Cancer Nanomedicine: From Drug Delivery To Imaging

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REVIEW

BIOENGINEERING

Cancer Nanomedicine: From Drug Delivery to Imaging


Edward Kai-Hua Chow1,2* and Dean Ho3,4,5,6,7*

Nanotechnology-based chemotherapeutics and imaging agents represent a new era of “cancer nanomedicine”
working to deliver versatile payloads with favorable pharmacokinetics and capitalize on molecular and cellular
targeting for enhanced specificity, efficacy, and safety. Despite the versatility of many nanomedicine-based
platforms, translating new drug or imaging agents to the clinic is costly and often hampered by regulatory
hurdles. Therefore, translating cancer nanomedicine may largely be application-defined, where materials are
adapted only toward specific indications where their properties confer unique advantages. This strategy may
also realize therapies that can optimize clinical impact through combinatorial nanomedicine. In this review, we
discuss how particular materials lend themselves to specific applications, the progress to date in clinical
translation of nanomedicine, and promising approaches that may catalyze clinical acceptance of nano.

ALL ABOUT THE APPLICATION tions, allowing for efficient penetration and retention (EPR) for improved
Nanomaterials come in many flavors. They include lipid-based vehicles localization (Fig. 1). Versatile surface chemistry and surface area–to–
(liposomes, solid lipid nanoparticles, and micelles) (1–3); polymer volume ratios of nanoparticles mediate potent drug binding or high
carriers, such as hydrogels, polymersomes, dendrimers, and nanofibers drug-loading capacities to improve retention, efficacy, and safety. The
(4–16); metallic nanoparticles (gold, silver, and titanium) (17–20); ability to cofunctionalize nanoparticles with targeting moieties, im-
carbon structures [nanotubes, nanohorns, nanodiamonds (NDs), and aging agents, and drug payloads enables the synthesis of multimodal
graphene] (21–30); and inorganic particles, such as silica (31–34). These complexes that may simultaneously provide patients with improved
nanomaterials have been envisioned as drug and imaging agent de- treatment specificity and highly sensitive imaging capabilities to mon-
livery vehicles (or even as drugs and imaging agents themselves). Nano- itor treatment progress and outcomes. Highlighting these attributes,
medicine applications range from cancer to inflammation and regenerative and discussing remaining challenges, which, when addressed, may lead
medicine—in essence, the gamut that is clinical medicine. It is becoming to the fruition of these approaches in the clinic, serves as the foundation
clear, however, that different classes of materials are optimal for spe- for a focused look at how nanotechnology can uniquely affect cancer.
cific applications. For example, albumin is an effective solubilization Currently, most nanomedicine studies focus on a single drug to treat
platform for hard-to-deliver chemotherapeutics, such as paclitaxel cancer, lower toxicity, etc., which can benefit the patient to an extent.
(ClinicalTrials.gov: NCT00733408). Metallic particles are promising Nanomedicine, however, will have the greatest impact when admin-
photothermal therapeutic agents (35). Nanocarbons have mediated un- istered in combination with traditional clinical therapies, like radiation,
precedented increases in magnetic resonance (MR) efficiency for im- cell therapy, and small-molecule and biological drugs. Current combi-
proving imaging-based diagnosis (36). In addition, the rational and nation therapy using unmodified drug compounds is often based on ex-
scalable design of targeted polymeric nanoparticles because of the abil- isting additive guidelines for single therapy treatment, which precludes
ity to tailor their chemical makeup and properties have enabled the optimization of combinatorial therapy treatment for maximal efficacy
stabilization of small interfering RNA (siRNA) therapies without and safety. For example, docetaxel in combination with doxorubicin,
the need for toxic polycationic agents, as well as small-molecule thera- followed by cyclophosphamide-methotrexate-fluorouracil (CMF) ther-
pies. These attributes have catalyzed their translation into the clinic apy, has been compared to other variations to this combination for
with improved efficacy over current standards. Recent studies have operable breast cancer treatment (NCT00174655). With regard to the
also examined how combining the innate attributes of varying classes potential of nanomedicine and combination therapy, a recent study in
of nanomaterials to both induce biological responses and respond to animals suggests that timed combination therapy—where an siRNA
biological signals can be used to shrink tumors (32, 37, 38). and a drug are released sequentially from liposomal nanoparticles—
More than 7 million people died of cancer in 2008 worldwide, and might even be the key to overcoming chemoresistance (38).
it continues to be the second leading cause of death in the United States This review will examine cancer nanomedicine progress toward the
(39, 40). Successful treatment and imaging of cancer relies on several clinic with the theme of application-dependent selection of nanoparticle
capabilities that can be uniquely addressed via nanomedicine. Nano- agents. One important way to rationally select nano platforms is struc-
material sizes match the length scales of tumor interendothelial junc- ture. For example, cyclodextrins can greatly improve nucleic acid stabil-
ity to improve transfection efficacy. Gold nanoparticles are particularly
1
Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, effective at crossing the blood-brain barrier (BBB), which can open new
Singapore. 2Department of Pharmacology, Yong Loo Lin School of Medicine, National avenues for glioblastoma treatment. Photoactive nanoparticles can
University of Singapore, Singapore 117599, Singapore. 3Division of Oral Biology and
Medicine and Division of Advanced Prosthodontics, University of California, Los Angeles
modulate changes in their own temperature for spatially controlled
(UCLA) School of Dentistry, Los Angeles, CA 90095, USA. 4The Jane and Jerry Weintraub therapy. Furthermore, faceted properties of materials such as NDs
Center for Reconstructive Biotechnology, UCLA, Los Angeles, CA 90095, USA. 5California can mediate marked enhancements in MR imaging (MRI) contrast.
NanoSystems Institute, UCLA, Los Angeles, CA 90095, USA. 6Jonsson Comprehensive Cancer This review will also examine how algorithm-defined strategies can
Center, UCLA, Los Angeles, CA 90095, USA. 7Department of Bioengineering, The Henry
Samueli School of Engineering and Applied Science, UCLA, Los Angeles, CA 90095, USA. simultaneously optimize for efficacy, safety, and pharmacokinetics
*Corresponding author. E-mail: csikce@nus.edu.sg (E.K.-H.C.); dean.ho@ucla.edu (D.H.) (PK), among other parameters. Ultimately, the rationally designed therapy

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 1


REVIEW

Leaky vasculature Tumor- or vasculature-targeting

Tumor
Tumor

Tumor
Drug-loaded NP

Normal
epithelia Tumor-targeting NP

Lymphatic
drainage

Impaired lymphatic drainage

Fig. 1. Passive versus targeted nanomedicine delivery and reten- highly expressed in tumors than healthy tissue, to actively target tumors. These
tion. Nanoparticles larger than 8 nm in size can passively target tumors proteins include epidermal growth factor receptor (EGFR/Her1), human epider-
through preferential passage through larger interendothelial junctions (40 nm mal growth factor receptor 2 (Her2), folate receptor, endoglin (CD105), prostate-
to 1 µm) compared to those of healthy tissue (≤8 nm). Large junctions are a key specific membrane antigen (PSMA), epithelial cell adhesion molecule (EpCAM),
characteristic of the irregular tumor vasculature. Nanoparticles can also be CD20, CD44, CD90, and CD133. Once nanoparticles enter tumors, defective lym-
conjugated with targeting agents, such as antibodies specific to proteins more phatic drainage of nanoparticles results in enhanced retention.

or imaging method could save time and energy in making sure nano- tially home to tumor tissue versus healthy tissue (Fig. 1) (1). This is due
CREDIT: C. BICKEL/SCIENCE TRANSLATIONAL MEDICINE

medicine will translate to those who need it most: the patients. to tumor interendothelial junctions ranging from 40 to 80 nm, on av-
erage, and as large as 1 mm, whereas interendothelial junctions of
healthy tissues are ≤8 nm wide (41). This preferential uptake combines
CONQUERING CANCER with defective lymphatic drainage of nanoparticle complexes to pro-
mote EPR in tumor tissue (Fig. 1) (1). With this particular approach,
The use of nanomaterials to treat and image cancer is arguably the efficient intratumoral localization of nanoparticles, and resulting im-
most active area of nanomedicine research. Intrinsic properties like provements in treatment specificity, can be mediated without function-
surface charge, structure, and biocompatibility can be harnessed for alizing the vehicle with a targeting moiety. However, harnessing the EPR
cancer therapy. Size, which can be controlled during synthesis, has been effect in drug development may be dependent on characteristics that in-
shown to play a major role in nanomedicine efficacy. In particular, fluence tumor vasculature such as tumor type, size, and organ location
extensive work with liposomal and polymer drug delivery vehicles (42). Therefore, EPR-mediated drug delivery may likely be indication-
has revealed that complexes greater than 8 nm in diameter preferen- dependent or reliant on combination therapy.

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 2


REVIEW

Although EPR mediates enhanced delivery of larger nano-drug com- Passive tumor targeting by nanoparticles has proven successful in
plexes to tumors compared with free small-molecule drugs, increased humans, with liposomal doxorubicin (Doxil) already being clinically
size can adversely affect nano-drug circulation half-life. Larger particles used for breast cancer (47). Yet, there are other exciting developments
are more easily recognized and cleared by the liver and phagocytic cells in the field of cancer nanomedicine that capitalize on the unique and
of the reticuloendothelial system (43). Rapid clearance can be partially intrinsic properties of certain materials for targeted delivery of thera-
alleviated by adding polymer coatings, like poly(ethylene glycol) (PEG). peutics (Fig. 2), including those that are difficult to solubilize, chal-
PEGylated liposomes 200 nm in diameter, however, are still cleared lenging to deliver, or unstable (that is, easily degraded) in vivo (48–52).
faster than 100-nm PEGylated liposomes (44). When these studies are Although the field is vast, some promising examples for translation are
taken together, optimal drug delivery complex size for cancer therapy described herein.
likely ranges from 8 to 100 nm, when relying on the EPR effect. These size
requirements are likely to extend from conventional liposomal and poly- Aiming at a target
meric complexes to other nanomedicine platforms, such as metal- or Drug-loaded nanoparticles can be targeted to surface molecules over-
carbon-based ones (45, 46). Targeted drug delivery complexes likely expressed in cancer cells or tumor vasculature. By adding a targeting com-
have a wider range of optimal size. Further studies, however, are needed ponent, nanoparticles can further improve their efficacy of tumor-specific
to determine how large these complexes can be and still maintain ac- drug delivery beyond EPR-mediated tumor homing (Fig. 1). Recently, a
ceptable body retention rates. poly(lactic-co-glycolic acid) (PLGA)/PEG–based nanoparticle, named
CREDIT: C. BICKEL/SCIENCE TRANSLATIONAL MEDICINE

Fig. 2. Designer nanomedicine. Nanomedicine platforms can take on any number of properties to tailor therapy or imaging, and improve chances of
translation.

