You are on page 1of 12

Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

Seminar article
New frontiers in nanotechnology for cancer treatment夡
Frank Alexisa,c, June-Wha Rheea,c, Jerome P. Richieb, Aleksandar F. Radovic-Morenoc,d,e,
Robert Langerc,d,e, Omid C. Farokhzada,c,*
a
Department of Anesthesiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
b
Department of Urology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
c
MIT-Harvard Center for Cancer Nanotechnology Excellence, Cambridge, MA 02139, USA
d
Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
e
Harvard-MIT Division of Health Sciences and Technology, Boston, MA 02115, USA

Abstract
Nanotechnology is a field of research at the crossroads of biology, chemistry, physics, engineering, and medicine. Design of multifunc-
tional nanoparticles capable of targeting cancer cells, delivering and releasing drugs in a regulated manner, and detecting cancer cells with
enormous specificity and sensitivity are just some examples of the potential application of nanotechnology to oncological diseases. In this
review we discuss the recent advances of cancer nanotechnology with particular attention to nanoparticle systems that are in clinical practice
or in various stages of development for cancer imaging and therapy. Published by Elsevier Inc.

Keywords: Cancer therapy; Cancer diagnosis; Nanocarriers; Targeting molecules

Introduction nents alone. Nanomaterials have a large surface area to


volume ratio and their physicochemical properties, such as
Cancer nanotechnology friction and interaction with other molecules, are distinct
from equivalent materials at a larger scale. The most com-
Nanotechnology—the engineering and manufacturing of mon use of nanotechnology in medicine has been in the
materials using atomic and molecular components—is ex- areas of developing novel therapeutic and imaging modal-
pected to benefit all branches of medicine, with oncology ities that have the potential to outperform the current state of
being an early and the most notable beneficiary to date [1,2]. the art in these areas. With the exponential introduction of
In its strictest definition from the National Nanotechnology novel nanotechnology platforms for life science applications,
Initiative (NNI; http://www.nano.gov), nanotechnology re- the potential application of nanotechnology in medicine ex-
fers to structures roughly in the 1 to 100 nm size in at least tends significantly beyond these early uses. The National Can-
one dimension. To put this size range in perspective, a small cer Institute in its 2004 commitment of $144 million toward
molecule, a virus, a bacterium, and the cross section of a the development of novel nanotechnologies for improving can-
human hair are around 1 nm, 100 nm, 1,000 nm, and cer mortality (http://nano.cancer.gov) defined the path of op-
100,000 nm, respectively. Nanotechnology more commonly portunities in six areas including: (1) detection of molecular
refers to structures that are up to several hundred nanome- changes responsible for disease pathogenesis; (2) disease di-
ters in size, and are developed by top-down or bottom-up agnosis and imaging; (3) drug delivery and therapy; (4) mul-
engineering of individual components (i.e., rational design) tifunctional systems for combined therapeutic and diagnostic
[1,3–5]. The resulting nanomaterials may have functional applications; (5) vehicles to report the in vivo efficacy of a
properties that are conferred through the precise assembly therapeutic agent; and (6) nanoscale enabling technologies,
of individual components but not by the individual compo- which will accelerate scientific discovery and basic research.
In this review, we will focus on the application of

nanotechnology to the development of smart drug deliv-
Supported by NIH grants CA119349 and EB003647.
* Corresponding author. Tel.: ⫹1-617-732-6093; fax: ⫹1-617-730-
ery vehicles for cancer therapeutic applications. The most
2801. common examples of these nanoscale delivery vehicles
E-mail address: ofarokhzad@partners.org (O.C. Farokhzad). (also referred to as nanocarriers) include polymeric nano-

1078-1439/08/$ – see front matter Published by Elsevier Inc.


doi:10.1016/j.urolonc.2007.03.017
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 75

particles, dendrimers, nanoshells, liposomes, nucleic acid- coming increasingly recognized with several examples of
based nanoparticles, magnetic nanoparticles, and virus first generation nanocarriers approved by the FDA for ther-
nanoparticles [1]. Nanocarriers have the potential to im- apeutic (Abraxane [11], Doxil [12], DaunoXome [13]) and
prove the therapeutic index of currently available drugs by diagnostic (Feridex [14]) applications (Table 1).
increasing drug efficacy, lowering drug toxicity, and achiev-
ing steady state therapeutic levels of drugs over an extended Targeted delivery of nanocarriers to tumors
period of time. Nanocarriers can also improve drug solubil-
ity and drug stability, allowing the development of poten- The vascularity of tumors is highly heterogeneous, rang-
tially effective new chemical entities that have been stalled ing from areas of vascular necrosis to areas which are
during the preclinical or clinical development because of densely vascularized in order to sustain the adequate supply
suboptimal pharmacokinetic or biochemical properties. Fi- of oxygen and nutrients to the growing tumor. Tumor blood
nally, nanocarriers may also facilitate the development of vessels have several abnormalities compared to normal
multifunctional systems for targeted drug delivery [6,7], blood vessels, including a high proportion of proliferating
combined therapies [8], or systems for simultaneous thera- endothelial cells with aberrant underlying basement mem-
peutic and diagnostic applications. The development of the brane, increased tortuosity of blood vessels, and a defi-
first nanocarrier dates back approximately 40 years, when ciency in pericytes [15]. Tumor microvessels demonstrate
the first example of a liposome was described [9,10]. Over enhanced permeability, which is regulated in part by abnor-
the past 2 decades an increasing number of nanoscale ve- mal secretion of vascular endothelium growth factor, bra-
hicles with distinct physical and biochemical properties dykinin, nitric oxide, prostaglandins, and matrix metallo-
have emerged for drug delivery applications. More recently, proteinases [15]. The transport of macromolecules across
the breakthrough potential of cancer nanotechnology is be- tumor microvasculature may occur through open interendo-
thelial junctions or transendothelial channels. The pore cut-
off size of these transport pathways in various models has
Table 1
been estimated to be in the ⬍1 ␮m range, and in vivo
Nanoparticle platforms for drug delivery
measurement of extravasation of liposomes into tumor
Nanoparticle Material Structure xenografts suggests a cutoff size in the 400 nm range [16].
Polymeric Poly(lactide-co-glycolide) In general, particle extravasation is inversely proportional to
Poly(lactide) size, and smaller particles (⬍200 nm size) would be most
Poly(caprolactone) effective for extravasating the tumor microvasculature
Poly(orthoester)
Poly(anhydride)
[16,17]. The tumor lymphatic system is also abnormal,
Poly(beta-aminoester) resulting in fluid retention in tumors and high interstitial
pressure with an outward convective interstitial fluid flow
[18]. This property is thought to promote tumor cell intra-
Liposome Doxil®/Caelyx®:
vasation, resulting in tumor metastasis and blockage of
PEG-DSPE:HSPC/Cholesterol nanocarrier extravasation from microvasculature into the
(5:56:39) tumor interstitium. However, the lack of an intact lymphatic
DaunoXome®: system also results in retention of the nanocarriers in the
DSPC/Cholesterol (2:1) tumor interstitium since these particles are not readily
cleared from the interstitium. When taken together, the
leaky microvasculature and the lack of intact lymphatic
system results in enhanced permeation and retention (EPR)
effect and “passive” cancer targeting through the accumu-
Dendrimer Poly(amidoamine)
Branched Poly(ethylenimine) lation of nanocarriers in the tumor at a higher concentration
Poly(peptide) that is present in the plasma and in other tissues [19]. The
release of drugs from nanocarriers in this case results in a
relatively higher intratumoral drug concentration translating
into enhanced tumor cytotoxicity. These nanocarriers may
be further modified for “active” cancer targeting by func-
tionalizing the surface of nanocarriers with ligands such as
antibodies, aptamers, peptides, or small molecules that rec-
Polymeric nanoparticles encapsulate both hydrophilic and hydrophobic ognize tumor-specific or tumor-associated antigens in the
small molecule and macromolecular drugs within a polymeric matrix. tumor microenvironment. When nanocarriers are targeted to
Liposomes encapsulate hydrophilic and hydrophobic drugs within the
the extracellular portion of transmembrane tumor antigens,
hollow liposome core and within their hydrophobic membrane, respec-
tively. Dendrimers are branched units with surface functional groups that they may be specifically taken up by cancer cells through
can be used to conjugate therapeutic or diagnostic payloads for subsequent receptor mediated endocytosis [20 –26]. The specific targeting,
release. intracellular uptake, and regulated therapeutic delivery of a
76 F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

