Recent advances in nanotechnology have established its importance in several areas including medicine. The myriad of applications in oncology range from detection and diagnosis to drug delivery and treatment. Although nanotechnology has attracted a lot of attention, the practical application of nanotechnology to clinical cancer care is still in its infancy. This review summarizes the role that nanotechnology has played in improving cancer therapy, its potential for affecting all aspects of cancer care, and the challenges that must be overcome to realize its full promise. Clin Cancer Res; 21(14); 3121–30. ©2015 AACR.

No potential conflicts of interest were disclosed.

The following editor(s) reported relevant financial relationships: J.R. Grandis—none.

The members of the planning committee have no real or apparent conflicts of interest to disclose.

Upon completion of this activity, the participant should have a better knowledge about the role of nanotechnology in various aspects of cancer therapy and the challenges that must be overcome to improve its transition to clinical practice.

This activity does not receive commercial support.

Nanomedicine offers unique opportunities for improving current ways of treating cancer and other diseases (Fig. 1). These stem from the potential of nanoformulations to improve drug delivery and achieve targeted delivery, thereby reducing systemic toxicity (1). Various nanoparticle formulations, such as quantum dots, liposomes, polymeric nanoparticles, carbon nanotubes, metallic nanoparticles, or dendrimers, have been investigated in preclinical and clinical settings for drug or gene delivery, photothermal therapy, immunotherapy, and imaging (Table 1). Although only a few formulations have been approved by the FDA (Table 2), the full potential of nanotechnology in the clinical setting is yet to be realized. Here, we review the successes of nanotechnology in cancer care and provide a critical appraisal of its future applications.

Figure 1.

Shown are some of the advantages of nanoparticles over conventional therapeutic modalities.

Figure 1.

Shown are some of the advantages of nanoparticles over conventional therapeutic modalities.

Close modal
Table 1.

Types of nanoparticles

NameFormulationAdvantagesDisadvantages and toxicityApplication
Liposome Self-assembled bilayers of phospholipid molecules 
  • Can incorporate hydrophobic molecules in the core and hydrophilic molecules in between the bilayer

  • Increased circulation time

  • Biodegradable

 
Cationic liposomes can lead to hemolysis and blood coagulation Mainly used for drug delivery (e.g., Doxil, DaunoXome) 
Polymeric nanoparticles 
  • Polymer–protein conjugation by chemical conjugation

  • Polymeric micelles by the emulsification or precipitation method

 
  • Biocompatible and biodegradable

  • Increased circulation time

  • Reduced toxicity

  • Surface can be conjugated with ligands

 
Toxicity at very high doses Drug delivery (Abraxane), siRNA delivery (currently in clinical trials) 
Dendrimers Highly branched polymeric molecules with a central core 
  • Chemical composition can be controlled, thus pharmacokinetic properties can be predicted

  • Ability to incorporate drug in the core or conjugate it to the surface

  • Multivalent structure allows sites for ligand conjugation for targeted delivery

 
  • Difficult to scale up

  • Cationic dendrimers lead to intravascular coagulation

 
Drug, siRNA, or plasmid delivery 
Gold nanoparticle Reduction and precipitation of ions lead to nanoformulations 
  • Biocompatible

  • Surface plasmon resonance

  • Increased sensitivity as a contrast agent

  • Can be used as imaging and therapeutic agent simultaneously

 
Some particle retention after therapy 
  • Currently in clinical trials for photothermal application

  • MRI

  • Drug delivery

 
Iron oxide nanoparticle Reduction and precipitation of ions lead to nanoformulation 
  • Supermagnetic property

  • Ease of synthesis

  • Can be used as a theranostic agent

 
Hydrophobic surface leads to particle aggregation 
  • MRI

  • Photothermal ablation

  • Drug delivery

 
Carbon nanotubes Single- or multiwalled cylindrical graphene sheets 
  • Ease of internalization by cells

  • Surface can be functionalized for attaching drugs/proteins/ligands, etc.

 
  • Inflammatory responses after aggregation

  • Pulmonary toxicities

  • Possible carcinogenic activity

 
  • Imaging

  • Drug delivery

 
Quantum dots 
  • Core-shell structure: most common are cadmium selenium and cadmium tellurium

  • Shell: zinc sulfide, zinc selenium

 
  • Ease of formulation

  • Higher brightness and photosensitivity than conventional dyes

  • Imaging and delivery agent

 
  • Limited tissue penetration

  • Low resolution of deeply situated tumor

  • Release of cadmium leads to toxicities

 
  • Imaging

  • Drug and gene delivery

 
NameFormulationAdvantagesDisadvantages and toxicityApplication
Liposome Self-assembled bilayers of phospholipid molecules 
  • Can incorporate hydrophobic molecules in the core and hydrophilic molecules in between the bilayer

  • Increased circulation time

  • Biodegradable

 
Cationic liposomes can lead to hemolysis and blood coagulation Mainly used for drug delivery (e.g., Doxil, DaunoXome) 
Polymeric nanoparticles 
  • Polymer–protein conjugation by chemical conjugation

  • Polymeric micelles by the emulsification or precipitation method

 
  • Biocompatible and biodegradable

  • Increased circulation time

  • Reduced toxicity

  • Surface can be conjugated with ligands

 
Toxicity at very high doses Drug delivery (Abraxane), siRNA delivery (currently in clinical trials) 
Dendrimers Highly branched polymeric molecules with a central core 
  • Chemical composition can be controlled, thus pharmacokinetic properties can be predicted

  • Ability to incorporate drug in the core or conjugate it to the surface

  • Multivalent structure allows sites for ligand conjugation for targeted delivery

 
  • Difficult to scale up

  • Cationic dendrimers lead to intravascular coagulation

 
Drug, siRNA, or plasmid delivery 
Gold nanoparticle Reduction and precipitation of ions lead to nanoformulations 
  • Biocompatible

  • Surface plasmon resonance

  • Increased sensitivity as a contrast agent

  • Can be used as imaging and therapeutic agent simultaneously

 
Some particle retention after therapy 
  • Currently in clinical trials for photothermal application

  • MRI

  • Drug delivery

 
Iron oxide nanoparticle Reduction and precipitation of ions lead to nanoformulation 
  • Supermagnetic property

  • Ease of synthesis

  • Can be used as a theranostic agent

 
Hydrophobic surface leads to particle aggregation 
  • MRI

  • Photothermal ablation

  • Drug delivery

 
Carbon nanotubes Single- or multiwalled cylindrical graphene sheets 
  • Ease of internalization by cells

  • Surface can be functionalized for attaching drugs/proteins/ligands, etc.

 
  • Inflammatory responses after aggregation

  • Pulmonary toxicities

  • Possible carcinogenic activity

 
  • Imaging

  • Drug delivery

 
Quantum dots 
  • Core-shell structure: most common are cadmium selenium and cadmium tellurium

  • Shell: zinc sulfide, zinc selenium

 
  • Ease of formulation

  • Higher brightness and photosensitivity than conventional dyes

  • Imaging and delivery agent

 
  • Limited tissue penetration

  • Low resolution of deeply situated tumor

  • Release of cadmium leads to toxicities

 
  • Imaging

  • Drug and gene delivery

 
Table 2.