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 3


REVIEW

BIND-014, was designed to target prostate cancer. The polymeric par- prior to U.S. Food and Drug Administration (FDA) approval for clin-
ticle, with a PSMA-targeting moiety on the surface and a docetaxel ical studies.
payload, has been tested in preclinical studies and in humans in early- A major hurdle of cancer therapy is intrinsic or acquired chemo-
phase clinical trials for metastatic lung cancer and tonsillar cancer (53). resistance, which contributes to treatment failure in more than 90% of
The first studies showed the importance of targeted drug delivery and metastatic cancers (64). A common mechanism of chemoresistance is
prolonged circulation toward improved therapeutic efficacy and safety the overexpression of adenosine triphosphate–binding cassette (ABC)
via an optimized multimodal polymer nanoparticle where BIND-014 transporter proteins that can efflux a wide range of small molecules, includ-
effective concentrations were only 20% of those needed with free ing chemotherapeutics (65). Conventional methods of overcoming ABC
docetaxel, which is an important consideration for reducing toxicity transporter protein chemoresistance have involved the development of in-
of chemotherapeutics. BIND-014 administration to prostate, breast, hibitors of these proteins. Clinical trials for such drugs have met little suc-
and lung xenografted tumors that normally (in humans) show little to cess as potent inhibitors of multiple transporter proteins and have been
no response to unmodified docetaxel treatment resulted in a significant associated with trial-ending toxic side effects (66). More specific inhibi-
therapeutic effect in animals (53). To date, clinical trials pertaining to tors struggle to find a balance between efficacy and tolerability (67).
examining the efficacy of BIND-014-001 in advanced and metastatic Moreover, administering a single inhibitor is unlikely to succeed because
cancers are active (NCT01300533), and another clinical trial pertaining chemoresistant tumors often express different ABC transporter proteins.
to the application of BIND-014-005 has been approved for non–small Delivery of anthracyclines—a class of DNA-damaging chemotherapeutics—
cell lung cancer (NCT01792479). by NDs has been shown to overcome ABC transporter drug efflux without
Transferrin receptor targeting using nanoparticles has also been targeting the transporter itself or another overexpressed cancer protein
explored clinically in pancreatic cancer, which overexpresses the trans- (26, 68). In mouse models of anthracycline-resistant cancer, delivery of the
ferrin receptor in as much as 93% of the tumor cells, whereas normal anthracycline doxorubicin by NDs (Fig. 2) resulted in increased tumor-
tissue did not stain positively for the transferrin receptor (54). This in- killing efficacy as well as lower systemic toxicity, including less myelo-
cludes recent developments in the clinical evaluation of transferrin suppression, compared with free doxorubicin (26). The combination of
receptor–targeted liposomal gene therapy that may improve standard safety and efficacy observed with the ND-doxorubicin agents was mediated
drug efficacy through restoration of tumor suppressor genes such as by potent drug binding, without the need for chemical modification of the
p53 (NCT00470613) and Rb94 (NCT01517464) (Table 1). This was drug or the ND surface. A targeting moiety was also not present, suggest-
particularly well demonstrated with SGT-53 (SynerGene Therapeutics ing that the efficacy of NDs was likely related to EPR.
Inc.), a transferrin receptor–targeting liposomal nanoparticle that delivers Although anthracycline delivery by NDs improved therapeutic efficacy
p53-containing plasmids to tumors that overexpress the transferrin re- compared to traditional chemotherapy—at least in mouse models—even
ceptor. Preclinical data demonstrated enhanced gene therapy to murine more impressive therapeutic gains can be achieved using a targeting com-
metastatic pancreatic tumors, improving gemcitabine response (55). ponent. Hybrid drug delivery complexes containing epirubicin (a chemo-
Clinical trials demonstrated that single-agent SGT-53 delivery is well toler- therapeutic) and an EGFR antibody (Fig. 2) were delivered to mice bearing
ated and the p53 transgene can be seen specifically in metastatic tumor spec- triple-negative breast cancer (TNBC) human breast cancer (MDA-MB-231)
imens and not in normal tissue. The trials have since been modified to xenografts (Table 1) (45). Drug-loaded EGFR-targeted complexes main-
include combinatorial therapy with docetaxel, as originally intended (56). tained the enhanced safety profile of their untargeted counterparts while
In addition to targeting cancer cells, targeting the tumor vasculature increasing homing to EGFR-positive mammary tumors. This resulted in
has also demonstrated to be a viable method for targeted drug delivery increased tumor response and, in some cases, complete regression of these
(Fig. 2 and Table 1). This is best exemplified by RGD-containing pep- highly aggressive tumors.
tides that target av integrins overexpressed in tumor vasculature (57). These studies demonstrate a clear clinical potential for untargeted and
Conjugation and coadministration of nanoparticles with internalizing targeted carbon nanomedicine. However, judging by the lack of nano-
RGD (iRGD), a peptide that combines RGD with a tumor-penetrating carbon clinical trials being conducted, continued toxicity and clearance as-
CendR motif, exhibited enhanced tumor targeting, penetration, and sessment in rodent and large animal models remains a critical precursor to
efficacy compared to nontargeted nanoparticles (58, 59). Clinical trials translation.
are currently under way to evaluate the effect of iRGD on tumor vascu-
lature and should pave the way to translation of iRGD-targeted delivery Silencing genes
of drugs and nanoparticles (NCT01741597). One area where the intrinsic properties of nanoparticles can improve
drug delivery is RNA interference (RNAi). RNAi is a promising method
Capitalizing on carbon for silencing cancer-causing genes, but is hampered by enzymatic deg-
Carbon nanomaterials, including nanotubes, nanohorns, fullerenols, radation of small RNAs in vivo. As the most clinically developed example,
graphene, and NDs, have been the subject of several promising preclinical cyclodextrin-based polymers (CDPs) are well tolerated and can self-
studies owing to their versatile surface properties. Carbon materials can assemble to protect and deliver siRNA to cancer cells (Fig. 2) (69–71).
deliver a wide spectrum of therapeutic compounds, often with little to no Following successful preclinical studies, these CDPs were used in the
modification to the drug or nanomaterial, owing to the ability to me- first siRNA-based cancer therapy clinical trial targeting patients with
diate p-p stacking with nanocarbon surfaces for noncovalent delivery recurrent or metastatic solid malignancies (NCT00689065) (69, 71).
(21, 24, 26, 27, 60–63). A major concern surrounding these materials is their Calando Pharmaceuticals developed CALAA-01, a CDP-siRNA com-
safety because of potential cardiopulmonary toxicity. However, multiple plex functionalized with a transferrin-targeting ligand for the treatment
reports show no apparent toxicity after nanocarbon administration of metastatic melanoma, PEG to promote stability and biocompatibility
in mouse models (24, 26, 45, 60, 62). Carbon nanomaterials have yet in vivo, and an siRNA to silence the gene RRM2 (ribonucleotide reductase
to be tested in humans, as continued safety assessments are ongoing M2 subunit) (Table 1). A striking element of this approach is the ability

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 4


REVIEW

Table 1. Example nanoparticle applications in cancer drug delivery. Table is organized by stage of translation.

Agent delivered Material(s) Translational status Key details of study References

Doxorubicin Pluronic block Phase 2 P-glycoprotein–targeted chemotherapeutic delivery used against (48, 49)
copolymers clinical trials human gastroesophageal adenocarcinoma
Acceptable safety profile and enhanced efficacy compared to
free doxorubicin in phase 2 clinical trials (SP1049C-202-UK)
Docetaxel PEG-PLGA Phase 1 PSMA-targeted small-molecule–treated mouse xenografts (53)
clinical trials of human prostate tumors
Ongoing clinical trials demonstrate favorable PK and initial efficacy
in human lung and tonsillar cancers (NCT01300533, NCT01792479)
p53 or Rb94 plasmid Lipid Phase 1 Transferrin receptor–targeted liposomes enhanced delivery and (55, 56)
clinical trials transgene expression in metastatic tumors in mouse metastatic
pancreatic cancer
Ongoing clinical trials in patients with advanced solid tumors
demonstrate favorable dose response and successful overexpression
of transgene in tumors compared to normal tissue (NCT00470613,
NCT01517464)
Paclitaxel Albumin Phase 1 Tumor vasculature–targeting and tumor-penetrating iRGD-coated Abraxane (59)
clinical trials (nanoparticle albumin–bound paclitaxel) exhibited enhanced tumor
uptake and tumor growth inhibition of mouse xenografts of human
breast cancer compared to Abraxane treatment
Ongoing clinical trials are investigating the ability of iRGD to
modify human metastatic breast cancer permeability as
measured by MRI (NCT01741597)
RRM2 siRNA Cyclodextrin-containing Phase 1 Transferrin-mediated tumor targeting allowed knockdown of (69–71)
polymers clinical trial RRM2, which inhibited growth of syngeneic graft murine
neuroblastoma tumors
Phase 1 clinical trials in humans are ongoing as of 2008 and
show evidence of RRM2 knockdown in metastatic human
melanoma tumors (NCT00689065)
Cisplatin Carbon Preclinical Drug-loaded carbon nanohorns enhanced efficacy in human (21)
lung cancer mouse xenograft tumors in vivo compared to
unmodified cisplatin
Paclitaxel Carbon Preclinical Drug-loaded single-walled carbon nanotubes enhanced circulation (24)
half-life, tumor killing, and overall survival in 4T1 murine breast
cancer model compared to unmodified paclitaxel
Doxorubicin Carbon Preclinical Drug-loaded NDs enhanced blood circulation, tumor killing, and (26)
animal survival in chemoresistant mouse breast and liver
tumors compared to unmodified doxorubicin
Epirubicin Carbon, lipid Preclinical EGFR-targeted, drug-loaded ND-lipid hybrid particle enhanced (45)
tumor homing and tumor regression in a mouse model of
human TNBC compared to free drug
ID4 siRNA Peptides Preclinical Tumor-penetrating peptide nanocomplexes loaded with (51)
siRNA-treated human ovarian tumors in mice
Proprietary siRNA Carbon Preclinical Multiwalled carbon nanotubes loaded with siRNA inhibited tumor growth (62)
and prolonged overall survival in human lung cancer xenografted mice
compared to siRNA alone or liposomal delivery of siRNA
Melittin peptide Carbon Preclinical Drug-loaded perfluorocarbon nanoparticles improved circulation (63)
and therapeutic efficacy against human breast cancer mouse
xenograft tumors and syngeneic graft mouse melanoma tumors
compared to free drug
Bcl2L12 siRNA Gold Preclinical Gold-based spherical nucleic acid (SNA) delivery crossed BBB and (73)
accumulated in human glioma tumors in mice to silence target
gene and reduce tumor burden

for self-assembly of the nanoparticles through the interaction of the pos- siRNA complex included stabilizing RNA therapeutics, manufacturing
itively charged cyclodextrin chains around the negatively charged siRNA scale-up [for example, chemistry, manufacturing, and controls (CMC)
to improve nucleic acid stability. The process of translating the CDP- compliance], and ensuring delivery specificity to melanoma solid tumors.