payload are properties that are derived through a rational de- ophthalmology [28], endocrinology, [29] orthopedics, [30]
sign of nanocarriers. The application of nanotechnology to pain medicine [31], and oncology [32]. Several examples of
cancer therapy, including the development of “smart” nano- these systems in clinical practice today include Atridox,
particles, is indeed an exciting and promising area of investi- Lupron Depot, Gliadel, Zoladex, Trelstar Depot, and San-
gation. In the following sections, we review some of the most dostatin LAR. The annual worldwide market of controlled
important breakthrough nanotechnology platforms for cancer release polymer systems, which extends beyond drug deliv-
therapeutic applications. ery, is now estimated at $60 billion, and these systems are
used by over 100 million people each year. “Controlled
release” occurs when a natural or synthetic polymer com-
Nanotechnology for cancer treatment bines with a drug in such a way that the drug is encapsulated
within the polymer system for subsequent release in a pre-
Biodegradable polymeric nanoparticles determined manner (Fig. 1). Polymeric drug delivery vehi-
cles can be designed as nanoparticles that release the en-
Virtually every branch of medicine has been dramati- capsulated drugs through surface or bulk erosion, diffusion,
cally impacted by controlled release polymer technologies or swelling, followed by diffusion in a time or condition
during the past four decades, including cardiology [27], dependent manner. The release of the active agent may be

Fig. 1. Schematic representation of nanoparticle targeting. (A) Nanoparticles are concentrated in the tumor interstitium via passive extravasation through the
leaky microvasculature shown as gaps in the endothelial layer (light orange). This process has been named the EPR effect. In this case the efficacy of
nanoparticles is largely mediated through the local release of the drug near the cancer cells. (B) Targeted nanoparticles similarly concentrate in the tumor
interstitium through the EPR effect but once there, nanoparticles are actively taken up by cancer cells after binding to their target antigens on the surface of
the cancer cells. In this case the drugs are released largely inside the cancer cells resulting in enhanced efficacy.
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 77

constant over a long period, cyclic over a long period, or it of degradation. Using the PLA and PLGA polymer systems,
may be triggered by environmental or other external events we have developed proof of concept docetaxel encapsulated
[33] such as changes in pH, [33–36] temperature, [37], or targeted nanoparticles, which target the prostate-specific
the presence of an analyte such as glucose [38]. In general, membrane antigen (PSMA) on the surface of prostate can-
controlled-release polymer systems can provide drug levels cer cells and are engulfed by cells that express the PSMA
in an optimum range over a longer period of time compared protein specifically and efficiently, in vitro and in vivo
to other drug delivery methods. Thus it increases the effi- [5,21,22]. We have shown that a single intratumoral injec-
cacy of the drug and maximizes patient compliance, while tion of these particles results in tumor eradication in 5/7 of
enhancing the ability to use highly toxic, poorly soluble, or the mice, with the remaining 2 mice having considerably
relatively unstable drugs. The primary consideration of drug smaller tumor volumes as compared to controls [22]. These
delivery is to achieve more effective therapies while elim- targeted nanoparticles release their payload directly inside
inating the potential for both under- and overdosing. Other the cancer cells, resulting in enhanced efficacy and decreased
advantages of using controlled release delivery systems systemic toxicity [22]. More recently, using the J591 antibody
include the maintenance of drug levels within a desired which recognizes the PSMA protein, J591-targeted controlled
range, the need for fewer administrations, optimal use of the release nanoparticles have been developed for effective gene
drug in question, and increased patient compliance. This is delivery to prostate cancer cells [52,53]. Our group has also
particularly relevant to cancer therapy in which the effec- developed HER2-targeted nanoparticles that may have util-
tiveness of the treatment is directly related to the ability to ity in the systemic therapy of breast, pancreatic, and ovarian
kill cancer cells while affecting as few healthy cells as cancers (Fig. 2). More recently, our group and other inves-
possible. By administering bolus doses of cytotoxic chemo- tigators have begun to explore computational modeling and
therapeutic drugs, adverse effects are commonly observed screening strategies to identify optimized physicochemical
and these may be sufficiently intense to limit continuation of properties of nanoparticles (i.e., size, charge, hydrophilicity,
the chemotherapy course. stability, drug release kinetics, and the density of targeting
Polymeric nanoparticles may represent the most effec- molecules on nanoparticle surface) for cancer therapeutic
tive nanocarriers for cancer chemotherapy [39,40]. These applications [54,55].
nanoparticles can be surface-functionalized to target cancer
cells specifically and have a prolonged systemic circulating
half-life to enhance their therapeutic efficiency. The nano- Dendrimers
particle surface is usually sterically stabilized by grafting,
conjugating, or adsorbing hydrophilic polymers such as Dendrimers represent a globular macromolecule defined
polyethyleneglycol (PEG) to its surface [41,42]. Polymeric by core and branched units and surface groups [56]. With
nanoparticles can be readily formulated to deliver hydro- well-controlled synthesis methods, new classes of dendrim-
philic or hydrophobic small drug molecules, and polymer ers are emerging for therapeutic and diagnostic applications.
systems have been developed and used to deliver macro- A single dendrimer molecule can carry a therapeutic drug, a
molecules such as proteins [43] and nucleic acids [44]. diagnostic agent, and an active targeting molecule. Den-
Biodegradable polymers are typically broken down into drimers with a hydrophobic core and hydrophilic surface
individual monomers, which are metabolized and removed groups are able to form micelles by hydrophobic/hydro-
from the body via normal metabolic pathways. The rate of philic self-assembly [57–59]. Although the rigidity and the
polymer degradation and subsequent drug release can be density of the branched units can affect the drug release rate
controlled by modification of the polymer side chain, de- [60], much effort is devoted to the design of hybrid struc-
velopment of novel polymers, or synthesis of copolymers. tures using pH or enzymatic sensitive linkages to disrupt
This subject has been an active area of investigation by our dendrimer micelles and trigger the site-specific release of
group and other investigators in academic and industry their payload. Taking advantage of the highly versatile
laboratories for several decades [45– 48]. The result has functionalities of dendrimers, hybrid structures of linear and
been an increasing arsenal of polymers with distinct encap- branched molecules (polymers or biomolecules) are being
sulation and release characteristics for a myriad of research, investigated [61,62]. Biodegradable polyester dendrimers
industrial and clinical applications [49,50]. PGA and PLA based on 2,2-bis(hydroxymethyl)-propanoic acid monomers
are common biocompatible polymers that are used for many have been developed for intracellular release of doxorubicin
biomedical applications. PGA is more hydrophilic since it after hydrolysis of hydrazone linkage [63]. The promising
lacks a methyl group and is more susceptible to hydrolysis, properties of polyester dendrimers as a drug delivery system
making this polymer easily degradable. Alternatively, PLA has led to further studies based on tunable architectures and
is relatively more stable in the body [51]. Through these molecular weights to optimize tumor accumulation [64,65].
unique properties, polymers such as PLGA have been de- The results show that higher molecular weight (ⱖ40 kDa)
rived that are made from both glycolic acid and lactic acid molecules have a longer half-circulation time. The degree of
components. The ability to change the ratio of these two branching also affects the circulation time and renal clear-
components of the polymer can then be used to alter the rate ance of the drug. In a comparative study with Doxil in C-26
78 F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

Fig. 2. (A) Schematic representation of a stealth targeted polymeric nanoparticle. (B) Scanning electron microscope (SEM) micrograph of PLGA
nanoparticles. The white represents 1 ␮m. (C) Comparative plasma drug concentration of polymeric controlled release drug delivery system vs. systemic
delivery of free drug. (D) Fluorescent micrograph of breast cancer cells treated with Her2 targeted nanoparticles loaded with rhodamine. The cell nuclei and
the actin cytoskeleton are stained with blue (DAPI), and green (Alexa-Flour phalloidin), respectively. The intracellular rhodamine-encapsulated Her2 targeted
nanoparticle bioconjugates are shown in red.

