied to the tumor to target the particles to

this site. Analysis of the particle biodistribution revealed that the majority of the radioactivity was localized within the tumors; demonstrating the potential of combinative antibody-based and magnetic targeting to induce tumoral localiza-tion of the targeted NP. This improved localiza-tion of the double-targeted iodine-131, encapsu-lated in VEGFR-targeted dextran magnetic NP, was evidenced by the improved tumor inhibi-tion rate (89%) compare with free iodine-131 (27%), or iodine-131 tartgeted only by anti-VEGFR antibody (77%) or dextran magnetic NPs (81%) [154]. This highlights the possible future application of this ‘double-targeting’ approach in clinical radioimmunotherapy.

� Prostate-specific membrane antigen Prostate-specific membrane antigen (PSMA) is a nonsecreted type II transmembrane glyco-protein expressed mainly in prostatic epithe-lium. However, it is only expressed in prostate cancer cells and its expression level increases dur-ing the progression of malignancy [112]. PSMA has also recently been detected in kidney and bladder carcinoma [155]. A radioimmunoscinti-graphic diagnostic method, using anti-PSMA radioactive indium conjugates (ProstaScint®; Cytogen Corporation, NJ, USA) is already widely used for diagnosing prostate cancer in patients [156]. PSMA has also been examined as a target in various therapeutic approaches for prostate cancer including cancer vaccine deliv-ery [157,158], antibody–drug conjugates [159–161] and radioimmunotherapy [162,163].

To investigate the application of anti-PSMA (clone J591) conjugated dendrimers as an effective targeted dual drug/imaging delivery system, in vitro studies were carried out using human prostate cancer LNCaP cells (PSMA+) incubated with nontargeted or J591-conjugated dendrimer. The presence of the J591 antibody on the particles led to preferential cellular adherence and internalization over controls. Moreover, competition assays using free J591 blocked this inter action, demonstrating the specificity of the binding of the conjugates to the surface PMSA [164]. Using the same antibody (J591) to conjugate PEGylated QD (QD-PSMA), Gao and coworkers performed an in vivo study with nude mice bearing PSMA+ human prostate cancer cells (C4–2) in sub-cutaneous xenografts in order to investigate the biodistribution of QD-PSMA compared with QDs with free carboxylic acid moieties (QD-COOH) or coated with PEG (QD-PEG). Upon tail vein administration, only targeted

Page 8: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)388 future science group

Review Fay & Scott

QD-PSMA induced a strong tumor signal, enabling the visualization of the tumor using a whole-body macro illumination system [165]. However, these studies also revealed a high sig-nal in the livers of treated animals, demonstrat-ing that further optimization of the QD bio-availability is required before clinical evaluation can begin.

� Transferrin receptorThe transferrin receptor (Tfr, CD71) is a mem-brane carrier protein that is involved in the reg-ulation of intracellular iron levels as it internal-izes the complexes formed by the blood plasma protein, transferrin, and iron ions [166]. As this receptor has been shown to be overexpressed in proliferating cells in comparison with resting cells [167], it has been examined as a target in tumor strategies. The main ligand developed to target the Tfr has been transferrin itself [168–170]; however, anti-transferrin antibody-conjugated NPs have also been developed and investigated. Using Tfr overexpressing human B-cell lym-phoma (Ramos) cells incubated with fluores-cent 200 nm size PEG-based NPs, Wang and coworkers demonstrated that 80% of the cells were labeled with the anti-Tfr NP after only 4 h, in comparison with 10% with control anti-body conjugates [102]. Interestingly, both anti-Tfr and even transferrin-coated NPs exhibited a cytotoxic effect, with 70% cell death observed, while control NPs did not show any toxicity, even though no drug was encapsulated. The cytotoxic effect of the anti-Tfr NP was fully suppressed by addition of ferric ammonium sulfate, suggesting that the toxicity was primar-ily caused by iron deprivation. However, these studies also revealed that the Tfr NP uptake correlated with receptor density and that the size of the particles was also crucial; incubation of anti-Tfr antibody coated with larger 5-µm microparticles resulted in no internalization and only negligible toxicity. This work clearly highlights that the size and composition of the particulate is also a key consideration in active targeting strategies.

As the Tfr is also constitutively expressed in the blood–brain barrier, Ulbrich and cowork-ers developed an analgesic (loperamide)-loaded NP, conjugated to either transferrin or anti-Tfr antibodies [103]. The analgesic effect of tail vein injection of various loperamide-loaded particle solutions was subsequently tested by monitor-ing mice responses to heat. While free analge-sic or loperamide-loaded NPs did not achieve any analgesic effect in vivo, both transferrin or

anti-Tfr antibody conjugated, drug-loaded NPs elicited significant effects, highlighting their future therapeutic utility.

� Dendritic cell receptorsDendritic cells (DCs) are key regulators in antigen-specific immunity, owing to their abil-ity to internalize, process and present antigens to T cells. Therefore, in order to increase T-cell-mediated immune responses in vivo, targeted delivery of antigens to DCs using antibodies has been examined. The main receptors targeted are endocytic transmembrane receptors such as C-type lectins (i.e., DC-SIGN and CD205), integrins (i.e., MACI and CD11c–CD18) or FcR (i.e., FcgRI and FcgRIII) families [171]. The in vivo application of antigen-loaded particles or liposomes conjugated with DC-targeted anti-CD205 has demonstrated the enhanced efficacy of DC-targeted carriers as a tumor immuno-therapy in animal studies [172]. In this work, mice were vaccinated against ovalbumin by tail injections of free ovalbumin or ovalbumin-loaded conjugated NPs conjugated to either anti-CD205 antibodies or isotype control. After 7 days, enhanced ovalbumin epitope-specific cytotoxic T-lymphocyte activity was measured in spleen cells isolated from mice vaccinated with the anti-CD205 monoclonal antibody conjugated ovalbumin-loaded NPs compared with free ovalbumin and ovalbumin-loaded NPs (conjugated with isotype control antibody). Similarly, targeting of DCs through application of a humanized DC-SIGN particles conjugate has been examined. Interestingly, analysis of the uptake and antigen presentation in the tar-geted DCs revealed that although DC-SIGN targeting increased intracellular accumula-tion and antigen presentation, the targeting of the larger macro particles did not significantly enhance either internalization or antigen pre-sentation. This again highlights that particle size is a critical parameter in these targeting strategies [173].

