Nanotechnology is emerging as a promising modality for cancer treatment; however, in the realm of cancer prevention, its full utility has yet to be determined. Here, we discuss the potential of integrating nanotechnology in cancer prevention to augment early diagnosis, precision targeting, and controlled release of chemopreventive agents, reduced toxicity, risk/response assessment, and personalized point-of-care monitoring. Cancer is a multistep, progressive disease; the functional and acquired characteristics of the early precancer phenotype are intrinsically different from those of a more advanced anaplastic or invasive malignancy. Therefore, applying nanotechnology to precancers is likely to be far more challenging than applying it to established disease. Frank cancers are more readily identifiable through imaging and biomarker and histopathologic assessment than their precancerous precursors. In addition, prevention subjects routinely have more rigorous intervention criteria than therapy subjects. Any nanopreventive agent developed to prevent sporadic cancers found in the general population must exhibit a very low risk of serious side effects. In contrast, a greater risk of side effects might be more acceptable in subjects at high risk for cancer. Using nanotechnology to prevent cancer is an aspirational goal, but clearly identifying the intermediate objectives and potential barriers is an essential first step in this exciting journey. Cancer Prev Res; 7(10); 973–92. ©2014 AACR.

Nanotechnology by definition involves the study and use of materials between 1 and 100 nanometers (nm) in size (Fig. 1). The idea of nanotechnology began as theoretical concepts posed by physicists and other scientists involving nanoscale assembly of materials (1, 2). The term nanotechnology was first used to describe semiconductor generation (3). The early discipline began with the invention of the scanning probe microscope and the discovery of molecular structures such as fullerenes (4, 5). The field has more recently expanded into various specialized areas of nanomaterials and nanomedicine (6–10).

Figure 1.

Understanding the relative nanotechnology scale. In terms of size, a nanometer (nm) equals one billionth of a meter (10−9 m). A proton in an elemental nucleus is 1 femtometer (fm). The nucleus of a Helium atom is 3.8 fm. The diameter of a glucose molecule is approaches 1 nm. The insulin protein approaches 5 nm in size. A nanoparticle 10 nm in size is 1,000-times smaller than the diameter of a human hair. A small microprocessor transistor is 22 nm. An adenovirus particle is between 60 and 90 nm. Liposome-based nanoparticles are often 100 nm in diameter. A typical bacterium is 1 μm in size. The average size of a human cell is 10 μm. As a reference, a billion inches is 15,783 miles, more than halfway around the earth.

Figure 1.

Understanding the relative nanotechnology scale. In terms of size, a nanometer (nm) equals one billionth of a meter (10−9 m). A proton in an elemental nucleus is 1 femtometer (fm). The nucleus of a Helium atom is 3.8 fm. The diameter of a glucose molecule is approaches 1 nm. The insulin protein approaches 5 nm in size. A nanoparticle 10 nm in size is 1,000-times smaller than the diameter of a human hair. A small microprocessor transistor is 22 nm. An adenovirus particle is between 60 and 90 nm. Liposome-based nanoparticles are often 100 nm in diameter. A typical bacterium is 1 μm in size. The average size of a human cell is 10 μm. As a reference, a billion inches is 15,783 miles, more than halfway around the earth.

Close modal

Existing nanomaterials include liposomes, natural polymers (chitosan), synthetic polymers [polyethylene glycol (PEG), etc.], carbon, carbon fullerenes such as buckyballs and carbon nanotubes (CNT), graphene, ceramic, crystals, metal, silica, and quantum dots (Fig. 2 and Table 1). They generally range in size between 5 and 200 nm, but some may exceed this range. When these technologies are coupled with molecular-based targeting methods, they can potentially achieve selective gene targeting. For example, coupling of chitosan with hydrogel technology and/or selective epitope targeting with siRNA-based gene silencing are exciting developments in selective gene targeting (11, 12). The use of thioaptamer-based molecular epitope recognition also holds significant promise for selective targeting (13). The use of phage display–based peptides as well can facilitate selectively “nanotargeting” cancers (14, 15). Selective targeting can also employ the use of specific promoter-based gene expression only in premalignant or malignant cells (16–20). Any single or combination approach for molecularly enhancing target recognition in conjunction with nanoscale payload delivery is likely to facilitate selectively targeting the various stages of cancer.

Figure 2.

Nanomaterials and nanodelivery. A, liposomes are vesicles formed into a lipid bilayer. They are commonly made of bipolar phospholipids that generally contain an aqueous core. Liposomes carrying drugs can readily fuse with plasma membranes of cells. B, synthetic polymers often begin with PEG backbones that exhibit a high degree of biocompatibility. PEG polymers can also be used in more complex multistage nanoparticles to improve biocompatibility. C, chitosan is a natural cationic polysaccharide made by the partial deacetylation of chitin. Chitosan is a positively charged hydrophilic polymer. Charge-based binding and release of drugs can involve physical or chemical stimuli, such as pH, ionic strength, temperature, and magnetic and biologic molecules. D, buckyballs are fullerenes that are made entirely of carbon. They form the shape of a hollow ball and are sparingly soluble in most solvents. They are often conjugated to amino acids like l-arginine and l-phenylalanine that enable amino acid transporters to bring them into cells. E, nanotubes are cylindrical fullerenes made of carbon. They are often only a few nanometers in diameter but can be very long, from micrometers to millimeters in length. F, mesoporous silica is a silicon-based molecule. It can be made from tetraethyl orthosilicate among other silicon-based molecules. Nanoparticles typically synthesized from silica generally have a large surface area of the pores that can be filled with a drug. Depending on the dimensions and synthesis process, silicon nanoparticles can be used in a multistage fashion to deliver other nanoparticles like liposomes or chitosan to target sites. G, quantum dots are semiconductor crystals that exhibit electronic characteristics, which are closely related to the size and shape of each individual crystal. They can either be grown as crystals or made using lithography. H, gold nanoparticles (colloidal gold) are produced using liquid chemical methods. Gold nanoparticles can be used to deliver drugs or to aid in noninvasive imaging.

Figure 2.

Nanomaterials and nanodelivery. A, liposomes are vesicles formed into a lipid bilayer. They are commonly made of bipolar phospholipids that generally contain an aqueous core. Liposomes carrying drugs can readily fuse with plasma membranes of cells. B, synthetic polymers often begin with PEG backbones that exhibit a high degree of biocompatibility. PEG polymers can also be used in more complex multistage nanoparticles to improve biocompatibility. C, chitosan is a natural cationic polysaccharide made by the partial deacetylation of chitin. Chitosan is a positively charged hydrophilic polymer. Charge-based binding and release of drugs can involve physical or chemical stimuli, such as pH, ionic strength, temperature, and magnetic and biologic molecules. D, buckyballs are fullerenes that are made entirely of carbon. They form the shape of a hollow ball and are sparingly soluble in most solvents. They are often conjugated to amino acids like l-arginine and l-phenylalanine that enable amino acid transporters to bring them into cells. E, nanotubes are cylindrical fullerenes made of carbon. They are often only a few nanometers in diameter but can be very long, from micrometers to millimeters in length. F, mesoporous silica is a silicon-based molecule. It can be made from tetraethyl orthosilicate among other silicon-based molecules. Nanoparticles typically synthesized from silica generally have a large surface area of the pores that can be filled with a drug. Depending on the dimensions and synthesis process, silicon nanoparticles can be used in a multistage fashion to deliver other nanoparticles like liposomes or chitosan to target sites. G, quantum dots are semiconductor crystals that exhibit electronic characteristics, which are closely related to the size and shape of each individual crystal. They can either be grown as crystals or made using lithography. H, gold nanoparticles (colloidal gold) are produced using liquid chemical methods. Gold nanoparticles can be used to deliver drugs or to aid in noninvasive imaging.

Close modal
Table 1.

Examples of nanoparticle targeting approaches in preclinical or clinical studies

Nanoparticle typeActive agentTumorClinical trialDeliveryTargetEffectRef
Liposomes (lipid) 
 ONC-TCS liposome Vincristine sulfate Hu Approved i.v. Non-Hodgkin lymph. Improved TI (39) 
 DepoCyt liposome Cytarabine ara-C Hu Phase III i.v. Lymphomatous meningitis High resp, QOL (40) 
 Myocet liposome Doxorubicin citrate Hu Approved i.v. Metastatic breast cancer Improved TI (37) 
 DaunoXome liposome Daunorubicin Hu Approved i.v. AIDS-related KS Skin necrosis (38) 
Liposomes (PEGylated/lipid) 
 Lipoplat-PEG-DSPE Cisplatin Hu Phase I i.v. Stage IV carcinomas Nephrotoxicity (43) 
 Doxil PEG-lipo Doxorubicin Hu Approved i.v. AIDS-related KS BMT (42) 
 Thermodox PEG-LS DoxThermo-bubble Hu Phase III i.v. Liver, breast cancer Improved TI (41) 
 NDC PEG-lipo Doxcurcumin comp Hu Preclinical i.v. Prostate cancer-PC3A Cardiotoxicity (44) 
siRNA liposomes 
 ALN-TTR01 & 2 siRNA transthyretin Hu Phase I i.v. Transthyretin amyloidosis Transthyretin (45) 
 ALN-PCS siRNA kexin type 9 Hu Phase I i.v. Fam. hypercholesterolemia LDL cholesterol (46) 
Albumin-bound nanoparticles 
 Abraxane Albumin-drug Paclitaxel Hu Approved i.v. Metastatic breast cancer Improved TI (48) 
Polymeric nanoparticles 
 PolyDL-lactide-PEG Paclitaxel Hu Phase II i.v. ARM Toxicity, OC (50) 
 Oncaspar PEG-L-aspar l-asparaginase Hu Approved i.v. ALL Remission (49) 
 DACH Plat poly prodrug Hu Phase II i.v. Ovarian cancer Tolerability, OC (51) 
 BIND-014 PN Docetaxel Hu Mo Phase II i.v. Animal tumors, PK-human PSA-targeted (52) 
Chitosan 
 Microcryst chitosan Absorb GI fat Hu Approved Oral High LDL cholesterol LDL cholosterol (53) 
 Glycated chitosan Laser immunotherm Hu Phase I i.v. Metastatic breast cancer Improved RR (55) 
 Chitosan Plasmid DNA Mo Preclinical Oral PIA Improved RR (56) 
 ExChitosan siRNA Mo Preclinical Nasal Inhibit gene expression Reduce EGFP XP (54) 
 Chitosan RGD-CH-NP siRNA-periostin Int Mo Preclinical i.v. Periostin ovarian tumors Decrease T. size (12) 
Fullerenes 
 C60(OH)24 Doxorubicin Ra Preclinical i.p. MNU induced rat tumors Dec hepato-tox (58) 
 C60-PEI-folate Docetaxel Mo Preclinical i.v. Sarcoma S180 Balb/c Decrease T. size (63) 
 C60-Hyaluronate NA Hu Mo Preclinical i.v. HCT-116 Decrease T. size (59) 
Nanotubes 
 PEG-SWNTs TNFR-(GITR) Mo Preclinical i.v. B16 melanoma Intratumor Tregs (61) 
 NGR-SWNTs-2ME Paclitaxel, taxol Hu Preclinical i.v. MCF-7 Decrease T. size (62) 
 SWNT-lipid-PTX 2-methox estradiol Mo Preclinical i.v. Sarcoma (S180) Balb/c Decrease T. size (57) 
 O-MWNTs-PEG-ANG Doxorubicin Ra/Mo Preclinical i.v. C6 glioma/Balb/c Decrease T. size (60) 
Mesoporous silica nanoparticles 
 C-dots 124I-RGDY-PEG Hu Mo Phase I i.v. αvβ3 Integrin–melanoma Real-time imaging (64) 
 Mag-Dye@MSN Fluores, magnetic Ra Preclinical i.v. Rat tumors Tumor imaging (66) 
 MSV/EphA2 siRNA EphA pac Hu Preclinical i.v. Ovarian tumor Decrease T. size (65) 
Quantum dots 
 QD-800 Anti-EGFRvIII Hu Preclinical i.v. Glioblastoma Tumor imaging (67) 
 QD-MUC1-DOX Doxorubicin Hu Preclinical i.p. Human ovarian Tumor imaging (69) 
 QD-antiVEGFR2 AntiVGFR2 Hu Preclinical i.v. Human PC3 prostate Tumor imaging (68) 
Metal nanoparticles 
 Gold, AuNPs Gold photo-activ. Ra Preclinical Cran F98 glioma-bearing rats Tumor imaging (71) 
 Immuno-magnetic EpCAM Mb Hu Preclinical CTC Identify CTCs CTCs in BrCa (72) 
 Gold, AuNP EMT target Hu Preclinical i.p. Ovarian cancer Decrease T. size (70) 
 Gold, AuNP-TNF TNFα Hu Preclinical i.v. Prostate cancer Improved therapy (73) 
Nanoparticle typeActive agentTumorClinical trialDeliveryTargetEffectRef
Liposomes (lipid) 
 ONC-TCS liposome Vincristine sulfate Hu Approved i.v. Non-Hodgkin lymph. Improved TI (39) 
 DepoCyt liposome Cytarabine ara-C Hu Phase III i.v. Lymphomatous meningitis High resp, QOL (40) 
 Myocet liposome Doxorubicin citrate Hu Approved i.v. Metastatic breast cancer Improved TI (37) 
 DaunoXome liposome Daunorubicin Hu Approved i.v. AIDS-related KS Skin necrosis (38) 
Liposomes (PEGylated/lipid) 
 Lipoplat-PEG-DSPE Cisplatin Hu Phase I i.v. Stage IV carcinomas Nephrotoxicity (43) 
 Doxil PEG-lipo Doxorubicin Hu Approved i.v. AIDS-related KS BMT (42) 
 Thermodox PEG-LS DoxThermo-bubble Hu Phase III i.v. Liver, breast cancer Improved TI (41) 
 NDC PEG-lipo Doxcurcumin comp Hu Preclinical i.v. Prostate cancer-PC3A Cardiotoxicity (44) 
siRNA liposomes 
 ALN-TTR01 & 2 siRNA transthyretin Hu Phase I i.v. Transthyretin amyloidosis Transthyretin (45) 
 ALN-PCS siRNA kexin type 9 Hu Phase I i.v. Fam. hypercholesterolemia LDL cholesterol (46) 
Albumin-bound nanoparticles 
 Abraxane Albumin-drug Paclitaxel Hu Approved i.v. Metastatic breast cancer Improved TI (48) 
Polymeric nanoparticles 
 PolyDL-lactide-PEG Paclitaxel Hu Phase II i.v. ARM Toxicity, OC (50) 
 Oncaspar PEG-L-aspar l-asparaginase Hu Approved i.v. ALL Remission (49) 
 DACH Plat poly prodrug Hu Phase II i.v. Ovarian cancer Tolerability, OC (51) 
 BIND-014 PN Docetaxel Hu Mo Phase II i.v. Animal tumors, PK-human PSA-targeted (52) 
Chitosan 
 Microcryst chitosan Absorb GI fat Hu Approved Oral High LDL cholesterol LDL cholosterol (53) 
 Glycated chitosan Laser immunotherm Hu Phase I i.v. Metastatic breast cancer Improved RR (55) 
 Chitosan Plasmid DNA Mo Preclinical Oral PIA Improved RR (56) 
 ExChitosan siRNA Mo Preclinical Nasal Inhibit gene expression Reduce EGFP XP (54) 
 Chitosan RGD-CH-NP siRNA-periostin Int Mo Preclinical i.v. Periostin ovarian tumors Decrease T. size (12) 
Fullerenes 
 C60(OH)24 Doxorubicin Ra Preclinical i.p. MNU induced rat tumors Dec hepato-tox (58) 
 C60-PEI-folate Docetaxel Mo Preclinical i.v. Sarcoma S180 Balb/c Decrease T. size (63) 
 C60-Hyaluronate NA Hu Mo Preclinical i.v. HCT-116 Decrease T. size (59) 
Nanotubes 
 PEG-SWNTs TNFR-(GITR) Mo Preclinical i.v. B16 melanoma Intratumor Tregs (61) 
 NGR-SWNTs-2ME Paclitaxel, taxol Hu Preclinical i.v. MCF-7 Decrease T. size (62) 
 SWNT-lipid-PTX 2-methox estradiol Mo Preclinical i.v. Sarcoma (S180) Balb/c Decrease T. size (57) 
 O-MWNTs-PEG-ANG Doxorubicin Ra/Mo Preclinical i.v. C6 glioma/Balb/c Decrease T. size (60) 
Mesoporous silica nanoparticles 
 C-dots 124I-RGDY-PEG Hu Mo Phase I i.v. αvβ3 Integrin–melanoma Real-time imaging (64) 
 Mag-Dye@MSN Fluores, magnetic Ra Preclinical i.v. Rat tumors Tumor imaging (66) 
 MSV/EphA2 siRNA EphA pac Hu Preclinical i.v. Ovarian tumor Decrease T. size (65) 
Quantum dots 
 QD-800 Anti-EGFRvIII Hu Preclinical i.v. Glioblastoma Tumor imaging (67) 
 QD-MUC1-DOX Doxorubicin Hu Preclinical i.p. Human ovarian Tumor imaging (69) 
 QD-antiVEGFR2 AntiVGFR2 Hu Preclinical i.v. Human PC3 prostate Tumor imaging (68) 
Metal nanoparticles 
 Gold, AuNPs Gold photo-activ. Ra Preclinical Cran F98 glioma-bearing rats Tumor imaging (71) 
 Immuno-magnetic EpCAM Mb Hu Preclinical CTC Identify CTCs CTCs in BrCa (72) 
 Gold, AuNP EMT target Hu Preclinical i.p. Ovarian cancer Decrease T. size (70) 
 Gold, AuNP-TNF TNFα Hu Preclinical i.v. Prostate cancer Improved therapy (73) 

Abbreviations: ALL, acute lymphocytic leukemia; ARM, advanced refractory malignancy; BMT, bone marrow toxicity; BrCa, breast cancer; CTC, circulating tumor cell; DACH, diaminocyclohexadine; Hu, human; KS, Kaposi sarcoma; LS, liposomes; MDR, multidrug resistance; Mo, mouse; MWNT, multiwall nanotubes; OC, outcome; PIA, peanut-induced allergy; PN, polynanoparticle; QD, quantum dot; QOL, quality of life; Ra, rat; RR, response rate; SWNT, single wall nanotubes; TI, therapeutic index.