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 5


REVIEW

Although the clinical trials are still ongoing, there is evidence of siRNA been commercialized by Nanospectra Biosciences as AuroLase, with clin-
activity in tumor biopsies from treated patients (69). ical trials in head and neck cancer (NCT00848042) and primary and meta-
In addition to CDP-based siRNA delivery, a previous clinical study static lung tumors (NCT01679470) already under way.
used liposomal bcr-abl siRNA for imatinib-resistant chronic myeloid In addition to photothermal therapy, several other triggered modal-
leukemia (CML) treatment that resulted in reduced bcr-abl gene expres- ities based on radio- or magneto-stimulus are being explored, including
sion in peripheral blood samples and reduced extramedullar CML node magnetic and radio-based methods for enhancing drug delivery and ef-
size (72). However, transfection efficiency decreased by the third round ficacy (Table 2) (79, 80). These methods may be more easily translated
of administration, in turn reducing the ability to affect bcr-abl mRNA because MRI and radiotherapy are already commonly used in cancer
levels. A stable and rationally designed delivery system could perhaps treatment. Radiotherapy is a standard method of tumor treatment that
resist serum ribonuclease activity that limits transfection efficiency. To relies on greater absorption of x-rays in tumor tissue compared to nearby
improve delivery and increase the circulatory half-life of siRNA, lipid- normal tissue. Radio-induced nanomedicine, specifically using hafnium
based carriers with tumor-targeting and tumor-penetrating moieties oxide nanoparticles, has demonstrated enhanced energy deposit in
have been developed (51) (Table 1). These “siRNA nanocomplexes” were nanoparticle-containing subcellular structures of cells, which results in
designed to display a cyclic nonapeptide on its surface, LyP-1. LyP-1 tar- enhanced energy release and subsequent localized cellular destruction
gets p32—a mitochondrial protein overexpressed on stressed tumor from nanoparticle clusters. Multiple xenograft tumor models demon-
and tumor-associated cells—and is also able to penetrate cells after being strated greater sensitivity to ionizing radiation treatment when pretreated
proteolytically processed by endogenous proteases. This ensured that with hafnium oxide nanoparticles compared to radiation treatment alone.
the particle was more toxic to tumors than to healthy tissue, because This approach is currently being evaluated clinically in patients with soft
the payload was only released once inside the cancer cell. The nano- tissue sarcoma of the extremity or advanced squamous cell carcinoma
complexes were loaded with siRNA against the oncogene inhibitor of of the oral cavity (NCT01946867 and NCT01433068) (80).
DNA binding 4 (ID4) and tested in mouse models of human ovarian
cancer. Tumor-penetrating nanoparticles functionalized with ID4- Immune system plays defense
silencing siRNA suppressed tumor growth by 82%, whereas control Immunotherapy involves the activation or suppression of immune re-
injections (saline and untargeted control nanoparticles) did not sup- sponses for disease treatment. In essence, immunotherapy capitalizes
press tumor growth. on our natural defense system to prevent the growth (or occurrence) of
As another example of how nanoparticles deliver siRNA—except cancer. Emerging immunotherapeutic strategies for cancer involve the de-
with the nucleic acids displayed on the outside rather than encapsulated— livery of agents, such as tumor-specific antigens (Fig. 2), that can prime or
Jensen et al. created gold nanoparticle–based SNAs (73). These SNAs stimulate one’s own immune system to actively seek out and destroy cancer
were recently used for RNAi against the Bcl2L12 oncogene in a mouse cells that have these antigens (Table 2). As a new area of research, nano-
glioblastoma model. SNAs exhibited no apparent toxicity, and owing particles may be used to modulate the immune response to tumors (81).
to their resistance to nuclease-triggered degradation, they mediated sta- Nanotechnology has made the passive and targeted delivery of antigens
ble and persistent gene transfection without the need for polycationic and adjuvants possible. In addition, nanomaterials have helped overcome
polymers. SNAs exhibited substantial accumulation in the brain tissue biological barriers that have stunted the development of more conventional
of healthy mice with an intact BBB after systemic injection, providing approaches to adjuvant delivery such as oral delivery. This is particularly
a strong basis for applications in brain cancer therapy, given the fact evident when delivering adjuvants and antigens orally through enhanced
that the presence of the BBB has blocked many candidate therapies uptake by mucosal immune cells, as well as delivering easily degradable
(Fig. 2). adjuvants, such as RNA (82, 83). Because of the preclinical successes of
nano-based immunotherapy, several clinical trials (NCT00291473 and
Pulling the trigger NCT00651703) are currently underway to translate these approaches to a
Photothermal therapy involves the use of heat to kill cancer cells, which has variety of human diseases ranging from cancer to allergies (84, 85) (Table 2).
gained interest because the inducible and noninvasive nature of this trig- Nanoparticles have been recently used in cancer immunotherapy
gered therapy can be applied to a wide range of solid tumors (Table 2). A vaccines because they can co-deliver multiple agents, such as an antigen
novel class of materials based on a gold nanoshell encapsulating a silica and adjuvant, in a single, biocompatible platform with enhanced uptake
core (Fig. 2) has been shown to be effective in near-infrared laser-induced and efficiency compared to conventional treatment [for example, standard
cancer therapy in multiple cancer models, including lung, head and vaccines and dendritic cell (DC) therapy]. Cancer vaccines impair cancer
neck, prostate, and gliomas (46, 74–76). Unique properties that make progression or prevent occurrence by delivering specific tumor antigens
these nanoshells clinically promising include biocompatibility of the par- that induce the generation of antigen-specific cytotoxic T lymphocytes
ticles as well as the tunability of the surface plasmon resonance of the (CTLs). Strong CTL response often requires the coadministration of
gold metal shell based on the modulation of the ratio of the dielectric adjuvants. Nanoparticles are well suited for this application because they
core radius to shell thickness. This architecture allows for highly effi- can ensure delivery of both an adjuvant and an antigen to the same im-
cient and localized conversion of light to heat. These two properties mune cell, whereas standard vaccine formulations are more inefficient and
are critical to developing a safe product that can be used for heat therapy require concentration-dependent levels of both adjuvant and antigen to
in a targeted area (as wide as 2 mm) with minimal laser power require- achieve similar effects. One of the first nanoparticle cancer vaccine de-
ments (10 to 100 mJ/cm2) (77, 78). In a recent mouse study, these gold- livery vehicles was based on a truncated Her2 protein complexed with
coated silica particles were labeled with VEGF to target the blood vessels hydrophobic polysaccharides into nanoparticles that enhanced the
of intracerebral tumors (35). Following administration and photo- uptake of Her2. Delivery of both an antigen and an adjuvant in a single
thermal ablation, the vasculature of diseased tissue was ruptured, nanoparticle in mice resulted in the stronger production of anti-Her2
whereas healthy tissue was not damaged. This nanoshell technology has antibodies compared to vaccination with Her2 protein alone as well as

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 6


REVIEW

Table 2. Example nanoparticle applications in cancer therapy. Table is organized by stage of translation.

Type of therapy Material(s) Translational status Key details of study References

Immunotherapy, vaccine Protein Phase 2 clinical trial Viral-like protein shell delivery of CpG-rich oligodeoxynucleotide (85)
(CpG-ODN) and melanocyte differentiation antigen resulted
in enhanced CD8+ T cell response in melanoma patients
compared to antigen alone (NCT00651703)
Photothermal Silica, gold Phase 1 clinical trials Vascular endothelial growth factor (VEGF)–targeted nanoshells (35)
for thermal ablation and vessel disruption in mouse
glioma model
Currently in clinical trials for head and neck cancer
(NCT00848042) and primary and/or metastatic lung
tumors (NCT01679470)
Radiotherapy Hafnium oxide Phase 1 clinical trials Nanoparticles for radio-induced tumor cell killing in (80)
crystals mesenchymal and epithelial tumor xenograft
mouse models
Phase 1 clinical trials are ongoing as of 2011
(NCT01946867, NCT01433068)
Immunotherapy, vaccine Polysaccharide Phase 1 clinical trial Her2 antigen cholesteryl mannan and cholesteryl pullulan (84, 86–88)
nanoparticle vaccines previously tested in Her2-expressing
murine sarcomas
Currently being evaluated in patients with advanced cancer
with Her2-expressing tumor cells (NCT00291473)
Immunotherapy, vaccine PLG Phase 1 clinical trial PLG matrices that co-deliver granulocyte-macrophage (90)
colony-stimulating factor (GM-CSF), CpG-ODN, and tumor
lysate antigen for recruitment of DCs to PLG matrices,
potent local and systemic antitumor CD8+ T cell response
in mouse melanoma model
Starting phase 1 study of implantable vaccine to treat
melanoma (NCT01753089)
Magneto-responsive Iron/cobalt, PLGA Preclinical Magnetically guided polymer carriers loaded with Fe-Co (79)
nanoparticle and doxorubicin for drug-nanoparticle
deposition in right or left liver lobe of rabbits
Immunotherapy, vaccine PEG-PLGA Preclinical Ovalbumin-loaded PEG-PLGA nanoparticles displaying (82)
RGD peptide for targeted M cell uptake and vaccination
in mice
Immunotherapy, RNA adjuvant Lipid Preclinical Lipidoid delivery of immunostimulatory RNA for antiviral (83)
and adjuvant activity in vivo in mice