Conclusion & future perspectiveTo date, developments in the field of antibody-conjugated NPs have demonstrated attractive results both in vitro and in vivo. The cell- specific receptor-mediated endocytosis of the NP at the site of disease provides an attractive feature for a variety of diagnostic and therapeutic strategies. Furthermore, an additional feature of receptor targeting is the possibility of inducing multi-valent receptor activation to trigger receptor-activated signaling, leading to downstream

Page 9: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 389future science group

Antibody-targeted nanoparticles for cancer therapy Review

Executive summary

� Antibody and antibody fragments can be efficiently conjugated to the surface of most types of nanoparticles by facile cross-linking approaches.

� Antibody-conjugated nanoparticles can target cells and be exploited for a range of diagnostic and therapeutic approaches, particularly in diseases such as cancer.

� For specific cell-surface antigens, the high density of antibody moieties on the nanoparticle surface can induce multivalent receptor activation, which can be exploited to trigger receptor-mediated internalization and other downstream signal transduction.

effects such as cell degranulation [146], apop-totic signaling [144,146,148] or iron depletion [147]. In addition, some of these studies have shown that NP size and surface ligand density are critical key features [144,146,147]. Therefore, optimizations of these parameters, as well as PEGylation and antibody density, are required to ensure that the functionality of each modi-fication is not compromised. As many antibod-ies are currently being developed towards novel antitumor targets or biomarkers, the ability to further exploit these molecules for targeting NPs is expected to be a rapidly expanding area. However, some key questions remain before targeted NPs are to be used clinically such as biodistribution, retention, metabolism and long-term toxicity of some particle materials [174]. In addition, the production of these particles to

a current good manufacturing practice stan-dard in a cost effective manner will also need to be addressed. Nonetheless, taking together the exciting preclinical efficacies that these NP conjugates have demonstrated, with the limited clinical evaluation that has been undertaken to date, highlights exciting opportunities for their commercialization and clinical application.

Financial & competing interests disclosureChristopher Scott owns shares in Fusion Antibodies Ltd, Belfast, UK. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

BibliographyPapers of special note have been highlighted as:� of interest�� of considerable interest

1 Cowens JW, Creaven PJ, Greco WR et al.: Initial clinical (Phase I) trial of TLC D-99 (doxorubicin encapsulated in liposomes). Cancer Res. 53, 2796–2802 (1993).

2 Chu D, Lacouture ME, Weiner E, Wu S: Risk of hand-foot skin reaction with the multitargeted kinase inhibitor sunitinib in patients with renal cell and non-renal cell carcinoma: a meta-analysis. Clin. Genitourin. Cancer 7, 11–19 (2009).

3 Darby SC, Cutter DJ, Boerma M et al.: Radiation-related heart disease: current knowledge and future prospects. Int. J. Radiat. Oncol. Biol. Phys. 76, 656–665 (2010).

4 Sparreboom A, de Jonge MJ, Verweij J: The use of oral cytotoxic and cytostatic drugs in cancer treatment. Eur. J. Cancer 38, 18–22 (2002).

5 van der Vijgh WJ: Clinical pharmacokinetics of carboplatin. Clin. Pharmacokinet. 21, 242–261 (1991).

6 Desilet N, Campbell TN, Choy FY: p53-based anti-cancer therapies: an empty promise? Curr. Issues Mol. Biol. 12, 143–146 (2010).

7 Vanniasinghe AS, Bender V, Manolios N: The potential of liposomal drug delivery for the treatment of inflammatory arthritis. Semin. Arthritis Rheum. 39, 182–196 (2009).

8 El-Sayed IH, Huang X, El-Sayed MA: Surface plasmon resonance scattering and absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics: applications in oral cancer. Nano Lett. 5, 829–834 (2005).

9 Huang X, Jain PK, El-Sayed IH, El-Sayed MA: Determination of the minimum temperature required for selective photothermal destruction of cancer cells with the use of immunotargeted gold nanoparticles. Photochem. Photobiol. 82, 412–417 (2006).

10 Conti PS, White C, Pieslor P, Molina A, Aussie J, Foster P: The role of imaging with 111In-ibritumomabtiuxetan in the ibritumomabtiuxetan (Zevalin) regimen: results from a Zevalin Imaging Registry. J. Nucl. Med. 46, 1812–1818 (2005).

11 Liu K, Anderson GP, Bozinovski S: DNA vector augments inflammation in epithelial cells via EGFR-dependent regulation of TLR4 and TLR2. Am. J. Respir. Cell Mol. Biol. 39, 305–311 (2008).

12 Yu C, Irudayaraj J: Multiplex biosensor using gold nanorods. Anal. Chem. 79, 572–579 (2007).

13 Pissuwan D, Cortie CH, Valenzuela S, Cortie MB: Gold nanosphere-antibody conjugates for hypothermal therapeutic applications. Gold Bulletin 40, 121 (2007).

14 Geso M: Gold nanoparticles: a new X-ray contrast agent. Br. J. Radiol. 80, 64–65; Author reply 65 (2007).

15 Pernodet N, Fang X, Sun Y et al.: Adverse effects of citrate/gold nanoparticles on human dermal fibroblasts. Small 2, 766–773 (2006).

16 Patra HK, Banerjee S, Chaudhuri U, Lahiri P, Dasgupta AK: Cell selective response to gold nanoparticles. Nanomedicine 3, 111–119 (2007).

17 Lewinski N, Colvin V, Drezek R: Cytotoxicity of nanoparticles. Small 4, 26–49 (2008).

18 Uboldi C, Bonacchi D, Lorenzi G et al.: Gold nanoparticles induce cytotoxicity in the alveolar type-II cell lines A549 and NCIH441. Part. Fibre Toxicol. 6, 18 (2009).

19 Libutti SK, Paciotti GF, Byrnes AA et al.: Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine. Clin. Cancer Res. 16, 6139–6149 (2010).

20 McDevitt MR, Chattopadhyay D, Jaggi JS et al.: PET imaging of soluble yttrium-86-labeled carbon nanotubes in mice. PLoS One 2, E907 (2007).