Knowledge about uptake and distribution of nanoparticles comes from studies involving various types of environmental exposures (21). Nanoparticle internalization depends on the exposure time, carrier vehicle, mode of access, and tissue interface involved in a given exposure (21). Tissue interfaces include dermal surfaces, any exposed mucous membrane, and the respiratory airways (21). The biodistribution of nanoparticles in the body typically depends on the material and its size, shape, and charge (21–24). In the case of dermal exposures, nanoparticle penetration typically occurs at hair follicles (25) and in flexed (26) and broken skin (27). As one example, 10 to 50 nm sized TiO2 nanoparticles found in sunscreen, when suspended in oil-in-water emulsions, can penetrate hairy skin at the hair follicle sites or pores when compared with water-based suspensions (28, 29). In the case of gastrointestinal (GI) mucous membranes, absorption depends on size, which diminishes for larger particles ranging from 50 to 1,000 nm (30), exposure time, and enterocyte involvement (31). As a potential consequence, GI uptake of dietary nanoparticles (100–1,000 nm) may also influence chronic inflammation in the colon (32). In the case of aerosol delivery to the lung in contrast, noncationic nanoparticles larger than 34 nm are retained within the lungs, whereas smaller nanoparticles enter the regional lymph nodes (33). In the same report, neutrally charged nanoparticles 6 nm or less entered the bloodstream from the alveolar air-spaces followed by renal clearance (33). Similarly, blood-borne particles 10 nm or less in size usually undergo elimination by renal clearance, whereas particles 100 nm or greater in size are taken up by the reticuloendothelial system (34). The shape and surface properties of nanoparticles can also influence uptake and distribution, which can be experimentally optimized at the molecular level to enhance targeting properties (35, 36).

Preclinical and clinical nanotherapeutics

The relative success of nanoparticle preclinical and clinical use has evolved with the introduction of technology (Table 1). The use of nanoencapsulated agents helps reduce the toxicity of chemotherapeutic drugs. Many clinically approved approaches involved the early introduction of lipid liposomes, including ONC-TCS, DepoCyt, Myocet, and DaunXome (37–40). Similarly, a number of PEGylated/lipid liposome–based approaches are either clinically approved or in trial, including Lipoplatin, Doxil, and Thermodox (41–43). One approach to overcome multidrug resistance integrates therapy and preventive approaches by combining doxorubicin–curcumin into nanoparticles known as NanoDoxCurc (NDC; ref. 44). The delivery of RNAi therapy using nanoparticles is another important use of nanotechnology. Alnylam Pharmaceuticals, for example, has two liposomal siRNA in non–cancer-related clinical trials (45, 46). Alnylam also produced polymeric nanoparticles for in vivo delivery of siRNA to target endothelial cells, primary tumor growth, and metastasis (47). Another nanoformulation consisting of albumin-based (Nab)–paclitaxel or Abraxane is the first drug delivery system approved for the treatment of metastatic breast cancer, metastatic non–small cell lung cancer (NSCLC) and first-line treatment of patients with metastatic pancreatic cancer (48). A variety of synthetic polymeric nanoparticles are also in clinical settings, such as Genexol-PM, Oncaspar, and Prolindac (49–51). In the targeted realm, BIND-14 is a nanoencapsulated composite that includes a controlled-release synthetic polymer containing docetaxel. This nanoparticle composite binds to the prostate-specific membrane antigen (PSMA) targets for treating solid tumors (52). In contrast, chitosan is a natural polymer used orally to decrease high-serum LDL cholesterol (53). Since chitosan was introduced, it has enjoyed numerous uses as a nanoparticle platform in preclinical and clinical studies (12, 54–56).

Inert carbon-based modalities are also receiving significant interest as drug, nucleotide, and protein-protective delivery approaches. Fullerenes and nanotubes are nanoparticles that hold much promise in preclinical settings (57–63). Mesoporous silica is another biologically inert platform for building complex nanoparticle identification and delivery of drug, nucleotide, and protein agents (64–66). Within the silica realm, Cornell (C)-dots received phase I approval for melanoma targeting (64). In contrast, uses of inert quantum dots are focused on targeted delivery and imaging due to their high quantum fluorescence yield (67–69).

In the heavy metal space, gold nanoparticles may be particularly useful for targeting tumors (70–73). In one study, unmodified gold nanoparticles inhibit tumor growth and metastasis through abrogation of MAPK signaling and reversal of the epithelial–mesenchymal transition in two preclinical mouse models of ovarian cancer (70). The authors suggested that these findings laid the foundation for further research in the use of inorganic nanoparticles as a class of antitumor and antimetastatic agents (70). In addition to serving as drug carriers, gold nanoparticles are finding use as photothermal agents, contrast agents, and radiosensitisers (74).

Other reports highlight some advantages of nanoparticles (75). Chauhan and Jain have applied nanodelivery methods to decrease the toxicity of therapeutic drugs (76). Inhalation of retinoids, steroids, or certain therapeutics used to treat lung cancer may also be more effective if applied in a nanoized form (77–80). Because a primary goal of nanoprevention is sustained release along with significant reduction in toxicity, the use of low-dose nanoized chemotherapeutic drugs might be useful in high-risk preventive settings.

Identifying biocompatible nontoxic nanomaterials is vital for the application of nanotechnology in prevention. The mode of delivery greatly influences uptake and toxicity (81). Nanotherapies are often injected directly via the blood stream or peritoneum (Table 1 and Fig. 3). Other common delivery methods include the skin, respiratory, or GI systems, as they are more acceptable for nanoprevention modalities (81). Mucous membrane surfaces of the eye, mouth, nose, upper GI tract, and vaginal surfaces can also take up nanoparticles. The particulate nature of nanomaterials leads to local accumulation followed by systemic dissemination via the cardiovascular or lymphatic systems. Among a variety of toxic effects, acute responses can begin with the generation of reactive oxygen species coupled with inflammatory reactions (82).

Figure 3.

Nanoparticles in the blood or lung. The biology and toxicity of nanoparticles are influenced by the mode of delivery. A, intravascular delivery of nanoparticles can initiate a variety of interactions in the blood stream. Nanoparticles can interact with circulating proteins to form active or inactive protein coronas. Active protein coronas promote can trigger platelet activation. Activating interactions can also stimulate various immune cells like macrophages, lymphocytes, neutrophils, and eosinophils, as well as antibodies, complement proteins, and coagulation factors. Nanoparticles with inactive coronas can undergo transcellular or transjunctional transport. B, inhaled nanoparticles are dispersed throughout the alveolar sacs of respiratory bronchioles and undergo processing in the lungs depending on the material size and charge. Nanoparticles smaller than or equal to 6 nm can readily enter the blood stream via transcellular processes. Nanoparticles larger than approximately 34 nm are engulfed and processed by alveolar macrophages.

Figure 3.

Nanoparticles in the blood or lung. The biology and toxicity of nanoparticles are influenced by the mode of delivery. A, intravascular delivery of nanoparticles can initiate a variety of interactions in the blood stream. Nanoparticles can interact with circulating proteins to form active or inactive protein coronas. Active protein coronas promote can trigger platelet activation. Activating interactions can also stimulate various immune cells like macrophages, lymphocytes, neutrophils, and eosinophils, as well as antibodies, complement proteins, and coagulation factors. Nanoparticles with inactive coronas can undergo transcellular or transjunctional transport. B, inhaled nanoparticles are dispersed throughout the alveolar sacs of respiratory bronchioles and undergo processing in the lungs depending on the material size and charge. Nanoparticles smaller than or equal to 6 nm can readily enter the blood stream via transcellular processes. Nanoparticles larger than approximately 34 nm are engulfed and processed by alveolar macrophages.

Close modal

The blood stream is often used for delivering nanotherapy (Fig. 3). It is a closed system that is not directly exposed to the environment. As a closed system, the blood stream has specific processes identifying and mitigating pathogens and foreign bodies, which are processed further by the liver or excreted by the kidneys (33, 83). Once in the blood stream, platelets are among the first responders to many foreign bodies that initiate emboli formation (84). Nanoparticles in flowing blood also interact with antibodies, circulating immune cells, coagulation factors, and the surfaces of endothelial cells (85, 86). Nanomaterials can bind proteins through noncovalent interactions to form a protein corona (87, 88). This corona influences biologic activity and interactions with platelets and probably liver clearance (87, 88). Chemically modifying CNT surfaces, for example, to preferentially bind albumin versus fibrinogen influences protein corona formation and platelet interactions (87). Similarly, nanoparticle protein coronas can activate the circulating macrophages and trigger inflammasome formation by immune cells (89). Preformed albumin nanoparticle coronas are another method of limiting toxicity in the blood by reducing complement activation (90). Other mechanisms of toxicity occur with metal and metal oxides that generate reactive oxygen species and proinflammatory oxidative stress (91, 92). Depending on the routes of exposure, metal and metal oxide nanoparticles can affect cells and organ pathophysiology (91, 92). Similarly, graphene-based (93) and other inorganic nanomaterials (94) exhibit various toxicities to consider in the exposure and safer design of these materials. Nanoparticles are also being used to facilitate vaccinations and immunotherapy (95). Functionalizing polymeric nanoparticles using membrane coating derived from cancer cells is a different approach to selectively target tumors, but any effects on cardiovascular toxicity remain to be determined (96). The structure, size, and surface properties that affect efficacy and toxicity should be carefully considered before using the blood stream as a portal for delivering nanomaterials.

Intraperitoneal (i.p.) injection is another common mode of nanoparticle delivery to a closed-body compartment (97, 98). Far less is known about intraperitoneal uptake and toxicity associated with this delivery method. Among the various abdominal-cell targets are peritoneal macrophages (99). After intraperitoneal injections in one study, silver nanoparticles accumulated most heavily in liver Kupffer cells and hepatocytes along with kidney podocytes and other cells in the peritoneum (100). These changes were accompanied by cell death and the infiltration of white blood cells, lymphocytes, granulocytes, and hemoglobin (100). Chemical modification of surface charges can minimize toxicity while enhancing biodistribution and tumor targeting of some intraperitoneal injected nanoparticles (101).

In contrast with the blood and peritoneum, the lungs, skin, and intestinal tract are in direct contact with the environment (102). The upper airway and lungs continuously interface with the atmosphere (ref. 103; Fig. 3). A well-developed immune system helps process inhaled atmospheric particulates (104). In the lung, inhaled nanoparticles initially encounter pulmonary surfactants and then pulmonary cells (105). The very thin air–blood barrier (<1 μm) combined with the collective lung-alveolar cell-surface area provides high systemic nanoparticle bioavailability (105). Once engaged by cell surfaces, caveolae-mediated endocytosis translocates nanoparticles into alveolar epithelial or immune cells (106). Depending on the type and size of inhaled nanoparticles (107), pyroptosis-based toxicity can occur (108). Pyroptosis is driven by alveolar macrophages that cause a specialized inflammasome-dependent form of cell death (108). Pulmonary effects are further influenced by the volumes of material inhaled and mass transfer (109). Clinicopathologic responses involve follicular hyperplasia, protein effusion, alveolar lipoproteinosis, and pulmonary capillary vessel hyperemia that lead to fibrosis and emphysema with sustained exposure (110). Modification of nanoparticle size and charge can reduce toxicity while providing rapid sustained release in the lung tissue, thereby resulting in reduced dosing frequency and improved patient compliance (104, 111, 112). As long as toxicity and side effects are minimized, inhalation is a highly efficient mode of nanomaterial delivery for preventive treatments.

Skin, in contrast, is the largest organ and protective barrier of the body that can serve as a topical, regional, and transdermal mode of drug delivery (113). The use of skin-based nanomedical delivery methods may help reduce toxicity, improve sustained release, and penetration (113, 114). Skin exposure to nanoparticles as pollution, antibacterials, and sun screen each have some toxicity concerns (115–117). Nanoparticle uptake measurements using quantum dots showed that penetration into the dermal layer is limited to the uppermost stratum corneum layers and the hair follicles (118). Those nanoparticles that enter the blood stream accumulate in the liver and kidney with poor clearance rates (118). Toxicity can directly affect skin cells by forming reactive oxygen species along with autophagic vacuole accumulation and mitochondria damage (119). Localized inflammatory responses lead to macrophage-, monocyte-, and dendritic-cell responses that cause cytotoxicity (120). Additional oxidative stress, Ca2+ flux and decreased mitochondrial membrane potential are accompanied by production of IL1beta and chemokine CXCL9 (120). A variety of approaches hold promise for decreasing dermal toxicity and improving delivery as potential preventive modalities (121–123). Similar approaches hold promise for directly improving melanoma treatment (124).

Oral delivery of nanomaterials is the most common and well-tolerated method for delivering preventive or other agents (125). Other than distribution studies, the toxicology of nanoparticles is not well understood (126). Although oral delivery of nanomaterials is well integrated into the food and drug industry, their long-term fate in the GI tract remains unclear (127, 128). Nanoparticle delivery methods are showing improvements in stabilizing oral delivery and survival of in low gastric pH (129, 130). The intestinal tract maintains elaborate mechanisms for simultaneously taking in nutrients while preserving healthy gut flora and controlling micro- and nanopathogens (131–134). The uptake and distribution of nanoparticles in the gut is also influenced by mucin produced as a protective barrier (135). The mucosal component of the GI tract includes the gut-associated lymphoid tissue that is responsible for antigen sampling (131, 136, 137). One of the primary endocytic pathways for pathogen sampling includes the follicle-associated epithelium of the Peyer's patches found in the upper GI tract (Fig. 4; refs. 131–134). Peyer's Patches contain specialized microfold (M) cells that actively internalize particulate material (131–134). This process involves endocytosis in clathrin-coated vesicles, actin-dependent phagocytosis, or macropinocytosis (131–134). Once internalized, M cells form “intraepithelial pockets” through an expanded basolateral domain. This transcytosis process culminates in shipment and presentation of pathogen-laden exosomes to immune cells (131–134). M-cell exosomal antigens are presented to subepithelial dendritic cells (DC) or are directly sampled by LysoM+ dome DCs. These antigens are subsequently presented to T- and B lymphocytes within Peyers' Patches (138, 139). Some efforts are under way to specifically target the M cells (131).