CTL-mediated repression of Her2-expressing murine tumors in vivo (TLR) ligands. For instance, polylactide-co-glycolide (PLG) was used to
(86). This work is currently in clinical trials (NCT00291473) where it form matrices that expressed the TLR9 ligand CpG-ODN and also re-
has been relatively well tolerated and elicits similar immune responses leased the cytokine GM-CSF as well as melanoma tumor antigens (90).
in humans (84, 87). Immune response was seen in 93% of patients, which is This work demonstrated that GM-CSF was capable of recruiting DCs to
compelling when compared to clinical trials with E75, an immunogenic the PLG matrices in vivo in mice, whereas CpG-ODN activated the re-
peptide from Her2/neu protein, that saw postvaccine delayed-type hyper- cruited DCs. Exposure of these activated DCs to melanoma tumor anti-
sensitivity response in 74% of treated patients (88). gens by PLG matrices resulted in a potent CD8+ CTL response and an
Antigen-presenting cells (APCs) are required for CTL response to enhanced impairment of melanoma tumor progression. Translation of
an antigen. DCs have been identified as the most potent APCs and have this work into the clinic should lead to a cheaper and faster DC-based
subsequently been used for vaccines. Conventional DC-based vaccines vaccine approach compared to current methods that require the re-
involve the isolation and expansion of antigen-exposed DCs in vitro, moval, ex vivo activation, and reimplantation of a patient’s DCs. Indeed,
followed by reintroduction of activated DCs into cancer patients. Al- phase 1 trials are underway (NCT01753089).
though DC vaccines are a promising strategy that has seen some success
in clinical trials, this procedure remains costly and inefficient. Several
studies suggest that, owing to their versatile surface chemistry and func- NANO GETS A BETTER VIEW
tionalization, nanoparticles can be used as modular platforms to ef-
ficiently deliver antigens and prime DCs for an anticancer immune Many of the advantages that are offered by nanotechnology for drug
response (89, 90). These studies often incorporate antigen exposure delivery, such as improved circulation half-life, EPR, and reducing tox-
with DC chemoattractants and DC activators, such as Toll-like receptor icity, are also beneficial to imaging. However, for nanomedicine to

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 7


REVIEW

benefit the imaging community in a translational setting, it is important 12-fold (36). This is among the highest reported per-Gd relaxivity values
that properties unique to the nano-imaging agents be harnessed. Con- owing to the faceted ND surface, which potently attracts water. The abil-
ventional approaches to making nano-imaging agents include high- ity of the NDs to facilitate water molecule interaction with Gd enables
density encapsulation of contrast agents such as gadolinium (Gd) or higher MR signal, and in turn, a lower Gd concentration is needed to
manganese oxide into liposomes and PLGA, or loading the contrast achieve the clinically required brightness and contrast. Clinically
agents onto the surface of nanoparticles, such as carbon nanotubes, speaking, one order of magnitude reduction in the amount of Gd
gold, or NDs. These strategies can improve circulation time and mediate needed would represent a game-changing advance in medicine because
tumor localization through EPR, thus improving imaging capabilities this would allow for simultaneous high-contrast imaging while also
(Table 3). Perhaps one of the most promising nano-imaging approaches markedly reducing the amount of contrast agent needed to achieve this.
is the use of superparamagnetic iron oxide nanoparticles, which are Furthermore, this may increase the population of Gd-amenable patients
currently in clinical trials for lymph node imaging and pancreatic can- who were previously precluded from Gd-based MRI because of renal
cer staging (NCT00920023) (91). Iron oxide nanoparticles are para- toxicity. In addition to ND-Gd, gadofullerenes, gadonanotubes, and
magnetic, which can be imaged after the application of an external silica-based imaging agents have demonstrated per-Gd relaxivity increases
magnetic field. Coating iron oxide nanoparticles with lipid-like mate- over the delivery of Gd alone (98, 99).
rials has resulted in improved control of particle size, dispersal, and, im- MRI is a standard of care for preoperative diagnostic imaging of
portantly, biocompatibility (92). Iron oxide degradation in vivo is tumors, yet intraoperative MRI has proven to be difficult and not very
also possible, which provides clear pathways for clearance after ad- accurate. Kircher et al. developed a trimodal MRI-photoacoustic-
ministration (93). Raman (MPR) nanoparticle imaging complex. This multimodal particle
Although established imaging platforms, such as iron oxide, have used both photoacoustic and Raman imaging for imaging during tumor
transitioned into clinical studies, emerging approaches using novel ma- resection combined with accurate MRI preoperative evaluation of
terials that can uniquely enhance imaging safety and efficacy are being tumors (100). MPRs accumulated preferentially in tumors via EPR and
developed. With regard to cancer nanomedicine (94, 95), nano-enhanced successfully delineated brain tumor margins preoperatively and intra-
imaging can be used to track therapy or diagnose disease. In particular, operatively with a single injection in a human glioblastoma mouse model.
nanoparticles have been designed to enhance contrast agent efficiency A particularly promising approach being developed for MRI is
(96). Some nanoparticles, such as self-assembling protein cages, can CEST, which uses polypeptides as biocompatible and biodegradable
be used to increase packaging of paramagnetic ions (97). Such systems MRI agents. Amide, amine, and hydroxyl protons and their unique
have demonstrated enhanced localized relaxivity over free paramagnetic exchange rates can be assigned specific colors to produce multicolor
ions. ND-based MRI is a revolutionary approach to MRI because the MR probes (101, 102). Recent studies have also demonstrated the ap-
ND surface mediates a per-Gd relaxivity (contrast efficiency) increase of plicability of CEST compounds as pH sensors to monitor cell transplan-

Table 3. Example nanoparticle applications in cancer imaging. Table is organized by translational status.

Imaging method Material Translational status Key details of study References

MR Iron oxide, dextran Phase 4 clinical trial Iron oxide (superparamagnetic) core within densely (91)
packed dextran nanoparticle allows for imaging of
lymph nodes in a clinical trial (NCT00920023)
Fluorescence Quantum dots (Qdots) Clinical samples Qdot conjugated to different antibodies allowed for (95)
multiplexed molecular mapping of benign and
metastatic prostate tumor cells in human prostatectomy
or needle biopsy prostate cancer specimens
Fluorescence Polystyrene Preclinical CD47 or “self”-peptide conjugation impaired macrophage (52)
clearance of nanobeads, thus enhancing tumor
near-infrared imaging compared to nontargeted
nanobeads in mice with human lung cancer
xenografts
Fluorescence Gelatin, PEG, Qdots Preclinical Gelatin core conjugated to PEG-Qdots increased blood (94)
circulation half-life and deeper tumor penetration
in vivo in human fibrosarcoma mouse xenograft
tumor models compared to silica Qdots
Fluorescence Gold, PEG Preclinical In vivo assembly of molecular contrast agent and gold (96)
nanoparticle for near-infrared imaging of human
breast cancer mouse xenografts in vivo
MR, photoacoustic, Raman Gold, silica Preclinical Gold core–silica shell nanoparticle for trimodal imaging (100)
before, during, and after surgery after single intravenous
injection into human glioblastoma xenografts in mice
MR Lipid Preclinical Diamagnetic chemical exchange saturation transfer (102)
(CEST) liposomes for “multicolor” imaging of lymph
nodes in mice

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 8


REVIEW

tation and viability (103). Here, mouse hepatocytes and liposomes specific outcome is not met (for example, optimal apoptosis and op-
loaded with L-arginine, a pH-sensitive CEST agent, were injected into timal safety), FSC is a broad approach that draws from a set of math-
mice. Reductions in pH after cell death resulted in decreased MR signal ematical search algorithms. These include Differential Evolution and
from the L-arginine–loaded liposomes, which served as a modality to the Gur Game, which select the next group of iterative drug adminis-
monitor murine hepatocyte viability after transplantation. Additional tration conditions to rapidly achieve desired phenotypes, even among
studies demonstrated the use of CEST functional liposomes that carry a prohibitively large testing parameter space. Although six drugs with
color signatures unique to the polypeptides being carried, which result 10 selected concentrations would each require 1,000,000 potential trials,
in spatially defined, multicolor imaging of popliteal lymph nodes (102). FSC “self-guides” a cellular population toward desired phenotypic out-
These studies exemplify how well-designed nanoparticle systems can comes that rapidly converge with only tens of iterations. In addition,
address clinical problems that more conventional approaches are unable because FSC can take PK/PD (pharmacodynamics) into account during
to achieve. the search process, it is easily adaptable toward the implementation of
nanotherapeutic combinations that are comprehensively optimized for
both efficacy and safety.
THE RIGHT COMBINATION In addition to FSC, additional large-scale, high-throughput methods
and databases can be unified to analyze interactions at the gene, sig-
Safety is of paramount importance in any drug’s translational roadmap. naling pathway, and whole systems level after the coadministration of
Although the highlighted approaches in this Review have demonstrated candidate drugs. CDD assesses these interactions to converge upon a
remarkable potential toward clinical translation, continued clinical trials directed dosage of multiple compounds that result in a targeted pheno-
of promising platforms and development of pathways toward approval typic outcome and simultaneously reduces off-target effects. Effective
of nanomaterials will continue to shape the landscape of how nano- CDD uses publicly available databases in parallel with computational/
medicine can affect clinical practice. The cancer applications described mathematic resources to evaluate varying types of drug interactions and
here, where nanomedicine is poised to improve the safety and efficacy test entire parameter spaces to arrive at a combinatorial dose that sat-
of treatment in a clinical setting—and is already doing so in clinical isfies designated criteria including optimized efficacy and safety (108).
trials—are by no means exhaustive, because enhanced drug delivery These include DrugBank (http://drugbank.ca) and Therapeutic Targets
and imaging can also benefit the fields of ophthalmology, regenerative Database (http://bidd.nus.edu.sg/group/cjttd/), which provide informa-
medicine, neurodegenerative diseases, sexually transmitted diseases, tion on thousands of drug compounds and targets. The National Insti-
infectious diseases, and global health. tutes of Health Clinical Collection (www.nihclinicalcollection.com)
The issue that follows now is the evaluation of the role nanomedicine provides information on more than 400 small molecules that are widely
will play, if any, in truly transforming the way that medicine is practiced. used in clinical trials. STRING (http://string-db.org) is a database of more
Nanoparticles can affect a wide range of diseases by improving efficacy than 5 million proteins that provides insight into protein-protein interac-
and safety, prolonging circulation time, and enhancing delivery speci- tions. These libraries can be paired with computational and modeling
ficity. For cancer, whether or not a single drug can truly lead to sustained tools such as flux balance analysis, which has been used to design drug
remission is a major question moving forward in nanomedicine design. combinations for cancer and other disorders by accounting for both drug
The concept of combinatorial treatment is gaining traction as a required activity and toxicity. Petri nets can assess drug-mediated effects upon
method of treatment, particularly in the field of cancer therapy (Fig. 2). gene expression and subsequently use this information to formulate
Furthermore, as cancer therapy begins to marry diagnostics and thera- synergistic approaches to enhance treatment potency.
peutics into a more personalized approach, optimal combinatorial ther- FSC and CDD serve as optimal dosing principles that can guide the
apy will likely differ for subsets of patients within a single type of cancer. packaging of directed drug combinations into nanomedicine delivery
With regard to clinical evaluation of the best methods of combina- vehicles to maximize their clinical efficacy. This is particularly applicable
torial treatment, these studies are often based on the therapeutic guidance for drugs, such as chemotherapeutics, that require improved circulatory
and additive combination of singular treatment with these drugs, re- stability, intratumoral retention in the cases of innate and acquired
sulting in only marginal improvements to efficacy and safety. As med- chemoresistance, and half-life. To address these challenges, promising
ical research begins to become more multidisciplinary, emerging methods, strategies are on the horizon. These include the aforementioned use of
such as feedback system control (FSC) and combinatorial drug design layer-by-layer co-delivery of siRNA and small molecules for TNBC treat-
(CDD), can be used to bring engineering approaches to rational CDD ment (38) to overcome chemoresistance. A recent study (109) combined
and optimization using nanomedicine. Real-time drug monitoring and liposomal and silica nanoparticle delivery to treat pancreatic ductal
personalized PK will enable more tailored nano-based therapies (104). adenocarcinoma by interfering with the formation of pericytes, which
In addition, emerging strategies including live cell interferometry can are cells that not only protect vascular endothelial cells but also typically
quantify changes in cellular mass and resulting fitness from populations block nanoparticle access to the vasculature. These studies demonstrate
of cells in response to drug dosing. This information can serve as a vital that combinatorial drug delivery through single or multinanoparticle
readout to monitor tissue response to combinatorial nanomedicine (105). platforms serves as a feasible and promising route toward translating
One of the emerging methods that may transform the way that rationally developed drug combinations into the clinic.
nanotherapy is administered and remove its barriers to acceptance is
the concept of FSC, a top-down approach that directs a cell or popula-
tion of cells toward a desired phenotype through control strategies that FUTURE STEPS FOR NANO
were conventionally reserved for nonbiological scenarios (106, 107).
When drug compounds with arbitrarily decided concentrations are Although a nanomaterial-modified therapeutic may be a stand-alone
applied to cells, their signaling pathways respond unpredictably. If a solution for cancer owing to improved safety and efficacy over an un-