Page 10: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)390 future science group

Review Fay & Scott

21 Welsher K, Liu Z, Daranciang D, Dai H: Selective probing and imaging of cells with single walled carbon nanotubes as near-infrared fluorescent molecules. Nano Lett. 8, 586–590 (2008).

22 Lei J, Ju H: Nanotubes in biosensing. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2, 496–509 (2010).

23 Nedosekin DA, Shashkov EV, Galanzha EI, Hennings L, Zharov VP: Photothermal multispectral image cytometry for quantitative histology of nanoparticles and micrometastasis in intact, stained and selectively burned tissues. Cytometry A. 77, 1049–1058 (2010).

24 de la Zerda A, Liu Z, Bodapati S et al.: Ultrahigh sensitivity carbon nanotube agents for photoacoustic molecular imaging in living mice. Nano Lett. 10, 2168–2172 (2010).

25 Liu S, Ng AK, Xu R et al.: Antibacterial action of dispersed single-walled carbon nanotubes on Escherichia coli and Bacillus subtilis investigated by atomic force microscopy. Nanoscale 2, 2744–2750 (2010).

26 Kim JW, Shashkov EV, Galanzha EI, Kotagiri N, Zharov VP: Photothermal antimicrobial nanotherapy and nanodiagnostics with self-assembling carbon nanotube clusters. Lasers Surg. Med. 39, 622–634 (2007).

27 Shao N, Lu S, Wickstrom E, Panchapakesan B: Integrated molecular targeting of IGF1R and HER2 surface receptors and destruction of breast cancer cells using single wall carbon nanotubes. Nanotechnology 18, 315101 (2007).

28 Huang N, Wang H, Zhao J, Lui H, Korbelik M, Zeng H: Single-wall carbon nanotubes assisted photothermal cancer therapy: animal study with a murine model of squamous cell carcinoma. Lasers Surg. Med. 42, 638–648 (2010).

29 Cai QY, Kim SH, Choi KS et al.: Colloidal gold nanoparticles as a blood-pool contrast agent for x-ray computed tomography in mice. Invest. Radiol. 42, 797–806 (2007).

30 Gao X: Multifunctional quantum dots for cellular and molecular imaging. Conf. Proc. IEEE Eng. Med. Biol. Soc. 2007, 524–525 (2007).

31 Gao J, Chen K, Miao Z et al.: Affibody-based nanoprobes for HER2-expressing cell and tumor imaging. Biomaterials 32(8), 2141–2148 (2011).

32 Gonda K, Watanabe TM, Ohuchi N, Higuchi H: In vivo nano-imaging of membrane dynamics in metastatic tumor cells using quantum dots. J. Biol. Chem. 285, 2750–2757 (2010).

33 Dubertret B, Skourides P, Norris DJ, Noireaux V, Brivanlou AH, Libchaber A: In vivo imaging of quantum dots encapsulated in phospholipid micelles. Science 298, 1759–1762 (2002).

34 Chang E, Thekkek N, Yu WW, Colvin VL, Drezek R: Evaluation of quantum dot cytotoxicity based on intracellular uptake. Small 2, 1412–1417 (2006).

35 Aillon KL, Xie Y, El-Gendy N, Berkland CJ, Forrest ML: Effects of nanomaterial physicochemical properties on in vivo toxicity. Adv. Drug Deliv. Rev. 61, 457–466 (2009).

36 KontermannRE: Immunoliposomes for cancer therapy. Curr. Opin. Mol. Ther. 8, 39–45 (2006).

37 Hatakeyama H, Akita H, Ishida E et al.: Tumor targeting of doxorubicin by anti-MT1-MMP antibody-modified PEG liposomes. Int. J. Pharm. 342, 194–200 (2007).

38 Nagae I, Koyanagi Y, Ito S, Tanabe Y, Unezaki S: Liposome drug delivery system for murine neuroblastoma. J. Pediatr. Surg. 33, 1521–1525 (1998).

39 Ranson MR, Cheeseman S, White S, Margison J: Caelyx (stealth liposomal doxorubicin) in the treatment of advanced breast cancer. Crit. Rev. Oncol. Hematol. 37, 115–120 (2001).

40 Andreopoulou E, Gaiotti D, Kim E et al.: Pegylated liposomal doxorubicin HCL (PLD; Caelyx/Doxil): experience with long-term maintenance in responding patients with recurrent epithelial ovarian cancer. Ann. Oncol. 18, 716–721 (2007).

41 Latagliata R, Breccia M, Fazi P et al.: Liposomal daunorubicin versus standard daunorubicin: long term follow-up of the GIMEMA GSI 103 AMLE randomized trial in patients older than 60 years with acute myelogenous leukaemia. Br. J. Haematol. 143, 681–689 (2008).

42 Camera A, Rinaldi CR, Palmieri S et al.: Sequential continuous infusion of fludarabine and cytarabine associated with liposomal daunorubicin (DaunoXome) (FLAD) in primary refractory or relapsed adult acute myeloid leukemia patients. Ann. Hematol. 88, 151–158 (2009).

43 Sparano JA, Makhson AN, Semiglazov VF et al.: Pegylated liposomal doxorubicin plus docetaxel significantly improves time to progression without additive cardiotoxicity compared with docetaxelmonotherapy in patients with advanced breast cancer previously treated with neoadjuvant-adjuvant anthracycline therapy: results from a randomized Phase III study. J. Clin. Oncol. 27, 4522–4529 (2009).

44 Duarte S, Faneca H, Pedroso de Lima MC: Non-covalent association of folate to lipoplexes: a promising strategy to improve gene delivery in the presence of serum. J. Control. Release DOI: 10.1016/j.jconrel.2010.10.032 (2010) (Epub ahead of print).

45 Kline CL, Shanmugavelandy SS, Kester M, Irby RB: Delivery of PAR-4 plasmid in vivo via nanoliposomes sensitizes colon tumor cells subcutaneously implanted into nude mice to 5-FU. Cancer Biol. Ther. 8, 1831–1837 (2009).

46 Chen Y, Bathula SR, Li J, Huang L: Multifunctional nanoparticles delivering small interfering RNA and doxorubicin overcome drug resistance in cancer. J. Biol. Chem. 285, 22639–22650 (2010).

47 Feng C, Wang T, Tang R et al.: Silencing of the MYCN gene by siRNA delivered by folate receptor-targeted liposomes in LA-N-5 cells. Pediatr. Surg. Int. 26, 1185–1191 (2010).