Figure 4.

Oral delivery and GI uptake. Oral delivery of nanoparticles depends primarily on GI uptake and sampling mechanisms. A, the GI tract contains 70% of the immune system and is heavily involved in uptake and processing of pathogens and foreign materials in preparation for defensive responses. Potential sampling of nanoparticles may include: (i) M-cell transport to subepithelial dome cells (SDDC) then to lymphocytes is a key immune sampling process; (ii) CXCR1+ LPC is another sapling mechanism; (iii) LPC movement across the epithelium is another sampling mechanism; (iv) LPC sampling of transepithelial endosomal and exosomal antigens can also occur; (v) GAPs to CD103+ DCs can also occur; (vi) pathologic breaches in the gut epithelium such as cancer formation can also allow for nanoparticle passage. B, vesicle processing may be a key aspect of nanoparticle migration across the gut epithelium. M cells form a series of prominent vesicles before transfer from SDDC to T- or B lymphocytes. The uptake of nanoparticles by microvillar enterocytes is often followed by endosome formation, the genesis of microvessicular bodies (MVB) and fusion with lysosomes and then transfer to the lamina propria is another mode of passage. Another mechanism enterocytes use for transepithelial transport involves uptake into endosomes, MVB formation, fusion with Golgi apparatus and the exosomal transfer to the lamina propria. Transport to the blood stream or lymphatic system can lead to further dissemination.

Figure 4.

Oral delivery and GI uptake. Oral delivery of nanoparticles depends primarily on GI uptake and sampling mechanisms. A, the GI tract contains 70% of the immune system and is heavily involved in uptake and processing of pathogens and foreign materials in preparation for defensive responses. Potential sampling of nanoparticles may include: (i) M-cell transport to subepithelial dome cells (SDDC) then to lymphocytes is a key immune sampling process; (ii) CXCR1+ LPC is another sapling mechanism; (iii) LPC movement across the epithelium is another sampling mechanism; (iv) LPC sampling of transepithelial endosomal and exosomal antigens can also occur; (v) GAPs to CD103+ DCs can also occur; (vi) pathologic breaches in the gut epithelium such as cancer formation can also allow for nanoparticle passage. B, vesicle processing may be a key aspect of nanoparticle migration across the gut epithelium. M cells form a series of prominent vesicles before transfer from SDDC to T- or B lymphocytes. The uptake of nanoparticles by microvillar enterocytes is often followed by endosome formation, the genesis of microvessicular bodies (MVB) and fusion with lysosomes and then transfer to the lamina propria is another mode of passage. Another mechanism enterocytes use for transepithelial transport involves uptake into endosomes, MVB formation, fusion with Golgi apparatus and the exosomal transfer to the lamina propria. Transport to the blood stream or lymphatic system can lead to further dissemination.

Close modal

In other areas of the gut, enterocytes absorb nutrients via microvilli of the brush border that can be disrupted by exposure to nanomaterials (140). Villous bearing areas of the gut also exhibit a different set of mechanisms for transepithelial antigen sampling or microbiota/antigen presentation to cells and vessels in the microenvironment of the lamina propria (Fig. 3). This process can occur directly as CX3CR1+ lamina propria cells (LPC) cross the epithelium in a basolateral-to-luminal direction (141). Another mechanism involves direct sampling from the gut lumen by mucus-producing goblet cells (142). After the goblet cells have sampled the gut lumen, goblet cell–associated antigen passages' (GAP) transfer various antigens to CD103(+) DCs in the lamina propria (142). These CD103(+) DCs within the lamina propria then induce T-cell responses (142). In other cases, CD11c+ DCs or CXCR1+ LPC cells in the lamina propria can extend transepithelial dendrites (TED) between enterocytes directly into the gut to sample luminal contents and ensnare microbiota (143–145). As a completely different mechanism of transcytosis, vesicle passage into the lamina propria also occurs directly through intestinal epithelial cells (138, 146). Once exosomes enter the lamina propria, they can gain access to the various cells, including myofibroblasts, pericytes, and capillaries or lacteal vessels of the lymphatic system (147–150). Functionalizing the outer nanoparticle surface can make them an ideal vector suited to traverse brush borders, mucosal lymphoid tissue, and other surfaces (140). Nanoparticles can also be used to deliver vaccines via the gut (151). Nanotargeting of chronic inflammation in the GI tract also holds promise for prevention (152). As with the other delivery methods, minimizing toxicity while maximizing tolerability are essential in generating acceptance of GI delivery approaches for prevention.

Vital differences exist between various precancerous and cancer lesions that can profoundly influence how they are targeted using nanotechnology. Morphologic differences based on gross pathology and histology are the most obvious distinctions (153). The site of origin influences the morphologic characteristics and biologic differences along with the unique characteristics of the individual tumor. Each type of cancer presents its own unique challenges for effective targeting (154). These challenges are influenced further by variations in the anatomic, physiologic, microenvironmental, cellular, and molecular features of the involved tissue site (155). The stage of progression is also critical (156). Advanced-stage cancers that have metastasized can originate in a totally different organ from the one in which they are discovered. In contrast, premalignancies are confined to the site of origin.

Complex multistep biology combined with long progression time frames provide multiple potential points for early detection or intervention using nanotechnology-based approaches. Nanotheranostic tools that simultaneously identify and selectively target premalignant lesions would be extremely useful. This is likely to require modification of probes that selectively identify specific premalignant lesions. Ideally, we will be able personalize theranostic approaches and nanotarget premalignancies by implementing “-omics” approaches for profiling as a first step and selective targeting as a second step (157–159). Some recent advances in theranostics incorporate magnetic resonance (MRI) imaging for early detection along with targeting (160, 161). A molecular imaging strategy using a new triple-modality MRI photoacoustic-Raman nanoparticle is a unique theranostic approach directed at brain tumors (162).

Premalignancies are generally considered the primary targets for cancer prevention (163). Several characteristics are used to distinguish between premalignant and malignant lesions. These characteristics can potentially affect the effectiveness of nanotechnology-based targeting (see Table 2). In addition to being the most common form of cancer, carcinomas arising from epithelial origins are the easiest to identify on the basis of the morphologic distinctions between precancers and cancers. They are often referred to as carcinoma in situ or intraepithelial neoplasias (IEN). Epithelial precancers are by nature organ-confined and generally remain restricted to ducts or the epithelial strata of tissues (164). Cancers, in contrast, are often identified as having breached or invaded through a fine fibrous complex that forms a sheet-like barrier known as the basement membrane (165). The basement membrane can undergo extensive remodeling or thickening during inflammatory responses (166, 167) or become disorganized in tumor vasculature (168) and in various cancers (169). How early this occurs is not clear, particularly with respect to precancerous lesions. Because the basement membrane serves as a structural base for normal epithelia/endothelia, but can potentially accrue abnormalities during carcinogenesis, its status demands adequate consideration when devising nanotargeting approaches.

Table 2.

Acquired characteristics of premalignant and malignant lesions

CharacteristicPremalignantMalignant
Genetic abnormalities Few Numerous 
Suppressor genes Semiactive growth suppression Shut off or fully autonomous 
Growth signals Homeostatic, semidependent Paracrine, autocrine, or fully autonomous 
Stem and progenitor cells Semiexpanded population Expanded population, enhanced chemoresistance 
Apoptosis Semifunctional Dysfunctional, ineffective 
Invasion status Noninvasive lesion Enhanced motility, invasion, and metastatic potential 
Basement membrane Intact Breached 
Cell morphology Hyperplasia, dysplasia Anaplasia 
Angiogenesis Semiactive Sustained dysfunction 
CharacteristicPremalignantMalignant
Genetic abnormalities Few Numerous 
Suppressor genes Semiactive growth suppression Shut off or fully autonomous 
Growth signals Homeostatic, semidependent Paracrine, autocrine, or fully autonomous 
Stem and progenitor cells Semiexpanded population Expanded population, enhanced chemoresistance 
Apoptosis Semifunctional Dysfunctional, ineffective 
Invasion status Noninvasive lesion Enhanced motility, invasion, and metastatic potential 
Basement membrane Intact Breached 
Cell morphology Hyperplasia, dysplasia Anaplasia 
Angiogenesis Semiactive Sustained dysfunction 

Premalignant IEN lesions are often accompanied by hyperplasia, or the physiologic proliferation of cells to a greater extent than normal (153). These precancerous lesions may also exhibit dysplasia or the abnormal maturation of cells. Along the continuum of progression from precancerous to cancerous lesions, the cells may begin to exhibit anaplasia. Anaplastic lesions contain cells that have lost their functional tissue identity and reverted to a more primitive or undifferentiated form. IEN lesions also can display pleomorphism that includes large, darkly stained nuclei, and the ratio of nuclear material to cytoplasmic material increases. These precancers are sometimes widely distributed and/or very small in extent and size and, thus, hard to identify until they grow larger (164). If IENs arise in a ductal structure, they can begin to fill in the vacant space or lumen cavity of the duct. Unless this proliferation is accompanied by the stimulation of angiogenesis, these lesions may begin to show signs of apoptosis (170). These IEN lesion characteristics are likely to influence the delivery of nanotechnology to the target site and should be considered during the design and analytic phases of any proposed prevention study (171).

Although angiogenesis may contribute to premalignancy (172), exactly how early it occurs and to what extent it plays a role in the genesis and progression of precancerous lesions is not fully understood (Fig. 5). The biomarkers used to immunochemically identify increased microvascular density include factor VIII, CD31, and CD34 (172). Another biomarker, CD105, is also used to identify newly formed vessels (173–177). More advanced tumor-mediated angiogenesis is highly deregulated and leads to disorganized, poorly formed, and leaky or highly permeable blood vessels (178).

Figure 5.

The angiogenic switch. The angiogenic switch can occur at different stages of carcinogenesis. A, normal tissue growth achieves a balance between cell growth and apoptosis that helps maintain normal homeostasis. B, premalignant disease may involve the earliest stages of angiogenesis. Chronic inflammation involving the accumulation of immune cells, cytokines, and prostaglandins during premalignancy helps to promote vessel dilation and the detachment of pericytes from blood vessels. During the early-stage conversion of premalignant to malignant tissues blood factors accumulate in the perivascular space. The upregulation of COX-2 and the accumulation of vascular endothelial cell growth factor (VGEF), basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF), epithelial growth factor (EGF), and angiopoietin 2 (Ang2) trigger the angiogenic switch. C, the onset of malignancy, heightened hypoxia, and apoptosis further increase vessel leakiness. The onset of angiogenic sprouting and tip cell–mediated migration into the growing tumor leads to further recruitment of immune cells and perivascular cells as part of the angiogenic switch.

Figure 5.

The angiogenic switch. The angiogenic switch can occur at different stages of carcinogenesis. A, normal tissue growth achieves a balance between cell growth and apoptosis that helps maintain normal homeostasis. B, premalignant disease may involve the earliest stages of angiogenesis. Chronic inflammation involving the accumulation of immune cells, cytokines, and prostaglandins during premalignancy helps to promote vessel dilation and the detachment of pericytes from blood vessels. During the early-stage conversion of premalignant to malignant tissues blood factors accumulate in the perivascular space. The upregulation of COX-2 and the accumulation of vascular endothelial cell growth factor (VGEF), basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF), epithelial growth factor (EGF), and angiopoietin 2 (Ang2) trigger the angiogenic switch. C, the onset of malignancy, heightened hypoxia, and apoptosis further increase vessel leakiness. The onset of angiogenic sprouting and tip cell–mediated migration into the growing tumor leads to further recruitment of immune cells and perivascular cells as part of the angiogenic switch.

Close modal

The “angiogenic switch” that initiates angiogenesis was shown by Folkman and colleagues to occur before cells achieve the invasive state (179). This switch was recently shown to involve microRNA, among other factors (180), and may serve as a useful target for cancer prevention (181). Recent evidence indicates that angiogenesis coincides with the onset of dysplasia during adenoma formation in colorectal cancer progression (182). In the breast, angiogenesis seems to begin with the onset of hyperplasia in the mammary duct and progresses through ductal carcinoma in situ and invasive disease (183). Whether angiogenesis starts as a normal inflammatory or tissue repair process during premalignancy and then progresses toward an abnormal state during tumorigenesis or begins as an abnormal process is not known.

Abnormal or leaky blood vessels that arise during tumor-initiated angiogenesis are likely to be attractive targets for nanotechnology-based interventions using vascular delivery approaches (184). However, further work is needed to determine whether vascular-based targeting will be a useful approach for cancer prevention.

Various nanotools could potentially help identify any connections that may exist between the angiogenic switch and premalignancy. Perhaps, nanotools might help identify early changes in endothelial cells and/or tumor cells that signify the beginning of neoangiogenesis. Optimally, early-stage biomarker recognition epitopes or ligands involved in neoangiogenesis will be incorporated onto nanotools as surface molecules that recognize premalignant changes. The involvement of interendothelial transport of modified nanotools may facilitate identifying not only the early changes in these lesions but also the interconnections between endothelial cells and tumor cells (185). These processes may also be identified by biologic changes in the pericytes (186, 187) or vascular endothelial tip cells (188, 189) that are heavily linked to angiogenesis and vascular sprouting. The ability to image and target early-stage changes in the microvasculature of premalignant lesions will help to significantly advance this emerging field.

Determining the usefulness of preventive interventions relies heavily on the therapeutic index of a given treatment (190). This index can be broken down into a risk–benefit ratio that assesses its effectiveness (191, 192). In the case of prevention, the risk to a given population is typically minimized to the greatest extent possible. Any risk assessment needs to be carefully examined, not only for the legal ramifications but also for cost effectiveness (193). It is also important to identify a reasonable balance between safety, efficacy, and affordability, particularly because nanoscale devices are often very complex and can add substantial production costs. Ultimately, a risk assessment also encompasses identifying a given agent's intended and unintended effects based on an individual's/cohort's susceptibilities to both disease development and the agent's effects. If a subset of patients is more susceptible to the adverse effects of a nanopreventive agent than the general population, these patients must be identified and excluded from any study. Moreover, any nanoscale preventive treatment with severe side effects would generally not be suitable for use. This type of intervention must also entail tolerable risk that is suitable for long-term use. However, if a nanopreventive approach heightens sustained local release at the target site, substantial benefit may be realized. In the final analysis, any intervention can exhibit risk, but the risk must be worth the reward.

Chemoprevention is a term describing the use of agents to reverse, suppress, or prevent carcinogenesis and malignancy (194). Subjects for chemoprevention trials are stratified into multiple levels of risk (195). The lowest level of risk encompasses subjects in the general population who are seemingly healthy but may contract sporadic cancers. The allowable risk increases for subjects who present with precancerous lesions, such as IENs (195). The more advanced the premalignancy, the higher the accompanying risk (195). Subjects at high risk, such as those with a genetic predisposition or familial inheritance, are more likely to tolerate more severe side effects. This class of subjects can also include cancer survivors or those at high risk for second cancers, in which case a higher risk of adverse effects becomes more tolerable (196). The same principles that apply to identifying and stratifying subjects for chemoprevention studies should apply to nanopreventive studies. The unique nature of any given nanopreventive intervention is likely to dictate how it is used. This is probably most applicable to nanodevices that might require special monitoring procedures to ensure clearance from the subject's system.

There is a growing number of “at-risk” individuals due to demographics, the aging of the general population, and the adoption of unhealthy lifestyles (196, 197). In addition, enhanced screening can lead to identification of more individuals with precancers; and the number of cancer survivors continues to grow with enhanced early detection and treatment (196). The public tends to have a limited understanding of cancer development, risk, and prevention, let alone nanotechnology (198). Educating the public should help improve the acceptance and adoption of both prevention and nanotechnology in fighting cancer. Once implemented, nanoprevention strategies may increase the need for insurance coverage/reimbursement for prevention, necessitating the passage of legislation.