colon carcinoma-bearing mice, the polyester dendrimer- layer. In addition, the surface of the nanoshells can be easily
drug conjugate shows similar efficacy to Doxil [66]. functionalized for targeting applications. pH-sensitive doxo-
Biocompatible polyamidoamine (PAMAM) dendrimers rubicin encapsulated nanoshells have been synthesized us-
demonstrate a remarkable molecular monodispersity and ing amphiphilic tercopolymerpoly(N-iso-propylacrylamide-
have been shown to be pH responsive. A variety of den- co-N,N-dimethylacrylamide-co-10-undecenoic acid) that
drimers with different cores (diamino derivatives and eth- can trigger intracellular drug release at pH 6.6. Metallic
ylenediamine) and surface functional groups (alcohol, nanoshells (⬃20 nm) are characterized by a dielectric core
amine, succinic acid, and carboxylic acid) are commercially coated with a thin metallic shell to improve their biocompat-
available. More recently, multifunctional PAMAM den- ibility and optical absorption [68]. These particles possess a
drimers with an imaging agent (fluorescein isothiocyanate highly tunable plasmon resonance mediated by the size of the
FITC), a cancer cell targeting molecule (folic acid) and a core and the thickness of the shell, which in turn determines
therapeutic drug (Taxol) have been described with impres- their absorbing and scattering properties over a broad range on
sive in vitro and in vivo results [67]. the spectrum from the near-ultraviolet to the mid-infrared [68].
Gold nanoshells have been developed for in vivo photothermal
Nanoshells therapy using near infrared light [69]. Similarly, thermally
sensitive polymeric hydrogels and optically active nanoshells
Polymeric nanoshells (20 to 60 nm) of diblock copolymers have been developed for the purpose of photothermally mod-
can assemble into a core/shell structure. These nanoshells can ulated drug delivery. Nanoshell particles with a magnetic core
be made by self-assembly of oppositely charged polymers (carbonyl iron) and a biodegradable poly(butylcyanoacrylate)
forming a thin multilayer structure. The characteristic of (PBCA) shell have also been developed for controlled release
nanoshells is defined by the layer-by-layer assembly of nano- of 5-fluorouracil [70]. The combination of magnetic nanoshells
particles, allowing a high drug loading efficiency. Therefore, and a drug encapsulated biodegradable polymer allows for
the drug release rate is controlled by the properties of the particle targeting to specific sites of the body in response to an
polymers and the diffusion coefficient through the polymeric externally applied magnetic field.
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 79

Liposomes a trivalent RNA core, RNA aptamers for targeting, and


siRNAs for therapeutic effect [83].
Liposomes form nanoparticles with an amphiphilic
unilamellar/multilamellar membrane of natural or synthetic
lipids [9]. Lipids are characterized by a hydrophilic head Targeting molecule platforms
group and a hydrophobic tail. Liposomes form lipid bilayers
through hydrophobic interactions and are able to carry hy- Antibodies or antibody fragments
drophilic and hydrophobic molecules simultaneously. The
hydrophobic and hydrophilic molecules can be encapsulated Antibodies are the first macromolecular ligands used for
into the lipid bilayer and hollow core respectively. This targeted delivery [84,85]. The use of monoclonal antibodies
unique property allows the formulation of a single system for (mAb) became widespread after the discovery of hybridoma
the combination therapy to treat cancer. Liposomes with mark- technology. Due to their inherent immunogenicity, murine
edly prolonged circulation and enhanced tumor accumulation monoclonal antibodies were not suitable for clinical appli-
have been developed by a series of modifications [9]. cations. Engineering antibody technologies led to the devel-
Liposomes have already been approved for routine uses opment of chimeric humanized and fully humanized anti-
in medicine [9,71]. Doxorubicin encapsulated liposome bodies. More recently, methods have been developed to
(Doxil) was the first to gain FDA approval in 1995 and has produce human immunoglobulins from transgenic mice
potent antineoplastic activity against a wide range of human [86]. Combinatorial phage display libraries (⬃1010 different
cancers including Kaposi’s sarcoma [72] and ovarian cancer antibody fragments) have also emerged as a powerful tool to
[73]. The surface of Doxil (100 nm) is functionalized with select novel protein ligands [87]. The latter approach relies
methoxypolyethylene glycol (MPEG, which provides a hy- on multiple selection and screening parameters to select
human antibodies against specific cell types or antigens.
drophilic “stealth” coating, allowing Doxil to circulate in
The selection process can be designed to improve the prop-
the blood stream for a prolonged period of time [74]. The
erties of the ligand such as stability, affinity, selectivity and
clinical success of Doxil has led to numerous new liposome
internalization. Most recently, this approach was used to
formulations to be FDA approved, including DaunoXome
isolate cancer targeting antibodies using live cancer patients
(daunorubicin liposomes), DepoDur (morphine liposomes),
in the selection process [88]. Antibody molecules show
and Ambisome (amphotericin B liposomes). DaunoXome, a
good stability but are limited by their large hydrodynamic
45 nm liposomal formulation, has been shown to be active
diameter (⬃20 nm; Mw ⱖ 150 kDa) to diffuse into tumor
against Kaposi’s sarcoma and other tumors [75]. Despite the tissues [89,90]. Recent advances in protein engineering
clinical success of liposomes, these nanocarriers are limited have led to the development of single chain antibodies,
by suboptimal stability and drug release profiles in vivo. antibody fragments (Fab or scFv) and dibodies [85]. Antibody
Thus, efforts have been focused on the development of fragments show less immune response and can be stabilized
stable and pH-sensitive liposomes that can unload the drug with disulfide bond or chemically crosslinked [91].
in an acidic environment [76 –78]. There are several examples of FDA approved antibodies
in clinical practice today, including Rituxan—a chimeric
Nucleic acid-based nanoparticles (DNA, RNAi, anti-sense) anti-CD20 antibody effective against CD20⫹ B-cell non-
Hodgkin’s lymphomas [92]. Other examples include anti-
HER2 Herceptin [93], anti-EGFR Erbitux [94], and anti-
Gene therapy refers to the transfer and expression of VEGF Avastin [95]. These antibodies function by antago
genes for therapeutic applications in the target cells. In nizing the signal transduction pathway that leads to un
contrast, RNAi [79] and anti-sense [80] therapies are controlled growth in cancer cells. Recent investigations [96]
based on oligonucleotides that will shut down the gene have focused on exploiting the affinity of these antibodies
expression of a target gene involved in a pathway of the for their respective antigens to “arm” them with chemother-
disease. Aptamers are nucleic acid ligands that bind to apy drugs (doxorubicin [97], methotrexate [98], and cali-
target proteins with high affinity and specificity, are anal- cheamicin [99]), toxins, or radionucleotides [100]. Despite
ogous to antibodies, and exert a biological effect [81]. much efforts, Mylotarg [99] is the only FDA approved
The greatest impact of these technologies is expected to antibody-toxin conjugate in practice today. The payload and
be in cancer therapy where there is a differentially ab- the chemical conjugation of the drug to antibodies remain
normal pattern of gene or protein expression. DNA and the major limitations of chemoimmunoconjugates. Targeted
RNA macromolecules can be used as substrates for de- drug delivery carriers are being functionalized with antibod-
veloping therapeutic and imaging nanocarriers. A multi- ies or antibody fragments to overcome some of these limi-
valent DNA delivery vehicle (100 nm) was recently re- tations [20,101–103]. Furthermore, the conjugation of mul-
ported for simultaneous targeted drug delivery, imaging, tiple antibodies to each nanocarrier enhances their avidity,
and gene therapy [82]. Targeted multifunctional RNA and nanocarriers can be surface functionalized with multiple
nanoparticles (25– 40 nm) have also been developed with distinct antibodies to overcome tumor heterogeneity. Long-
80 F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