48 Pardridge WM: shRNA and siRNA delivery to the brain. Adv. Drug Deliv. Rev. 59, 141–152 (2007).

49 Geusens B, Lambert J, De Smedt SC, Buyens K, Sanders NN, Van Gele M: Ultradeformable cationic liposomes for delivery of small interfering RNA (siRNA) into human primary melanocytes. J. Control. Release 133(3), 214–220 (2009).

50 Mendonca LS, Moreira JN, de Lima MC, Simoes S: Co-encapsulation of anti-BCR-ABL siRNA and imatinibmesylate in transferrin receptor-targeted sterically stabilized liposomes for chronic myeloid leukemia treatment. Biotechnol. Bioeng. 107, 884–893 (2010).

51 Dufes C, Gaillard F, Uchegbu IF, Schatzlein AG, Olivier JC, Muller JM: Glucose-targeted niosomes deliver vasoactive intestinal peptide (VIP) to the brain. Int. J. Pharm. 285, 77–85 (2004).

52 Conacher M, Alexander J, Brewer JM: Niosomes as immunological adjuvants. In: Synthetic Surfactant Vesicles. Uchegbu I (Ed.). International Publishers Distrubtors, Singapore, 185–205 (2000).

53 Arora R, Jain CP: Advances in niosome as a drug carrier: a review. Asian J. Pharm. 1, 29–39 (2007).

54 Tomalia DA, Reyna LA, Svenson S: Dendrimers as multi-purpose nanodevices for oncology drug delivery and diagnostic imaging. Biochem. Soc. Trans. 35, 61–67 (2007).

55 Shukla R, Thomas TP, Peters JL et al.: HER2 specific tumor targeting with dendrimer conjugated anti-HER2 mAb. Bioconjug. Chem. 17, 1109–1115 (2006).

Page 11: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 391future science group

Antibody-targeted nanoparticles for cancer therapy Review

391www.futuremedicine.com

56 Ke W, Shao K, Huang R et al.: Gene delivery targeted to the brain using an angiopep-conjugated polyethyleneglycol-modified polyamidoaminedendrimer. Biomaterials 30, 6976–6985 (2009).

57 Santos JL, Oliveira H, Pandita D et al.: Functionalization of poly(amidoamine) dendrimers with hydrophobic chains for improved gene delivery in mesenchymal stem cells. J. Control. Release 144, 55–64 (2010).

58 Ofek P, Fischer W, Calderon M, Haag R, Satchi-Fainaro R: In vivo delivery of small interfering RNA to tumors and their vasculature by novel dendritic nanocarriers. FASEB J. 24(9), 3122–3134 (2010).

59 Dutta T, Burgess M, McMillan NA, Parekh HS: Dendrosome-based delivery of siRNA against E6 and E7 oncogenes in cervical cancer. Nanomedicine 6(3), 463–470 (2010).

60 Lim J, Chouai A, Lo ST, Liu W, Sun X, Simanek EE: Design, synthesis, characterization, and biological evaluation of triazinedendrimers bearing paclitaxel using ester and ester/disulfide linkages. Bioconjug. Chem. 20, 2154–2161 (2009).

61 Matsumoto S, Christie RJ, Nishiyama N et al.: Environment-responsive block copolymer micelles with a disulfide cross-linked core for enhanced siRNA delivery. Biomacromolecules 10, 119–127 (2009).

62 Takae S, Miyata K, Oba M et al.: PEG-detachable polyplex micelles based on disulfide-linked block catiomers as bioresponsivenonviral gene vectors. J. Am. Chem. Soc. 130, 6001–6009 (2008).

63 Luo Y, Wang A, Yuan J, Gao Q: Preparation, characterization and drug release behavior of polyion complex micelles. Int. J. Pharm. 374, 139–144 (2009).

64 Elbayoumi TA, Pabba S, Roby A, Torchilin VP: Antinucleosome antibody-modified liposomes and lipid-core micelles for tumor-targeted delivery of therapeutic and diagnostic agents. J. Liposome Res. 17, 1–14 (2007).

65 Yoo HS, Park TG: Folate receptor targeted biodegradable polymeric doxorubicin micelles. J. Control. Release 96, 273–283 (2004).

66 Koo OM, Rubinstein I, Onyuksel H: Camptothecin in sterically stabilized phospholipid micelles: a novel nanomedicine. Nanomedicine 1, 77–84 (2005).

67 Kawano K, Watanabe M, Yamamoto T et al.: Enhanced antitumor effect of camptothecin loaded in long-circulating polymeric micelles. J. Control. Release 112, 329–332 (2006).

68 Talelli M, Pispas S: Complexes of cationic block copolymer micelles with DNA: histone/DNA complex mimetics. Macromol. Biosci. 8, 960–967 (2008).

69 Yin L, Ding J, He C, Cui L, Tang C, Yin C: Drug permeability and mucoadhesion properties of thiolatedtrimethyl chitosan nanoparticles in oral insulin delivery. Biomaterials 30, 5691–5700 (2009).

70 Rekha MR, Sharma CP: Synthesis and evaluation of lauryl succinyl chitosan particles towards oral insulin delivery and absorption. J. Control. Release 135, 144–151 (2009).

71 George M, Abraham TE: Polyionic hydrocolloids for the intestinal delivery of protein drugs: alginate and chitosan – a review. J. Control. Release 114, 1–14 (2006).

72 Layre AM, Couvreur P, Chacun H et al.: Busulfan loading into poly(alkyl cyanoacrylate) nanoparticles: physico-chemistry and molecular modeling. J. Biomed. Mater. Res. B Appl. Biomater . 79, 254–262 (2006).

73 Kreuter J: Nanoparticles – a historical perspective. Int. J. Pharm. 331, 1–10 (2007).

74 Fay F, Quinn DJ, Gilmore BF, McCarron PA, Scott CJ: Gene delivery using dimethyldidodecylammonium bromide-coated PLGA nanoparticles. Biomaterials 31, 4214–4222 (2010).

75 Li S: Hydrolytic degradation characteristics of aliphatic polyesters derived from lactic and glycolic acids. J. Biomed. Mater. Res. 48, 342–353 (1999).