Nanoformulations currently in clinical trials

NameFormulationIndicationPhase statusTrial number (if applicable)
Drug delivery 
Myocet (Sopherion) Liposomal doxorubicin Metastastic breast cancer Approved  
DaunoXome (Galen) Liposomal daunorubicin Kaposi sarcoma Approved  
Doxil (Janssen) PEGylated liposomal doxorubicin 
  • Kaposi sarcoma

  • Recurrent ovarian cancer

  • Metastatic breast cancer

  • Multiple myeloma

 
Approved  
Marqibo (Spectrum/Talon) Liposomal vincristine Acute lymphoblastic leukemia Approved  
Abraxane (Celgene) Albumin-bound paclitaxel 
  • Breast cancer

  • Non–small cell lung cancer

  • Pancreatic cancer

 
Approved  
Paclitaxel poliglumex Polyamino acid bound paclitaxel 
  • Head and neck cancer

  • Ovarian cancer

  • Glioma

  • Non–small cell lung cancer

 
  • Phase I/II

  • Phase III

  • Phase II

 
  • NCT00660218

  • NCT00269828

  • NCT01402063

  • NCT00045682

 
Zinostatin stimalamer Neocarzinostatin SMANCS (polymer–protein conjugate) Hepatocellular carcinoma Approved  
Oncaspar (Sigma-Tau) PEG-L-asparaginase Acute lymphoblastic leukemia Approved  
siRNA delivery 
CALAA-01 Transferrin-targeted cyclodextrin nanoparticle with siRRM2 Solid tumor Phase I NCT00689065 
Atu027 Cationic liposome-siPKN3 Solid tumor Phase I 
  • NCT00938574

  • NCT01808638

 
TKM 080301 SNALP-siPLK1 Solid tumor Phase I NCT01262235 
ALN-VSP02 SNALP-siVEGF and siKSP Solid tumors Phase I NCT01158079 
Epharna (unlicensed) EphA2 siRNA-DOPC Solid tumor Phase I NCT01591356 
Immunotherapeutic agents 
CHP-HER2 and CHP-NY-ESO-01 Cholesterol-bearing hydrophobized pullulan HER2 protein 146 (CHP-HER2) and NY-ESO-1 protein (CHP-NY-ESO-1) in combination with OK-432 
  • Esophageal cancer

  • Lung cancer

  • Stomach cancer

  • Breast cancer

  • Ovarian cancer

 
Phase I NCT00291473 
CYT004-MelQbG10 Virus-like nanoparticle with antigens Melan-A/MART-1 and adjuvant CpG-oligonucleotide (ODn) Malignant melanoma Phase II NCT00651703 
Photothermal (Nanospectra) and radiotherapy application 
AuroLase (Nanospectra) Silica core with gold metal shell and near IR laser 
  • Head and neck cancer

  • Lung cancer

 
Phase I NCT00848042, NCT01679470 
NBTXR3 Hafnium oxide nanocrystals Soft tissue sarcoma Phase I NCT01946867, NCT01433068 
Imaging agents 
SPIO MRI Ultrasmall superparamagnetic iron oxide MRI Pancreatic cancer Phase IV NCT00920023 
Fluorescent cRGDY-PEG-Cy5.5-C dots RGD-labeled silica nanoparticle with Cy5.5 dye Solid tumor Phase 0 NCT02106598 
Carbon nanoparticles Carbon nanoparticles Advanced gastric cancer Phase III NCT02123407 
NameFormulationIndicationPhase statusTrial number (if applicable)
Drug delivery 
Myocet (Sopherion) Liposomal doxorubicin Metastastic breast cancer Approved  
DaunoXome (Galen) Liposomal daunorubicin Kaposi sarcoma Approved  
Doxil (Janssen) PEGylated liposomal doxorubicin 
  • Kaposi sarcoma

  • Recurrent ovarian cancer

  • Metastatic breast cancer

  • Multiple myeloma

 
Approved  
Marqibo (Spectrum/Talon) Liposomal vincristine Acute lymphoblastic leukemia Approved  
Abraxane (Celgene) Albumin-bound paclitaxel 
  • Breast cancer

  • Non–small cell lung cancer

  • Pancreatic cancer

 
Approved  
Paclitaxel poliglumex Polyamino acid bound paclitaxel 
  • Head and neck cancer

  • Ovarian cancer

  • Glioma

  • Non–small cell lung cancer

 
  • Phase I/II

  • Phase III

  • Phase II

 
  • NCT00660218

  • NCT00269828

  • NCT01402063

  • NCT00045682

 
Zinostatin stimalamer Neocarzinostatin SMANCS (polymer–protein conjugate) Hepatocellular carcinoma Approved  
Oncaspar (Sigma-Tau) PEG-L-asparaginase Acute lymphoblastic leukemia Approved  
siRNA delivery 
CALAA-01 Transferrin-targeted cyclodextrin nanoparticle with siRRM2 Solid tumor Phase I NCT00689065 
Atu027 Cationic liposome-siPKN3 Solid tumor Phase I 
  • NCT00938574

  • NCT01808638

 
TKM 080301 SNALP-siPLK1 Solid tumor Phase I NCT01262235 
ALN-VSP02 SNALP-siVEGF and siKSP Solid tumors Phase I NCT01158079 
Epharna (unlicensed) EphA2 siRNA-DOPC Solid tumor Phase I NCT01591356 
Immunotherapeutic agents 
CHP-HER2 and CHP-NY-ESO-01 Cholesterol-bearing hydrophobized pullulan HER2 protein 146 (CHP-HER2) and NY-ESO-1 protein (CHP-NY-ESO-1) in combination with OK-432 
  • Esophageal cancer

  • Lung cancer

  • Stomach cancer

  • Breast cancer

  • Ovarian cancer

 
Phase I NCT00291473 
CYT004-MelQbG10 Virus-like nanoparticle with antigens Melan-A/MART-1 and adjuvant CpG-oligonucleotide (ODn) Malignant melanoma Phase II NCT00651703 
Photothermal (Nanospectra) and radiotherapy application 
AuroLase (Nanospectra) Silica core with gold metal shell and near IR laser 
  • Head and neck cancer

  • Lung cancer

 
Phase I NCT00848042, NCT01679470 
NBTXR3 Hafnium oxide nanocrystals Soft tissue sarcoma Phase I NCT01946867, NCT01433068 
Imaging agents 
SPIO MRI Ultrasmall superparamagnetic iron oxide MRI Pancreatic cancer Phase IV NCT00920023 
Fluorescent cRGDY-PEG-Cy5.5-C dots RGD-labeled silica nanoparticle with Cy5.5 dye Solid tumor Phase 0 NCT02106598 
Carbon nanoparticles Carbon nanoparticles Advanced gastric cancer Phase III NCT02123407 

Small-molecule drug delivery

Chemotherapy drugs are used for many cancer types, but conventional chemotherapy is nonspecific and can lead to intolerable toxicities, compromising patients' quality of life. Nanotechnology has the potential to overcome such hurdles. Targeted delivery, reduced toxicity, improved pharmacokinetics, and bioavailability are some of the potential advantages offered by nanotechnology.

Among various nanoparticle platforms, liposomes are the most advanced with regard to integration into clinical care. Liposomal incorporation of doxorubicin and daunorubicin increase plasma concentration and reduce clearance rate and volume of distribution, thus increasing bioavailability of the drug (2, 3). Moreover, there is a substantial decrease in cardiac and other toxicities with liposomal doxorubicin as compared with free doxorubicin (4). Further improvement in the safety and pharmacokinetics was achieved by using polyethylene glycol (PEG) to coat liposomes (5–7).

Polymeric nanoparticles have also been instrumental in improving the therapeutic window of conventional drugs. For instance, the use of Cremophor (Sigma-Aldrich) with paclitaxel contributes to hypersensitivity reactions and neuropathy, but albumin nanoparticle-based formulation of paclitaxel facilitates endothelial transcytosis to achieve significant accumulation in the tumor (8). Phase I evaluation established that maximum tolerated dose (MTD) of such nanoparticles was about 70% higher than traditional paclitaxel (9). This formulation is associated with lower neutropenia and hypersensitivity while achieving a higher response rate than standard paclitaxel (10). Paclitaxel poliglumex poly(L-glutamic acid)-paclitaxel is another polymeric formulation with increased water solubility of paclitaxel, increased plasma half-life, tumor uptake, increased antitumor activity, and an improved safety profile compared with free paclitaxel (11, 12).

The nanoparticle platforms discussed above rely predominantly on passive accumulation of nanoparticles at tumor sites based on the enhanced permeability and retention (EPR) effect. Tumor selectivity can be further enhanced by attaching tumor-specific ligands (e.g., folic acid, HER2 antibody, aptamers, and transferrin) to nanoparticles to enhance tumor accumulation, increased cellular internalization and increased antitumor effects (13–17). For example, MCC-465 (PEGylated immunoliposome conjugated with F(ab′)2 fragment of GAH and encapsulates doxorubicin) was well tolerated in preclinical and early clinical testing (18). MM-302 is a HER-2–targeted PEGylated liposome containing doxorubicin that has shown an improved cardiac toxicity profile in combination with trastuzumab (19). Phase I testing of cetuximab-conjugated doxorubicin liposome was also well tolerated (20). Additional formulations, such as MBP-426 and SGT53 (p53), are in clinical testing (21).