New research venues and funding opportunities are needed to improve the insights and options for the nano-based prevention of cancer. Advances in genomics, proteomics, lipidomics, metabolomics, and biospecimen-based risk assessment and prevention have had limited impact to date, perhaps because of concerns about the risk:benefit balance (199). A clear demonstration of enhanced benefit with minimized risk is needed to move this area forward at both the basic and translational levels.

Cancer prevention involves overcoming more complex biology in comparison with successes in the cardiovascular prevention area. In cancer prevention, multiple disease sites, each with their own unique challenges, must be taken into account, along with the difficulty of identifying premalignant lesions. Nanotechnology-based preventive interventions may help solve some of these issues, if they improve the early detection and targetability of premalignancies and cancers as well as the delivery and efficacy of preventive agents.

The U.S. Food and Drug Administration approved various agents for treatment of precancerous lesions or cancer risk reduction (Table 3). These vary in the type of agent and mechanism of action, depending upon the target site. There may be opportunities to make these treatments more effective by applying nanotechnology to achieve more targeted release or uptake of some of the smaller molecules or to enhance the immune response to some of the more complex molecules.

Table 3.

Approved agents for treatment of precancerous lesions or cancer risk reduction—2012

AgentTargeted cohortIndication
Tamoxifen 
  • Women with DCIS following breast surgery and radiation

 
Reduce the risk of invasive breast cancer 
 
  • Women at high risk for breast cancer

 
Reduce the incidence of breast cancer 
Raloxifene 
  • Postmenopausal women at high risk for invasive breast cancer

 
Reduction in risk of invasive breast cancer 
Cervarix 
  • Females 9 through 25 years of age

 
Prevention of the following, caused by HPV types 16 and 18:
  • cervical cancer

  • cervical intraepithelial neoplasia grade 2 or worse and adenocarcinoma in situ

  • cervical intraepithelial neoplasia grade 1

 
Gardasil Girls and women 9 through 26 years of age Prevention of cervical, vulvar, vaginal, and anal cancer caused by HPV types 16 and 18; and the following precancerous or dysplastic lesions caused by HPV types 6, 11, 16, and 18:
  • Cervical intraepithelial neoplasia grade 2/3 and cervical adenocarcinoma in situ

  • Cervical intraepithelial neoplasia grade 1

  • Vulvar intraepithelial neoplasia grade 2 and grade 3

  • Vaginal intraepithelial neoplasia grade 2 and grade 3

  • Anal intraepithelial neoplasia grades 1, 2, and 3

 
 Boys and men 9 through 26 years of age Prevention of anal cancer caused by HPV types 16 and 18; and anal intraepithelial neoplasia grades 1, 2, and 3 caused by HPV types 6, 11, 16, and 18: 
Photodynamic Therapy with Photofrin Males and females with high-grade dysplasia in Barrett esophagus. Ablation of high-grade dysplasia in Barrett esophagus patients who do not undergo esophagectomy 
Celecoxiba Males and females ≥18 years old with familial adenomatous polyposis Reduction in the number of adenomatous colorectal polyps in FAP, as an adjunct o usual care (e.g., endoscopic surveillance, surgery) 
Bacillus-Calmette- Guerin(BCG) Males and females with carcinoma in situ of the urinary bladder Intravesical use in the treatment and prophylaxis of carcinoma in situ of the urinary bladder and for the prophylaxis of primary or recurrent stage Ta and/or T1 papillary tumors following transurethral resection 
Valrubicin Males and females with Bacillus- Calmette-Guerin–refractory carcinoma in situ Intravesical therapy of BCG-refractory carcinoma in situ of the urinary bladder in patients for whom immediate cystectomy would be associated with unacceptable morbidity or mortality. 
Fluorouracil Males and females with multiple actinic or solar keratoses Topical treatment of multiple actinic or solar keratoses 
Diclofenac sodium Males and females with actinic keratoses Topical treatment of actinic keratoses 
Photodynamic Therapy with 5- aminolevulinic acid Males and females with actinic keratoses of the face or scalp Topical treatment of minimally to moderately thick actinic keratoses of the face or scalp. 
Masoprocolb Males and females with actinic (solar) keratoses Topical treatment of actinic keratoses 
Ingenol mebutate Those with actinic keratoses on the face, scalp, trunk and extremities Topical treatment of actinic keratoses 
AgentTargeted cohortIndication
Tamoxifen 
  • Women with DCIS following breast surgery and radiation

 
Reduce the risk of invasive breast cancer 
 
  • Women at high risk for breast cancer

 
Reduce the incidence of breast cancer 
Raloxifene 
  • Postmenopausal women at high risk for invasive breast cancer

 
Reduction in risk of invasive breast cancer 
Cervarix 
  • Females 9 through 25 years of age

 
Prevention of the following, caused by HPV types 16 and 18:
  • cervical cancer

  • cervical intraepithelial neoplasia grade 2 or worse and adenocarcinoma in situ

  • cervical intraepithelial neoplasia grade 1

 
Gardasil Girls and women 9 through 26 years of age Prevention of cervical, vulvar, vaginal, and anal cancer caused by HPV types 16 and 18; and the following precancerous or dysplastic lesions caused by HPV types 6, 11, 16, and 18:
  • Cervical intraepithelial neoplasia grade 2/3 and cervical adenocarcinoma in situ

  • Cervical intraepithelial neoplasia grade 1

  • Vulvar intraepithelial neoplasia grade 2 and grade 3

  • Vaginal intraepithelial neoplasia grade 2 and grade 3

  • Anal intraepithelial neoplasia grades 1, 2, and 3

 
 Boys and men 9 through 26 years of age Prevention of anal cancer caused by HPV types 16 and 18; and anal intraepithelial neoplasia grades 1, 2, and 3 caused by HPV types 6, 11, 16, and 18: 
Photodynamic Therapy with Photofrin Males and females with high-grade dysplasia in Barrett esophagus. Ablation of high-grade dysplasia in Barrett esophagus patients who do not undergo esophagectomy 
Celecoxiba Males and females ≥18 years old with familial adenomatous polyposis Reduction in the number of adenomatous colorectal polyps in FAP, as an adjunct o usual care (e.g., endoscopic surveillance, surgery) 
Bacillus-Calmette- Guerin(BCG) Males and females with carcinoma in situ of the urinary bladder Intravesical use in the treatment and prophylaxis of carcinoma in situ of the urinary bladder and for the prophylaxis of primary or recurrent stage Ta and/or T1 papillary tumors following transurethral resection 
Valrubicin Males and females with Bacillus- Calmette-Guerin–refractory carcinoma in situ Intravesical therapy of BCG-refractory carcinoma in situ of the urinary bladder in patients for whom immediate cystectomy would be associated with unacceptable morbidity or mortality. 
Fluorouracil Males and females with multiple actinic or solar keratoses Topical treatment of multiple actinic or solar keratoses 
Diclofenac sodium Males and females with actinic keratoses Topical treatment of actinic keratoses 
Photodynamic Therapy with 5- aminolevulinic acid Males and females with actinic keratoses of the face or scalp Topical treatment of minimally to moderately thick actinic keratoses of the face or scalp. 
Masoprocolb Males and females with actinic (solar) keratoses Topical treatment of actinic keratoses 
Ingenol mebutate Those with actinic keratoses on the face, scalp, trunk and extremities Topical treatment of actinic keratoses 

aFDA labeling voluntarily withdrawn by Pfizer, February 2011.

bWithdrawn from the U.S. market June 1996.

Multiple areas of cancer prevention can benefit from the application of nanotechnology. The easiest application to implement and the most useful for helping to collect scientific data is probably early detection. Nanoscale devices that effectively identify premalignancies or cancer signatures hold great promise.

Early forays into device development have targeted blood analysis. A prime example is the barcode chip (200). Devices such as these that integrate microfluidics and a barcoded protein-biomarker capture mechanism on a single chip are ideal for point-of-care detection using a finger-prick sample. Other systems use single-walled CNTs as multicolor Raman labels to achieve highly sensitive, multiplexed protein detection (201). Still other systems have coupled nanoporous silica chips that selectively enrich and stabilize low-molecular-weight peptides with mass spectrometry for highly sensitive biomarker detection (202). Coupling these nanotechnologies with stabile isotope mass spectrometry–based quantitation methods and/or specific gene subsets to plasma analyses exhibiting predictive or preventive value may significantly advance this field (203, 204). One example of this approach was used to monitor the progression of metastatic breast cancer in mouse xenografts (205). Analysis of urine also is under intensive development. Evaluation of urine biomarkers has been accomplished using near-infrared fluorescent core-shell silica-based nanoparticles (C dots; ref. 206). Similar approaches have been applied to feces (207) and saliva (208). Obtaining these samples is minimally invasive and their use will afford effective monitoring, but are, for the most part, indirect. More direct measures include isolating circulating cells or DNA from blood coupled with next-generation sequencing to identify inherent risk.

Nanotechnology may also be directly applied to the analysis of tissue biopsies or exfoliated cells for early detection. The key advantage to having cellular material is the presence of a primary source of DNA, RNA, and proteins. However, obtaining tissue biopsies is generally invasive and involves substantial discomfort and risk to the patient. Thus, obtaining biopsies is usually reserved for high-risk subjects. Another source of cellular material is exfoliated cells. The early detection of oral cancer using exfoliated cells is one example. A nanobiochip sensor technique was applied to exfoliative cytology specimens; these studies illustrate the advantage of targeting both biochemical and morphologic changes associated with early oral tumorigenesis (209). Other sources of exfoliated cells include the colon (210–212), lung (213, 214), bladder (215), cervix (216, 217), and breast (218). Ideally, the source of cells themselves or factors they produce would be suited to point-of-care monitoring (219).

Nanotechnology-based procedures are expected to significantly improve the imaging of cancer (220), including early precancerous lesions. In addition to the imaging uses already described (Table 1), other examples include magnetic nanoparticles (221), Q-dots (222), gold nanoparticles (223), nanoshells (224), and nanotubes (225). When these technologies are coupled with the modification of cell-surface chemistry, adding epitope recognition or antibody/ligand binding sites can enable targeted homing. Coupling homing with unique magnetic signatures, tunable absorption and emission spectral properties, and advanced synthesis and physical characteristics can enhance the usefulness of nanoparticles as probes. A particularly exciting breakthrough involves hyperpolarized MRI of select molecules and/or nanoparticles at up to 20,000-fold greater signal level for early detection of metabolic changes that occur in both premalignancies and cancer. (226, 227).

Achieving targeted imaging using nanoparticles in many cases affords the advantage of also delivering a site-specific chemopreventive payload. The selectiveness of these targeting systems can be driven on two levels. The first level includes the surface ligands involved in homing to the lesion site, and the second level relies on the specificity of released molecules that act directly on the target. In some cases, optimized delivery of novel nanoparticles to the tumor microenvironment may take advantage of unique or abnormal vasculature (178). In other cases, the presence of fibrotic stroma may enhance uptake and delivery (228). Furthermore, other cells such as mesenchymal stem cells may be harnessed to act as a targeted delivery system (229).

Another potential approach involves multistage nanovector delivery (230). The rationale for the use of these more complex delivery systems involves many factors. They are expected to overcome endothelial and epithelial barriers or to be taken up by the reticuloendothelial system (231), are biodegradable, and exhibit favorable hemorheology characteristics in circulation (201). The largest component of these nanovectors acts as a porous shuttle that bears the payload to a particular site. This porous shuttle is loaded with smaller nanoliposomes that contain the targeted payload, such as siRNA (232). This type of delivery enables the sustained release of an agent at a specific target site.

The immune system is capable of recognizing, homing to, and killing premalignant and malignant cells. Harnessing the immune system and enhancing the immune response is another important application of nanotechnology. Nanotechnology can enhance the antibody response (233) and the response of specific immune cells, such as T-helper-17 cells, cytokines (234), and DCs (235). Enhancing the effectiveness of vaccines is another objective (236). These efforts include nanoencapsulation to achieve transcutaneous delivery (237) as well as mucosal immunization (238). Targeted immune system recruitment may also enhance the antitumor immune response (239). These and other strategies may involve personalized regimens to eliminate tumors.

Vaccines can be very effective preventive measures. The prevention of cervical cancer through vaccine development against human papilloma virus (HPV) and early immunization is a good example. However, those individuals already infected with HPV may need other options for controlling or eliminating disease. The delivery of siRNA against NFκB through “photothermal transfection” was used to successfully treat nude mice bearing HeLa cervical cancer xenografts (240). This involved using hollow gold nanospheres, to deliver siRNA in conjunction with near-infrared light irradiation to elicit a photothermal effect along with micro-positron emission tomography/computed tomography imaging (240). Engineering viral nanoparticles may also be effective in treating cervical cancer. The use of RNAi/RGD-based mimoretrovirus to target the Zbtb7 gene is one example (241). This mimoretrovirus exhibited excellent antitumor capacity in vivo in a nude mouse model of cervical cancer (241). The utility of using nanotools to target human HPV–infected tissues for preventive management or virus eradication remains to be fully tested.

Nanoparticle-mediated delivery is expected to limit the toxicity of chemopreventive agents while simultaneously enhancing bioavailability and sustained release. Various success stories continue to emerge involving the use of nanodelivery approaches. The application of nano-epigallocatechin-3-gallate (EGCG) is one example (242). EGCG encapsulated in polylactic acid–PEG nanoparticles enhanced the biologic effectiveness of EGCG at inducing apoptosis and reducing angiogenesis by 10 fold. The use of EGCG in a sustained food release setting has proven useful in a cancer nanochemoprevention setting (243).

Chemoprevention using nonsteroidal anti-inflammatory drugs (NSAID) has shown proven efficacy in treating high-risk colorectal cancer patients (244, 245). Cyclooxygenase-2 (COX-2) selective inhibitors, such as celecoxib, are associated with increased cardiovascular risk (246). Various attempts have been made to incorporate celecoxib into nanoparticles to control release and minimize GI and cardiovascular toxicity (150, 247–251). When applied to arthritis, the compartmentalized injection of a nanolipid–celecoxib formulation directly into joints enabled a localized and gradual sustained release of celecoxib, without any significant increase in cardiovascular drug levels (251). These nanoized formulations of celecoxib also show promise for treating cancer (250). Other investigators used an ibuprofen-conjugated phosphatidylcholine formulation to enhance GI safety and analgesic efficacy in osteoarthritic patients (252). Combinatorial nanoencapsulation of NSAIDs with other drugs may also be effective. For example, when aspirin, folic acid, and calcium (AFAC) were incorporated into nanoparticles, the combination greatly reduced the formation of premalignant aberrant crypt foci in the colon tissues of azoxymethane-treated rats (156). Encapsulation of various NSAIDs or combination treatments continues to progress as promising approaches for the future of nanochemoprevention.

Another example of a combinatorial nanoparticle approach has also had a major impact on pancreatic cancer prevention. Grandhi and colleagues generated an aspirin, curcumin, and sulforaphane (ACS) combination in solid liquid nanoparticles (SLN; ref. 253). These ACS/SLNs were used to perform multimodal targeting of pancreatic cancer. A hamster pancreatic cancer model was generated using N-nitrosobis (2-oxopropyl) amine (BOP) treatment that developed pancreatic intraepithelial neoplasms (PanIN) and tumors. Nanoencapsulated ACS regimens reduced tumor incidence by as high as 75% at doses 10-times lower than free-drug combinations (253).

Other success stories involve nanoencapsulation of proven chemopreventive agents that help solve poor bioavailability problems. Curcumin, the active component of turmeric, is a prime example. Polymeric nanoparticle–encapsulated curcumin (NanoCurc; ref. 254) inhibited tumor growth and systemic metastases in orthotopic pancreatic cancer xenograft models. This approach was also effective in delivering cocktails containing ACS to treat pancreatic cancer (253). Similarly, other forms of curcumin nanoparticles were effective in treating human lung tumor xenografts (255, 256). Enhancing bioavailability and the sustained release of poorly bioavailable compounds may serve as a successful formula for nanoprevention.