76 Owens DE 3rd, Peppas NA: Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int. J. Pharm. 307, 93–102 (2006).

77 Panagi Z, Beletsi A, Evangelatos G, Livaniou E, Ithakissios DS, Avgoustakis K: Effect of dose on the biodistribution and pharmacokinetics of PLGA and PLGA–mPEG nanoparticles. Int. J. Pharm. 221, 143–152 (2001).

78 Durand R, Paul M, Rivollet D, Houin R, Astier A, Deniau M: Activity of pentamidine-loaded methacrylate nanoparticles against Leishmaniainfantum in a mouse model. Int. J. Parasitol. 27, 1361–1367 (1997).

79 Ruehm SG, Corot C, Vogt P, Kolb S, Debatin JF: Magnetic resonance imaging of atherosclerotic plaque with ultrasmallsuperparamagnetic particles of iron oxide in hyperlipidemic rabbits. Circulation 103, 415–422 (2001).

80 Kim JY, Kim JK, Park JS, Byun Y, Kim CK: The use of PEGylated liposomes to prolong circulation lifetimes of tissue plasminogen activator. Biomaterials 30, 5751–5756 (2009).

81 Hu Y, Xie J, Tong YW, Wang CH: Effect of PEG conformation and particle size on the cellular uptake efficiency of nanoparticles with the HepG2 cells. J. Control. Release 118, 7–17 (2007).

82 Alexis F, Pridgen E, Molnar LK, Farokhzad OC: Factors affecting the clearance and biodistribution of polymeric nanoparticles. Mol. Pharm. 5, 505–515 (2008).

83 Moffatt S, Cristiano RJ: Uptake characteristics of NGR-coupled stealth PEI/pDNA nanoparticles loaded with PLGA–PEG–PLGA tri-block copolymer for targeted delivery to human monocyte-derived dendritic cells. Int. J. Pharm. 321, 143–154 (2006).

84 Garinot M, Fievez V, Pourcelle V et al.: PEGylated PLGA-based nanoparticles targeting M cells for oral vaccination. J. Control. Release 120, 195–204 (2007).

85 Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ: Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug–PLGA–PEG nanoparticles. Proc. Natl Acad. Sci. USA 105, 17356–17361 (2008).

86 Garg A, Tisdale AW, Haidari E, Kokkoli E: Targeting colon cancer cells using PEGylated liposomes modified with a fibronectin-mimetic peptide. Int. J. Pharm. 366, 201–210 (2009).

87 Kim SH, Jeong JH, Chun KW, Park TG: Target-specific cellular uptake of PLGA nanoparticles coated with poly(l-lysine)–poly(ethylene glycol)–folate conjugate. Langmuir 21, 8852–8857 (2005).

88 Brandhonneur N, Chevanne F, Vie V et al.: Specific and non-specific phagocytosis of ligand-grafted PLGA microspheres by macrophages. Eur. J. Pharm. Sci. 36(4–5), 474–485 (2009).

89 Lu W, Huang Q, Ku G et al.: Photoacoustic imaging of living mouse brain vasculature using hollow gold nanospheres. Biomaterials 31, 2617–2626 (2010).

90 Kim JH, Kim YS, Park K et al.: Antitumor efficacy of cisplatin-loaded glycol chitosan nanoparticles in tumor-bearing mice. J. Control. Release 127, 41–49 (2008).

91 Chytil P, Etrych T, Konak C et al.: New HPMA copolymer-based drug carriers with covalently bound hydrophobic substituents for solid tumour targeting. J. Control. Release 127, 121–130 (2008).

92 Elbayoumi TA, Torchilin VP: Tumor-specific antibody-mediated targeted delivery of Doxil reduces the manifestation of auricular erythema side effect in mice. Int. J. Pharm. 357, 272–279 (2008).

Page 12: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)392 future science group

Review Fay & Scott

93 Gupta PN, Khatri K, Goyal AK, Mishra N, Vyas SP: M-cell targeted biodegradable PLGA nanoparticles for oral immunization against hepatitis B. J. Drug Target 15, 701–713 (2007).

94 Zhang Z, Huey Lee S, Feng SS: Folate-decorated poly(lactide-co-glycolide)-vitamin E TPGS nanoparticles for targeted drug delivery. Biomaterials 28, 1889–1899 (2007).

95 Eliaz RE, Szoka FC Jr: Liposome-encapsulated doxorubicin targeted to CD44: a strategy to kill CD44-overexpressing tumor cells. Cancer Res. 61, 2592–2601 (2001).

96 Farokhzad OC, Cheng J, Teply BA et al.: Targeted nanoparticle-aptamerbioconjugates for cancer chemotherapy in vivo. Proc. Natl Acad. Sci. USA 103, 6315–6320 (2006).

97 Lu J, Shi M, Shoichet MS: Click chemistry functionalized polymeric nanoparticles target corneal epithelial cells through RGD-cell surface receptors. Bioconjug. Chem. 20, 87–94 (2009).

98 Singh SR, Grossniklaus HE, Kang SJ, Edelhauser HF, Ambati BK, Kompella UB: Intravenous transferrin, RGD peptide and dual-targeted nanoparticles enhance anti-VEGF intraceptor gene delivery to laser-induced CNV. Gene Ther. 16, 645–659 (2009).

99 Hu Z, Luo F, Pan Y et al.: Arg-Gly-Asp (RGD) peptide conjugated poly(lactic acid)–poly(ethylene oxide) micelle for targeted drug delivery. J. Biomed. Mater. Res. A 85, 797–807 (2008).

100 Gao X, Chen J, Tao W et al.: UEA I-bearing nanoparticles for brain delivery following intranasal administration. Int. J. Pharm. 340(1–2), 207–215 (2007).

101 Gupta PN, Mahor S, Rawat A, Khatri K, Goyal A, Vyas SP: Lectin anchored stabilized biodegradable nanoparticles for oral immunization 1. Development and in vitro evaluation. Int. J. Pharm. 318, 163–173 (2006).

102 Wang J, Tian S, Petros RA, Napier ME, Desimone JM: The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies. J. Am. Chem. Soc. 132, 11306–11313 (2010).

103 Ulbrich K, Hekmatara T, Herbert E, Kreuter J: Transferrin- and transferrin-receptor-antibody-modified nanoparticles enable drug delivery across the blood–brain barrier (BBB). Eur. J. Pharm. Biopharm. 71, 251–256 (2009).