Several nanoparticle strategies have been developed for targeting stromal populations such as endothelial cells, macrophages, and cancer stem cells. Paclitaxel loaded into poly (lactic-co-glycolic) acid PLGA nanoparticles decorated with CD133 antibody resulted in enhanced survival in preclinical cancer models (22). Combination therapy with epigenetic-targeted decitabine and doxorubicin nanoparticles targeting cancer stem cells was shown to be more beneficial than free decitabine and doxorubicin in chemoresistant breast cancer models (23). Chitosan nanoparticles decorated with RGD peptides localize to the tumor vasculature and exert antiangiogenic effects (24). The next-generation nanoparticles aim to achieve further selectivity by allowing spatiotemporal control over drug release. These nanoparticles are designed to selectively release drugs in response to stimuli such as an alternating magnetic field, UV or near IR radiation, or low pH in the tumor microenvironment (25–29). However, issues related to tumor heterogeneity, cost considerations and changes in characteristics of a nanoparticle after ligand conjugation will require careful consideration during drug development.

Nucleotide delivery

Nucleotide therapies hold an important place in cancer therapy because many of the undruggable genes can be targeted using antisense oligonucleotides (ASO) or siRNAs. Several ASOs are now in clinical trials, but the success has been modest (30). SiRNAs may be a better alternative to ASOs due to ease of synthesis and ability to achieve greater silencing at lower concentration than ASOs. However, several challenges associated with siRNA (e.g., enzymatic degradation in plasma, inefficient uptake by cells, and immunostimulation) must be overcome. Several nanoparticle platforms have been investigated to overcome these hurdles in siRNA delivery. Although some cationic liposomes are efficacious, these carriers can cause toxicities (e.g., activation of complement system and inflammatory responses) (31–33). Formulations such as AtuPLEX showed that toxicity can be reduced by incorporation of helper neutral lipids and PEGylation (34). The lipoplex Atu027 containing siPKN3 is currently in clinical trials for advanced solid cancers. Although preclinical studies showed antitumor effect, it should be noted that the PKN3 mRNA reduction was more pronounced in liver and lung compared with tumor (35). Stable nucleic acid lipid particle (SNALP) formulations such as ALN-VSP02 (first-generation SNALP containing siVEGF and siKSP) showed moderate gene knockdown (36), and some liver and spleen toxicities were noted. The next-generation SNALP, TKM-080301, was formulated with more stable PEG-lipids in the nanoparticles. The clinical trial with siPLK1 showed a better immune profile along with increased drug exposure compared with the earlier generation of SNALPs (37). Neutral nanoliposomes (e.g., DOPC) have shown improved delivery (approximately 10-fold) of siRNA and antitumor effects with systemic delivery (38). Moreover, in a hepatocarcinoma mouse model, neutral liposome containing doxorubicin showed a better biodistribution profile and antitumor efficacy compared with its cationic counterparts (39).

Once inside the tumor cells, it is important to overcome barriers such as endosomal uptake (40). Systems such as the polymer-based dynamic polyconjugate (DPC) delivery system, which contains endosomolytic N-acetylgalactosamine–conjugated melittin-like peptide, may allow specific endosomal release of siRNA from its nanocarrier, thus lowering siRNA the half-maximal effective concentration (37, 41).

One of the unmet needs in the field of immunotherapy is the lack of efficient delivery systems for cytokines and antigens. Success of systemic administration of cytokines has been limited because of their early degradation, nonspecific binding to proteins, quick excretion, and undesired toxic effects. Gold nanoparticles conjugated with TNFα are currently being investigated in clinical trials (42). Early results suggest that such a formulation may be safer with higher MTD than free recombinant TNFα (43). In addition, studies with IL2 and IL12 have also shown that nanoparticle incorporation increased plasma retention time (44, 45).

Successful antigen vaccination can be achieved by ensuring (i) sufficient concentration of antigen in antigen-presenting cells (APC); (ii) sustained release of antigen for prolonged exposure to APCs; and (iii) cytoplasmic delivery of antigen for MHC class I processing. Incorporation of an antigen into target-specific nanoparticles can increase the concentration of antigens in dendritic cells (DC; refs. 46–48). Nanoparticles can also incorporate adjuvants and antigens in the same vehicle (46, 47, 49). For example, conjugation of polyribocytidylic acid (adjuvant) with DOTAP containing a tumor lysate (antigen) not only increased Toll-like receptor signaling, but also led to increased DC maturation and enhanced anti-immune response (47). Because DOTAP can lead to reactive oxygen species (ROS) production and apoptosis of DC cells (50), safer and more effective systems are needed.

Poly-lactic-co-glycolic acid nanoparticles have been shown to act as intracellular antigen reservoirs for DCs, minimizing degradation of antigens and the need of repeated dosing (51). Such sustained antigen presentation leads to stronger immune responses and consequently better antitumor effects. However, precise knowledge of degradation and release kinetics is essential to formulate effective sustained release of nanoparticles. Once inside the cells, it is important that the antigens are released in the cytosol for preferential processing through the MHC class I pathway to prime CD8+ T cells. Use of poly-(gamma–glutamic acid), conjugation of pH responsive peptide bonds, or incorporation of cell penetrating peptide R8 are some of the strategies to achieve enhanced cytoplasmic release of antigens (52–54). Immunogenicity of certain nanomaterials is a potential concern (e.g., PEG coated nanoparticles can activate the complement system and lead to PEG-specific IgM antibodies; refs. 55, 56); strategies to overcome these unexpected side effects are needed.

Photothermal ablation for tumoricidal effect

Photothermal ablation involves exposure of tissues to high temperature for membrane lysis and subsequent cell death (57). Increased susceptibility of cancer cells to hyperthermia is due to their higher metabolic rates than normal cells (58); however, this selectivity is minimal. The main concern with photothermal therapy (PTT) is the nonspecific effect on surrounding normal tissues. Localized heating, enabled with the use of nanoparticles, can avoid toxicity to normal cells. Blood and tissues are relatively transmissive in near-infrared (NIR) range. NIR has thus been effective for PTT as it achieves optimal tissue penetration to reach deeply localized tumor tissues. Initial preclinical studies were conducted using FDA-approved NIR-free dyes (e.g., indocyanine green). Although this strategy showed an antitumor effect, the strategy mainly suffered because of the low circulation time of indocyanine green (ICG) (3 minutes) and damage to normal tissues (59). Incorporation of these dyes into polymeric nanoparticles improved solubility and stability and increased photothermal ablation while keeping toxicity at a minimum (60–62).

Gold nanoparticles are widely used for PTT (57, 63, 64). Nanoshells with silica core coated with a thin gold layer have been investigated extensively in preclinical studies and are currently in clinical trials for head and neck and metastatic lung cancer (65, 66). The temperatures achieved by these nanoparticles (ΔT = 37.4 ± 6.6°C) were significantly higher than those achieved by laser treatment alone (ΔT < 10°C; ref. 65). In the treatment arm, tumor growth was significantly lower and survival was significantly higher. Further studies demonstrated that malignant cells required less than half of the laser density (∼20 W/cm2) for ablation compared with normal cells (57 W/cm2) when incubated with EGFR conjugated gold nanoshells (67, 68). Smaller hollow gold nanospheres have also been developed for simultaneous laser triggered drug delivery of doxorubicin, with significantly better antitumor effects compared with PTT alone (69), and have a favorable safety profile (70). Copper sulfide (CuS) nanoparticles are also being investigated for PTT. PEG-CuS nanoparticles plus laser treatment resulted in significantly higher tumor tissue necrosis (∼65%) compared with saline plus laser treatment (∼5%; ref. 71). Several attractive features [smaller size (<15 nm), better renal clearance, ease of synthesis, and low cost] make them promising candidates for PTT (71, 72).

Clinical applicability of a nanoparticle can be further improved if it can also be used as an imaging agent for MRI and spatiotemporal monitoring of the nanoparticles (73). Similar to previously discussed nanoparticles, gold and iron oxide nanoparticles also have certain toxicity issues (74–77) that will require additional work.