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 9


REVIEW

modified compound, the issue of whether or not a single nanoparticle In summary, a clearer roadmap toward solidifying how nano-
agent can effectively cure a patient of cancer should be raised. The need medicine can move from enhancing treatment efficacy to eliminating
for an effective combination of small-molecule drugs, biologics, nu- the major diseases of our generation is coming to fruition. To this end,
cleic acids, and/or other compounds may be necessary to mediate harnessing the highlighted unique features of nanomaterial platforms
comprehensive cancer management, or potentially a cure. The diverse will inspire the rational design of combinatorial nanotherapeutics,
properties of nanomaterials should be able to facilitate combinatorial which, combined with powerful new imaging agents, will underscore
treatment, and optimization methods such as FSC and CDD can improve the true catalysis of nanomedicine translation.
the likelihood of such combinatorial therapies ending up in the clinic. Fur-
thermore, if nanoparticles are needed as carriers, a determination of which REFERENCES AND NOTES
nanoparticle would be best suited for each compound or imaging agent is
required. As reviewed here, nanoparticles can have varying properties, such 1. M. E. Fox, F. C. Szoka, J. M. Fréchet, Soluble polymer carriers for the treatment of cancer:
as aspect ratios, surface properties, and capacity for and interaction with The importance of molecular architecture. Acc. Chem. Res. 42, 1141–1151 (2009).
external and biological stimuli, that will determine the best nanoparticle 2. C. Schwarz, W. Mehnert, J. S. Lucks, R. H. Müller, Solid lipid nanoparticles (SLN) for
controlled drug delivery. I. Production, characterization and sterilization. J. Control. Release
for a particular application (Fig. 2). 30, 83–96 (1994).
Bringing nanomedicine to market is a multidisciplinary effort, and 3. S. K. Sahoo, V. Labhasetwar, Nanotech approaches to drug delivery and imaging. Drug
brings unique challenges and opportunities at the intersection of manu- Discov. Today 8, 1112–1120 (2003).
facturing, regulation, and funding. Regulatory bodies from all over the 4. V. P. Torchilin, Structure and design of polymeric surfactant-based drug delivery systems.
J. Control. Release 73, 137–172 (2001).
world, including the FDA, Pharmaceuticals and Medical Devices Agency
5. B. M. Discher, Y. Y. Won, D. S. Ege, J. C. Lee, F. S. Bates, D. E. Discher, D. A. Hammer,
of Japan, and European Medicines Agency, have been actively developing Polymersomes: Tough vesicles made from diblock copolymers. Science 284, 1143–1146
procedures for the consideration of new drugs or imaging agents that (1999).
are delivered via nanotechnology, and the general requirements for trans- 6. A. K. Patri, I. J. Majoros, J. R. Baker Jr., Dendritic polymer macromolecular carriers for drug
lation into clinical studies have been established (Fig. 2) (110–112). Be- delivery. Curr. Opin. Chem. Biol. 6, 466–471 (2002).
7. R. Langer, N. A. Peppas, Advances in biomaterials, drug delivery, and bionanotechnology.
fore the filing of an Investigative New Drug (IND) application, which is AIChE J. 49, 2990–3006 (2003).
important for both the commencement of clinical trials and any sub- 8. R. K. O’Reilly, C. J. Hawker, K. L. Wooley, Cross-linked block copolymer micelles: Functional
sequent product development, these requirements include compliance nanostructures of great potential and versatility. Chem. Soc. Rev. 35, 1068–1083 (2006).
with quality control guidelines [for example, good laboratory practice 9. C. J. Hawker, K. L. Wooley, The convergence of synthetic organic and polymer chemistries.
Science 309, 1200–1205 (2005).
(GLP) and good manufacturing practice (GMP)] to ensure material uni-
10. L. M. Ensign, B. C. Tang, Y. Y. Wang, T. A. Tse, T. Hoen, R. Cone, J. Hanes, Mucus-penetrating
formity and reproducibility. Absorption, distribution, metabolism, and nanoparticles for vaginal drug delivery protect against herpes simplex virus. Sci. Transl.
excretion (ADME) and toxicity studies account for acute and chronic Med. 4, 138ra79 (2012).
material safety. CMC guidelines, which include analytical testing and 11. E. A. Nance, G. F. Woodworth, K. A. Sailor, T. Y. Shih, Q. Xu, G. Swaminathan, D. Xiang,
process controls, serve as the foundation for synthesis at the kilogram C. Eberhart, J. Hanes, A dense poly(ethylene glycol) coating improves penetration of
large polymeric nanoparticles within brain tissue. Sci. Transl. Med. 4, 149ra119 (2012).
and larger quantity scales, as well as achieving the selected formulation 12. N. A. Peppas, J. Z. Hilt, A. Khademhosseini, R. Langer, Hydrogels in biology and medicine:
required for administration (for example, intramuscular, intravenous, From molecular principles to bionanotechnology. Adv. Mater. 18, 1345–1360 (2006).
and oral). 13. O. C. Farokhzad, S. Jon, A. Khademhosseini, T. N. Tran, D. A. Lavan, R. Langer, Nanoparticle-
The completion of these and other requirements for FDA IND aptamer bioconjugates: A new approach for targeting prostate cancer cells. Cancer Res. 64,
7668–7672 (2004).
filings and subsequent clinical trials represents an ambitious threshold. 14. A. Khademhosseini, R. Langer, Nanobiotechnology—Drug delivery and tissue engineering.
Key barriers in completing these requirements include the significant Chem. Eng. Prog. 102, 38–42 (2006).
costs of manufacturing scale-up, completing GLP-compliant preclinical 15. D. S. Katti, K. W. Robinson, F. K. Ko, C. T. Laurencin, Bioresorbable nanofiber-based
safety, and toxicity and efficacy studies that are adequately designed so systems for wound healing and drug delivery: Optimization of fabrication parameters.
J. Biomed. Mater. Res. B Appl. Biomater. 70, 286–296 (2004).
that findings can be accepted by the FDA. Despite these important chal- 16. J. D. Hartgerink, E. Beniash, S. I. Stupp, Peptide-amphiphile nanofibers: A versatile scaffold for
lenges that must be addressed, the cancer nanomedicine field is starting the preparation of self-assembling materials. Proc. Natl. Acad. Sci. U.S.A. 99, 5133–5138 (2002).
to experience success in navigating the complex roadmap needed to 17. X. Q. Zhang, X. Xu, R. Lam, D. Giljohann, D. Ho, C. A. Mirkin, Strategy for increasing drug
realize its impact in the clinic. As previously mentioned, funding is a solubility and efficacy through covalent attachment to polyvalent DNA-nanoparticle
conjugates. ACS Nano 5, 6962–6970 (2011).
necessary part of this roadmap, and support from government agencies, 18. I. H. El-Sayed, X. Huang, M. A. El-Sayed, Selective laser photo-thermal therapy of epithelial
venture firms, and pharmaceutical companies has served as a catalyst carcinoma using anti-EGFR antibody conjugated gold nanoparticles. Cancer Lett. 239,
for the translation of nanomedicines described in this Review. This is 129–135 (2006).
exemplified by partnerships between BIND Therapeutics [for example, 19. T. Paunesku, T. Rajh, G. Wiederrecht, J. Maser, S. Vogt, N. Stojićević, M. Protić, B. Lai, J. Oryhon,
M. Thurnauer, G. Woloschak, Biology of TiO2–oligonucleotide nanocomposites. Nat. Mater. 2,
BIND-014 (53)] and AstraZeneca, Amgen, and Pfizer. With these 343–346 (2003).
requirements in mind, success in achieving accelerated drug approval 20. J. S. Kim, E. Kuk, K. N. Yu, J. H. Kim, S. J. Park, H. J. Lee, S. H. Kim, Y. K. Park, Y. H. Park,
through the FDA 505(b)(2) pathway remains to be seen for cancer C. Y. Hwang, Y. K. Kim, Y. S. Lee, D. H. Jeong, M. H. Cho, Antimicrobial effects of silver
nanomedicines in the pipeline. The 505(b)(2) approval of Abraxane, nanoparticles. Nanomedicine 3, 95–101 (2007).
21. K. Ajima, T. Murakami, Y. Mizoguchi, K. Tsuchida, T. Ichihashi, S. Iijima, M. Yudasaka, En-
a paclitaxel-albumin nanoparticle for the treatment of non–small cell hancement of in vivo anticancer effects of cisplatin by incorporation inside single-wall
lung cancer in combination with carboplatin, represents a positive step carbon nanohorns. ACS Nano 2, 2057–2064 (2008).
forward for fast-track approvals of nanotherapeutics. This, coupled with 22. K. Ajima, M. Yudasaka, T. Murakami, A. Maigné, K. Shiba, S. Iijima, Carbon nanohorns as
the existing approvals for imaging drugs such as iron oxide (Feridex) anticancer drug carriers. Mol. Pharm. 2, 475–480 (2005).
23. A. Bianco, K. Kostarelos, M. Prato, Applications of carbon nanotubes in drug delivery. Curr.
and Gd-diethylenetriamine pentaacetic acid (Gd-DTPA/Magnevist), Opin. Chem. Biol. 9, 674–679 (2005).
may further support the translation of nanotechnology-modified im- 24. Z. Liu, K. Chen, C. Davis, S. Sherlock, Q. Cao, X. Chen, H. Dai, Drug delivery with carbon
aging compounds (Table 3). nanotubes for in vivo cancer treatment. Cancer Res. 68, 6652–6660 (2008).