104 Cirstoiu-Hapca A, Buchegger F, Lange N, Bossy L, Gurny R, Delie F: Benefit of anti-HER2-coated paclitaxel-loaded immuno-nanoparticles in the treatment of

disseminated ovarian cancer: therapeutic efficacy and biodistribution in mice. J. Control. Release 144, 324–331 (2010).

105 Scott CJ, Marouf WM, Quinn DJ et al.: Immunocolloidal targeting of the endocytotic siglec-7 receptor using peripheral attachment of siglec-7 antibodies to poly(lactide-co-glycolide) nanoparticles. Pharm. Res. 25, 135–146 (2008).

106 Gleysteen JP, Newman JR, Chhieng D, Frost A, Zinn KR, Rosenthal EL: Fluorescent labeled anti-EGFR antibody for identification of regional and distant metastasis in a preclinical xenograft model. Head Neck 30(6), 782–789 (2008).

107 Lode HN, Xiang R, Becker JC, Gillies SD, Reisfeld RA: Immunocytokines: a promising approach to cancer immunotherapy. Pharmacol. Ther. 80, 277–292 (1998).

108 Kerr DE, Vrudhula VM, Svensson HP, Siemers NO, Senter PD: Comparison of recombinant and synthetically formed monoclonal antibody-b-lactamase conjugates for anticancer prodrug activation. Bioconjug. Chem. 10, 1084–1089 (1999).

109 Wolfe LA, Mullin RJ, Laethem R et al.: Antibody-directed enzyme prodrug therapy with the T268G mutant of human carboxypeptidase A1: in vitro and in vivo studies with prodrugs of methotrexate and the thymidylate synthase inhibitors GW1031 and GW1843. Bioconjug. Chem. 10, 38–48 (1999).

110 Kawakami K, Nakajima O, Morishita R, Nagai R: Targeted anticancer immunotoxins and cytotoxic agents with direct killing moieties. ScientificWorldJournal 6, 781–790 (2006).

111 Hamann PR, Hinman LM, Hollander I et al.: Gemtuzumabozogamicin, a potent and selective anti-CD33 antibody–calicheamicin conjugate for treatment of acute myeloid leukemia. Bioconjug. Chem. 13, 47–58 (2002).

112 Henry MD, Wen S, Silva MD, Chandra S, Milton M, Worland PJ: A prostate-specific membrane antigen-targeted monoclonal antibody–chemotherapeutic conjugate designed for the treatment of prostate cancer. Cancer Res. 64, 7995–8001 (2004).

113 Sorokin P: Mylotarg approved for patients with CD33+ acute myeloid leukemia. Clin. J. Oncol. Nurs. 4, 279–280 (2000).

114 Jacobs SA: Yttrium ibritumomabtiuxetan in the treatment of non-Hodgkin’s lymphoma: current status and future prospects. Biologics 1, 215–227 (2007).

115 Vitolo U, Barosi G, Fanti S et al.: Consensus conference on the use of 90-yttrium-ibritumomab tiuxetantherapy in clinical

practice. A project of the Italian Society of Hematology. Am. J. Hematol. 85, 147–155 (2010).

116 Park JW, Kirpotin DB, Hong K et al.: Tumor targeting using anti-Her2 immunoliposomes. J. Control. Release 74, 95–113 (2001).

117 Tuffin G, Waelti E, Huwyler J, Hammer C, Marti HP: Immunoliposome targeting to mesangial cells: a promising strategy for specific drug delivery to the kidney. J. Am. Soc. Nephrol. 16, 3295–3305 (2005).

118 McCarron PA, Marouf WM, Donnelly RF, Scott C: Enhanced surface attachment of protein-type targeting ligands to poly(lactide-co-glycolide) nanoparticles using variable expression of polymeric acid functionality. J. Biomed. Mater. Res. A 87(4), 873–884 (2008).

119 Herbst RS, Fukuoka M, Baselga J: Gefitinib – a novel targeted approach to treating cancer. Nat. Rev. Cancer 4, 956–965 (2004).

120 Acharya S, Dilnawaz F, Sahoo SK: Targeted epidermal growth factor receptor nanoparticle bioconjugates for breast cancer therapy. Biomaterials 30, 5737–5750 (2009).

121 Hanawa M, Suzuki S, Dobashi Y et al.: EGFR protein overexpression and gene amplification in squamous cell carcinomas of the esophagus. Int. J. Cancer 118, 1173–1180 (2006).

122 Bhargava R, Gerald WL, Li AR et al.: EGFR gene amplification in breast cancer: correlation with epidermal growth factor receptor mRNA and protein expression and HER-2 status and absence of EGFR-activating mutations. Mod. Pathol. 18, 1027–1033 (2005).

123 Kocbek P, Obermajer N, Cegnar M, Kos J, Kristl J: Targeting cancer cells using PLGA nanoparticles surface modified with monoclonal antibody. J. Control. Release 120, 18–26 (2007).

124 Tejani MA, Cohen RB, Mehra R: The contribution of cetuximab in the treatment of recurrent and/or metastatic head and neck cancer. Biologics 4, 173–185 (2010).

125 Fakih M, Wong R: Efficacy of the monoclonal antibody EGFR inhibitors for the treatment of metastatic colorectal cancer. Curr. Oncol. 17(Suppl. 1), S3–S17 (2010).

126 Talavera A, Friemann R, Gomez-Puerta S et al.: Nimotuzumab, an antitumor antibody that targets the epidermal growth factor receptor, blocks ligand binding while permitting the active receptor conformation. Cancer Res. 69, 5851–5859 (2009).

127 Gupta B, Torchilin VP: Monoclonal antibody 2C5-modified doxorubicin-loaded liposomes with significantly enhanced therapeutic activity against intracranial human brain U-87 MG tumorxenografts in nude mice. Cancer Immunol. Immunother. 56, 1215–1223 (2007).

Page 13: Antibody-targeted nanoparticles for cancer therapy

www.futuremedicine.com 393future science group

Antibody-targeted nanoparticles for cancer therapy Review

393www.futuremedicine.com

128 Mamot C, Ritschard R, Kung W, Park JW, Herrmann R, Rochlitz CF: EGFR-targeted immunoliposomes derived from the monoclonal antibody EMD72000 mediate specific and efficient drug delivery to a variety of colorectal cancer cells. J. Drug Target 14, 215–223 (2006).