Tumor imaging

The limitations of current imaging modalities, such as iodine and gadolinium based CT, X-ray, and MRI scans, are lack of sensitivity in detecting small tumor nodules, lack of specificity, short imaging time, and toxic effects. Novel nanoparticle platforms may help to overcome these limitations. For example, ferumoxtran-10, an iron oxide nanoparticle, showed significantly higher sensitivity (90.5% vs. 35.4%) in detecting lymph node metastasis as compared with conventional MRI scans (78, 79). Iron oxide nanoparticles, when compared with gadolinium chelates, showed lower diffusion from tumor site, increased internalization by cancer cells, and enhanced detection of lesions in the brain (80). Polymeric dendrimers used as nanocarriers for gadolinium proved to be a better tool for detecting lymph nodes compared with free gadolinium chelates; such sensitivity was achieved at 1/2,500th of the molar concentration of the clinical gadolinium dose (81). Owing to the high atomic number and electron density, gold nanoparticles have a higher absorption coefficient than conventional iodine and are better contrast agents for PET scans and X-rays (82). Ability to coat them with PEG and functionalize the surface with targeting ligands makes it possible to increase circulation time and achieve high tumor cell specificity (83, 84). Nanoparticle formulations have also been used to reduce the toxicity of conventional contrast agents. Gadolinium-containing agents can cause nephrogenic systemic fibrosis, which can be prevented with nanoparticle incorporation (85, 86). Certain nanoparticles, such as self-assembled nanocages, facilitate interaction between water molecules and Gd, allowing a higher magnetic resonance signal at a much lower gadolinium concentration (87). This was also the case with free NIR dyes, where nonspecific accumulation of dyes in lungs and testicles had raised concerns, but nanoparticle formulations improved biodistribution and significantly reduced toxicity (88). Carbon nanotubes are being investigated for X-ray imaging, but may be limited by potential carcinogenic effects (89).

Oncolytic viruses are natural or genetically modified viruses that specifically kill cancer cells either by intensive cytopathic effect or inducing a strong immune response in the tumor microenvironment. JX-594 poxvirus, genetically modified to express GM-CSF, has been shown to increase antitumor immune response. The intratumoral injections had limited side effects and resulted in partial remission or stable disease in patients with liver cancer (90). Talimogene laherparepvec (T-vec) is a Herpes simplex virus expressing GM-CSF that has shown promising results in patients with advanced melanoma. ONYX-015, an adenovirus specifically targeting tumors with inactivated p53 has also shown promising results in phase I and II clinical trials (91). A similar virus (H101) has already gained approval for the treatment of head and neck cancer in China. Systemic administration can lead to the production of neutralizing antibodies, which may require immunosuppressive treatments prior to viral therapy (92). Combination of oncolytic viral therapy with chemotherapy or radiation may further enhance its activity (91).

Nanotechnology has transformed the field of medicine by crafting promising avenues in therapeutics and diagnosis (Fig. 2), but there is clearly room for further improvement. Considering the heterogeneity of tumor, extent of hypoxia or expression of specific enzymes required for drug release may not be the same at all metastatic sites, potentially making drug release unpredictable. A possible solution to increase tumor specificity is to use dual stimuli–responsive triggers (93–96), but particular attention must be given to characterizing these systems further and improving the scalability of the formulations. Regarding the multidrug-carrying nanoparticles, optimized ratiometric loading and compatibility of their efficacy and toxicity profiles are important aspects to be considered. For theranostic nanoparticles, care must be taken to avoid compromising imaging quality or therapeutic efficacy. Pharmacokinetic (PK) and pharamacodynamic (PD) requirements are also different for imaging and drug delivery vehicles. For example, long circulation times that are ideal for effective drug delivery may not be suitable for imaging purposes and will give high background signal (97, 98). Collectively, several factors must be considered to improve the translation of nanomedicines from bench to bedside. Here, we discuss key issues and ways to accelerate the development of clinically feasible nanosystems.

Figure 2.

Nanotechnology offers a wide array of applications for drug delivery, nucleotide delivery, photothermal therapy, immunotherapy, and imaging. Shown are some of the commonly used nanoformulations for each of the applications: ligand conjugated, target-specific PEGylated liposomes for small-molecule drug delivery; PEGylated SNALPs for nucleotide (e.g., siRNA) delivery; gold nanoshells for photothermal therapy; ligand conjugated, target-specific antigen carrying polymeric nanoparticles for immunotherapy and super paramagnetic iron oxide nanoparticles (SPIO) for imaging. DCs are immune cells that process and present antigens to T cells.

Figure 2.

Nanotechnology offers a wide array of applications for drug delivery, nucleotide delivery, photothermal therapy, immunotherapy, and imaging. Shown are some of the commonly used nanoformulations for each of the applications: ligand conjugated, target-specific PEGylated liposomes for small-molecule drug delivery; PEGylated SNALPs for nucleotide (e.g., siRNA) delivery; gold nanoshells for photothermal therapy; ligand conjugated, target-specific antigen carrying polymeric nanoparticles for immunotherapy and super paramagnetic iron oxide nanoparticles (SPIO) for imaging. DCs are immune cells that process and present antigens to T cells.

Close modal

Use of relevant preclinical models to predict EPR effect

Most nanoparticles are thought to rely on the EPR effect to accumulate in tumors. Reliable methods for assessing delivery are needed. A recent study with near-infrared fluorescence–labeled polymeric nanoparticles showed that tumors with high vascularity accumulated a greater density of nanoparticles (99). Although the study used fairly small nanoparticles (10 nm), the concept of predicting EPR effects by simple ultrasound imaging of vasculature should be explored further in relevant preclinical and clinical models.

Discrepancies between preclinical studies and clinical trials

It is now recognized that in vitro models may not reliably predict the utility of nanoparticles. For example, a dual targeted (Tf and mAb 2C5) nanoparticle system failed to reproduce the in vitro effectiveness when tested in mouse models (100). Whether 3D biomimetic tumor models can help bridge this gap to some extent is not fully understood (101). The current 3D systems could potentially be improved by incorporating relevant stromal cells, extracellular matrix proteins, or even relevant mechanical forces. These factors will help to closely simulate the tumor microenvironment and will make 3D systems a reliable platform for designing subsequent preclinical studies.

Testing the biodistribution and efficacy of nanoparticles in relevant animal models is crucial to move the therapy into the clinic. Ideally, animal models that are reflective of human disease should be used for such studies. Subcutaneous models are likely to be the least reliable due to aberrant stromal and vascular biology compared with the orthotopic sites (102). Design of preclinical trials is crucial for predicting the efficacy and safety of the nanoparticles. Moreover, it is also important to assess immunologic parameters (e.g., changes in cytokine levels or number of immune cells) during preclinical testing. Such comprehensive analysis will help in predicting the efficacy and toxicity profile of an individual nanotherapy in patients. In addition, phase 0 studies should be used to improve clinical translatability of nanoparticles. PK profile and tumor localization potential of a given nanocarrier in humans can be assessed in a timely manner in phase 0 trials. They are much less expensive to conduct compared with phase I trials and researchers can also obtain feedback on the clinical feasibility of a given nanosystem much more quickly.

Selection of a clinically relevant route of administration

Nanodrugs should ideally be administered the same way in the preclinical models as it is expected to be delivered in patients. For instance, preclinical studies with oncolytic viral therapy typically use intratumoral injections (103). Many other studies using nonviral nanocarriers also try to prove better efficacy using intratumoral injections (104, 105). However, this would have limited utility in patients with widely metastatic disease. Unlike intratumoral injections; i.v. injections can expose the particles to various biologic barriers, and thus will not be as effective as intartumoral route.

Choosing the right ligand for targeted delivery

Ligand-targeted therapies have been shown to be superior in terms of tumor specificity and low off-target effects. (106–108). In preclinical studies, greater accumulation of nanoparticles can be achieved by choosing a tumor model overexpressing the specific receptor (105). However, there is heterogeneity in receptor expression in tumors (109, 110), and a single ligand could ultimately lead to selection of cells that lack target expression. A multiligand approach has been shown to be more specific and leads to better uptake of nanoparticles (111–113). With increased specificity, multiligand nanoparticles are less likely to be taken up by normal cells, and thus have fewer toxicity issues (112, 114).

In summary, the versatility of formulations, targeted delivery, and biocompatibility have garnered a lot of interest in nanotechnology. However, we must first address several practical issues. Every new nanomaterial and added complexities require additional controls and toxicity checks, making FDA approval potentially more difficult. Batch-to-batch variations in these cases further complicate scaling up the production. Thus, the clinical benefit and toxicity profile must be far superior compared with the conventional drugs to justify the cost. Moreover, it is important to understand the intricacies of nanotechnology in vivo and predict the behavior, distribution, and kinetics with certainty. Then, we can develop strategies for scaling up production and distribution with the ultimate goal of direct clinical translation and patient benefit.