circulating doxorubicin encapsulated immunoliposomes spleen and liver tissues due to macrophage clearance [121].
conjugated to human HER2 antibodies have been developed Ex-vivo selection of targeted peptide on biopsy specimens
and evaluated in vivo [20]. After intravenous administra- have been developed and shown to successfully isolate
tion, nontargeted and targeted liposomes similarly accumu- potent peptide ligands. In vivo phage library selection was
lated in the tumors; however, targeted liposomes had a signif- pioneered by Arap et al. [114] to further optimize the se-
icantly higher anti-tumor efficacy, presumably due to the lection parameters. The discovery of potent peptide se-
HER2 mediated internalization of targeted liposomes and in- quences depends on the design of the selection protocols
tracellular drug release vs. the nontargeted liposomes that re- and it is recognized that it is critical for the isolation of most
leased the drug locally into the extracellular space [20]. promising molecules. The ultimate conditions should be as
close as possible to the human physiological environment
Peptides and tumor structure. Thus, the in-vivo phage is designed to
select peptides that bind tumor vasculatures and to allow
Research using antibodies for targeting applications has distinction between common vascular cell receptors and
led to a better understanding of critical factors affecting specific tissues’ receptor-binding peptides. Possible limita-
targeting. Peptides and antibody fragments have been de- tions of using animal models that might not completely
veloped to overcome some of the shortcomings of antibod- represent the human vasculature profile have led to the
ies, and several examples of these ligands are now under selection of peptide libraries directly on patients. This ap-
clinical development. It is now well accepted that the bind- proach has already been reported for various cancers. Pep-
ing affinity, stability, and the size of the ligand play a tide sequence binding to human interleukin (IL-11) receptor
critical role for successful targeting. Peptides are small, on prostate cancers was characterized. However, the sur-
synthetic molecules that can be manufactured in large quan- vival time was only extended by 20 days. The development
tities with excellent quality control. Peptides are more stable of selection protocols, ethical guidelines, and nanotechnolo-
than antibodies and unlikely to be immunogenic. The dis- gies will lead to the clinical translation of new imaging and
covery of new peptide targeting domains has been success- therapeutic technologies.
ful due to the development of peptide library screening Other peptide-based targeting technologies have been
methods (e.g., phage display; ⬃1011 different peptides) developed that provide several new platforms for generating
[88,104,105]. Display technologies are powerful tools to target specificity. Using the A-domain of various native
isolate novel ligands for therapeutic and diagnostic applica- human receptors as a platform for modification, single-
tions. Twenty five years ago, Scott and Smith reported the chain proteins that can bind to multiple regions of target
first peptide phage library [106]. Combinatorial peptide molecules using multiple domains have been developed and
libraries are defined by a large pool of random sequences or named avidity multimers or avimers [122]. In a proof of
based on a backbone sequence with random mutations. concept study, an avimer specific for IL-6 (an acute phase
Large libraries of linear or constrained peptides are cur- inflammatory mediator) was demonstrated to be non-immu-
rently available [107]. In general, circular stabilized or mul- nogenic in mice, as well as an inhibitor of IL-6 function in
tivalent peptides show higher binding affinity due to en- vivo. More generally, this highlights the potential of this
hanced rigidity and interaction with cell membrane class of ligand to antagonize proteins, which may ultimately
antigens. In most cases, short peptides are selected after find application in cancer therapy. Functional, single-do-
affinity screening of the library on a target protein, whole main heavy chain antibodies, also known as nanobodies,
cell, ex vivo tissues or live animals and even humans have been raised against cancer targets, which either antag-
[104,108 –110]. A large number of high affinity peptides onize receptor function [123] or deliver an enzyme for
(Kd in uM–nM range) with potential for targeting applica- prodrug activation [124], both for therapeutic benefit. Yet
tion have been isolated against a variety of cell surface another important strategy employing phage display tech-
antigens [108,111–113]. Peptides that contain RGD (Arg- nology used a 58 residue ␣-helical domain of staphylococ-
Gly-Asp) domains can preferentially bind cells in tumor cal protein A as a platform to develop polypeptide targeting
microvasculature that express the ␣(V) ␤3 integrin ligands named affibodies [125]. Affibodies against a variety
[109,114]. This strategy has been used experimentally to of cancer-related targets have been developed and are now
deliver chemotherapy and radioactive molecules selectively commercially available, including: EGFR, HER2, and
to tumors [115–117]. Moreover, cyclic RGD peptide (EMD transferrin.
121974 or Cilengitide) is now under phase II clinical eval-
uation for the treatment of several tumors including andro- Small molecule targeting
gen independent prostate cancer and recurrent Glioblastoma
multiforme [118,119]. However, RGD sequences also act In general, small molecules are very attractive as target-
as adhesive molecules and can non-specifically bind tissues ing ligands due to their low cost and ease to conjugate with
that also express its integrin complement. Integrin receptors drugs such as imaging probes (quantum dots, etc.) and
are also expressed on the cell membrane of macrophages nanoparticles [126,127]. The small size of the targeting
[120] and it is shown that RGD bioconjugates aggregate in ligand allows the functionalization of multiple molecules on
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 81

single nanoparticles. In this regard, folic acid and sugar Systemic evolution of ligands by exponential enrichment
molecules have been extensively used. The vitamin folic (SELEX) (⬃1015 different sequences) has been shown to be
acid is required for cell survival and folate receptors are very powerful for the selection of high affinity targeted
homogeneously over-expressed in many cancer cells ligands, and multiple selection protocols are reviewed else-
[128,129]. Folic acid receptors (FR) interact with high af- where [143]. The unique antigen-aptamer binding mecha-
finity folic acid conjugates and transport the conjugates via nism has led to the isolation of ligands with high binding
receptor mediated endocytosis, before it recycles back to the affinity. It was shown that aptamer-antigen interaction oc-
cell surface [130]. Thus, it is not surprising that folate- curs by a complex mechanism involving the folding of the
targeted nanoparticles have shown to be effective in a num- aptamer [144] on its antigen like a “paper clip”. In the short
ber of tumors using liposomes [35,131–133] or polyplexed time since Szostak and coworkers [145] and Tuerk and Gold
nanosystems [134,135]. However, immunochemistry stud- [146] independently described the ground-breaking meth-
ies have shown the overexpression of folate receptors in odology for in vitro evolution of aptamers, this class of
normal tissues like placenta and kidneys. This raises some ligands has emerged as an important arsenal in research and
issues for possible clinical translation of folate bioconju- clinical medicine. Aptamers offer the substantial advantage
gates. In addition, folate-targeted stealth nanoparticles need of synthetic materials that can be produced in preparative
to be carefully designed to avoid possible steric hindrance scale with high purity and reproduction. The consideration
of free PEG chains. Ideally, the folate ligand should be of such favorable characteristics of aptamers has resulted in
accessible to enhance the specific binding of folate biocon- their rapid progress into clinical practice. We began to
jugates [136]. Cell surface lectins have been shown to be exploit this class of molecules for targeted delivery of con-
overexpressed on numerous cancer cells [137,138] and to trolled drug releasing polymer vehicle. Recently we de-
internalize glycoconjugates via a receptor mediated endo- scribed the first proof-of-concept drug delivery vehicles
cytosis mechanism. The presence of cell surface membrane utilizing aptamers for targeted delivery of drug encapsulated
lectins, which recognize specific sugar molecules (lactose, nanoparticles [21], and subsequently showed the efficacy of
galactose, mannose) represents an interesting approach for similarly designed nanoparticles against prostate cancer tu-
targeted delivery or imaging. However, to compensate for mors in vivo [22]. The efficacy study shows that a single
the weak binding affinity of carbohydrates, multiple or intratumoral injection of aptamer targeted nanoparticles
multivalent molecules should be conjugated to the nanocar- loaded with docetaxel allowed all the treated mice to sur-
rier. Targeting efficacy of galactosylated macromolecular vive more than three months in contrast to other controls.
carriers were shown to depend on galactose ligand concen- More recently, we reported a novel strategy for targeted
tration [139]. In addition, the conjugation of galactoside doxorubicin delivery to cancer cells using an aptamer-Dox
residues with N-(2-hydroxypropyl) methacrylamide copol- physical conjugate [147]. Doxorubicin is known to interca-
ymers (HPMA) resulted in a significant increase in three late within the DNA strand due to the presence of flat
different colon cancer cells [140]. Surprisingly, the trivalent aromatic rings of this molecule. We demonstrated that
galactoside bioconjugates did not show higher binding af- doxorubicin could be intercalated in aptamers and released
finity. Recently, Weissleder et al. [54] have demonstrated over a period of 6 hours and, most importantly, without
that small molecules can change the biological affinity of altering its affinity to bind to PSMA antigens.
nanoparticles used for imaging specific tissues. Using a Other strategies involving aptamers in cancer therapy
high-throughput screening based on surface chemistry, 146 have included targeting the growth factors PDGF [148,149]
different small molecules (ⱕ500 Da) were conjugated to and VEGF [150] for delivery of therapeutic or diagnostic
nanoparticles and the results showed that very small mole- agents, discovering new cancer-associated antigens using
cules can drastically affect the binding of nanoparticles to cell-SELEX [151], and delivering toxins [152] or siRNA
different cell lines and pancreatic tumor xenografts. [153].