We are just beginning our journey into the realm of nanoprevention. A good first step is to clearly identify reasonable goals for this fledgling area of research. High-priority areas where an impact could be made will depend on the type of premalignancy targeted. Finding ways to effectively couple “-omics” into rapid profiling and selective targeting of nanotools for individualized treatment is an ambitious long-term goal. In the near term, improving theranostics based on more generalized molecular recognition signatures such as neoangiogenesis or viral infection may be a good start. For example, imaging and treatment of HPV as a high-risk factor for cervical or oral dysplasia/cancer by nanoparticle-mediated delivery of RNAi may be achievable. As a measurable outcome, this approach might effectively lead to complete clearance of HPV, thereby preventing subsequent disease. As another potential near-term goal, improving methods for nanoencapsulation might enhance the sustained localized release of RNAi or anti-inflammatory agents. For example, one might envision a macro–micro–nano–encapsulation approach that achieves targeted release at the appropriate site in the GI tract. For the field to be successful, it is also critical to not only identify realizable goals but also potential barriers. Ultimately, we must merge the best ideas in prevention with the most effective and least risky nanotechnology.

No potential conflicts of interest were disclosed.

The authors thank Karen L. Colbert for editorial improvements in this article.

This research was supported in part by the Boone Pickens Distinguished Chair for Early Prevention of Cancer (to E.T. Hawk) and from CPRIT RP100969 (to D.G. Menter). Additional support included 5U54CA151668-03 (to D.G. Menter, C.D. Logsdon, and A.K. Sood) and Cancer Center Support grant 5P30CA016672-37 from the NIH.