K.M. Gharpure is supported by the Altman Goldstein Discovery Fellowship. S.Y. Wu is supported by the Ovarian Cancer Research Fund, Foundation for Women's Cancer, and Cancer Prevention Research Institute of Texas (CPRIT) training grants (RP101502, RP101489, and RP110595). C. Li and A.K. Sood are supported by the NIH under award number U54CA151668. A.K. Sood is supported by the NIH under award numbers P50CA083639, CA109298, P50CA098258, UH2TR000943, CA016672, U54CA96300, and U54CA96297; CPRIT grants (RP110595 and RP120214); an Ovarian Cancer Research Fund Program Project Development Grant; U.S. Department of Defense grants (OC120547 and OC093416); the Betty Ann Asche Murray Distinguished Professorship; the RGK Foundation; the Gilder Foundation; the Judi A. Rees Ovarian Cancer Research Fund; the Chapman Foundation; the Meyer and Ida Gordon Foundation; and the Blanton-Davis Ovarian Cancer Research Program.

1.
Parveen
S
,
Misra
R
,
Sahoo
SK
. 
Nanoparticles: a boon to drug delivery, therapeutics, diagnostics, and imaging
.
Nanomedicine
2012
;
8
:
147
66
.
2.
Swenson
CE
,
Bolcsak
LE
,
Batist
G
,
Guthrie
TH
 Jr
,
Tkaczuk
KH
,
Boxenbaum
H
, et al
Pharmacokinetics of doxorubicin administered i.v. as Myocet (TLC D-99; liposome-encapsulated doxorubicin citrate) compared with conventional doxorubicin when given in combination with cyclophosphamide in patients with metastatic breast cancer
.
Anticancer Drugs
2003
;
14
:
239
46
.
3.
Gill
PS
,
Espina
BM
,
Muggia
F
,
Cabriales
S
,
Tulpule
A
,
Esplin
JA
, et al
Phase I/II clinical and pharmacokinetic evaluation of liposomal daunorubicin
.
J Clin Oncol
1995
;
13
:
996
1003
.
4.
Batist
G
,
Ramakrishnan
G
,
Rao
CS
,
Chandrasekharan
A
,
Gutheil
J
,
Guthrie
T
, et al
Reduced cardiotoxicity and preserved antitumor efficacy of liposome-encapsulated doxorubicin and cyclophosphamide compared with conventional doxorubicin and cyclophosphamide in a randomized, multicenter trial of metastatic breast cancer
.
J Clin Oncol
2001
;
19
:
1444
54
.
5.
Gabizon
A
,
Catane
R
,
Uziely
B
,
Kaufman
B
,
Safra
T
,
Cohen
R
, et al
Prolonged circulation time and enhanced accumulation in malignant exudates of doxorubicin encapsulated in polyethylene-glycol coated liposomes
.
Cancer Res
1994
;
54
:
987
92
.
6.
O'Brien
ME
,
Wigler
N
,
Inbar
M
,
Rosso
R
,
Grischke
E
,
Santoro
A
, et al
Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer
.
Ann Oncol
2004
;
15
:
440
9
.
7.
Gabizon
A
,
Martin
F
. 
Polyethylene glycol-coated (pegylated) liposomal doxorubicin. Rationale for use in solid tumours
.
Drugs
1997
;
4
:
15
21
.
8.
Miele
E
,
Spinelli
GP
,
Miele
E
,
Tomao
F
,
Tomao
S
. 
Albumin-bound formulation of paclitaxel (Abraxane(®) ABI-007) in the treatment of breast cancer
.
Int J Nanomedicine
2009
;
4
:
99
105
.
9.
Ibrahim
NK
,
Desai
N
,
Legha
S
,
Soon-Shiong
P
,
Theriault
RL
,
Rivera
E
, et al
Phase I and pharmacokinetic study of ABI-007, a Cremophor-free, protein-stabilized, nanoparticle formulation of paclitaxel
.
Clin Cancer Res
2002
;
8
:
1038
44
.
10.
Gradishar
WJ
,
Tjulandin
S
,
Davidson
N
,
Shaw
H
,
Desai
N
,
Bhar
P
, et al
Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer
.
J Clin Oncol
2005
;
23
:
7794
803
.
11.
Li
C
,
Wallace
S
. 
Polymer-drug conjugates: recent development in clinical oncology
.
Adv Drug Deliv Rev
2008
;
60
:
886
98
.
12.
Li
C
,
Yu
DF
,
Newman
RA
,
Cabral
F
,
Stephens
LC
,
Hunter
N
, et al
Complete regression of well-established tumors using a novel water-soluble poly(L-glutamic acid)-paclitaxel conjugate
.
Cancer Res
1998
;
58
:
2404
9
.
13.
Hrkach
J
,
Von Hoff
D
,
Mukkaram Ali
M
,
Andrianova
E
,
Auer
J
,
Campbell
T
, et al
Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile
.
Sci Transl Med
2012
;
4
:
128ra39
.
14.
Ireson
CR
,
Kelland
LR
. 
Discovery and development of anticancer aptamers
.
Mol Cancer Ther
2006
;
5
:
2957
62
.
15.
Park
JW
,
Hong
K
,
Kirpotin
DB
,
Colbern
G
,
Shalaby
R
,
Baselga
J
, et al
Anti-HER2 immunoliposomes: enhanced efficacy attributable to targeted delivery
.
Clin Cancer Res
2002
;
8
:
1172
81
.
16.
Werner
ME
,
Karve
S
,
Sukumar
R
,
Cummings
ND
,
Copp
JA
,
Chen
RC
, et al
Folate-targeted nanoparticle delivery of chemo- and radiotherapeutics for the treatment of ovarian cancer peritoneal metastasis
.
Biomaterials
2011
;
32
:
8548
54
.
17.
Zheng
Y
,
Yu
B
,
Weecharangsan
W
,
Piao
L
,
Darby
M
,
Mao
Y
, et al
Transferrin-conjugated lipid-coated PLGA nanoparticles for targeted delivery of aromatase inhibitor 7alpha-APTADD to breast cancer cells
.
Int J Pharm
2010
;
390
:
234
41
.
18.
Hamaguchi
T
,
Matsumura
Y
,
Nakanishi
Y
,
Muro
K
,
Yamada
Y
,
Shimada
Y
, et al
Antitumor effect of MCC-465, pegylated liposomal doxorubicin tagged with newly developed monoclonal antibody GAH, in colorectal cancer xenografts
.
Cancer Sci
2004
;
95
:
608
13
.
19.
Reynolds
JG
,
Geretti
E
,
Hendriks
BS
,
Lee
H
,
Leonard
SC
,
Klinz
SG
, et al
HER2-targeted liposomal doxorubicin displays enhanced anti-tumorigenic effects without associated cardiotoxicity
.
Toxicol Appl Pharmacol
2012
;
262
:
1
10
.
20.
Mamot
C
,
Ritschard
R
,
Wicki
A
,
Stehle
G
,
Dieterle
T
,
Bubendorf
L
, et al
Tolerability, safety, pharmacokinetics, and efficacy of doxorubicin-loaded anti-EGFR immunoliposomes in advanced solid tumours: a phase 1 dose-escalation study
.
Lancet Oncol
2012
;
13
:
1234
41
.
21.
Senzer
N
,
Nemunaitis
J
,
Nemunaitis
D
,
Bedell
C
,
Edelman
G
,
Barve
M
, et al
Phase I study of a systemically delivered p53 nanoparticle in advanced solid tumors
.
Mol Ther
2013
;
21
:
1096
103
.
22.
Jin
C
,
Yang
Z
,
Yang
J
,
Li
H
,
He
Y
,
An
J
, et al
Paclitaxel-loaded nanoparticles decorated with anti-CD133 antibody: a targeted therapy for liver cancer stem cells
.
J Nanopart Res
2013
;
16
:
1
15
.
23.
Li
SY
,
Sun
R
,
Wang
HX
,
Shen
S
,
Liu
Y
,
Du
XJ
, et al
Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells
.
J Control Release
2014
;
15
:
00748
2
.
24.
Han
HD
,
Mangala
LS
,
Lee
JW
,
Shahzad
MM
,