Aptamers
Summary
Nucleic acid aptamers are single stranded DNA, RNA or
unnatural oligonucleotides that fold into unique structures The principal goals of cancer nanotechnology are to
capable of binding to specific targets with high affinity and develop safer yet more effective diagnostic and therapeutic
specificity [141]. Nucleic acid aptamers are a novel class of modalities for the fight against cancer. Drug encapsulated
ligands [142] that are small (10 –20 kDa), non-immuno- targeted nanoparticles are a promising class of therapeutics
genic, easy to isolate, and exhibit high specificity and af- with the potential to improve efficacy and safety of cur-
finity for their target antigen, at best in the picomolar scale. rently available drugs. There are also opportunities to create
In addition, aptamers are synthetic molecules that can be entirely novel therapeutics using nanotechnology. There is
easily modified and scaled up for synthesis. The utility of an increasing arsenal of nanotechnology platforms under
aptamers for possible clinical translation has been possible evaluation including polymeric nanoparticles, liposomes,
due to chemical modification allowing a higher stability. nanoshells, dendrimers, and polynucleotide nanoparticles.
82 F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

There are also increasing targeting molecule platforms that [11] Green MR, Manikhas GM, Orlov S, et al. Abraxane, a novel Cre-
have emerged for use in the engineering of nanocarriers. mophor-free, albumin-bound particle form of paclitaxel for the treat-
ment of advanced non-small-cell lung cancer. Ann Oncol 2006;17:
These include antibody and antibody fragments, peptides, 1263– 8.
aptamers, small molecules, and carbohydrates. The intrinsic [12] Ellerhorst JA, Bedikian A, Ring S, et al. Phase II trial of Doxil for
properties of the nanocarriers including the density of li- patients with metastatic melanoma refractory to frontline therapy.
gands on their surface might affect the treatment efficacy Oncol Rep 1999;6:1097–9.
and should be optimized. Targets within the vasculature are [13] Fassas A, Anagnostopoulos A. The use of liposomal daunorubicin
(DaunoXome) in acute myeloid leukemia. Leuk Lymphoma 2005;
readily accessible for bioconjugates to be internalized, but 46:795– 802.
physical and physiological barriers in solid tumors might [14] Wang YX, Hussain SM, Krestin G. Superparamagnetic iron oxide
limit the diffusion of therapeutic nanocarriers to reach the contrast agents: Physicochemical characteristics and applications in
target cells. Each class of nanoparticles might be useful for MR imaging. Eur Radiol 2001;11:2319 –31.
a particular application based on specific physiological en- [15] Jain RK. Delivery of molecular and cellular medicine to solid
tumors. Adv Drug Deliv Rev 2001;46:149 – 68.
vironment and clinical requirements. Hence, multifunc- [16] Yuan F, Dellian M, Fukumura D, et al. Vascular permeability in a
tional nanocarriers are now emerging to combine radiother- human tumor xenograft: Molecular size dependence and cutoff size.
apy and chemotherapy, with or without imaging properties. Cancer Res 1995;55:3752– 6.
To move these technologies forward, novel approaches are [17] Kong G, Braun RD, Dewhirst M. Hyperthermia enables tumor-
needed to accelerate the discovery process, such as synthe- specific nanoparticle delivery: Effect of particle size. Cancer Res
2000;60:4440 –5.
sis and screening automation. Future areas of nanotechnol- [18] Jain RK. Transport of molecules in the tumor interstitium: A review.
ogy innovation for cancer treatment include the develop- Cancer Res 1987;47:3039 –51.
ment of bioresponsive and self-regulated delivery systems. [19] Maeda H, Wu J, Sawa T, et al. Tumor vascular permeability and the
These are truly exciting times for medicine and, with con- EPR effect in macromolecular therapeutics: A review. J Control
tinual support, medicine will continue to be a major bene- Release 2000;65:271– 84.
[20] Kirpotin DB, Drummond DC, Shao Y, et al. Antibody targeting of
ficiary of nanotechnology for years to come.
long-circulating lipidic nanoparticles does not increase tumor local-
ization but does increase internalization in animal models. Cancer
Res 2006;66:6732– 40.
Acknowledgments [21] Farokhzad OC, Jon S, Khademhosseini A, et al. Nanoparticle-
aptamer bioconjugates: A new approach for targeting prostate can-
cer cells. Cancer Res 2004;64:7668 –72.
This work was supported by NIH/NCI CA119349, NIH/ [22] Farokhzad OC, Cheng J, Teply BA, et al. Targeted nanoparticle-
NIBIB EB003647, and a development grant from the DFHCC aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl
Prostate SPORE. Acad Sci USA 2006;103:6315–20.
[23] Chiu SJ, Ueno NT, Lee RJ. Tumor-targeted gene delivery via anti-
HER2 antibody (trastuzumab, Herceptin) conjugated poly ethyleni-
mine. J Control Release 2004;97:357– 69.
References [24] Cheng J, Teply BA, Sherifi I, et al. Formulation of functionalized
PLGA-PEG nanoparticles for in vivo targeted drug delivery. Bio-
[1] Ferrari M. Cancer nanotechnology: Opportunities and challenges. materials 2007;28:869 –76.
Nat Rev Cancer 2005;5:161–71. [25] Bagalkot V, Farokhzad OC, Langer R, et al. An aptamer-doxorubi-
[2] Farokhzad OC, Langer R. Nanomedicine: Developing smarter ther- cin physical conjugate as a novel targeted drug-delivery platform.
apeutic and diagnostic modalities. Adv Drug Deliv Rev 2006;58: Angew Chem Int Ed Engl 2006;45:8149 –52.
1456 –9. [26] Arencibia JM, Schally AV, Krupa M, et al. Targeting of doxorubicin
[3] Whitesides GM. The “right” size in nanobiotechnology. Nat Bio- to ES-2 human ovarian cancers in nude mice by linking to an analog
technol 2003;21:1161–5. of luteinizing hormone-releasing hormone improves its effective-
[4] LaVan DA, McGuire T, Langer R. Small-scale systems for in vivo ness. Int J Oncol 2001;19:571–7.
drug delivery. Nat Biotechnol 2003;21:1184 –91. [27] Guzman LA, Labhasetwar V, Song C, et al. Local intraluminal
[5] Farokhzad OC, Karp JM, Langer R. Nanoparticle-aptamer bioconju- infusion of biodegradable polymeric nanoparticles. A novel ap-
gates for cancer targeting. Expert Opin Drug Deliv 2006;3:311–24. proach for prolonged drug delivery after balloon angioplasty. Cir-
[6] Bae Y, Jang WD, Nishiyama N, et al. Multifunctional polymeric culation 1996;94:1441– 8.
micelles with folate-mediated cancer cell targeting and pH-triggered [28] Bourlais CL, Acar L, Zia H, et al. Ophthalmic drug delivery sys-
drug releasing properties for active intracellular drug delivery. Mol tems—recent advances. Prog Retin Eye Res 1998;17:33–58.
Biosyst 2005;1:242–50. [29] Eerikainen H, Kauppinen EI. Preparation of polymeric nanoparticles
[7] Nasongkla N, Bey E, Ren J, et al. Multifunctional polymeric mi- containing corticosteroid by a novel aerosol flow reactor method. Int
celles as cancer-targeted, MRI-ultrasensitive drug delivery systems. J Pharmacol 2003;263:69 – 83.
Nano Lett 2006;6:2427–30. [30] Holland TA, Mikos AG. Biodegradable polymeric scaffolds. Im-
[8] Larina IV, Evers BM, Ashitkov TV, et al. Enhancement of drug provements in bone tissue engineering through controlled drug de-
delivery in tumors by using interaction of nanoparticles with ultra- livery. Adv Biochem Eng Biotechnol 2006;102:161– 85.
sound radiation. Technol Cancer Res Treat 2005;4:217–26. [31] Schroder U, Sabel BA. Nanoparticles, a drug carrier system to pass
[9] Torchilin VP. Recent advances with liposomes as pharmaceutical the blood-brain barrier, permit central analgesic effects of i.v. dalar-
carriers. Nat Rev Drug Discov 2005;4:145– 60. gin injections. Brain Res 1996;710:121– 4.
[10] Bangham AD, Standish MM, Watkins JC. Diffusion of univalent [32] Moghimi SM. Recent developments in polymeric nanoparticle en-
ions across the lamellae of swollen phospholipids. J Mol Biol 1965; gineering and their applications in experimental and clinical oncol-
13:238 –52. ogy. Anticancer Agents Med Chem 2006;6:553– 61.
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 83