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 10


REVIEW

25. Z. Liu, S. M. Tabakman, Z. Chen, H. Dai, Preparation of carbon nanotube bioconjugates for 50. A. L. van de Ven, P. Kim, O. Haley, J. R. Fakhoury, G. Adriani, J. Schmulen, P. Moloney,
biomedical applications. Nat. Protoc. 4, 1372–1382 (2009). F. Hussain, M. Ferrari, X. Liu, S. H. Yun, P. Decuzzi, Rapid tumoritropic accumulation of
26. E. K. Chow, X. Q. Zhang, M. Chen, R. Lam, E. Robinson, H. Huang, D. Schaffer, E. Osawa, systemically injected plateloid particles and their biodistribution. J. Control. Release 158,
A. Goga, D. Ho, Nanodiamond therapeutic delivery agents mediate enhanced chemo- 148–155 (2012).
resistant tumor treatment. Sci. Transl. Med. 3, 73ra21 (2011). 51. Y. Ren, H. W. Cheung, G. von Maltzhan, A. Agrawal, G. S. Cowley, B. A. Weir, J. S. Boehm,
27. H. Huang, E. Pierstorff, E. Osawa, D. Ho, Active nanodiamond hydrogels for chemo- P. Tamayo, A. M. Karst, J. F. Liu, M. S. Hirsch, J. P. Mesirov, R. Drapkin, D. E. Root, J. Lo, V. Fogal,
therapeutic delivery. Nano Lett. 7, 3305–3314 (2007). E. Ruoslahti, W. C. Hahn, S. N. Bhatia, Targeted tumor-penetrating siRNA nanocomplexes
28. H. Huang, E. Pierstorff, E. Osawa, D. Ho, Protein-mediated assembly of nanodiamond hy- for credentialing the ovarian cancer oncogene ID4. Sci. Transl. Med. 4, 147ra112 (2012).
drogels into a biocompatible and biofunctional multilayer nanofilm. ACS Nano 2, 203–212 52. P. L. Rodriguez, T. Harada, D. A. Christian, D. A. Pantano, R. K. Tsai, D. E. Discher, Minimal
(2008). “self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles.
29. V. N. Mochalin, O. Shenderova, D. Ho, Y. Gogotsi, The properties and applications of Science 339, 971–975 (2013).
nanodiamonds. Nat. Nanotechnol. 7, 11–23 (2011). 53. J. Hrkach, D. Von Hoff, M. Mukkaram Ali, E. Andrianova, J. Auer, T. Campbell, D. De Witt,
30. K. K. Liu, W. W. Zheng, C. C. Wang, Y. C. Chiu, C. L. Cheng, Y. S. Lo, C. Chen, J. I. Chao, M. Figa, M. Figueiredo, A. Horhota, S. Low, K. McDonnell, E. Peeke, B. Retnarajan, A. Sabnis,
Covalent linkage of nanodiamond-paclitaxel for drug delivery and cancer therapy. Nano- E. Schnipper, J. J. Song, Y. H. Song, J. Summa, D. Tompsett, G. Troiano, T. Van Geen Hoven,
technology 21, 315106 (2010). J. Wright, P. LoRusso, P. W. Kantoff, N. H. Bander, C. Sweeney, O. C. Farokhzad, R. Langer,
31. M. Liong, J. Lu, M. Kovochich, T. Xia, S. G. Ruehm, A. E. Nel, F. Tamanoi, J. I. Zink, Multi- S. Zale, Preclinical development and clinical translation of a PSMA-targeted docetaxel
functional inorganic nanoparticles for imaging, targeting, and drug delivery. ACS Nano 2, nanoparticle with a differentiated pharmacological profile. Sci. Transl. Med. 4, 128ra39 (2012).
889–896 (2008). 54. E. Ryschich, G. Huszty, H. P. Knaebel, M. Hartel, M. W. Büchler, J. Schmidt, Transferrin
32. H. Meng, W. X. Mai, H. Zhang, M. Xue, T. Xia, S. Lin, X. Wang, Y. Zhao, Z. Ji, J. I. Zink, A. E. Nel, receptor is a marker of malignant phenotype in human pancreatic cancer and in
Codelivery of an optimal drug/siRNA combination using mesoporous silica nanoparticles to neuroendocrine carcinoma of the pancreas. Eur. J. Cancer. 40, 1418–1422 (2004).
overcome drug resistance in breast cancer in vitro and in vivo. ACS Nano 7, 994–1005 (2013). 55. E. R. Camp, C. Wang, E. C. Little, P. M. Watson, K. F. Pirollo, A. Rait, D. J. Cole, E. H. Chang,
33. H. Meng, M. Xue, T. Xia, Z. Ji, D. Y. Tarn, J. I. Zink, A. E. Nel, Use of size and a copolymer D. K. Watson, Transferrin receptor targeting nanomedicine delivering wild-type p53 gene
design feature to improve the biodistribution and the enhanced permeability and retention sensitizes pancreatic cancer to gemcitabine therapy. Cancer Gene Ther. 20, 222–228 (2013).
effect of doxorubicin-loaded mesoporous silica nanoparticles in a murine xenograft tumor 56. N. Senzer, J. Nemunaitis, D. Nemunaitis, C. Bedell, G. Edelman, M. Barve, R. Nunan, K. F. Pirollo,
model. ACS Nano 5, 4131–4144 (2011). A. Rait, E. H. Chang, Phase I study of a systemically delivered p53 nanoparticle in advanced
34. J. Kim, J. E. Lee, J. Lee, J. H. Yu, B. C. Kim, K. An, Y. Hwang, C. H. Shin, J. G. Park, J. Kim, T. Hyeon, solid tumors. Mol. Ther. 21, 1096–1103 (2013).
Magnetic fluorescent delivery vehicle using uniform mesoporous silica spheres embedded 57. E. Ruoslahti, Specialization of tumour vasculature. Nat. Rev. Cancer 2, 83–90 (2002).
with monodisperse magnetic and semiconductor nanocrystals. J. Am. Chem. Soc. 128, 58. K. N. Sugahara, T. Teesalu, P. P. Karmali, V. R. Kotamraju, L. Agemy, D. R. Greenwald,
688–689 (2006). E. Ruoslahti, Coadministration of a tumor-penetrating peptide enhances the efficacy
35. E. S. Day, L. Zhang, P. A. Thompson, J. A. Zawaski, C. C. Kaffes, M. W. Gaber, S. M. Blaney, of cancer drugs. Science 328, 1031–1035 (2010).
J. L. West, Vascular-targeted photothermal therapy of an orthotopic murine glioma 59. K. N. Sugahara, T. Teesalu, P. P. Karmali, V. R. Kotamraju, L. Agemy, O. M. Girard, D. Hanahan,
model. Nanomedicine 7, 1133–1148 (2012). R. F. Mattrey, E. Ruoslahti, Tissue-penetrating delivery of compounds and nanoparticles into
36. L. M. Manus, D. J. Mastarone, E. A. Waters, X. Q. Zhang, E. A. Schultz-Sikma, K. W. Macrenaris, tumors. Cancer Cell 16, 510–520 (2009).
D. Ho, T. J. Meade, Gd(III)-nanodiamond conjugates for MRI contrast enhancement. Nano Lett. 60. P. Chaudhuri, A. Paraskar, S. Soni, R. A. Mashelkar, S. Sengupta, Fullerenol-cytotoxic con-
10, 484–489 (2010). jugates for cancer chemotherapy. ACS Nano 3, 2505–2514 (2009).