Kim
HS
,
Shen
D
, et al
Targeted gene silencing using RGD-labeled chitosan nanoparticles
.
Clin Cancer Res
2010
;
16
:
3910
22
.
25.
Amstad
E
,
Kohlbrecher
J
,
Muller
E
,
Schweizer
T
,
Textor
M
,
Reimhult
E
. 
Triggered release from liposomes through magnetic actuation of iron oxide nanoparticle containing membranes
.
Nano Lett
2011
;
11
:
1664
70
.
26.
Derfus
AM
,
von Maltzahn
G
,
Harris
TJ
,
Duza
T
,
Vecchio
KS
,
Ruoslahti
E
, et al
Remotely triggered release from magnetic nanoparticles
.
Adv Mater
2007
;
19
:
3932
6
.
27.
Dromi
S
,
Frenkel
V
,
Luk
A
,
Traughber
B
,
Angstadt
M
,
Bur
M
, et al
Pulsed-high intensity focused ultrasound and low temperature-sensitive liposomes for enhanced targeted drug delivery and antitumor effect
.
Clin Cancer Res
2007
;
13
:
2722
7
.
28.
Fomina
N
,
McFearin
C
,
Sermsakdi
M
,
Edigin
O
,
Almutairi
A
. 
UV and near-IR triggered release from polymeric nanoparticles
.
J Am Chem Soc
2010
;
132
:
9540
2
.
29.
Giri
S
,
Trewyn
BG
,
Stellmaker
MP
,
Lin
VSY
. 
Stimuli-responsive controlled-release delivery system based on mesoporous silica nanorods capped with magnetic nanoparticles
.
Angew Chem Int Ed
2005
;
44
:
5038
44
.
30.
Farooqi
AA
,
Rehman
Zu
,
Muntane
J
. 
Antisense therapeutics in oncology: current status
.
OncoTargets Ther
2014
;
7
:
2035
42
.
31.
Schreier
H
,
Gagne
L
,
Bock
T
,
Erdos
GW
,
Druzgala
P
,
Conary
JT
, et al
Physicochemical properties and in vitro toxicity of cationic liposome cDNA complexes
.
Pharm Acta Helv
1997
;
72
:
215
23
.
32.
Guo
S
,
Huang
L
. 
Nanoparticles escaping RES and endosome: challenges for siRNA delivery for cancer therapy
.
J Nanomaterials
2011
;
2011
:
12
.
33.
Filion
MC
,
Phillips
NC
. 
Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells
.
Biochim Biophys Acta
1997
;
1329
:
345
56
.
34.
Schultheis
B
,
Strumberg
D
,
Santel
A
,
Vank
C
,
Gebhardt
F
,
Keil
O
, et al
First-in-human phase I study of the liposomal RNA interference therapeutic Atu027 in patients with advanced solid tumors
.
J Clin Oncol
2014
;
32
:
4141
8
.
35.
Aleku
M
,
Schulz
P
,
Keil
O
,
Santel
A
,
Schaeper
U
,
Dieckhoff
B
, et al
Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression
.
Cancer Res
2008
;
68
:
9788
98
.
36.
Tabernero
J
,
Shapiro
GI
,
LoRusso
PM
,
Cervantes
A
,
Schwartz
GK
,
Weiss
GJ
, et al
First-in-man trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement
.
Cancer Discov
2013
;
3
:
406
17
.
37.
Wu
SY
,
Lopez-Berestein
G
,
Calin
GA
,
Sood
AK
. 
RNAi therapies: drugging the undruggable
.
Sci Transl Med
2014
;
6
:
240ps7
.
38.
Landen
CN
 Jr
,
Chavez-Reyes
A
,
Bucana
C
,
Schmandt
R
,
Deavers
MT
,
Lopez-Berestein
G
, et al
Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery
.
Cancer Res
2005
;
65
:
6910
8
.
39.
Zhao
W
,
Zhuang
S
,
Qi
XR
. 
Comparative study of the in vitro and in vivo characteristics of cationic and neutral liposomes
.
Int J Nanomedicine
2011
;
6
:
3087
98
.
40.
Gilleron
J
,
Querbes
W
,
Zeigerer
A
,
Borodovsky
A
,
Marsico
G
,
Schubert
U
, et al
Image-based analysis of lipid nanoparticle-mediated siRNA delivery, intracellular trafficking and endosomal escape
.
Nat Biotech
2013
;
31
:
638
46
.
41.
Wong
SC
,
Klein
JJ
,
Hamilton
HL
,
Chu
Q
,
Frey
CL
,
Trubetskoy
VS
, et al
Co-injection of a targeted, reversibly masked endosomolytic polymer dramatically improves the efficacy of cholesterol-conjugated small interfering RNAs in vivo
.
Nucleic Acid Ther
2012
;
22
:
380
90
.
42.
Ghosh
P
,
Han
G
,
De
M
,
Kim
CK
,
Rotello
VM
. 
Gold nanoparticles in delivery applications
.
Adv Drug Deliv Rev
2008
;
60
:
1307
15
.
43.
Libutti
SK
,
Paciotti
GF
,
Byrnes
AA
,
Alexander
HR
,
Gannon
WE
,
Walker
M
, et al
Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine
.
Clin Cancer Res
2010
;
16
:
6139
49
.
44.
Shimizu
T
,
Kishida
T
,
Hasegawa
U
,
Ueda
Y
,
Imanishi
J
,
Yamagishi
H
, et al
Nanogel DDS enables sustained release of IL-12 for tumor immunotherapy
.
Biochem Biophys Res Commun
2008
;
367
:
330
5
.
45.
Yao
H
,
Ng
SS
,
Huo
LF
,
Chow
BK
,
Shen
Z
,
Yang
M
, et al
Effective melanoma immunotherapy with interleukin-2 delivered by a novel polymeric nanoparticle
.
Mol Cancer Ther
2011
;
10
:
1082
92
.
46.
Mansour
M
,
Pohajdak
B
,
Kast
WM
,
Fuentes-Ortega
A
,
Korets-Smith
E
,
Weir
GM
, et al
Therapy of established B16-F10 melanoma tumors by a single vaccination of CTL/T helper peptides in VacciMax
.
J Transl Med
2007
;
5
:
20
.
47.
Wang
C
,
Zhuang
Y
,
Zhang
Y
,
Luo
Z
,
Gao
N
,
Li
P
, et al
Toll-like receptor 3 agonist complexed with cationic liposome augments vaccine-elicited antitumor immunity by enhancing TLR3-IRF3 signaling and type I interferons in dendritic cells
.
Vaccine
2012
;
30
:
4790
9
.
48.
Zhang
Z
,
Tongchusak
S
,
Mizukami
Y
,
Kang
YJ
,
Ioji
T
,
Touma
M
, et al
Induction of anti-tumor cytotoxic T cell responses through PLGA-nanoparticle mediated antigen delivery
.
Biomaterials
2011
;
32
:
3666
78
.
49.
Diwan
M
,
Tafaghodi
M
,
Samuel
J
. 
Enhancement of immune responses by co-delivery of a CpG oligodeoxynucleotide and tetanus toxoid in biodegradable nanospheres
.
J Control Release
2002
;
85
:
247
62
.
50.
Yan
W
,
Chen
W
,
Huang
L
. 
Reactive oxygen species play a central role in the activity of cationic liposome based cancer vaccine
.
J Control Release
2008
;
130
:
22
8
.
51.
Shen
H
,
Ackerman
AL
,
Cody
V
,
Giodini
A
,
Hinson
ER
,
Cresswell
P
, et al
Enhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles
.
Immunology
2006
;
117
:
78
88
.
52.
Mukai
Y
,
Yoshinaga
T
,
Yoshikawa
M
,
Matsuo
K
,
Yoshikawa
T
,
Niki
K
, et al
Induction of endoplasmic reticulum-endosome fusion for antigen cross-presentation induced by poly (gamma-glutamic acid) nanoparticles
.
J Immunol
2011
;
187
:
6249
55
.
53.
Nakamura
T
,
Moriguchi
R
,
Kogure
K
,
Shastri
N
,
Harashima
H
. 
Efficient MHC class I presentation by controlled intracellular trafficking of antigens in octaarginine-modified liposomes
.
Mol Ther
2008
;
16
:
1507
14
.
54.
Yuba
E
,
Kojima
C
,
Harada
A
,
Tana
,