[33] Kost J, Langer R. Responsive polymeric delivery systems. Adv [58] Lee CC, MacKay JA, Frechet JM, et al. Designing dendrimers for
Drug Deliv Rev 2001;46:125– 48. biological applications. Nat Biotechnol 2005;23:1517–26.
[34] Shenoy DB, Amiji MM. Poly(ethylene oxide)-modified poly(␧-ca- [59] Liu M, Kono K, Frechet JM. Water-soluble dendritic unimolecular
prolactone) nanoparticles for targeted delivery of tamoxifen in micelles: Their potential as drug delivery agents. J Control Release
breast cancer. Int J Pharmacol 2005;293:261–70. 2000;65:121–31.
[35] Shmeeda H, Mak L, Tzemach D, et al. Intracellular uptake and [60] Gillies ER, Jonsson TB, Frechet JM. Stimuli-responsive supramo-
intracavitary targeting of folate-conjugated liposomes in a mouse lecular assemblies of linear-dendritic copolymers. J Am Chem Soc
lymphoma model with up-regulated folate receptors. Mol Cancer 2004;126:11936 – 43.
Ther 2006;5:818 –24. [61] Gupta U, Agashe HB, Asthana A, et al. Dendrimers: Novel poly-
[36] Yin X, Hoffman AS, Stayton PS. Poly(N-isopropylacrylamide-co- meric nanoarchitectures for solubility enhancement. Biomacromol-
propylacrylic acid) copolymers that respond sharply to temperature ecules 2006;7:649 –58.
and pH. Biomacromolecules 2006;7:1381–5. [62] Qiu LY, Bae YH. Polymer architecture and drug delivery. Pharma-
[37] Furgeson DY, Dreher MR, Chilkoti A. Structural optimization of a col Res 2006;23:1–30.
“smart” doxorubicin-polypeptide conjugate for thermally targeted [63] Padilla De Jesus OL, Ihre HR, et al. Polyester dendritic systems for
delivery to solid tumors. J Control Release 2006;110:362–9. drug delivery applications: In vitro and in vivo evaluation. Biocon-
[38] Brown LR, Edelman ER, Fischel-Ghodsian F, et al. Characterization jugate Chem 2002;13:453– 61.
of glucose-mediated insulin release from implantable polymers. [64] Gillies ER, Dy E, Frechet JM, et al. Biological evaluation of poly-
J Pharmacol Sci 1996;85:1341–5. ester dendrimer: Poly(ethylene oxide) “bow-tie” hybrids with tun-
[39] Gref R, Minamitake Y, Peracchia MT, et al. Biodegradable long- able molecular weight and architecture. Mol Pharmacol 2005;2:
circulating polymeric nanospheres. Science 1994;263:1600 –3. 129 –38.
[40] Moghimi SM, Hunter AC, Murray JC. Long-circulating and target- [65] Gillies ER, Frechet JM. Designing macromolecules for therapeutic
specific nanoparticles: Theory to practice. Pharmacol Rev 2001;53: applications: Polyester dendrimer-poly(ethylene oxide) “bow-tie”
283–318. hybrids with tunable molecular weight and architecture. J Am Chem
[41] Gref R, Luck M, Quellec P, et al. “Stealth” corona-core nanopar- Soc 2002;124:14137– 46.
ticles surface modified by polyethylene glycol (PEG): Influences of [66] Lee CC, Gillies ER, Fox ME, et al. A single dose of doxorubicin-
the corona (PEG chain length and surface density) and of the core functionalized bow-tie dendrimer cures mice bearing C-26 colon
composition on phagocytic uptake and plasma protein adsorption.
carcinomas. Proc Natl Acad Sci USA 2006;103:16649 –54.
Colloids Surf B Biointerfaces 2000;18:301–13.
[67] Majoros IJ, Myc A, Thomas T, et al. PAMAM dendrimer-based
[42] Owens DE III, Peppas NA. Opsonization, biodistribution, and phar-
multifunctional conjugate for cancer therapy: Synthesis, character-
macokinetics of polymeric nanoparticles. Int J Pharmacol 2006;307:
ization, and functionality. Biomacromolecules 2006;7:572–9.
93–102.
[68] Hirsch LR, Gobin AM, Lowery AR, et al. Metal nanoshells. Ann
[43] Tobio M, Gref R, Sanchez A, et al. Stealth PLA-PEG nanoparticles
Biomed Eng 2006;34:15–22.
as protein carriers for nasal administration. Pharmacol Res 1998;15:
[69] Hirsch LR, Stafford RJ, Bankson JA, et al. Nanoshell-mediated
270 –5.
near-infrared thermal therapy of tumors under magnetic resonance
[44] Perez C, Sanchez A, Putnam D, et al. Poly(lactic acid)-poly(ethylene
guidance. Proc Natl Acad Sci USA 2003;100:13549 –54.
glycol) nanoparticles as new carriers for the delivery of plasmid
[70] Arias JL, Gallardo V, Linares-Molinero F, et al. Preparation and
DNA. J Control Release 2001;75:211–24.
characterization of carbonyl iron/poly(butylcyanoacrylate) core/
[45] Langer R, Tirrell DA. Designing materials for biology and medicine.
shell nanoparticles. J Colloid Interface Sci 2006;299:599 – 607.
Nature 2004;428:487–92.
[71] Lian T, Ho RJ. Trends and developments in liposome drug delivery
[46] Langer R, Folkman J. Polymers for the sustained release of proteins
and other macromolecules. Nature 1976;263:797– 800. systems. J Pharmacol Sci 2001;90:667– 80.
[47] Langer R. Drug delivery and targeting. Nature 1998;392:5–10. [72] James ND, Coker RJ, Tomlinson D, et al. Liposomal doxorubicin
[48] Langer R. New methods of drug delivery. Science 1990;249: (Doxil): An effective new treatment for Kaposi’s sarcoma in AIDS.
1527–33. Clin Oncol (R Coll Radiol) 1994;6:294 – 6.
[49] Langer R. Drug delivery. Drugs on target. Science 2001;293:58 –9. [73] Gibbs DD, Pyle L, Allen M, et al. A phase I dose-finding study of a
[50] Langer R, Peppas NA. Advances in biomaterials, drug delivery, and combination of pegylated liposomal doxorubicin (Doxil), carboplatin
bionanotechnology. AICHE J 2003;49:2990 –3006. and paclitaxel in ovarian cancer. Br J Cancer 2002;86:1379 – 84.
[51] Matsusue Y, Hanafusa S, Yamamuro T, et al. Tissue reaction of [74] Gabizon A, Shmeeda H, Barenholz Y. Pharmacokinetics of pegy-
bioabsorbable ultra high strength poly (L-lactide) rod. A long-term lated liposomal Doxorubicin: Review of animal and human studies.
study in rabbits. Clin Orthop Relat Res 1995;246 –53. Clin Pharmacokinet 2003;42:419 –36.
[52] Moffatt S, Papasakelariou C, Wiehle S, et al. Successful in vivo [75] Sadava D, Coleman A, Kane SE. Liposomal daunorubicin over-
tumor targeting of prostate-specific membrane antigen with a highly comes drug resistance in human breast, ovarian, and lung carcinoma
efficient J591/PEI/DNA molecular conjugate. Gene Ther 2006;13: cells. J Liposome Res 2002;12:301–9.
761–72. [76] Reddy JA, Low PS. Enhanced folate receptor mediated gene therapy
[53] Moffatt S, Cristiano RJ. Pegylated J591 mAb loaded in PLGA-PEG- using a novel pH-sensitive lipid formulation. J Control Release
PLGA tri-block copolymer for targeted delivery: In vitro evaluation 2000;64:27–37.
in human prostate cancer cells. Int J Pharmacol 2006;317:10 –3. [77] Slepushkin VA, Simoes S, Dazin P, et al. Sterically stabilized
[54] Weissleder R, Kelly K, Sun EY, et al. Cell-specific targeting of pH-sensitive liposomes. Intracellular delivery of aqueous contents
nanoparticles by multivalent attachment of small molecules. Nat and prolonged circulation in vivo. J Biol Chem 1997;272:2382– 8.
Biotechnol 2005;23:1418 –23. [78] Turk MJ, Reddy JA, Chmielewski JA, et al. Characterization of a
[55] Decuzzi P, Lee S, Bhushan B, et al. A theoretical model for the novel pH-sensitive peptide that enhances drug release from folate-
margination of particles within blood vessels. Ann Biomed Eng targeted liposomes at endosomal pH. Biochim Biophys Acta 2002;
2005;33:179 –90. 1559:56 – 68.
[56] Sampathkumar SG, Yarema KJ. Targeting cancer cells with den- [79] Cheng JC, Moore TB, Sakamoto KM. RNA interference and human
drimers. Chem Biol 2005;12:5– 6. disease. Mol Genet Metab 2003;80:121– 8.
[57] Gillies ER, Frechet JM. Dendrimers and dendritic polymers in drug [80] Chi KN, Gleave ME. Antisense approaches in prostate cancer.
delivery. Drug Discov Today 2005;10:35– 43. Expert Opin Biol Ther 2004;4:927–36.
84 F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85