37. G. von Maltzahn, J. H. Park, K. Y. Lin, N. Singh, C. Schwöppe, R. Mesters, W. E. Berdel, 61. A. S. Paraskar, S. Soni, K. T. Chin, P. Chaudhuri, K. W. Muto, J. Berkowitz, M. W. Handlogten,
E. Ruoslahti, M. J. Sailor, S. N. Bhatia, Nanoparticles that communicate in vivo to amplify N. J. Alves, B. Bilgicer, D. M. Dinulescu, R. A. Mashelkar, S. Sengupta, Harnessing structure-
tumour targeting. Nat. Mater. 10, 545–552 (2011). activity relationship to engineer a cisplatin nanoparticle for enhanced antitumor efficacy.
38. Z. J. Deng, S. W. Morton, E. Ben-Akiva, E. C. Dreaden, K. E. Shopsowitz, P. T. Hammond, Proc. Natl. Acad. Sci. U.S.A. 107, 12435–12440 (2010).
Layer-by-layer nanoparticles for systemic codelivery of an anticancer drug and siRNA for 62. J. E. Podesta, K. T. Al-Jamal, M. A. Herrero, B. Tian, H. Ali-Boucetta, V. Hegde, A. Bianco,
potential triple-negative breast cancer treatment. ACS Nano 7, 9571–9584 (2013). M. Prato, K. Kostarelos, Antitumor activity and prolonged survival by carbon-nanotube-
39. World Health Organization, GLOBOCAN 2008 cancer fact sheet; http://globocan.iarc.fr/. mediated therapeutic siRNA silencing in a human lung xenograft model. Small 5, 1176–1185
40. American Cancer Society, Cancer facts and figures 2013; http://www.cancer.org/acs/ (2009).
groups/content/@epidemiologysurveilance/documents/document/acspc-036845.pdf. 63. N. R. Soman, S. L. Baldwin, G. Hu, J. N. Marsh, G. M. Lanza, J. E. Heuser, J. M. Arbeit,
41. R. K. Jain, Transport of molecules across tumor vasculature. Cancer Metastasis Rev. 6, 559–93 S. A. Wickline, P. H. Schlesinger, Molecularly targeted nanocarriers deliver the cytolytic
(1987). peptide melittin specifically to tumor cells in mice, reducing tumor growth. J. Clin. Invest.
42. U. Prabhakar, H. Maeda, R. K. Jain, E. M. Sevick-Muraca, W. Zamboni, O. C. Farokhzad, 119, 2830–2842 (2009).
S. T. Barry, A. Gabizon, P. Grodzinski, D. C. Blakey, Challenges and key considerations 64. D. B. Longley, P. G. Johnston, Molecular mechanisms of drug resistance. J. Pathol. 205,
of the enhanced permeability and retention effect for nanomedicine drug delivery in 275–292 (2005).
oncology. Cancer Res. 73, 2412–2417 (2013). 65. L. N. Abdullah, E. K. Chow, Mechanisms of chemoresistance in cancer stem cells. Clin.
43. J. Senior, J. C. Crawley, G. Gregoriadis, Tissue distribution of liposomes exhibiting long half- Transl. Med. 2, 3 (2013).
lives in the circulation after intravenous injection. Biochim. Biophys. Acta 839, 1–8 (1985). 66. W. S. Dalton, J. J. Crowley, S. S. Salmon, T. M. Grogan, L. R. Laufman, G. R. Weiss, J. D. Bonnet,
44. M. C. Woodle, K. K. Matthay, M. S. Newman, J. E. Hidayat, L. R. Collins, C. Redemann, F. J. Martin, A phase III randomized study of oral verapamil as a chemosensitizer to reverse drug
D. Papahadjopoulos, Versatility in lipid compositions showing prolonged circulation with resistance in patients with refractory myeloma. A Southwest Oncology Group study. Cancer
sterically stabilized liposomes. Biochim. Biophys. Acta 1105, 193–200 (1992). 75, 815–820 (1995).
45. L. Moore, E. K. Chow, E. Osawa, J. M. Bishop, D. Ho, Diamond-lipid hybrids enhance chemo- 67. C. Lhommé, F. Joly, J. L. Walker, A. A. Lissoni, M. O. Nicoletto, G. M. Manikhas, M. M. Baekelandt,
therapeutic tolerance and mediate tumor regression. Adv. Mater. 25, 3532–3541 (2013). A. N. Gordon, P. M. Fracasso, W. L. Mietlowski, G. J. Jones, M. H. Dugan, Phase III study of
46. A. M. Gobin, M. H. Lee, N. J. Halas, W. D. James, R. A. Drezek, J. L. West, Near-infrared valspodar (PSC 833) combined with paclitaxel and carboplatin compared with paclitaxel
resonant nanoshells for combined optical imaging and photothermal cancer therapy. and carboplatin alone in patients with stage IV or suboptimally debulked stage III epithelial
Nano Lett. 7, 1929–1934 (2007). ovarian cancer or primary peritoneal cancer. J. Clin. Oncol. 26, 2674–2682 (2008).
47. N. J. Robert, C. L. Vogel, I. C. Henderson, J. A. Sparano, M. R. Moore, P. Silverman, B. A. Overmoyer, 68. H. B. Man, H. Kim, H. J. Kim, E. Robinson, W. K. Liu, E. K. Chow, D. Ho, Synthesis of nanodiamond–
C. L. Shapiro, J. W. Park, G. T. Colbern, E. P. Winer, A. A. Gabizon, The role of the liposomal daunorubicin conjugates to overcome multidrug chemoresistance in leukemia. Nanomedicine
anthracyclines and other systemic therapies in the management of advanced breast cancer. 10.1016/j.nano.2013.07.014 (2013).
Semin. Oncol. 31, 106–146 (2004). 69. M. E. Davis, J. E. Zuckerman, C. H. Choi, D. Seligson, A. Tolcher, C. A. Alabi, Y. Yen, J. D. Heidel,
48. J. W. Valle, A. Armstrong, C. Newman, V. Alakhov, G. Pietrzynski, J. Brewer, S. Campbell, A. Ribas, Evidence of RNAi in humans from systemically administered siRNA via targeted
P. Corrie, E. K. Rowinsky, M. Ranson, A phase 2 study of SP1049C, doxorubicin in P-glycoprotein- nanoparticles. Nature 464, 1067–1070 (2010).
targeting pluronics, in patients with advanced adenocarcinoma of the esophagus and 70. M. E. Davis, The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin
gastroesophageal junction. Invest. New Drugs 29, 1029–1037 (2011). polymer-based nanoparticle: From concept to clinic. Mol. Pharm. 6, 659–668 (2009).
49. S. Danson, D. Ferry, V. Alakhov, J. Margison, D. Kerr, D. Jowle, M. Brampton, G. Halbert, 71. J. D. Heidel, J. Y. Liu, Y. Yen, B. Zhou, B. S. Heale, J. J. Rossi, D. W. Bartlett, M. E. Davis,
M. Ranson, Phase I dose escalation and pharmacokinetic study of pluronic polymer-bound Potent siRNA inhibitors of ribonucleotide reductase subunit RRM2 reduce cell prolifera-
doxorubicin (SP1049C) in patients with advanced cancer. Br. J. Cancer 90, 2085–2091 (2004). tion in vitro and in vivo. Clin. Cancer Res. 13, 2207–2215 (2007).