129 Wu G, Yang W, Barth RF et al.: Molecular targeting and treatment of an epidermal growth factor receptor-positive glioma using boronatedcetuximab. Clin. Cancer Res. 13, 1260–1268 (2007).

� In vivo study of anti-EGF receptor monoclonal antibody-labeled dendrimers.

130 Mallidi S, Larson T, Aaron J, Sokolov K, Emelianov S: Molecular specific optoacoustic imaging with plasmonic nanoparticles. Opt. Express 15, 6583–6588 (2007).

131 El-Sayed IH, Huang X, El-Sayed MA: Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett. 239, 129–135 (2006).

132 Glazer ES, Massey KL, Zhu C, Curley SA: Pancreatic carcinoma cells are susceptible to noninvasive radio frequency fields after treatment with targeted gold nanoparticles. Surgery 148, 319–324 (2010).

133 Peus D, Hamacher L, Pittelkow MR: EGF-receptor tyrosine kinase inhibition induces keratinocyte growth arrest and terminal differentiation. J. Invest. Dermatol. 109, 751–756 (1997).

134 Voldborg BR, Damstrup L, Spang-Thomsen M, Poulsen HS: Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann. Oncol. 8, 1197–1206 (1997).

135 Sainsbury JR, Farndon JR, Needham GK, Malcolm AJ, Harris AL: Epidermal-growth-factor receptor status as predictor of early recurrence of and death from breast cancer. Lancet 1, 1398–1402 (1987).

136 Minner S, Jessen B, Stiedenroth L et al.: Low level HER2 overexpression is associated with rapid tumor cell proliferation and poor prognosis in prostate cancer. Clin. Cancer Res. 16, 1553–1560 (2010).

137 Farzadnia M, Meibodi NT, Shandiz FH et al.: Evaluation of HER2/neuoncoprotein in serum and tissue samples of women with breast cancer: correlation with clinicopathological parameters. Breast 19, 489–492 (2010).

138 Goldhirsch A, Ingle JN, Gelber RD et al.: Thresholds for therapies: highlights of the St Gallen International Expert Consensus on the primary therapy of early breast cancer 2009. Ann. Oncol. 20, 1319–1329 (2009).

139 Hall PS, Cameron DA: Current perspective – trastuzumab. Eur. J. Cancer 45, 12–18 (2009).

140 Nielsen DL, Andersson M, Kamby C: HER2-targeted therapy in breast cancer. Monoclonal antibodies and tyrosine kinase inhibitors. Cancer Treat. Rev. 35, 121–136 (2009).

141 Lievens Y: Cost-effectiveness of adjuvant trastuzumab in early breast cancer: is less better? Belg. J. Med. Oncol. 4, 33–37 (2010).

142 Banerjee S, Smith IE: Management of small HER2-positive breast cancers. Lancet Oncol. 11, 1193–1199 (2010).

143 Cirstoiu-Hapca A, Bossy-Nobs L, Buchegger F, Gurny R, Delie F: Differential tumor cell targeting of anti-HER2 (Herceptin) and anti-CD20 (Mabthera) coupled nanoparticles. Int. J. Pharm. 331, 190–196 (2007).

144 Jiang W, Kim BY, Rutka JT, Chan WC: Nanoparticle-mediated cellular response is size-dependent. Nat. Nanotechnol. 3, 145–150 (2008).

�� Reports that cross-linking by antibody–nanoparticle conjugates is size dependent.

145 Chiu GN, Edwards LA, Kapanen AI et al.: Modulation of cancer cell survival pathways using multivalent liposomal therapeutic antibody constructs. Mol. Cancer Ther. 6, 844–855 (2007).

146 Huang YF, Liu H, Xiong X, Chen Y, Tan W: Nanoparticle-mediated IgE-receptor aggregation and signaling in RBL mast cells. J. Am. Chem. Soc. 131, 17328–17334 (2009).

� Along with [147,148], discusses antibody-conjugated nanoparticles inducing receptor cross-linking-triggered regulation pathways.

147 Wang J, Tian S, Petros RA, Napier ME, Desimone JM: The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies. J. Am. Chem. Soc. 132, 11306–11313 (2010).

� Along with [146,148], discusses antibody-conjugated nanoparticles inducing receptor cross-linking-triggered regulation pathways.

148 McCarron PA, Marouf WM, Quinn DJ et al.: Antibody targeting of camptothecin-loaded PLGA nanoparticles to tumor cells. Bioconjug. Chem. 19, 1561–1569 (2008).

� Along with [146,147], discusses antibody-conjugated nanoparticles inducing receptor cross-linking-triggered regulation pathways.

149 Ferrara N, Gerber HP, LeCouter J: The biology of VEGF and its receptors. Nat. Med. 9, 669–676 (2003).

150 Hendrix MJ, Seftor EA, Kirschmann DA, Quaranta V, Seftor RE: Remodeling of the microenvironment by aggressive melanoma tumor cells. Ann. NY Acad. Sci. 995, 151–161 (2003).

151 Soman C, Giorgio T: Sensitive and multiplexed detection of proteomic antigens via quantum dot aggregation. Nanomedicine 5, 402–409 (2009).

152 Towner RA, Smith N, Asano Y et al.: Molecular magnetic resonance imaging approaches used to aid in the understanding of angiogenesis in vivo: implications for tissue engineering. Tissue Eng. Part A 16, 357–364 (2010).

153 Mukherjee P, Bhattacharya R, Bone N et al.: Potential therapeutic application of gold nanoparticles in B-chronic lymphocytic leukemia (BCLL): enhancing apoptosis. J. Nanobiotechnol. 5, 4 (2007).

154 Chen J, Wu H, Han D, Xie C: Using anti-VEGF McAb and magnetic nanoparticles as double-targeting vector for the radioimmunotherapy of liver cancer. Cancer Lett. 231, 169–175 (2006).

155 Mhawech-Fauceglia P, Smiraglia DJ, Bshara W et al.: Prostate-specific membrane antigen expression is a potential prognostic marker in endometrial adenocarcinoma. Cancer Epidemiol. Biomarkers Prev. 17, 571–577 (2008).