Watarai
S
,
Kono
K
. 
pH-Sensitive fusogenic polymer-modified liposomes as a carrier of antigenic proteins for activation of cellular immunity
.
Biomaterials
2010
;
31
:
943
51
.
55.
Ishida
T
,
Kiwada
H
. 
Accelerated blood clearance (ABC) phenomenon upon repeated injection of PEGylated liposomes
.
Int J Pharm
2008
;
354
:
56
62
.
56.
Pham
CT
,
Mitchell
LM
,
Huang
JL
,
Lubniewski
CM
,
Schall
OF
,
Killgore
JK
, et al
Variable antibody-dependent activation of complement by functionalized phospholipid nanoparticle surfaces
.
J Biol Chem
2011
;
286
:
123
30
.
57.
Jing-Liang
L
,
Gu
M
. 
Gold-nanoparticle-enhanced cancer photothermal therapy
.
IEEE J Sel Top Quantum Electron
2010
;
16
:
989
96
.
58.
Huff
TB
,
Tong
L
,
Zhao
Y
,
Hansen
MN
,
Cheng
JX
,
Wei
A
. 
Hyperthermic effects of gold nanorods on tumor cells
.
Nanomedicine
2007
;
2
:
125
32
.
59.
Yuan
A
,
Wu
J
,
Tang
X
,
Zhao
L
,
Xu
F
,
Hu
Y
. 
Application of near-infrared dyes for tumor imaging, photothermal, and photodynamic therapies
.
J Pharm Sci
2013
;
102
:
6
28
.
60.
Peng
CL
,
Shih
YH
,
Lee
PC
,
Hsieh
TM
,
Luo
TY
,
Shieh
MJ
. 
Multimodal image-guided photothermal therapy mediated by 188Re-labeled micelles containing a cyanine-type photosensitizer
.
ACS Nano
2011
;
5
:
5594
607
.
61.
Yu
J
,
Javier
D
,
Yaseen
MA
,
Nitin
N
,
Richards-Kortum
R
,
Anvari
B
, et al
Self-assembly synthesis, tumor cell targeting, and photothermal capabilities of antibody-coated indocyanine green nanocapsules
.
J Am Chem Soc
2010
;
132
:
1929
38
.
62.
Zheng
X
,
Xing
D
,
Zhou
F
,
Wu
B
,
Chen
WR
. 
Indocyanine green-containing nanostructure as near infrared dual-functional targeting probes for optical imaging and photothermal therapy
.
Mol Pharm
2011
;
8
:
447
56
.
63.
Huang
X
,
El-Sayed
MA
. 
Gold nanoparticles: optical properties and implementations in cancer diagnosis and photothermal therapy
.
J Adv Res
2010
;
1
:
13
28
.
64.
Shao
J
,
Griffin
RJ
,
Galanzha
EI
,
Kim
J-W
,
Koonce
N
,
Webber
J
, et al
Photothermal nanodrugs: potential of TNF-gold nanospheres for cancer theranostics
.
Sci Rep
2013
;
3
:
1293
.
65.
Hirsch
LR
,
Stafford
RJ
,
Bankson
JA
,
Sershen
SR
,
Rivera
B
,
Price
RE
, et al
Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance
.
Proc Natl Acad Sci U S A
2003
;
100
:
13549
54
.
66.
O'Neal
DP
,
Hirsch
LR
,
Halas
NJ
,
Payne
JD
,
West
JL
. 
Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles
.
Cancer Lett
2004
;
209
:
171
6
.
67.
El-Sayed
IH
,
Huang
X
,
El-Sayed
MA
. 
Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles
.
Cancer Lett
2006
;
239
:
129
35
.
68.
Huang
X
,
El-Sayed
IH
,
Qian
W
,
El-Sayed
MA
. 
Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods
.
J Am Chem Soc
2006
;
128
:
2115
20
.
69.
You
J
,
Zhang
R
,
Zhang
G
,
Zhong
M
,
Liu
Y
,
Van Pelt
CS
, et al
Photothermal-chemotherapy with doxorubicin-loaded hollow gold nanospheres: a platform for near-infrared light-trigged drug release
.
J Control Release
2012
;
158
:
319
28
.
70.
You
J
,
Zhou
J
,
Zhou
M
,
Liu
Y
,
Robertson
J
,
Liang
D
, et al
Pharmacokinetics, clearance, and biosafety of polyethylene glycol-coated hollow gold nanospheres
.
Part Fibre Toxicol
2014
;
11
:
26
.
71.
Zhou
M
,
Zhang
R
,
Huang
M
,
Lu
W
,
Song
S
,
Melancon
MP
, et al
A chelator-free multifunctional [64Cu]CuS nanoparticle platform for simultaneous micro-PET/CT imaging and photothermal ablation therapy
.
J Am Chem Soc
2010
;
132
:
15351
8
.
72.
Li
Y
,
Lu
W
,
Huang
Q
,
Huang
M
,
Li
C
,
Chen
W
. 
Copper sulfide nanoparticles for photothermal ablation of tumor cells
.
Nanomedicine
2010
;
5
:
1161
71
.
73.
Melancon
MP
,
Elliott
A
,
Ji
X
,
Shetty
A
,
Yang
Z
,
Tian
M
, et al
Theranostics with multifunctional magnetic gold nanoshells: photothermal therapy and t2* magnetic resonance imaging
.
Invest Radiol
2011
;
46
:
132
40
.
74.
Alkilany
AM
,
Murphy
CJ
. 
Toxicity and cellular uptake of gold nanoparticles: what we have learned so far?
J Nanopart Res
2010
;
12
:
2313
33
.
75.
Murphy
CJ
,
Gole
AM
,
Stone
JW
,
Sisco
PN
,
Alkilany
AM
,
Goldsmith
EC
, et al
Gold nanoparticles in biology: beyond toxicity to cellular imaging
.
Acc Chem Res
2008
;
41
:
1721
30
.
76.
Gupta
AK
,
Gupta
M
. 
Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications
.
Biomaterials
2005
;
26
:
3995
4021
.
77.
Pisanic Ii
TR
,
Blackwell
JD
,
Shubayev
VI
,
Fiñones
RR
,
Jin
S
. 
Nanotoxicity of iron oxide nanoparticle internalization in growing neurons
.
Biomaterials
2007
;
28
:
2572
81
.
78.
Harisinghani
MG
,
Barentsz
J
,
Hahn
PF
,
Deserno
WM
,
Tabatabaei
S
,
van de Kaa
CH
, et al
Noninvasive detection of clinically occult lymph-node metastases in prostate cancer
.
N Engl J Med
2003
;
348
:
2491
9
.
79.
Ross
RW
,
Zietman
AL
,
Xie
W
,
Coen
JJ
,
Dahl
DM
,
Shipley
WU
, et al
Lymphotropic nanoparticle-enhanced magnetic resonance imaging (LNMRI) identifies occult lymph node metastases in prostate cancer patients prior to salvage radiation therapy
.
Clin Imaging
2009
;
33
:
301
5
.
80.
Neuwelt
EA
,
Varallyay
P
,
Bago
AG
,
Muldoon
LL
,
Nesbit
G
,
Nixon
R
. 
Imaging of iron oxide nanoparticles by MR and light microscopy in patients with malignant brain tumours
.
Neuropathol Appl Neurobiol
2004
;
30
:
456
71
.
81.
Kobayashi
H
,
Kawamoto
S
,
Sakai
Y
,
Choyke
PL
,
Star
RA
,
Brechbiel
MW
, et al
Lymphatic drainage imaging of breast cancer in mice by micro-magnetic resonance lymphangiography using a nano-size paramagnetic contrast agent
.
J Natl Cancer Inst
2004
;
96
:
703
8
.
82.
Jain
S
,
Hirst
DG
,
O'Sullivan
JM
. 
Gold nanoparticles as novel agents for cancer therapy
.
Brit J Radiol
2012
;
85
:
101
13
.
83.
Popovtzer
R
,
Agrawal
A
,
Kotov
NA
,
Popovtzer
A
,
Balter
J
,
Carey
TE
, et al
Targeted gold nanoparticles enable molecular CT imaging of cancer
.
Nano Lett
2008
;
8
:
4593
6
.
84.
Wang
Z
. 
Plasmon—resonant gold nanoparticles for cancer optical imaging
.
Sci China Phys, Mech Astron
2013
;
56
:
506
13
.
85.
Liu
Y
,
Chen
Z
,
Liu
C
,
Yu
D
,
Lu
Z
,
Zhang
N
. 