[81] Wilson DS, Szostak JW. In vitro selection of functional nucleic [105] Newton JR, Kelly KA, Mahmood U, et al. In vivo selection of phage
acids. Annu Rev Biochem 1999;68:611– 47. for the optical imaging of PC-3 human prostate carcinoma in mice.
[82] Li Y, Tseng YD, Kwon SY, et al. Controlled assembly of den- Neoplasia 2006;8:772– 80.
drimer-like DNA. Nat Mater 2004;3:38 – 42. [106] Scott JK, Smith GP. Searching for peptide ligands with an epitope
[83] Khaled A, Guo S, Li F, et al. Controllable self-assembly of nano- library. Science 1990;249:386 –90.
particles for specific delivery of multiple therapeutic molecules to [107] Landon LA, Zou J, Deutscher SL. Is phage display technology on
cancer cells using RNA nanotechnology. Nano Lett 2005;5:1797– target for developing peptide-based cancer drugs? Curr Drug Discov
808. Technol, 2004;1:113–32.
[84] Maynard J, Georgiou G. Antibody engineering. Annu Rev Biomed [108] Kelly KA, Nahrendorf M, Yu AM, et al. In vivo phage display
Eng, 2000;2:339 –76. selection yields atherosclerotic plaque targeted peptides for imaging.
[85] Weiner LM, Adams GP. New approaches to antibody therapy. Mol Imaging Biol 2006;8:201–7.
Oncogene 2000;19:6144 –51. [109] Pasqualini R, Koivunen E, Ruoslahti E. ␣-V integrins as receptors
[86] Lonberg N. Human antibodies from transgenic animals. Nat Bio- for tumor targeting by circulating ligands. Nat Biotechnol 1997;15:
technol 2005;23:1117–25. 542– 6.
[87] McWhirter JR, Kretz-Rommel A, Saven A, et al. Antibodies se- [110] Krag DN, Shukla GS, Shen G-P, et al. Selection of tumor-binding
lected from combinatorial libraries block a tumor antigen that plays ligands in cancer patients with phage display libraries. Cancer Res
a key role in immunomodulation. Proc Natl Acad Sci USA 2006; 2006;66:7724 –33.
103:1041– 6. [111] Wolf P, Gierschner D, Buhler P, et al. A recombinant PSMA-
[88] Krag DN, Shukla GS, Shen GP, et al. Selection of tumor-binding specific single-chain immunotoxin has potent and selective toxicity
ligands in cancer patients with phage display libraries. Cancer Res against prostate cancer cells. Cancer Immunol Immunother 2006;
2006;66:7724 –33. 55:1367–73.
[89] Clauss MA, Jain RK. Interstitial transport of rabbit and sheep antibodies [112] Welply JK, Steininger CN, Caparon M, et al. A peptide isolated by
in normal and neoplastic tissues. Cancer Res 1990;50:3487–92. phage display binds to ICAM-1 and inhibits binding to LFA-1.
[90] Graff CP, Wittrup KD. Theoretical analysis of antibody targeting of Proteins 1996;26:262–70.
tumor spheroids: Importance of dosage for penetration, and affinity [113] Belimezi MM, Papanastassiou D, Merkouri E, et al. Growth inhi-
for retention. Cancer Res 2003;63:1288 –96. bition of breast cancer cell lines overexpressing Her2/neu by a novel
[91] Presta LG. Engineering of therapeutic antibodies to minimize im-
internalized fully human Fab antibody fragment. Cancer Immunol
munogenicity and optimize function. Adv Drug Deliv Rev 2006;58:
Immunother 2006;55:1091–9.
640 –56.
[114] Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by targeted
[92] Nowakowski GS, Witzig TE. Radioimmunotherapy for B-cell non-
drug delivery to tumor vasculature in a mouse model. Science
Hodgkin lymphoma. Clin Adv Hematol Oncol 2006;4:225–31.
1998;279:377– 80.
[93] Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab
[115] Bibby DC, Talmadge JE, Dalal MK, et al. Pharmacokinetics and
after adjuvant chemotherapy in HER2-positive breast cancer. N Engl
biodistribution of RGD-targeted doxorubicin-loaded nanoparticles
J Med 2005;353:1659 –72.
in tumor-bearing mice. Int J Pharmacol 2005;293:281–90.
[94] Reck M, Kranich AL, Steinbach AK, et al. EGFR/HER1 inhibition:
[116] Mitra A, Mulholland J, Nan A, et al. Targeting tumor angiogenic
An example of new targeted therapies in non-small cell lung cancer.
vasculature using polymer-RGD conjugates. J Control Release
Med Klin, Munich 2005;100:785–93.
2005;102:191–201.
[95] Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the
[117] Schiffelers RM, Koning GA, ten Hagen TL, et al. Antitumor effi-
VEGF-specific antibody bevacizumab has antivascular effects in
human rectal cancer. Nat Med 2004;10:145– 47. cacy of tumor vasculature-targeted liposomal doxorubicin. J Control
[96] Schrama D, Reisfeld RA, Becker JC. Antibody targeted drugs as Release 2003;91:115–22.
cancer therapeutics. Nat Rev Drug Discov 2006;5:147–59. [118] Beekman KW, Colevas AD, Cooney K, et al. Phase II evaluations of
[97] Inoh K, Muramatsu H, Torii S, et al. Doxorubicin-conjugated anti- cilengitide in asymptomatic patients with androgen-independent
midkine monoclonal antibody as a potential anti-tumor drug. Jpn prostate cancer: Scientific rationale and study design. Clin Genito-
J Clin Oncol 2006;36:207–11. urin Cancer 2006;4:299 –302.
[98] Vitols KS, Haag-Zeino B, Baer T, et al. Methotrexate-␣-phenylal- [119] Eskens FA, Dumez H, Hoekstra R, et al. Phase I and pharmacoki-
anine: Optimization of methotrexate prodrug for activation by car- netic study of continuous twice weekly intravenous administration
boxypeptidase A-monoclonal antibody conjugate. Cancer Res 1995; of Cilengitide (EMD 121974), a novel inhibitor of the integrins
55:478 – 81. ␣V␤3 and ␣V␤5 in patients with advanced solid tumors. Eur J
[99] Sievers EL, Linenberger M. Mylotarg: Antibody-targeted chemo- Cancer, 2003;39:917–26.
therapy comes of age. Curr Opin Oncol 2001;13:522–7. [120] Savill J, Dransfield I, Hogg N, et al. Vitronectin receptor-mediated
[100] Sharkey RM, Goldenberg DM. Targeted therapy of cancer: New phagocytosis of cells undergoing apoptosis. Nature 1990;343:170 –3.
prospects for antibodies and immunoconjugates. CA Cancer J Clin [121] Xiong XB, Huang Y, Lu WL, et al. Intracellular delivery of doxo-
2006;56:226 – 43. rubicin with RGD-modified sterically stabilized liposomes for an
[101] Nielsen UB, Kirpotin DB, Pickering EM, et al. Therapeutic efficacy improved antitumor efficacy: In vitro and in vivo. J Pharmacol Sci
of anti-ErbB2 immunoliposomes targeted by a phage antibody se- 2005;94:1782–93.
lected for cellular endocytosis. Biochim Biophys Acta 2002;1591: [122] Silverman J, Liu Q, Bakker A, et al. Multivalent avimer proteins
109 –18. evolved by exon shuffling of a family of human receptor domains.
[102] Park JW, Hong K, Kirpotin DB, et al. Anti-HER2 immunolipo- Nat Biotechnol 2005;23:1556 – 61.
somes: Enhanced efficacy attributable to targeted delivery. Clin [123] Roovers RC, Laeremans T, Huang L, et al. Efficient inhibition of
Cancer Res 2002;8:1172– 81. EGFR signaling and of tumour growth by antagonistic anti-EGFR
[103] Steinhauser I, Spankuch B, Strebhardt K, et al. Trastuzumab-mod- Nanobodies. Cancer Immunol Immunother 2007;56:303–17.
ified nanoparticles: Optimization of preparation and uptake in cancer [124] Cortez-Retamozo V, Backmann N, Senter PD, et al. Efficient cancer
cells. Biomaterials 2006;27:4975– 83. therapy with a nanobody-based conjugate. Cancer Res 2004;64:
[104] Brissette R, Prendergast JK, Goldstein NI. Identification of cancer 2853–7.
targets and therapeutics using phage display. Curr Opin Drug Discov [125] Hansson M, Ringdahl J, Robert A, et al. An in vitro selected binding
Devel 2006;9:363– 69. protein (Affibody) shows conformation-dependent recognition of
F. Alexis et al. / Urologic Oncology: Seminars and Original Investigations 26 (2008) 74 – 85 85