1.
Feynman
R
. 
There's Plenty of Room at the Bottom
.
Eng Sci
1960
;
23
:
5
.
2.
Drexler
E
. 
Nanosystems: Molecular Machinery, Manufacturing and Computation
; 
1992
.
3.
Taniguchi
N.
On the basic concept of ‘Nano-Technology’.
Proc. Intl. Conf. Prod. Eng. Tokyo, Part II, Japan Society of Precision Engineering
, 
1974
;
5
10
.
4.
Binnig
B
,
Rohrer
H
,
Gerber
C
,
Weibel
E
. 
Tunneling through a controllable vacuum gap
.
Appl Phys Lett
1982
;
40
:
178
81
.
5.
Kroto
H
,
Heath
J
,
O'Brien
S
,
Curl
R
,
Smalley
R
. 
C60: Buckminsterfullerene
.
Nature
1985
;
318
:
162
3
.
6.
Higgins
P
,
Dawson
J
,
Walters
M
. 
Nanomedicine: Nanotubes reduce stroke damage
.
Nat Nanotechnol
2011
;
6
:
83
4
.
7.
Ohno
H
,
Kobayashi
T
,
Kabata
R
,
Endo
K
,
Iwasa
T
,
Yoshimura
SH
, et al
Synthetic RNA-protein complex shaped like an equilateral triangle
.
Nat Nanotechnol
2011
;
6
:
116
20
.
8.
Guo
P
. 
The emerging field of RNA nanotechnology
.
Nat Nanotechnol
2010
;
5
:
833
42
.
9.
Zemp
RJ
. 
Nanomedicine: detecting rare cancer cells
.
Nat Nanotechnol
2009
;
4
:
798
9
.
10.
Sanhai
WR
,
Sakamoto
JH
,
Canady
R
,
Ferrari
M
. 
Seven challenges for nanomedicine
.
Nat Nanotechnol
2008
;
3
:
242
4
.
11.
Han
HD
,
Mora
EM
,
Roh
JW
,
Nishimura
M
,
Lee
SJ
,
Stone
RL
, et al
Chitosan hydrogel for localized gene silencing
.
Cancer Biol Ther
2011
;
11
:
839
45
.
12.
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
.
13.
Mann
AP
,
Tanaka
T
,
Somasunderam
A
,
Liu
X
,
Gorenstein
DG
,
Ferrari
M
. 
E-selectin-targeted porous silicon particle for nanoparticle delivery to the bone marrow
.
Adv Mater
2011
;
23
:
H278
82
.
14.
Petrenko
VA
,
Jayanna
PK
. 
Phage protein-targeted cancer nanomedicines
.
FEBS Lett
2014
;
588
:
341
9
.
15.
Loi
M
,
Di Paolo
D
,
Soster
M
,
Brignole
C
,
Bartolini
A
,
Emionite
L
, et al
Novel phage display-derived neuroblastoma-targeting peptides potentiate the effect of drug nanocarriers in preclinical settings
.
J Control Release
2013
;
170
:
233
41
.
16.
Xie
X
,
Xia
W
,
Li
Z
,
Kuo
HP
,
Liu
Y
,
Ding
Q
, et al
Targeted expression of BikDD eradicates pancreatic tumors in noninvasive imaging models
.
Cancer Cell
2007
;
12
:
52
65
.
17.
Li
Z
,
Ding
Q
,
Li
Y
,
Miller
SA
,
Abbruzzese
JL
,
Hung
MC
. 
Suppression of pancreatic tumor progression by systemic delivery of a pancreatic-cancer-specific promoter driven Bik mutant
.
Cancer Lett
2006
;
236
:
58
63
.
18.
Lopez
MV
,
Rivera
AA
,
Viale
DL
,
Benedetti
L
,
Cuneo
N
,
Kimball
KJ
, et al
A tumor-stroma targeted oncolytic adenovirus replicated in human ovary cancer samples and inhibited growth of disseminated solid tumors in mice
.
Mol Ther
2012
;
20
:
2222
33
.
19.
Sarkar
D
,
Su
ZZ
,
Park
ES
,
Vozhilla
N
,
Dent
P
,
Curiel
DT
, et al
A cancer terminator virus eradicates both primary and distant human melanomas
.
Cancer Gene Ther
2008
;
15
:
293
302
.
20.
Ulasov
IV
,
Zhu
ZB
,
Tyler
MA
,
Han
Y
,
Rivera
AA
,
Khramtsov
A
, et al
Survivin-driven and fiber-modified oncolytic adenovirus exhibits potent antitumor activity in established intracranial glioma
.
Hum Gene Ther
2007
;
18
:
589
602
.
21.
Buzea
C
,
Pacheco
II
,
Robbie
K
. 
Nanomaterials and nanoparticles: sources and toxicity
.
Biointerphases
2007
;
2
:
MR17
71
.
22.
Chen
D
,
Liu
W
,
Shen
Y
,
Mu
H
,
Zhang
Y
,
Liang
R
, et al
Effects of a novel pH-sensitive liposome with cleavable esterase-catalyzed and pH-responsive double smart mPEG lipid derivative on ABC phenomenon
.
Int J Nanomedicine
2011
;
6
:
2053
61
.
23.
Riviere
JE
. 
Pharmacokinetics of nanomaterials: an overview of carbon nanotubes, fullerenes and quantum dots
.
Wiley Interdiscip Rev Nanomed Nanobiotechnol
2009
;
1
:
26
34
.
24.
Sarlo
K
,
Blackburn
KL
,
Clark
ED
,
Grothaus
J
,
Chaney
J
,
Neu
S
, et al
Tissue distribution of 20 nm, 100 nm and 1000 nm fluorescent polystyrene latex nanospheres following acute systemic or acute and repeat airway exposure in the rat
.
Toxicology
2009
;
263
:
117
26
.
25.
Toll
R
,
Jacobi
U
,
Richter
H
,
Lademann
J
,
Schaefer
H
,
Blume-Peytavi
U
. 
Penetration profile of microspheres in follicular targeting of terminal hair follicles
.
J Invest Dermatol
2004
;
123
:
168
76
.
26.
Tinkle
SS
,
Antonini
JM
,
Rich
BA
,
Roberts
JR
,
Salmen
R
,
DePree
K
, et al
Skin as a route of exposure and sensitization in chronic beryllium disease
.
Environ Health Perspect
2003
;
111
:
1202
8
.
27.
Oberdorster
G
,
Oberdorster
E
,
Oberdorster
J
. 
Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles
.
Environ Health Perspect
2005
;
113
:
823
39
.
28.
Tsuji
JS
,
Maynard
AD
,
Howard
PC
,
James
JT
,
Lam
CW
,
Warheit
DB
, et al
Research strategies for safety evaluation of nanomaterials, part IV: risk assessment of nanoparticles
.
Toxicol Sci
2006
;
89
:
42
50
.
29.
Bennat
C
,
Muller-Goymann
CC
. 
Skin penetration and stabilization of formulations containing microfine titanium dioxide as physical UV filter
.
Int J Cosmet Sci
2000
;
22
:
271
83
.
30.
Jani
P
,
Halbert
GW
,
Langridge
J
,
Florence
AT
. 
Nanoparticle uptake by the rat gastrointestinal mucosa: quantitation and particle size dependency
.
J Pharm Pharmacol
1990
;
42
:
821
6
.
31.
Hoet
PH
,
Bruske-Hohlfeld
I
,
Salata
OV
. 
Nanoparticles - known and unknown health risks
.
J Nanobiotechnology
2004
;
2
:
12
.
32.
Lomer
MC
,
Cook
WB
,
Jan-Mohamed
HJ
,
Hutchinson
C
,
Liu
DY
,
Hider
RC
, et al
Iron requirements based upon iron absorption tests are poorly predicted by haematological indices in patients with inactive inflammatory bowel disease
.
Br J Nutr
2012
;
107
:
1806
11
.
33.
Choi
HS
,
Ashitate
Y
,
Lee
JH
,
Kim
SH
,
Matsui
A
,
Insin
N
, et al
Rapid translocation of nanoparticles from the lung airspaces to the body
.
Nature biotechnology
2010
;
28
:
1300
3
.
34.
Davis
ME
,
Chen
ZG
,
Shin
DM
. 
Nanoparticle therapeutics: an emerging treatment modality for cancer
.
Nat Rev Drug Discov
2008
;
7
:
771
82
.
35.
Adriani
G
,
de Tullio
MD
,
Ferrari
M
,
Hussain
F
,
Pascazio
G
,
Liu
X
, et al
The preferential targeting of the diseased microvasculature by disk-like particles
.
Biomaterials
2012
;
33
:
5504
13
.
36.
Boso
DP
,
Lee
SY
,
Ferrari
M
,
Schrefler
BA
,
Decuzzi
P
. 
Optimizing particle size for targeting diseased microvasculature: from experiments to artificial neural networks
.
Int J Nanomedicine
2011
;
6
:
1517
26
.
37.
Batist
G
,
Barton
J
,
Chaikin
P
,
Swenson
C
,
Welles
L
. 
Myocet (liposome-encapsulated doxorubicin citrate): a new approach in breast cancer therapy
.
Expert Opin Pharmacother
2002
;
3
:
1739
51
.
38.
Cabriales
S
,
Bresnahan
J
,
Testa
D
,
Espina
BM
,
Scadden
DT
,
Ross
M
, et al
Extravasation of liposomal daunorubicin in patients with AIDS-associated Kaposi's sarcoma: a report of four cases
.
Oncol Nurs Forum
1998
;
25
:
67
70
.
39.
Gelmon
KA
,
Tolcher
A
,
Diab
AR
,
Bally
MB
,
Embree
L
,
Hudon
N
, et al
Phase I study of liposomal vincristine
.
J Clin Oncol
1999
;
17
:
697
705
.
40.
Glantz
MJ
,
LaFollette
S
,
Jaeckle
KA
,
Shapiro
W
,
Swinnen
L
,
Rozental
JR
, et al
Randomized trial of a slow-release versus a standard formulation of cytarabine for the intrathecal treatment of lymphomatous meningitis
.
J Clin Oncol
1999
;
17
:
3110
6
.
41.
Chen
KJ
,
Chaung
EY
,
Wey
SP
,
Lin
KJ
,
Cheng
F
,
Lin
CC
, et al
Hyperthermia-mediated local drug delivery by a bubble-generating liposomal system for tumor-specific chemotherapy
.
ACS Nano
2014
;
8
:
5105
15
.
42.
Hengge
UR
,
Brockmeyer
NH
,
Baumann
M
,
Reimann
G
,
Goos
M
. 
Liposomal doxorubicin in AIDS-related Kaposi's sarcoma
.
Lancet
1993
;
342
:
497
.
43.
Stathopoulos
GP
,
Boulikas
T
,
Vougiouka
M
,
Deliconstantinos
G
,
Rigatos
S
,
Darli
E
, et al
Pharmacokinetics and adverse reactions of a new liposomal cisplatin (Lipoplatin): phase I study
.
Oncol Rep
2005
;
13
:
589
95
.
44.
Pramanik
D
,
Campbell
NR
,
Das
S
,
Gupta
S
,
Chenna
V
,
Bisht
S
, et al
A composite polymer nanoparticle overcomes multidrug resistance and ameliorates doxorubicin-associated cardiomyopathy
.
Oncotarget
2012
;
3
:
640
50
.
45.
Coelho
T
,
Adams
D
,
Silva
A
,
Lozeron
P
,
Hawkins
PN
,
Mant
T
, et al
Safety and efficacy of RNAi therapy for transthyretin amyloidosis
.
N Engl J Med
2013
;
369
:
819
29
.
46.
Fitzgerald
K
,
Frank-Kamenetsky
M
,
Shulga-Morskaya
S
,
Liebow
A
,
Bettencourt
BR
,
Sutherland
JE
, et al
Effect of an RNA interference drug on the synthesis of proprotein convertase subtilisin/kexin type 9 (PCSK9) and the concentration of serum LDL cholesterol in healthy volunteers: a randomised, single-blind, placebo-controlled, phase 1 trial
.
Lancet
2014
;
383
:
60
8
.
47.
Dahlman
JE
,
Barnes
C
,
Khan
O
,
Thiriot
A
,
Jhunjunwala
S
,
Shaw
TE
, et al
In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight
.
Nat Nanotechnol
2014
;
9
:
648
55
.
48.
Moreno-Aspitia
A
,
Perez
EA
. 
Nanoparticle albumin-bound paclitaxel (ABI-007): a newer taxane alternative in breast cancer
.
Future Oncol
2005
;
1
:
755
62
.
49.
Ettinger
LJ
,
Kurtzberg
J
,
Voute
PA
,
Jurgens
H
,
Halpern
SL
. 
An open-label, multicenter study of polyethylene glycol-L-asparaginase for the treatment of acute lymphoblastic leukemia
.
Cancer
1995
;
75
:
1176
81
.
50.
Kim
TY
,
Kim
DW
,
Chung
JY
,
Shin
SG
,
Kim
SC
,
Heo
DS
, et al
Phase I and pharmacokinetic study of Genexol-PM, a cremophor-free, polymeric micelle-formulated paclitaxel, in patients with advanced malignancies
.
Clin Cancer Res
2004
;
10
:
3708
16
.
51.
Nowotnik
DP
,
Cvitkovic
E
. 
ProLindac (AP5346): a review of the development of an HPMA DACH platinum Polymer Therapeutic
.
Adv Drug Deliv Rev
2009
;
61
:
1214
9
.
52.
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 Translational Med
2012
;
4
:
128ra39
.
53.
Wuolijoki
E
,
Hirvela
T
,
Ylitalo
P
. 
Decrease in serum LDL cholesterol with microcrystalline chitosan
.
Methods Find Exp Clin Pharmacol
1999
;
21
:
357
61
.
54.
Howard
KA
,
Rahbek
UL
,
Liu
X
,
Damgaard
CK
,
Glud
SZ
,
Andersen
MO
, et al
RNA interference in vitro and in vivo using a novel chitosan/siRNA nanoparticle system
.
Mol Ther
2006
;
14
:
476
84
.
55.
Li
X
,
Ferrel
GL
,
Guerra
MC
,
Hode
T
,
Lunn
JA
,
Adalsteinsson
O
, et al
Preliminary safety and efficacy results of laser immunotherapy for the treatment of metastatic breast cancer patients
.
Photochem Photobiol Sci
2011
;
10
:
817
21
.
56.
Roy
K
,
Mao
HQ
,
Huang
SK
,
Leong
KW
. 
Oral gene delivery with chitosan–DNA nanoparticles generates immunologic protection in a murine model of peanut allergy
.
Nat Med
1999
;
5
:
387
91
.
57.
Chen
C
,
Zhang
H
,
Hou
L
,
Shi
J
,
Wang
L
,
Zhang
C
, et al
Single-walled carbon nanotubes mediated neovascularity targeted antitumor drug delivery system
.
J Pharm Pharm Sci
2013
;
16
:
40
51
.
58.
Injac
R
,
Perse
M
,
Obermajer
N
,
Djordjevic-Milic
V
,
Prijatelj
M
,
Djordjevic
A
, et al
Potential hepatoprotective effects of fullerenol C60(OH)24 in doxorubicin-induced hepatotoxicity in rats with mammary carcinomas
.
Biomaterials
2008
;
29
:
3451
60
.
59.
Kwag
DS
,
Park
K
,
Oh
KT
,
Lee
ES
. 
Hyaluronated fullerenes with photoluminescent and antitumoral activity
.
Chem Commun
2013
;
49
:
282
4
.
60.
Ren
J
,
Shen
S
,
Wang
D
,
Xi
Z
,
Guo
L
,
Pang
Z
, et al
The targeted delivery of anticancer drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with angiopep-2
.
Biomaterials
2012
;
33
:
3324
33
.
61.
Sacchetti
C
,
Rapini
N
,
Magrini
A
,
Cirelli
E
,
Bellucci
S
,
Mattei
M
, et al
In vivo targeting of intratumor regulatory T cells using PEG-modified single-walled carbon nanotubes
.
Bioconjug Chem
2013
;
24
:
852
8
.
62.
Shao
W
,
Paul
A
,
Zhao
B
,
Lee
C
,
Rodes
L
,
Prakash
S
. 
Carbon nanotube lipid drug approach for targeted delivery of a chemotherapy drug in a human breast cancer xenograft animal model
.
Biomaterials
2013
;
34
:
10109
19
.
63.
Shi
J
,
Zhang
H
,
Wang
L
,
Li
L
,
Wang
H
,
Wang
Z
, et al
PEI-derivatized fullerene drug delivery using folate as a homing device targeting to tumor
.
Biomaterials
2013
;
34
:
251
61
.
64.
Benezra
M
,
Penate-Medina
O
,
Zanzonico
PB
,
Schaer
D
,
Ow
H
,
Burns
A
, et al
Multimodal silica nanoparticles are effective cancer-targeted probes in a model of human melanoma
.
J Clin Invest
2011
;
121
:
2768
80
.
65.
Shen
H
,
Rodriguez-Aguayo
C
,
Xu
R
,
Gonzalez-Villasana
V
,
Mai
J
,
Huang
Y
, et al
Enhancing chemotherapy response with sustained EphA2 silencing using multistage vector delivery
.
Clin Cancer Res
2013
;
19
:
1806
15
.
66.
Wu
SH
,
Lin
YS
,
Hung
Y
,
Chou
YH
,
Hsu
YH
,
Chang
C
, et al
Multifunctional mesoporous silica nanoparticles for intracellular labeling and animal magnetic resonance imaging studies
.
Chembiochem
2008
;
9
:
53
7
.
67.
Fatehi
D
,
Baral
TN
,
Abulrob
A
. 
In vivo imaging of brain cancer using epidermal growth factor single domain antibody bioconjugated to near-infrared quantum dots
.
J Nanosci Nanotechnol
2014
;
14
:
5355
62
.
68.
Kwon
H
,
Lee
J
,
Song
R
,
Hwang
SI
,
Lee
J
,
Kim
YH
, et al
In vitro and in vivo imaging of prostate cancer angiogenesis using anti-vascular endothelial growth factor receptor 2 antibody-conjugated quantum dot
.
Korean J Radiol
2013
;
14
:
30
7
.
69.
Savla
R
,
Taratula
O
,
Garbuzenko
O
,
Minko
T
. 
Tumor targeted quantum dot-mucin 1 aptamer-doxorubicin conjugate for imaging and treatment of cancer
.
J Control Release
2011
;
153
:
16
22
.
70.
Arvizo
RR
,
Saha
S
,
Wang
E
,
Robertson
JD
,
Bhattacharya
R
,
Mukherjee
P
. 
Inhibition of tumor growth and metastasis by a self-therapeutic nanoparticle
.
Proc Natl Acad Sci U S A
2013
;
110
:
6700
5
.
71.
Bobyk
L
,
Edouard
M
,
Deman
P
,
Vautrin
M
,
Pernet-Gallay
K
,
Delaroche
J
, et al
Photoactivation of gold nanoparticles for glioma treatment
.
Nanomedicine
2013
;
9
:
1089
97
.
72.
Li
FR
,
Li
Q
,
Zhou
HX
,
Qi
H
,
Deng
CY
. 
Detection of circulating tumor cells in breast cancer with a refined immunomagnetic nanoparticle enriched assay and nested-RT-PCR
.
Nanomedicine
2013
;
9
:
1106
13
.
73.
Shenoi
MM
,
Iltis
I
,
Choi
J
,
Koonce
NA
,
Metzger
GJ
,
Griffin
RJ
, et al
Nanoparticle delivered vascular disrupting agents (VDAs): use of TNF-alpha conjugated gold nanoparticles for multimodal cancer therapy
.
Mol Pharm
2013
;
10
:
1683
94
.
74.
Jain
S
,
Hirst
DG
,
O'Sullivan
JM
. 
Gold nanoparticles as novel agents for cancer therapy
.
Br J Radiol
2012
;
85
:
101
13
.
75.
Heath
JR
,
Davis
ME
. 
Nanotechnology and cancer
.
Annu Rev Med
2008
;
59
:
251
65
.
76.
Chauhan
VP
,
Jain
RK
. 
Strategies for advancing cancer nanomedicine
.
Nat Mater
2013
;
12
:
958
62
.
77.
Otterson
GA
,
Villalona-Calero
MA
,
Hicks
W
,
Pan
X
,
Ellerton
JA
,
Gettinger
SN
, et al
Phase I/II study of inhaled doxorubicin combined with platinum-based therapy for advanced non-small cell lung cancer
.
Clin Cancer Res
2010
;
16
:
2466
73
.
78.
Lam
S
,
leRiche
JC
,
McWilliams
A
,
Macaulay
C
,
Dyachkova
Y
,
Szabo
E
, et al
A randomized phase IIb trial of pulmicort turbuhaler (budesonide) in people with dysplasia of the bronchial epithelium
.
Clin Cancer Res
2004
;
10
:
6502
11
.
79.
Dahl
AR
,
Grossi
IM
,
Houchens
DP
,
Scovell
LJ
,
Placke
ME
,
Imondi