Gadolinium-loaded polymeric nanoparticles modified with anti-VEGF as multifunctional MRI contrast agents for the diagnosis of liver cancer
.
Biomaterials
2011
;
32
:
5167
76
.
86.
Zhang
HW
,
Wang
LQ
,
Xiang
QF
,
Zhong
Q
,
Chen
LM
,
Xu
CX
, et al
Specific lipase-responsive polymer-coated gadolinium nanoparticles for MR imaging of early acute pancreatitis
.
Biomaterials
2014
;
35
:
356
67
.
87.
Manus
LM
,
Mastarone
DJ
,
Waters
EA
,
Zhang
X-Q
,
Schultz-Sikma
EA
,
MacRenaris
KW
, et al
Gd(III)-nanodiamond conjugates for MRI contrast enhancement
.
Nano Lett
2009
;
10
:
484
9
.
88.
Reul
R
,
Tsapis
N
,
Hillaireau
H
,
Sancey
L
,
Mura
S
,
Recher
M
, et al
Near infrared labeling of PLGA for in vivo imaging of nanoparticles
.
Polym Chem
2012
;
3
:
694
702
.
89.
Magrez
A
,
Kasas
S
,
Salicio
V
,
Pasquier
N
,
Seo
JW
,
Celio
M
, et al
Cellular toxicity of carbon-based nanomaterials
.
Nano Lett
2006
;
6
:
1121
5
.
90.
Heo
J
,
Reid
T
,
Ruo
L
,
Breitbach
CJ
,
Rose
S
,
Bloomston
M
, et al
Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer
.
Nat Med
2013
;
19
:
329
36
.
91.
Galanis
E
,
Okuno
SH
,
Nascimento
AG
,
Lewis
BD
,
Lee
RA
,
Oliveira
AM
, et al
Phase I-II trial of ONYX-015 in combination with MAP chemotherapy in patients with advanced sarcomas
.
Gene Ther
2005
;
12
:
437
45
.
92.
Lichty
BD
,
Breitbach
CJ
,
Stojdl
DF
,
Bell
JC
. 
Going viral with cancer immunotherapy
.
Nat Rev Cancer
2014
;
14
:
559
67
.
93.
Gao
W
,
Xiang
B
,
Meng
TT
,
Liu
F
,
Qi
XR
. 
Chemotherapeutic drug delivery to cancer cells using a combination of folate targeting and tumor microenvironment-sensitive polypeptides
.
Biomaterials
2013
;
34
:
4137
49
.
94.
Soppimath
KS
,
Liu
LH
,
Seow
WY
,
Liu
SQ
,
Powell
R
,
Chan
P
, et al
Multifunctional core/shell nanoparticles self-assembled from pH-induced thermosensitive polymers for targeted intracellular anticancer drug delivery
.
Adv Funct Mater
2007
;
17
:
355
62
.
95.
Xiao
D
,
Jia
H-Z
,
Zhang
J
,
Liu
C-W
,
Zhuo
R-X
,
Zhang
X-Z
. 
A dual-responsive mesoporous silica nanoparticle for tumor-triggered targeting drug delivery
.
Small
2014
;
10
:
591
8
.
96.
Huang
S
,
Shao
K
,
Liu
Y
,
Kuang
Y
,
Li
J
,
An
S
, et al
Tumor-targeting and microenvironment-responsive smart nanoparticles for combination therapy of antiangiogenesis and apoptosis
.
ACS Nano
2013
;
7
:
2860
71
.
97.
Cheng
Z
,
Al Zaki
A
,
Hui
JZ
,
Muzykantov
VR
,
Tsourkas
A
. 
Multifunctional nanoparticles: cost versus benefit of adding targeting and imaging capabilities
.
Science
2012
;
338
:
903
10
.
98.
Theek
B
,
Rizzo
LY
,
Ehling
J
,
Kiessling
F
,
Lammers
T
. 
The theranostic path to personalized nanomedicine
.
Clin Transl Imaging
2014
;
2
:
66
76
.
99.
Theek
B
,
Gremse
F
,
Kunjachan
S
,
Fokong
S
,
Pola
R
,
Pechar
M
, et al
Characterizing EPR-mediated passive drug targeting using contrast-enhanced functional ultrasound imaging
.
J Control Release
2014
;
182
:
83
9
.
100.
Sawant
RR
,
Jhaveri
AM
,
Koshkaryev
A
,
Qureshi
F
,
Torchilin
VP
. 
The effect of dual ligand-targeted micelles on the delivery and efficacy of poorly soluble drug for cancer therapy
.
J Drug Target
2013
;
21
:
630
8
.
101.
Goodman
TT
,
Ng
CP
,
Pun
SH
. 
3-D tissue culture systems for the evaluation and optimization of nanoparticle-based drug carriers
.
Bioconjug Chem
2008
;
19
:
1951
9
.
102.
Francia
G
,
Cruz-Munoz
W
,
Man
S
,
Xu
P
,
Kerbel
RS
. 
Mouse models of advanced spontaneous metastasis for experimental therapeutics
.
Nat Rev Cancer
2011
;
11
:
135
41
.
103.
Bartlett
D
,
Liu
Z
,
Sathaiah
M
,
Ravindranathan
R
,
Guo
Z
,
He
Y
, et al
Oncolytic viruses as therapeutic cancer vaccines
.
Mol Cancer
2013
;
12
:
103
.
104.
Al-Ghananeem
AM
,
Malkawi
AH
,
Muammer
YM
,
Balko
JM
,
Black
EP
,
Mourad
W
, et al
Intratumoral delivery of Paclitaxel in solid tumor from biodegradable hyaluronan nanoparticle formulations
.
AAPS PharmSciTech
2009
;
10
:
410
7
.
105.
Chattopadhyay
N
,
Fonge
H
,
Cai
Z
,
Scollard
D
,
Lechtman
E
,
Done
SJ
, et al
Role of antibody-mediated tumor targeting and route of administration in nanoparticle tumor accumulation in vivo
.
Mol Pharm
2012
;
9
:
2168
79
.
106.
Sahoo
SK
,
Ma
W
,
Labhasetwar
V
. 
Efficacy of transferrin-conjugated paclitaxel-loaded nanoparticles in a murine model of prostate cancer
.
Int J Cancer
2004
;
112
:
335
40
.
107.
Mitra
S
,
Gaur
U
,
Ghosh
PC
,
Maitra
AN
. 
Tumour targeted delivery of encapsulated dextran-doxorubicin conjugate using chitosan nanoparticles as carrier
.
J Control Release
2001
;
74
:
317
23
.
108.
Farokhzad
OC
,
Cheng
J
,
Teply
BA
,
Sherifi
I
,
Jon
S
,
Kantoff
PW
, et al
Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo
.
Proc Natl Acad Sci U S A
2006
;
103
:
6315
20
.
109.
Jabbour
M
,
Massad
C
,
Boulos
F
. 
Variability in hormone and growth factor receptor expression in primary versus recurrent, metastatic, and post-neoadjuvant breast carcinoma
.
Breast Cancer Res Treat
2012
;
135
:
29
37
.
110.
Gasch
C
,
Bauernhofer
T
,
Pichler
M
,
Langer-Freitag
S
,
Reeh
M
,
Seifert
AM
, et al
Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer
.
Clin Chem
2013
;
59
:
252
60
.
111.
Xu
Q
,
Liu
Y
,
Su
S
,
Li
W
,
Chen
C
,
Wu
Y
. 
Anti-tumor activity of paclitaxel through dual-targeting carrier of cyclic RGD and transferrin conjugated hyperbranched copolymer nanoparticles
.
Biomaterials
2012
;
33
:
1627
39
.
112.
Ko
HY
,
Choi
KJ
,
Lee
CH
,
Kim
S
. 
A multimodal nanoparticle-based cancer imaging probe simultaneously targeting nucleolin, integrin alphavbeta3 and tenascin-C proteins
.
Biomaterials
2011
;
32
:
1130
8
.
113.
Kluza
E
,
van der Schaft
DWJ
,
Hautvast
PAI
,
Mulder
WJM
,
Mayo
KH
,
Griffioen
AW
, et al
Synergistic targeting of αvβ3 integrin and galectin-1 with heteromultivalent paramagnetic liposomes for combined MR imaging and treatment of angiogenesis
.
Nano Lett
2010
;
10
:
52
8
.
114.
Saul
JM
,
Annapragada
AV
,
Bellamkonda
RV
. 
A dual-ligand approach for enhancing targeting selectivity of therapeutic nanocarriers
.
J Control Release
2006
;
114
:
277
87
.