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 11


REVIEW

72. M. Koldehoff, N. K. Steckel, D. W. Beelen, A. H. Elmaagacli, Therapeutic application of small 94. C. Wong, T. Stylianopoulos, J. Cui, J. Martin, V. P. Chauhan, W. Jiang, Z. Popovic, R. K. Jain,
interfering RNA directed against bcr-abl transcripts to a patient with imatinib-resistant chron- M. G. Bawendi, D. Fukumura, Multistage nanoparticle delivery system for deep penetration
ic myeloid leukaemia. Clin. Exp. Med. 7, 47–55 (2007). into tumor tissue. Proc. Natl. Acad. Sci. U.S.A. 108, 2426–2431 (2011).
73. S. A. Jensen, E. S. Day, C. H. Ko, L. A. Hurley, J. P. Luciano, F. M. Kouri, T. J. Merkel, A. J. Luthi, 95. J. Liu, S. K. Lau, V. A. Varma, R. A. Moffitt, M. Caldwell, T. Liu, A. N. Young, J. A. Petros,
P. C. Patel, J. I. Cutler, W. L. Daniel, A. W. Scott, M. W. Rotz, T. J. Meade, D. A. Giljohann, A. O. Osunkoya, T. Krogstad, B. Leyland-Jones, M. D. Wang, S. Nie, Molecular mapping of
C. A. Mirkin, A. H. Stegh, Spherical nucleic acid nanoparticle conjugates as an RNAi-based tumor heterogeneity on clinical tissue specimens with multiplexed quantum dots. ACS
therapy for glioblastoma. Sci. Transl. Med. 5, 209ra152 (2013). Nano 4, 2755–2765 (2010).
74. C. Loo, A. Lowery, N. Halas, J. West, R. Drezek, Immunotargeted nanoshells for integrated 96. S. D. Perrault, W. C. Chan, In vivo assembly of nanoparticle components to improve targeted
cancer imaging and therapy. Nano Lett. 5, 709–711 (2005). cancer imaging. Proc. Natl. Acad. Sci. U.S.A. 107, 11194–11199 (2010).
75. L. B. Carpin, L. R. Bickford, G. Agollah, T. K. Yu, R. Schiff, Y. Li, R. A. Drezek, Immunoconjugated 97. B. Sana, E. Johnson, K. Sheah, C. L. Poh, S. Lim, Iron-based ferritin nanocore as a contrast
gold nanoshell-mediated photothermal ablation of trastuzumab-resistant breast cancer cells. agent. Biointerphases 5, FA48–FA52 (2010).
Breast Cancer Res. Treat. 125, 27–34 (2011). 98. E. Tóth, R. D. Bolskar, A. Borel, G. González, L. Helm, A. E. Merbach, B. Sitharaman, L. J. Wilson,
76. X. Hu, X. Gao, Multilayer coating of gold nanorods for combined stability and bio- Water-soluble gadofullerenes: Toward high-relaxivity, pH-responsive MRI contrast agents.
compatibility. Phys. Chem. Chem. Phys. 13, 10028–10035 (2011). J. Am. Chem. Soc. 127, 799–805 (2004).
77. R. K. Kannadorai, Q. Liu, Optimization in interstitial plasmonic photothermal therapy for 99. J. S. Ananta, B. Godin, R. Sethi, L. Moriggi, X. Liu, R. E. Serda, R. Krishnamurthy, R. Muthupillai,
treatment planning. Med. Phys. 40, 103301 (2013). R. D. Bolskar, L. Helm, M. Ferrari, L. J. Wilson, P. Decuzzi, Geometrical confinement of
78. C. L. Chen, L. R. Kuo, S. Y. Lee, Y. K. Hwu, S. W. Chou, C. C. Chen, F. H. Chang, K. H. Lin, gadolinium-based contrast agents in nanoporous particles enhances T1 contrast.
D. H. Tsai, Y. Y. Chen, Photothermal cancer therapy via femtosecond-laser-excited FePt Nat. Nanotechnol. 5, 815–821 (2010).
nanoparticles. Biomaterials 34, 1128–1134 (2013). 100. M. F. Kircher, A. de la Zerda, J. V. Jokerst, C. L. Zavaleta, P. J. Kempen, E. Mittra, K. Pitter,
79. P. Pouponneau, J. C. Leroux, G. Soulez, L. Gaboury, S. Martel, Co-encapsulation of mag- R. Huang, C. Campos, F. Habte, R. Sinclair, C. W. Brennan, I. K. Mellinghoff, E. C. Holland,
netic nanoparticles and doxorubicin into biodegradable microcarriers for deep tissue S. S. Gambhir, A brain tumor molecular imaging strategy using a new triple-modality
targeting by vascular MRI navigation. Biomaterials 32, 3481–3486 (2011). MRI-photoacoustic-Raman nanoparticle. Nat. Med. 18, 829–834 (2012).
80. L. Maggiorella, G. Barouch, C. Devaux, A. Pottier, E. Deutsch, J. Bourhis, E. Borghi, L. Levy, 101. P. C. M. van Zijl, N. N. Yadav, Chemical exchange saturation transfer (CEST): What is in a
Nanoscale radiotherapy with hafnium oxide nanoparticles. Future Oncol. 8, 1167–1181 name and what isn’t? Magn. Reson. Med. 65, 927–948 (2011).
(2012). 102. G. Liu, M. Moake, Y. E. Har-el, C. M. Long, K. W. Chan, A. Cardona, M. Jamil, P. Walczak,
81. P. R. Dal Monte, F. C. Szoka Jr., Antigen presentation by B cells and macrophages of A. A. Gilad, G. Sgouros, P. C. van Zijl, J. W. Bulte, M. T. McMahon, In vivo multicolor mo-
cytochrome c and its antigenic fragment when conjugated to the surface of liposomes. lecular MR imaging using diamagnetic chemical exchange saturation transfer liposomes.
Vaccine 7, 401–408 (1989). Magn. Reson. Med. 67, 1106–1113 (2012).
82. M. Garinot, V. Fiévez, V. Pourcelle, F. Stoffelbach, A. des Rieux, L. Plapied, I. Theate, H. Freichels, 103. K. W. Chan, G. Liu, X. Song, H. Kim, T. Yu, D. R. Arifin, A. A. Gilad, J. Hanes, P. Walczak,
C. Jérôme, J. Marchand-Brynaert, Y. J. Schneider, V. Préat, PEGylated PLGA-based nanoparticles P. C. van Zijl, J. W. Bulte, M. T. McMahon, MRI-detectable pH nanosensors incorporated
targeting M cells for oral vaccination. J. Control. Release 120, 195–204 (2007). into hydrogels for in vivo sensing of transplanted-cell viability. Nat. Mater. 12, 268–275
83. D. N. Nguyen, K. P. Mahon, G. Chikh, P. Kim, H. Chung, A. P. Vicari, K. T. Love, M. Goldberg, (2013).
S. Chen, A. M. Krieg, J. Chen, R. Langer, D. G. Anderson, Lipid-derived nanoparticles for 104. B. S. Ferguson, D. A. Hoggarth, D. Maliniak, K. Ploense, R. J. White, N. Woodward, K. Hsieh,
immunostimulatory RNA adjuvant delivery. Proc. Natl. Acad. Sci. U.S.A. 109, E797–E803 (2012). A. J. Bonham, M. Eisenstein, T. E. Kippin, K. W. Plaxco, H. T. Soh, Real-time, aptamer-based
84. S. Kageyama, S. Kitano, M. Hirayama, Y. Nagata, H. Imai, T. Shiraishi, K. Akiyoshi, A. M. Scott, tracking of circulating therapeutic agents in living animals. Sci. Transl. Med. 5, 213ra165
R. Murphy, E. W. Hoffman, L. J. Old, N. Katayama, H. Shiku, Humoral immune responses in (2013).
patients vaccinated with 1–146 HER2 protein complexed with cholesteryl pullulan nanogel. 105. J. Reed, J. Chun, T. A. Zangle, S. Kalim, J. S. Hong, S. E. Pefley, X. Zheng, J. K. Gimzewski,
Cancer Sci. 99, 601–607 (2008). M. A. Teitell, Rapid, massively parallel single-cell drug response measurements via live
85. S. M. Goldinger, R. Dummer, P. Baumgaertner, D. Mihic-Probst, K. Schwarz, A. Hammann-Haenni, cell interferometry. Biophys. J. 101, 1025–1031 (2011).
J. Willers, C. Geldhof, J. O. Prior, T. M. Kündig, O. Michielin, M. F. Bachmann, D. E. Speiser, 106. C. P. Sun, T. Usui, F. Yu, I. Al-Shyoukh, J. Shamma, R. Sun, C. M. Ho, Integrative systems
Nano-particle vaccination combined with TLR-7 and -9 ligands triggers memory and control approach for reactivating Kaposi’s sarcoma-associated herpesvirus (KSHV) with
effector CD8+ T-cell responses in melanoma patients. Eur. J. Immunol. 42, 3049–3061 (2012). combinatory drugs. Integr. Biol. 1, 123–130 (2009).
86. X. G. Gu, M. Schmitt, A. Hiasa, Y. Nagata, H. Ikeda, Y. Sasaki, K. Akiyoshi, J. Sunamoto, 107. P. K. Wong, F. Yu, A. Shahangian, G. Cheng, R. Sun, C. M. Ho, Closed-loop control of
H. Nakamura, K. Kuribayashi, H. Shiku, A novel hydrophobized polysaccharide/oncoprotein cellular functions using combinatory drugs guided by a stochastic search algorithm. Proc.
complex vaccine induces in vitro and in vivo cellular and humoral immune responses against Natl. Acad. Sci. U.S.A. 105, 5105–5110 (2008).
HER2-expressing murine sarcomas. Cancer Res. 58, 3385–3390 (1998). 108. X. Sun, S. Vilar, N. P. Tatonetti, High-throughput methods for combinatorial drug discov-
87. S. Kitano, S. Kageyama, Y. Nagata, Y. Miyahara, A. Hiasa, H. Naota, S. Okumura, H. Imai, ery. Sci. Transl. Med. 5, 205rv1 (2013).
T. Shiraishi, M. Masuya, M. Nishikawa, J. Sunamoto, K. Akiyoshi, T. Kanematsu, A. M. Scott, 109. H. Meng, Y. Zhao, J. Dong, M. Xue, Y. S. Lin, Z. Ji, W. X. Mai, H. Zhang, C. H. Chang, C. J. Brinker,
R. Murphy, E. W. Hoffman, L. J. Old, H. Shiku, HER2-specific T-cell immune responses in J. I. Zink, A. E. Nel, Two-wave nanotherapy to target the stroma and optimize gemcitabine
patients vaccinated with truncated HER2 protein complexed with nanogels of cholesteryl delivery to a human pancreatic cancer model in mice. ACS Nano 7, 10048–10065 (2013).
pullulan. Clin. Cancer Res. 12, 7397–7405 (2006). 110. Food and Drug Administration Draft Guidance, Considering whether an FDA-regulated product
88. G. E. Peoples, J. P. Holmes, M. T. Hueman, E. A. Mittendorf, A. Amin, S. Khoo, Z. A. Dehqanzada, involves the application of nanotechnology; www.fda.gov/regulatoryinformation/guidances/
J. M. Gurney, M. M. Woll, G. B. Ryan, C. E. Storrer, D. Craig, C. G. Ioannides, S. Ponniah, Combined ucm257698.htm.
clinical trial results of a HER2/neu (E75) vaccine for the prevention of recurrence in high-risk 111. Food and Drug Administration Consumer Update, FDA continues dialogue on ‘nano’
breast cancer patients: U.S. Military Cancer Institute Clinical Trials Group Study I-01 and I-02. regulation; www.fda.gov/forconsumers/consumerupdates/ucm258462.htm.
Clin. Cancer Res. 14, 797–803 (2008). 112. European Medicines Agency Reflection Paper, Joint MHLW/EMA reflection paper on the
89. E. A. Scott, A. Stano, M. Gillard, A. C. Maio-Liu, M. A. Swartz, J. A. Hubbell, Dendritic cell development of block copolymer micelle medicinal products; www.ema.europa.eu/docs/
activation and T cell priming with adjuvant- and antigen-loaded oxidation-sensitive poly- en_GB/document_library/Scientific_guideline/2013/02/WC500138390.pdf.
mersomes. Biomaterials 33, 6211–6219 (2012).
90. O. A. Ali, D. Emerich, G. Dranoff, D. J. Mooney, In situ regulation of DC subsets and T cells Acknowledgments: We would like to acknowledge the studies that we were unable to in-
mediates tumor regression in mice. Sci. Transl. Med. 1, 8ra19 (2009). clude in this Review. Competing interests: D.H. is listed as an inventor on pending patents
91. R. W. Ross, A. L. Zietman, W. Xie, J. J. Coen, D. M. Dahl, W. U. Shipley, D. S. Kaufman, T. Islam, pertaining to ND drug delivery and imaging.
A. R. Guimaraes, R. Weissleder, M. Harisinghani, Lymphotropic nanoparticle-enhanced
magnetic resonance imaging (LNMRI) identifies occult lymph node metastases in pros- Submitted 5 February 2013
tate cancer patients prior to salvage radiation therapy. Clin. Imaging 33, 301–305 (2009). Accepted 27 November 2013
92. S. Jiang, A. A. Eltoukhy, K. T. Love, R. Langer, D. G. Anderson, Lipidoid-coated iron oxide Published 18 December 2013
nanoparticles for efficient DNA and siRNA delivery. Nano Lett. 13, 1059–1064 (2013). 10.1126/scitranslmed.3005872
93. M. Levy, N. Luciani, D. Alloyeau, D. Elgrabli, V. Deveaux, C. Pechoux, S. Chat, G. Wang, N. Vats,
F. Gendron, C. Factor, S. Lotersztajn, A. Luciani, C. Wilhelm, F. Gazeau, Long term in vivo Citation: E. K.-H. Chow, D. Ho, Cancer nanomedicine: From drug delivery to imaging. Sci.
biotransformation of iron oxide nanoparticles. Biomaterials 32, 3988–3999 (2011). Transl. Med. 5, 216rv4 (2013).

www.ScienceTranslationalMedicine.org 18 December 2013 Vol 5 Issue 216 216rv4 12

You might also like