AR
, et al
Inhaled isotretinoin (13-cis retinoic acid) is an effective lung cancer chemopreventive agent in A/J mice at low doses: a pilot study
.
Clin Cancer Res
2000
;
6
:
3015
24
.
80.
Mulshine
JL
,
Hirsch
FR
. 
Lung cancer chemoprevention: moving from concept to a reality
.
Lung cancer
2003
;
41
:
S163
74
.
81.
Kammona
O
,
Kiparissides
C
. 
Recent advances in nanocarrier-based mucosal delivery of biomolecules
.
J Control Release
2012
;
161
:
781
94
.
82.
Manke
A
,
Wang
L
,
Rojanasakul
Y
. 
Mechanisms of nanoparticle-induced oxidative stress and toxicity
.
Bio Med Res Int
2013
;
2013
:
942916
.
83.
Su
CK
,
Sun
YC
. 
In vivo monitoring of distributional transport kinetics and extravasation of quantum dots in living rat liver
.
Nanotechnology
2013
;
24
:
165101
.
84.
Menter
DG
,
Tucker
SC
,
Kopetz
S
,
Sood
AK
,
Crissman
JD
,
Honn
KV
. 
Platelets and cancer: a casual or causal relationship: revisited
.
Cancer Metastasis Rev
2014 Apr 3
.
[Epub ahead of print]
.
85.
Ilinskaya
AN
,
Dobrovolskaia
MA
. 
Nanoparticles and the blood coagulation system. Part I: benefits of nanotechnology
.
Nanomedicine
2013
;
8
:
773
84
.
86.
Samuel
SP
,
Jain
N
,
O'Dowd
F
,
Paul
T
,
Kashanin
D
,
Gerard
VA
, et al
Multifactorial determinants that govern nanoparticle uptake by human endothelial cells under flow
.
Int J Nanomedicine
2012
;
7
:
2943
56
.
87.
De Paoli
SH
,
Diduch
LL
,
Tegegn
TZ
,
Orecna
M
,
Strader
MB
,
Karnaukhova
E
, et al
The effect of protein corona composition on the interaction of carbon nanotubes with human blood platelets
.
Biomaterials
2014
;
35
:
6182
94
.
88.
Grozovsky
R
,
Hoffmeister
KM
,
Falet
H
. 
Novel clearance mechanisms of platelets
.
Curr Opin Hematol
2010
;
17
:
585
9
.
89.
Fadeel
B
. 
Clear and present danger? Engineered nanoparticles and the immune system
.
Swiss Med Wkly
2012
;
142
:
w13609
.
90.
Peng
Q
,
Zhang
S
,
Yang
Q
,
Zhang
T
,
Wei
XQ
,
Jiang
L
, et al
Preformed albumin corona, a protective coating for nanoparticles based drug delivery system
.
Biomaterials
2013
;
34
:
8521
30
.
91.
Sarkar
A
,
Ghosh
M
,
Sil
PC
. 
Nanotoxicity: oxidative stress mediated toxicity of metal and metal oxide nanoparticles
.
J Nanosci Nanotechnol
2014
;
14
:
730
43
.
92.
Fu
PP
,
Xia
Q
,
Hwang
HM
,
Ray
PC
,
Yu
H
. 
Mechanisms of nanotoxicity: generation of reactive oxygen species
.
J Food Drug Anal
2014
;
22
:
64
75
.
93.
Seabra
AB
,
Paula
AJ
,
de Lima
R
,
Alves
OL
,
Duran
N
. 
Nanotoxicity of graphene and graphene oxide
.
Chem Res Toxicol
2014
;
27
:
159
68
.
94.
Li
J
,
Chang
X
,
Chen
X
,
Gu
Z
,
Zhao
F
,
Chai
Z
, et al
Toxicity of inorganic nanomaterials in biomedical imaging
.
Biotechnol Adv
2014
;
32
:
727
43
.
95.
McCarthy
DP
,
Hunter
ZN
,
Chackerian
B
,
Shea
LD
,
Miller
SD
. 
Targeted immunomodulation using antigen-conjugated nanoparticles
.
Wiley Interdiscip Rev Nanomed Nanobiotechnol
2014
;
6
:
298
315
.
96.
Fang
RH
,
Hu
CM
,
Luk
BT
,
Gao
W
,
Copp
JA
,
Tai
Y
, et al
Cancer cell membrane-coated nanoparticles for anticancer vaccination and drug delivery
.
Nano Lett
2014
;
14
:
2181
8
.
97.
Abdelhalim
MA
. 
Uptake of gold nanoparticles in several rat organs after intraperitoneal administration in vivo: a fluorescence study
.
BioMed Res Int
2013
;
2013
:
353695
.
98.
Hallaj-Nezhadi
S
,
Dass
CR
,
Lotfipour
F
. 
Intraperitoneal delivery of nanoparticles for cancer gene therapy
.
Future Oncol
2013
;
9
:
59
68
.
99.
Nakamura
M
,
Miyamoto
K
,
Hayashi
K
,
Awaad
A
,
Ochiai
M
,
Ishimura
K
. 
Time-lapse fluorescence imaging and quantitative single cell and endosomal analysis of peritoneal macrophages using fluorescent organosilica nanoparticles
.
Nanomedicine
2013
;
9
:
274
83
.
100.
Sarhan
OM
,
Hussein
RM
. 
Effects of intraperitoneally injected silver nanoparticles on histological structures and blood parameters in the albino rat
.
Int J Nanomedicine
2014
;
9
:
1505
17
.
101.
Arvizo
RR
,
Miranda
OR
,
Moyano
DF
,
Walden
CA
,
Giri
K
,
Bhattacharya
R
, et al
Modulating pharmacokinetics, tumor uptake and biodistribution by engineered nanoparticles
.
PLoS ONE
2011
;
6
:
e24374
.
102.
Zhang
Y
,
Bai
Y
,
Jia
J
,
Gao
N
,
Li
Y
,
Zhang
R
, et al
Perturbation of physiological systems by nanoparticles
.
Chem Soc Rev
2014
;
43
:
3762
809
.
103.
Yang
W
,
Peters
JI
,
Williams
RO
 III
. 
Inhaled nanoparticles–a current review
.
Int J Pharm
2008
;
356
:
239
47
.
104.
Roberts
RA
,
Shen
T
,
Allen
IC
,
Hasan
W
,
DeSimone
JM
,
Ting
JP
. 
Analysis of the murine immune response to pulmonary delivery of precisely fabricated nano- and microscale particles
.
PLoS ONE
2013
;
8
:
e62115
.
105.
Schleh
C
,
Rothen-Rutishauser
B
,
Kreyling
WG
. 
The influence of pulmonary surfactant on nanoparticulate drug delivery systems
.
Eur J Pharm Biopharm
2011
;
77
:
350
2
.
106.
Naota
M
,
Shimada
A
,
Morita
T
,
Yamamoto
Y
,
Inoue
K
,
Takano
H
. 
Caveolae-mediated endocytosis of intratracheally instilled gold colloid nanoparticles at the air-blood barrier in mice
.
Toxicol Pathol
2013
;
41
:
487
96
.
107.
Muhlfeld
C
,
Rothen-Rutishauser
B
,
Blank
F
,
Vanhecke
D
,
Ochs
M
,
Gehr
P
. 
Interactions of nanoparticles with pulmonary structures and cellular responses
.
Am J Physiol Lung Cell Mol Physiol
2008
;
294
:
L817
29
.
108.
Reisetter
AC
,
Stebounova
LV
,
Baltrusaitis
J
,
Powers
L
,
Gupta
A
,
Grassian
VH
, et al
Induction of inflammasome-dependent pyroptosis by carbon black nanoparticles
.
J Biol Chem
2011
;
286
:
21844
52
.
109.
Kolanjiyil
AV
,
Kleinstreuer
C
. 
Nanoparticle mass transfer from lung airways to systemic regions–Part I: Whole-lung aerosol dynamics
.
J Biomech Eng
2013
;
135
:
121003
.
110.
Zhu
MT
,
Feng
WY
,
Wang
B
,
Wang
TC
,
Gu
YQ
,
Wang
M
, et al
Comparative study of pulmonary responses to nano- and submicron-sized ferric oxide in rats
.
Toxicology
2008
;
247
:
102
11
.
111.
Jones
MC
,
Jones
SA
,
Riffo-Vasquez
Y
,
Spina
D
,
Hoffman
E
,
Morgan
A
, et al
Quantitative assessment of nanoparticle surface hydrophobicity and its influence on pulmonary biocompatibility
.
J Control Release
2014
;
183
:
94
104
.
112.
Zarogoulidis
P
,
Chatzaki
E
,
Porpodis
K
,
Domvri
K
,
Hohenforst-Schmidt
W
,
Goldberg
EP
, et al
Inhaled chemotherapy in lung cancer: future concept of nanomedicine
.
Int J Nanomedicine
2012
;
7
:
1551
72
.
113.
Garg
T
,
Rath
G
,
Goyal
AK
. 
Comprehensive review on additives of topical dosage forms for drug delivery
.
Drug Deliv
2014 Jan 23
.
[Epub ahead of print]
.
114.
DeLouise
LA
. 
Applications of nanotechnology in dermatology
.
J Invest Dermatol
2012
;
132
:
964
75
.
115.
Konieczny
P
,
Goralczyk
AG
,
Szmyd
R
,
Skalniak
L
,
Koziel
J
,
Filon
FL
, et al
Effects triggered by platinum nanoparticles on primary keratinocytes
.
Int J Nanomedicine
2013
;
8
:
3963
75
.
116.
Kaur
J
,
Tikoo
K
. 
Evaluating cell specific cytotoxicity of differentially charged silver nanoparticles
.
Food Chem Toxicol
2013
;
51
:
1
14
.
117.
Hackenberg
S
,
Kleinsasser
N
. 
Dermal toxicity of ZnO nanoparticles: a worrying feature of sunscreen
?
Nanomedicine
2012
;
7
:
461
3
.
118.
Tang
L
,
Zhang
C
,
Song
G
,
Jin
X
,
Xu
Z
. 
In vivo skin penetration and metabolic path of quantum dots
.
Sci China Life Sci
2013
;
56
:
181
8
.
119.
Yu
KN
,
Yoon
TJ
,
Minai-Tehrani
A
,
Kim
JE
,
Park
SJ
,
Jeong
MS
, et al
Zinc oxide nanoparticle induced autophagic cell death and mitochondrial damage via reactive oxygen species generation
.
Toxicol In Vitro
2013
;
27
:
1187
95
.
120.
Vandebriel
RJ
,
De Jong
WH
. 
A review of mammalian toxicity of ZnO nanoparticles
.
Nanotechnology Sci Appl
2012
;
5
:
61
71
.
121.
Gao
W
,
Vecchio
D
,
Li
J
,
Zhu
J
,
Zhang
Q
,
Fu
V
, et al
Hydrogel containing nanoparticle-stabilized liposomes for topical antimicrobial delivery
.
ACS Nano
2014
;
8
:
2900
7
.
122.
Eke
G
,
Kuzmina
AM
,
Goreva
AV
,
Shishatskaya
EI
,
Hasirci
N
,
Hasirci
V
. 
In vitro and transdermal penetration of PHBV micro/nanoparticles
.
J MatSsc Mat Med
2014
;
25
:
1471
81
.
123.
Cho
HK
,
Cho
JH
,
Jeong
SH
,
Cho
DC
,
Yeum
JH
,
Cheong
IW
. 
Polymeric vehicles for topical delivery and related analytical methods
.
Arch Pharm Res
2014
;
37
:
423
34
.
124.
Wadajkar
AS
,
Bhavsar
Z
,
Ko
CY
,
Koppolu
B
,
Cui
W
,
Tang
L
, et al
Multifunctional particles for melanoma-targeted drug delivery
.
Acta Biomaterialia
2012
;
8
:
2996
3004
.
125.
Yun
Y
,
Cho
YW
,
Park
K
. 
Nanoparticles for oral delivery: targeted nanoparticles with peptidic ligands for oral protein delivery
.
Adv Drug Deliv Rev
2013
;
65
:
822
32
.
126.
Landsiedel
R
,
Fabian
E
,
Ma-Hock
L
,
van Ravenzwaay
B
,
Wohlleben
W
,
Wiench
K
, et al
Toxico-/biokinetics of nanomaterials
.
Arch Toxicol
2012
;
86
:
1021
60
.
127.
Wang
H
,
Du
LJ
,
Song
ZM
,
Chen
XX
. 
Progress in the characterization and safety evaluation of engineered inorganic nanomaterials in food
.
Nanomedicine
2013
;
8
:
2007
25
.
128.
Bernkop-Schnurch
A
. 
Nanocarrier systems for oral drug delivery: do we really need them?
Eur J Pharm Sci
2013
;
49
:
272
7
.
129.
Shahbazi
MA
,
Santos
HA
. 
Improving oral absorption via drug-loaded nanocarriers: absorption mechanisms, intestinal models and rational fabrication
.
Curr Drug Metab
2013
;
14
:
28
56
.
130.
Feng
C
,
Wang
Z
,
Jiang
C
,
Kong
M
,
Zhou
X
,
Li
Y
, et al
Chitosan/o-carboxymethyl chitosan nanoparticles for efficient and safe oral anticancer drug delivery: in vitro and in vivo evaluation
.
Int J Pharm
2013
;
457
:
158
67
.
131.
Kim
SH
,
Lee
KY
,
Jang
YS
. 
Mucosal immune system and M cell-targeting strategies for oral mucosal vaccination
.
Immune Netw
2012
;
12
:
165
75
.
132.
Knoop
KA
,
Miller
MJ
,
Newberry
RD
. 
Transepithelial antigen delivery in the small intestine: different paths, different outcomes
.
Curr Opin Gastroenterol
2013
;
29
:
112
8
.
133.
Menard
S
,
Cerf-Bensussan
N
,
Heyman
M
. 
Multiple facets of intestinal permeability and epithelial handling of dietary antigens
.
Mucosal Immunol
2010
;
3
:
247
59
.
134.
Schulz
O
,
Pabst
O
. 
Antigen sampling in the small intestine
.
Trends Immunol
2013
;
34
:
155
61
.
135.
Crater
JS
,
Carrier
RL
. 
Barrier properties of gastrointestinal mucus to nanoparticle transport
.
Macromolecular Biosci
2010
;
10
:
1473
83
.
136.
Bergin
IL
,
Witzmann
FA
. 
Nanoparticle toxicity by the gastrointestinal route: evidence and knowledge gaps
.
Int J Biomed Nanosci Nanotechnol
2013
;
3
:
1
2
.
137.
Kunisawa
J
,
Kurashima
Y
,
Kiyono
H
. 
Gut-associated lymphoid tissues for the development of oral vaccines
.
Adv Drug Deliv Rev
2012
;
64
:
523
30
.
138.
Mallegol
J
,
Van Niel
G
,
Lebreton
C
,
Lepelletier
Y
,
Candalh
C
,
Dugave
C
, et al
T84-intestinal epithelial exosomes bear MHC class II/peptide complexes potentiating antigen presentation by dendritic cells
.
Gastroenterology
2007
;
132
:
1866
76
.
139.
Yoshida
M
,
Claypool
SM
,
Wagner
JS
,
Mizoguchi
E
,
Mizoguchi
A
,
Roopenian
DC
, et al
Human neonatal Fc receptor mediates transport of IgG into luminal secretions for delivery of antigens to mucosal dendritic cells
.
Immunity
2004
;
20
:
769
83
.
140.
Faust
JJ
,
Masserano
BM
,
Mielke
AH
,
Abraham
A
,
Capco
DG
. 
Engineered nanoparticles induced brush border disruption in a human model of the intestinal epithelium
.
Adv Exp Med Biol
2014
;
811
:
55
72
.
141.
Arques
JL
,
Hautefort
I
,
Ivory
K
,
Bertelli
E
,
Regoli
M
,
Clare
S
, et al
Salmonella induces flagellin- and MyD88-dependent migration of bacteria-capturing dendritic cells into the gut lumen
.
Gastroenterology
2009
;
137
:
579
87
,
87 e1
2
.
142.
McDole
JR
,
Wheeler
LW
,
McDonald
KG
,
Wang
B
,
Konjufca
V
,
Knoop
KA
, et al
Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine
.
Nature
2012
;
483
:
345
9
.
143.
Rescigno
M
,
Urbano
M
,
Valzasina
B
,
Francolini
M
,
Rotta
G
,
Bonasio
R
, et al
Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria
.
Nat Immunol
2001
;
2
:
361
7
.
144.
Niess
JH
,
Brand
S
,
Gu
X
,
Landsman
L
,
Jung
S
,
McCormick
BA
, et al
CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance
.
Science
2005
;
307
:
254
8
.
145.
Chieppa
M
,
Rescigno
M
,
Huang
AY
,
Germain
RN
. 
Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement
.
J Exp Med
2006
;
203
:
2841
52
.
146.
Van Niel
G
,
Mallegol
J
,
Bevilacqua
C
,
Candalh
C
,
Brugiere
S
,
Tomaskovic-Crook
E
, et al
Intestinal epithelial exosomes carry MHC class II/peptides able to inform the immune system in mice
.
Gut
2003
;
52
:
1690
7
.
147.
Mifflin
RC
,
Pinchuk
IV
,
Saada
JI
,
Powell
DW
. 
Intestinal myofibroblasts: targets for stem cell therapy
.
Am J Physiol Gastrointest Liver Physiol
2011
;
300
:
G684
96
.
148.
Pinchuk
IV
,
Mifflin
RC
,
Saada
JI
,
Powell
DW
. 
Intestinal mesenchymal cells
.
Curr Gastroenterol Rep
2010
;
12
:
310
8
.
149.
Powell
DW
,
Pinchuk
IV
,
Saada
JI
,
Chen
X
,
Mifflin
RC
. 
Mesenchymal cells of the intestinal lamina propria
.
Annu Rev Physiol
2011
;
73
:
213
37
.
150.
Zhao
P
,
Jiang
H
,
Jiang
T
,
Zhi
Z
,
Wu
C
,
Sun
C
, et al
Inclusion of celecoxib into fibrous ordered mesoporous carbon for enhanced oral bioavailability and reduced gastric irritancy
.
Eur J Pharm Sci
2012
;
45
:
639
47
.
151.
Zhu
Q
,
Talton
J
,
Zhang
G
,
Cunningham
T
,
Wang
Z
,
Waters
RC
, et al
Large intestine-targeted, nanoparticle-releasing oral vaccine to control genitorectal viral infection
.
Nat Med
2012
;
18
:
1291
6
.
152.
Laroui
H
,
Sitaraman
SV
,
Merlin
D
. 
Gastrointestinal delivery of anti-inflammatory nanoparticles
.
Methods Enzymol
2012
;
509
:
101
25
.
153.
Cote
R
,
Suster
S
,
Weiss
L
,
Weidne
N
. 
Modern Surgical Pathology
.
Philadelphia, PA
:
Saunders
; 
2009
.
154.
Dreaden
EC
,
Austin
LA
,
Mackey
MA
,
El-Sayed
MA
. 
Size matters: gold nanoparticles in targeted cancer drug delivery
.
Therapeutic Deliv
2012
;
3
:
457
78
.
155.
Upreti
M
,
Jyoti
A
,
Sethi
P
. 
Tumor microenvironment and nanotherapeutics
.
Translational Cancer Res
2013
;
2
:
309
19
.
156.
Chaudhary
A
,
Sutaria
D
,
Huang
Y
,
Wang
J
,
Prabhu
S
. 
Chemoprevention of colon cancer in a rat carcinogenesis model using a novel nanotechnology-based combined treatment system
.
Cancer Prev Res
2011
;
4
:
1655
64
.
157.
Soundararajan
V
,
Warnock
K
,
Sasisekharan
R
. 
Multifunctional nanoscale platforms for targeting of the cancer cell immortality spectrum
.
Macromol Rapid Commun
2010
;
31
:
202
16
.
158.
Daka
A
,
Peer
D
. 
RNAi-based nanomedicines for targeted personalized therapy
.
Adv Drug Deliv Rev
2012
;
64
:
1508
21
.
159.
Maojo
V
,
Fritts
M
,
Martin-Sanchez
F
,
De la Iglesia
D
,
Cachau
RE
,
Garcia-Remesal
M
, et al
Nanoinformatics: developing new computing applications for nanomedicine
.
Comput Sci Eng
2012
;
94
:
521
39
.
160.
Casciaro
S
. 
Theranostic applications: Non-ionizing cellular and molecular imaging through innovative nanosystems for early diagnosis and therapy
.
World J Radiol
2011
;
3
:
249
55
.
161.
Yoo
D
,
Lee
JH
,
Shin
TH
,
Cheon
J
. 
Theranostic magnetic nanoparticles
.
Acc Chem Res
2011
;
44
:
863
74
.
162.
Kircher
MF
,
de la Zerda
A
,
Jokerst
JV
,
Zavaleta
CL
,
Kempen
PJ
,
Mittra
E
, et al