the respiratory syncytial virus (RSV) G protein. Immunotechnology [139] Managit C, Kawakami S, Nishikawa M, et al. Targeted and sus-
1999;4:237–52. tained drug delivery using pegylated galactosylated liposomes. Int
[126] Gabizon A, Shmeeda H, Horowitz AT, et al. Tumor cell targeting of J Pharmacol 2003;266:77– 84.
liposome-entrapped drugs with phospholipid-anchored folic acid- [140] David A, Kopeckova P, Kopecek J, et al. The role of galactose,
PEG conjugates. Adv Drug Deliv Rev 2004;56:1177–92. lactose, and galactose valency in the biorecognition of N-(2-hy-
[127] Leamon CP, DePrince RB, Hendren RW. Folate-mediated drug droxypropyl)methacrylamide copolymers by human colon adeno-
delivery: Effect of alternative conjugation chemistry. J Drug Target carcinoma cells. Pharmacol Res 2002;19:1114 –22.
1999;7:157– 69. [141] Nimjee SM, Rusconi CP, Sullenger BA. Aptamers: An emerging
[128] Ross JF, Chaudhuri PK, Ratnam M. Differential regulation of folate class of therapeutics. Annu Rev Med 2005;56:555– 83.
receptor isoforms in normal and malignant tissues in vivo and in [142] Farokhzad OC, Karp JM, Langer R. Nanoparticle-aptamer bio-
established cell lines. Physiologic and clinical implications. Cancer conjugates for cancer targeting. Expert Opin Drug Deliv 2006;
1994;73:2432– 43. 3:311–24.
[129] Weitman SD, Lark RH, Coney LR, et al. Distribution of the folate [143] Gopinath SC. Methods developed for SELEX. Anal Bioanal Chem,
receptor GP38 in normal and malignant cell lines and tissues. Cancer 2007;387:171– 82.
Res 1992;52:3396 – 401. [144] Hermann T, Patel DJ. Adaptive recognition by nucleic acid aptam-
[130] Kamen BA, Capdevila A. Receptor-mediated folate accumulation is ers. Science 2000;287:820 –5.
regulated by the cellular folate content. Proc Natl Acad Sci USA [145] Holeman LA, Robinson SL, Szostak JW, et al. Isolation and char-
acterization of fluorophore-binding RNA aptamers. Fold Des 1998;
1986;83:5983–7.
3:423–31.
[131] Aronov O, Horowitz AT, Gabizon A, et al. Folate-targeted PEG as
[146] Tuerk C, Gold L. Systemic evolution of ligands by exponential
a potential carrier for carboplatin analogs. Synthesis and in vitro
enrichment: RNA ligands to bacteriophage T4 DNA polymerase.
studies. Bioconjugate Chem 2003;14:563–74.
Science 1990;249:505–10.
[132] Chiu SJ, Liu S, Perrotti D, et al. Efficient delivery of a Bcl-2-specific
[147] Bagalkot V, Farokhzad OC, Langer R, et al. An aptamer-doxorubi-
antisense oligodeoxyribonucleotide (G3139) via transferrin recep-
cin physical conjugate as a novel targeted drug-delivery platform.
tor-targeted liposomes. J Control Release 2006;112:199 –207.
Angew Chem Int Ed Engl, 2006;45:8149 –52.
[133] Gabizon A, Horowitz AT, Goren D, et al. In vivo fate of folate-
[148] Zhou C, Jiang Y, Hou S, et al. Detection of oncoprotein platelet-
targeted polyethylene-glycol liposomes in tumor-bearing mice. Clin
derived growth factor using a fluorescent signaling complex of an
Cancer Res 2003;9:6551–9. aptamer and TOTO. Anal Bioanal Chem 2006;384:1175– 80.
[134] Park EK, Lee SB, Lee YM. Preparation and characterization of [149] Pietras K, Ostman A, Sjoquist M, et al. Inhibition of platelet-derived
methoxy poly(ethylene glycol)/poly(␧-caprolactam) amphiphilic growth factor receptors reduces interstitial hypertension and in-
block copolymeric nanospheres for tumor-specific folate-mediated creases transcapillary transport in tumors. Cancer Res 2001;61:
targeting of anticancer drugs. Biomaterials 2005;26:1053– 61. 2929 –34.
[135] Quintana A, Raczka E, Piehler L, et al. Design and function of a [150] Kim ES, Serur A, Huang J, et al. Potent VEGF blockade causes
dendrimer-based therapeutic nanodevice targeted to tumor cells regression of coopted vessels in a model of neuroblastoma. Proc
through the folate receptor. Pharmacol Res 2002;19:1310 – 6. Natl Acad Sci USA 2002;99:11399 – 404.
[136] Gabizon A, Horowitz AT, Goren D, et al. Targeting folate receptor [151] Shangguan D, Li Y, Tang Z, et al. Aptamers evolved from live cells
with folate linked to extremities of poly(ethylene glycol)-grafted as effective molecular probes for cancer study. Proc Natl Acad Sci
liposomes: In vitro studies. Bioconjugate Chem 1999;10:289 –98. USA 2006;103:11838 – 43.
[137] Ohannesian DW, Lotan D, Thomas P, et al. Carcinoembryonic [152] Chu TC, Marks JW III, Lavery LA, et al. Aptamer toxin conjugates
antigen and other glycoconjugates act as ligands for galectin-3 in that specifically target prostate tumor cells. Cancer Res 2006;66:
human colon carcinoma cells. Cancer Res 1995;55:2191–9. 5989 –92.
[138] Zubieta MR, Furman D, Barrio M, et al. Galectin-3 expression [153] McNamara JO II, Andrechek ER, Wang Y, et al. Cell type-specific
correlates with apoptosis of tumor-associated lymphocytes in human delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol
melanoma biopsies. Am J Pathol 2006;168:1666 –75. 2006;24:1005–15.

You might also like