156 Minner S, Wittmer C, Graefen M et al.: High level PSMA expression is associated with early PSA recurrence in surgically treated prostate cancer. Prostate 71(3), 281–288 (2011).

157 Tjoa BA, Simmons SJ, Bowes VA et al.: Evaluation of Phase I/II clinical trials in prostate cancer with dendritic cells and PSMA peptides. Prostate 36, 39–44 (1998).

158 Naylor PH, Hernandez KE, Nixon AE et al.: IRX-2 increases the T cell-specific immune response to protein/peptide vaccines. Vaccine 28(43), 7054–7062 (2010).

159 Henry MD, Wen S, Silva MD, Chandra S, Milton M, Worland PJ: A prostate-specific membrane antigen-targeted monoclonal antibody–chemotherapeutic conjugate designed for the treatment of prostate cancer. Cancer Res. 64, 7995–8001 (2004).

160 Ma D, Hopf CE, Malewicz AD et al.: Potent antitumor activity of an auristatin-conjugated, fully human monoclonal antibody to prostate-specific membrane antigen. Clin. Cancer Res. 12, 2591–2596 (2006).

161 Kuroda K, Liu H, Kim S, Guo M, Navarro V, Bander NH: Saporin toxin-conjugated monoclonal antibody targeting prostate-specific membrane antigen has potent anticancer activity. Prostate 70, 1286–1294 (2010).

Page 14: Antibody-targeted nanoparticles for cancer therapy

Immunotherapy (2011) 3(3)394 future science group

Review Fay & Scott

162 Milowsky MI, Nanus DM, Kostakoglu L, Vallabhajosula S, Goldsmith SJ, Bander NH: Phase I trial of yttrium-90-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 for androgen-independent prostate cancer. J. Clin. Oncol. 22, 2522–2531 (2004).

163 Smith-Jones PM, Vallabhajosula S, Navarro V, Bastidas D, Goldsmith SJ, Bander NH: Radiolabeled monoclonal antibodies specific to the extracellular domain of prostate-specific membrane antigen: preclinical studies in nude mice bearing LNCaP human prostate tumor. J. Nucl. Med. 44, 610–617 (2003).

164 Thomas TP, Patri AK, Myc A et al.: In vitro targeting of synthesized antibody-conjugated dendrimer nanoparticles. Biomacromolecules 5, 2269–2274 (2004).

165 Gao X, Cui Y, Levenson RM, Chung LW, Nie S: In vivo cancer targeting and imaging with semiconductor quantum dots. Nat. Biotechnol. 22, 969–976 (2004).

� In vivo use of anti-prostate-specific membrane antigen antibody-conjugated quantum dots.

166 Cheng Y, Zak O, Aisen P, Harrison SC, Walz T: Structure of the human transferrin receptor–transferrin complex. Cell 116, 565–576 (2004).

167 Gatter KC, Brown G, Trowbridge IS, Woolston RE, Mason DY: Transferrin receptors in human tissues: their distribution and possible clinical relevance. J. Clin. Pathol. 36, 539–545 (1983).

168 Davis ME, Zuckerman JE, Choi CH et al.: Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464, 1067–1070 (2010).

169 Yoon DJ, Kwan BH, Chao FC et al.: Intratumoral therapy of glioblastomamultiforme using genetically engineered transferrin for drug delivery. Cancer Res. 70, 4520–4527 (2010).

170 Koppu S, Oh YJ, Edrada-Ebel R et al.: Tumor regression after systemic administration of a novel tumor-targeted gene delivery system carrying a therapeutic plasmid DNA. J. Control. Release 143, 215–221 (2010).

171 Tacken PJ, de Vries IJ, Torensma R, Figdor CG: Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 7, 790–802 (2007).

172 Kwon YJ, James E, Shastri N, Frechet JM: In vivo targeting of dendritic cells for activation of cellular immunity using vaccine carriers based on pH-responsive microparticles. Proc. Natl Acad. Sci. USA 102, 18264–18268 (2005).

� In vivo use of antibody-targeted nanoparticles as vaccine carriers.

173 Cruz LJ, Tacken PJ, Fokkink R et al.: Targeted PLGA nano- but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. J. Control. Release 144, 118–126 (2010).

174 Skotland T, Iversen TG, Sandvig K: New metal-based nanoparticles for intravenous use: requirements for clinical success with focus on medical imaging. Nanomedicine 6,730–737 (2010).

175 Cannon GJ, Swanson JA: The macrophage capacity for phagocytosis. J. Cell. Sci. 101(Pt 4), 907–913 (1992).

176 Limburg PC, Bijl M: Autoantibodies to opsonins of apoptotic cells in systemic lupus erythematosus. Ann. Rheum. Dis. 64, 1669–1670 (2005).

177 Nobes C, Marsh M: Dendritic cells: new roles for Cdc42 and Rac in antigen uptake? Curr. Biol. 10, R739–R741 (2000).

178 Kolpak AL, Jiang J, Guo D et al.: Negative guidance factor-induced macropinocytosis in the growth cone plays a critical role in repulsive axon turning. J. Neurosci. 29, 10488–10498 (2009).

179 Dauty E, Remy JS, Zuber G, Behr JP: Intracellular delivery of nanometric DNA particles via the folate receptor. Bioconjug. Chem. 13, 831–839 (2002).

180 Pelkmans L, Helenius A: Endocytosis via caveolae. Traffic 3, 311–320 (2002).

181 Conner SD, Schmid SL: Regulated portals of entry into the cell. Nature 422, 37–44 (2003).

182 Wolfe BL, Trejo J: Clathrin-dependent mechanisms of G protein-coupled receptor endocytosis. Traffic 8, 462–470 (2007).

183 Zwang Y, Yarden Y: Systems biology of growth factor-induced receptor endocytosis. Traffic 10, 349–363 (2009).

184 Sun B, Ranganathan B, Feng SS: Multifunctional poly(d,l-lactide-co-glycolide)/montmorillonite (PLGA/MMT) nanoparticles decorated by trastuzumab for targeted chemotherapy of breast cancer. Biomaterials 29, 475–486 (2008).

185 Bernardi RJ, Lowery AR, Thompson PA, Blaney SM, West JL: Immunonanoshells for targeted photothermal ablation in medulloblastoma and glioma: an in vitro evaluation using human cell lines. J. Neurooncol. 86, 165–172 (2008).