A brain tumor molecular imaging strategy using a new triple-modality MRI-photoacoustic-Raman nanoparticle
.
Nat Med
2012
;
18
:
829
34
.
163.
Delahunt
B
,
Scolyer
RA
,
Srigley
JR
. 
Premalignancy: the lull before the storm
.
Pathology
2013
;
45
:
207
8
.
164.
Kelloff
GJ
,
Sullivan
DC
,
Baker
H
,
Clarke
LP
,
Nordstrom
R
,
Tatum
JL
, et al
Workshop on imaging science development for cancer prevention and preemption
.
Cancer Biomark
2007
;
3
:
1
33
.
165.
Vincent
TL
,
Gatenby
RA
. 
An evolutionary model for initiation, promotion, and progression in carcinogenesis
.
Int J Oncol
2008
;
32
:
729
37
.
166.
Westergren-Thorsson
G
,
Larsen
K
,
Nihlberg
K
,
Andersson-Sjoland
A
,
Hallgren
O
,
Marko-Varga
G
, et al
Pathological airway remodelling in inflammation
.
Clin Respir J
2010
;
4
:
1
8
.
167.
Sveinsson
OA
,
Orvar
KB
,
Birgisson
S
,
Jonasson
JG
. [ 
Microscopic colitis - review]
.
Laeknabladid
2008
;
94
:
363
70
.
168.
di Tomaso
E
,
Capen
D
,
Haskell
A
,
Hart
J
,
Logie
JJ
,
Jain
RK
, et al
Mosaic tumor vessels: cellular basis and ultrastructure of focal regions lacking endothelial cell markers
.
Cancer Res
2005
;
65
:
5740
9
.
169.
Eyden
B
,
Tzaphlidou
M
. 
Structural variations of collagen in normal and pathological tissues: role of electron microscopy
.
Micron
2001
;
32
:
287
300
.
170.
Krop
I
,
Marz
A
,
Carlsson
H
,
Li
X
,
Bloushtain-Qimron
N
,
Hu
M
, et al
A putative role for psoriasin in breast tumor progression
.
Cancer Res
2005
;
65
:
11326
34
.
171.
Yu
J
,
Monaco
SE
,
Onisko
A
,
Bhargava
R
,
Dabbs
DJ
,
Cieply
KM
, et al
A validation study of quantum dot multispectral imaging to evaluate hormone receptor status in ductal carcinoma in situ of the breast
.
Hum Pathol
2013
;
44
:
394
401
.
172.
Menakuru
SR
,
Brown
NJ
,
Staton
CA
,
Reed
MW
. 
Angiogenesis in pre-malignant conditions
.
Br J Cancer
2008
;
99
:
1961
6
.
173.
Fox
SB
,
Harris
AL
. 
Histological quantitation of tumour angiogenesis
.
APMIS
2004
;
112
:
413
30
.
174.
Smith
SJ
,
Tilly
H
,
Ward
JH
,
Macarthur
DC
,
Lowe
J
,
Coyle
B
, et al
CD105 (Endoglin) exerts prognostic effects via its role in the microvascular niche of paediatric high grade glioma
.
Acta Neuropathol
2012
;
124
:
99
110
.
175.
Ratajczak
P
,
Leboeuf
C
,
Wang
L
,
Briere
J
,
Loisel-Ferreira
I
,
Thieblemont
C
, et al
BCL2 expression in CD105 positive neoangiogenic cells and tumor progression in angioimmunoblastic T-cell lymphoma
.
Mod Pathol
2012
;
25
:
805
14
.
176.
Dubinski
W
,
Gabril
M
,
Iakovlev
VV
,
Scorilas
A
,
Youssef
YM
,
Faragalla
H
, et al
Assessment of the prognostic significance of endoglin (CD105) in clear cell renal cell carcinoma using automated image analysis
.
Hum Pathol
2012
;
43
:
1037
43
.
177.
Marioni
G
,
Staffieri
A
,
Manzato
E
,
Ralli
G
,
Lionello
M
,
Giacomelli
L
, et al
A higher CD105-assessed microvessel density and worse prognosis in elderly patients with laryngeal carcinoma
.
Arch Otolaryngol Head Neck Surg
2011
;
137
:
175
80
.
178.
Fang
J
,
Nakamura
H
,
Maeda
H
. 
The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect
.
Adv Drug Deliv Rev
2011
;
63
:
136
51
.
179.
Folkman
J
,
Watson
K
,
Ingber
D
,
Hanahan
D
. 
Induction of angiogenesis during the transition from hyperplasia to neoplasia
.
Nature
1989
;
339
:
58
61
.
180.
Bergers
G
,
Benjamin
LE
. 
Tumorigenesis and the angiogenic switch
.
Nat Rev Cancer
2003
;
3
:
401
10
.
181.
Grandis
JR
,
Argiris
A
. 
Targeting angiogenesis from premalignancy to metastases
.
Cancer Prev Res
2009
;
2
:
291
4
.
182.
Staton
CA
,
Chetwood
AS
,
Cameron
IC
,
Cross
SS
,
Brown
NJ
,
Reed
MW
. 
The angiogenic switch occurs at the adenoma stage of the adenoma carcinoma sequence in colorectal cancer
.
Gut
2007
;
56
:
1426
32
.
183.
Bluff
JE
,
Menakuru
SR
,
Cross
SS
,
Higham
SE
,
Balasubramanian
SP
,
Brown
NJ
, et al
Angiogenesis is associated with the onset of hyperplasia in human ductal breast disease
.
Br J Cancer
2009
;
101
:
666
72
.
184.
Decuzzi
P
,
Causa
F
,
Ferrari
M
,
Netti
PA
. 
The effective dispersion of nanovectors within the tumor microvasculature
.
Ann Biomed Eng
2006
;
34
:
633
41
.
185.
Ferrati
S
,
Shamsudeen
S
,
Summers
HD
,
Rees
P
,
Abbey
JV
,
Schmulen
J
, et al
Inter-endothelial transport of microvectors using cellular shuttles and tunneling nanotubes
.
Small
2012
;
8
:
3151
60
.
186.
Cooke
VG
,
LeBleu
VS
,
Keskin
D
,
Khan
Z
,
O'Connell
JT
,
Teng
Y
, et al
Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway
.
Cancer Cell
2012
;
21
:
66
81
.
187.
Barlow
KD
,
Sanders
AM
,
Soker
S
,
Ergun
S
,
Metheny-Barlow
LJ
. 
Pericytes on the Tumor Vasculature: Jekyll or Hyde
?
Cancer Microenviron
2013
;
6
:
1
17
.
188.
Virgintino
D
,
Rizzi
M
,
Errede
M
,
Strippoli
M
,
Girolamo
F
,
Bertossi
M
, et al
Plasma membrane-derived microvesicles released from tip endothelial cells during vascular sprouting
.
Angiogenesis
2012
;
15
:
761
9
.
189.
Ribatti
D
,
Crivellato
E
. 
“Sprouting angiogenesis”, a reappraisal
.
Dev Biol
2012
;
372
:
157
65
.
190.
Lammers
T
,
Hennink
WE
,
Storm
G
. 
Tumour-targeted nanomedicines: principles and practice
.
Br J Cancer
2008
;
99
:
392
7
.
191.
Kostarelos
K
,
Bianco
A
,
Prato
M
. 
Promises, facts and challenges for carbon nanotubes in imaging and therapeutics
.
Nat Nanotechnol
2009
;
4
:
627
33
.
192.
Donaldson
K
,
Seaton
A
. 
The Janus faces of nanoparticles
.
J Nanosci Nanotechnol
2007
;
7
:
4607
11
.
193.
Faunce
T
,
Shats
K
. 
Researching safety and cost-effectiveness in the life cycle of nanomedicine
.
J Law Med
2007
;
15
:
128
35
.
194.
Lippman
SM
,
Hawk
ET
. 
Cancer prevention: from 1727 to milestones of the past 100 years
.
Cancer Res
2009
;
69
:
5269
84
.
195.
Kelloff
GJ
,
Lippman
SM
,
Dannenberg
AJ
,
Sigman
CC
,
Pearce
HL
,
Reid
BJ
, et al
Progress in chemoprevention drug development: the promise of molecular biomarkers for prevention of intraepithelial neoplasia and cancer–a plan to move forward
.
Clin Cancer Res
2006
;
12
:
3661
97
.
196.
Ganz
PA
. 
Survivorship: adult cancer survivors
.
Primary care
2009
;
36
:
721
41
.
197.
Myint
PK
,
Smith
RD
,
Luben
RN
,
Surtees
PG
,
Wainwright
NW
,
Wareham
NJ
, et al
Lifestyle behaviours and quality-adjusted life years in middle and older age
.
Age Ageing
2011
;
40
:
589
95
.
198.
Nagahara
LA
,
Lee
JS
,
Molnar
LK
,
Panaro
NJ
,
Farrell
D
,
Ptak
K
, et al
Strategic workshops on cancer nanotechnology
.
Cancer Res
2010
;
70
:
4265
8
.
199.
Wild
CP
,
Scalbert
A
,
Herceg
Z
. 
Measuring the exposome: a powerful basis for evaluating environmental exposures and cancer risk
.
Environ Mol Mutagen
2013
;
54
:
480
99
.
200.
Fan
R
,
Vermesh
O
,
Srivastava
A
,
Yen
BK
,
Qin
L
,
Ahmad
H
, et al
Integrated barcode chips for rapid, multiplexed analysis of proteins in microliter quantities of blood
.
Nat Biotechnol
2008
;
26
:
1373
8
.
201.
Tasciotti
E
,
Liu
X
,
Bhavane
R
,
Plant
K
,
Leonard
AD
,
Price
BK
, et al
Mesoporous silicon particles as a multistage delivery system for imaging and therapeutic applications
.
Nat Nanotechnol
2008
;
3
:
151
7
.
202.
Bouamrani
A
,
Hu
Y
,
Tasciotti
E
,
Li
L
,
Chiappini
C
,
Liu
X
, et al
Mesoporous silica chips for selective enrichment and stabilization of low molecular weight proteome
.
Proteomics
2010
;
10
:
496
505
.
203.
Wang
H
,
Hanash
S
. 
Intact-protein analysis system for discovery of serum-based disease biomarkers
.
Methods Mol Biol
2011
;
728
:
69
85
.
204.
Lu
H
,
Ladd
J
,
Feng
Z
,
Wu
M
,
Goodell
V
,
Pitteri
SJ
, et al
Evaluation of known oncoantibodies, HER2, p53, and cyclin B1, in prediagnostic breast cancer sera
.
Cancer Prev Res
2012
;
5
:
1036
43
.
205.
Fan
J
,
Deng
X
,
Gallagher
JW
,
Huang
H
,
Huang
Y
,
Wen
J
, et al
Monitoring the progression of metastatic breast cancer on nanoporous silica chips
.
Philos Transact A Math Phys Eng Sci
2012
;
370
:
2433
47
.
206.
Burns
AA
,
Vider
J
,
Ow
H
,
Herz
E
,
Penate-Medina
O
,
Baumgart
M
, et al
Fluorescent silica nanoparticles with efficient urinary excretion for nanomedicine
.
Nano Lett
2009
;
9
:
442
8
.
207.
Li
Y
,
Xu
W
. 
Highly sensitive detection of Shigella flexneri using fluorescent silica nanoparticles
.
New Microbiol
2009
;
32
:
377
83
.
208.
Luther
T
,
Carrion
CF
,
Cobb
N
,
Le
G
,
Edwards
C
,
Schwartz
S
, et al
Methods for analyzing saliva proteins for systemic disease detection
.
Gen Dent
2010
;
58
:
110
3
.
209.
Weigum
SE
,
Floriano
PN
,
Redding
SW
,
Yeh
CK
,
Westbrook
SD
,
McGuff
HS
, et al
Nano-bio-chip sensor platform for examination of oral exfoliative cytology
.
Cancer Prev Res
2010
;
3
:
518
28
.
210.
Nagasaka
T
,
Tanaka
N
,
Cullings
HM
,
Sun
DS
,
Sasamoto
H
,
Uchida
T
, et al
Analysis of fecal DNA methylation to detect gastrointestinal neoplasia
.
J Natl Cancer Inst
2009
;
101
:
1244
58
.
211.
White
V
,
Scarpini
C
,
Barbosa-Morais
NL
,
Ikelle
E
,
Carter
S
,
Laskey
RA
, et al
Isolation of stool-derived mucus provides a high yield of colonocytes suitable for early detection of colorectal carcinoma
.
Cancer Epidemiol Biomarkers Prev
2009
;
18
:
2006
13
.
212.
Loktionov
A
,
Bandaletova
T
,
Llewelyn
AH
,
Dion
C
,
Lywood
HG
,
Lywood
RC
, et al
Colorectal cancer detection by measuring DNA from exfoliated colonocytes obtained by direct contact with rectal mucosa
.
Int J Oncol
2009
;
34
:
301
11
.
213.
Varella-Garcia
M
,
Schulte
AP
,
Wolf
HJ
,
Feser
WJ
,
Zeng
C
,
Braudrick
S
, et al
The detection of chromosomal aneusomy by fluorescence in situ hybridization in sputum predicts lung cancer incidence
.
Cancer Prev Res
2010
;
3
:
447
53
.
214.
Qiu
Q
,
Todd
NW
,
Li
R
,
Peng
H
,
Liu
Z
,
Yfantis
HG
, et al
Magnetic enrichment of bronchial epithelial cells from sputum for lung cancer diagnosis
.
Cancer
2008
;
114
:
275
83
.
215.
Negraes
PD
,
Favaro
FP
,
Camargo
JL
,
Oliveira
ML
,
Goldberg
J
,
Rainho
CA
, et al
DNA methylation patterns in bladder cancer and washing cell sediments: a perspective for tumor recurrence detection
.
BMC Cancer
2008
;
8
:
238
.
216.
Tsai
HT
,
Tsai
YM
,
Yang
SF
,
Lee
CH
,
Lin
LY
,
Lee
S
, et al
A notable accessory screening program for detection of cervical intraepithelial neoplasia
.
Pathol Biol
2009
;
57
:
477
82
.
217.
Wang
SS
,
Smiraglia
DJ
,
Wu
YZ
,
Ghosh
S
,
Rader
JS
,
Cho
KR
, et al
Identification of novel methylation markers in cervical cancer using restriction landmark genomic scanning
.
Cancer Res
2008
;
68
:
2489
97
.
218.
Cazzaniga
M
,
Decensi
A
,
Bonanni
B
,
Luini
A
,
Gentilini
O
. 
Biomarkers for risk assessment and prevention of breast cancer
.
Curr Cancer Drug Targets
2009
;
9
:
482
99
.
219.
Rusling
JF
,
Kumar
CV
,
Gutkind
JS
,
Patel
V
. 
Measurement of biomarker proteins for point-of-care early detection and monitoring of cancer
.
Analyst
2010
;
135
:
2496
511
.
220.
Nune
SK
,
Gunda
P
,
Thallapally
PK
,
Lin
YY
,
Forrest
ML
,
Berkland
CJ
. 
Nanoparticles for biomedical imaging
.
Expert Opin Drug Deliv
2009
;
6
:
1175
94
.
221.
Shubayev
VI
,
Pisanic
TR
 II
,
Jin
S
. 
Magnetic nanoparticles for theragnostics
.
Adv Drug Deliv Rev
2009
;
61
:
467
77
.
222.
Bentolila
LA
,
Ebenstein
Y
,
Weiss
S
. 
Quantum dots for in vivo small-animal imaging
.
J Nucl Med
2009
;
50
:
493
6
.
223.
Boisselier
E
,
Astruc
D
. 
Gold nanoparticles in nanomedicine: preparations, imaging, diagnostics, therapies and toxicity
.
Chem Soc Rev
2009
;
38
:
1759
82
.
224.
LaRocque
J
,
Bharali
DJ
,
Mousa
SA
. 
Cancer detection and treatment: the role of nanomedicines
.
Mol Biotechnol
2009
;
42
:
358
66
.
225.
Hartman
KB
,
Wilson
LJ
. 
Carbon nanostructures as a new high-performance platform for MR molecular imaging
.
Adv Exp Med Biol
2007
;
620
:
74
84
.
226.
Ross
BD
,
Bhattacharya
P
,
Wagner
S
,
Tran
T
,
Sailasuta
N
. 
Hyperpolarized MR imaging: neurologic applications of hyperpolarized metabolism
.
AJNR Am J Neuroradiol
2010
;
31
:
24
33
.
227.
Zacharias
NM
,
Chan
HR
,
Sailasuta
N
,
Ross
BD
,
Bhattacharya
P
. 
Real-time molecular imaging of tricarboxylic acid cycle metabolism in vivo by hyperpolarized 1-(13)C diethyl succinate
.
J Am Chem Soc
2012
;
134
:
934
43
.
228.
Eck
W
,
Craig
G
,
Sigdel
A
,
Ritter
G
,
Old
LJ
,
Tang
L
, et al
PEGylated gold nanoparticles conjugated to monoclonal F19 antibodies as targeted labeling agents for human pancreatic carcinoma tissue
.
ACS Nano
2008
;
2
:
2263
72
.
229.
Roger
M
,
Clavreul
A
,
Venier-Julienne
MC
,
Passirani
C
,
Sindji
L
,
Schiller
P
, et al
Mesenchymal stem cells as cellular vehicles for delivery of nanoparticles to brain tumors
.
Biomaterials
2010
;
31
:
8393
401
.
230.
Serda
RE
,
Godin
B
,
Blanco
E
,
Chiappini
C
,
Ferrari
M
. 
Multi-stage delivery nano-particle systems for therapeutic applications
.
Biochim Biophys Acta
2011
;
1810
:
317
29
.
231.
Serda
RE
,
Ferrati
S
,
Godin
B
,
Tasciotti
E
,
Liu
X
,
Ferrari
M
. 
Mitotic trafficking of silicon microparticles
.
Nanoscale
2009
;
1
:
250
9
.
232.
Tanaka
T
,
Mangala
LS
,
Vivas-Mejia
PE
,
Nieves-Alicea
R
,
Mann
AP
,
Mora
E
, et al
Sustained small interfering RNA delivery by mesoporous silicon particles
.
Cancer Res
2010
;
70
:
3687
96
.
233.
Chen
YS
,
Hung
YC
,
Lin
WH
,
Huang
GS
. 
Assessment of gold nanoparticles as a size-dependent vaccine carrier for enhancing the antibody response against synthetic foot-and-mouth disease virus peptide
.
Nanotechnology
2010
;
21
:
195101
.
234.
Bielinska
AU
,
Gerber
M
,
Blanco
LP
,
Makidon
PE
,
Janczak
KW
,
Beer
M
, et al
Induction of Th17 cellular immunity with a novel nanoemulsion adjuvant
.
Crit Rev Immunol
2010
;
30
:
189
99
.
235.
Klippstein
R
,
Pozo
D
. 
Nanotechnology-based manipulation of dendritic cells for enhanced immunotherapy strategies
.
Nanomedicine
2010
;
6
:
523
9
.
236.
Sheng
WY
,
Huang
L
. 
Cancer Immunotherapy and Nanomedicine
.
Pharm Res
2011
;
28
:
200
14
.
237.
Mattheolabakis
G
,
Lagoumintzis
G
,
Panagi
Z
,
Papadimitriou
E
,
Partidos
CD
,
Avgoustakis
K
. 
Transcutaneous delivery of a nanoencapsulated antigen: induction of immune responses
.
Int J Pharm
2010
;
385
:
187
93
.
238.
Chadwick
S
,
Kriegel
C
,
Amiji
M
. 
Nanotechnology solutions for mucosal immunization
.
Adv Drug Deliv Rev
2010
;
62
:
394
407
.
239.
Ge
W
,
Hu
PZ
,
Huang
Y
,
Wang
XM
,
Zhang
XM
,
Sun
YJ
, et al
The antitumor immune responses induced by nanoemulsion-encapsulated MAGE1-HSP70/SEA complex protein vaccine following different administration routes
.
Oncol Rep
2009
;
22
:
915
20
.
240.
Lu
W
,
Zhang
G
,
Zhang
R
,
Flores
LG
 II
,
Huang
Q
,
Gelovani
JG
, et al
Tumor site-specific silencing of NF-kappaB p65 by targeted hollow gold nanosphere-mediated photothermal transfection
.
Cancer Res
2010
;
70
:
3177
88
.
241.
Tian
Z
,
Wang
H
,
Jia
Z
,
Shi
J
,
Tang
J
,
Mao
L
, et al
Tumor-targeted inhibition by a novel strategy - mimoretrovirus expressing siRNA targeting the Pokemon gene
.
Curr Cancer Drug Targets
2010
;
10
:
932
41
.
242.
Siddiqui
IA
,
Adhami
VM
,
Bharali
DJ
,
Hafeez
BB
,
Asim
M
,
Khwaja
SI
, et al
Introducing nanochemoprevention as a novel approach for cancer control: proof of principle with green tea polyphenol epigallocatechin-3-gallate
.
Cancer Res
2009
;
69
:
1712
6
.
243.
Siddiqui
IA
,
Adhami
VM
,
Ahmad
N
,
Mukhtar
H
. 
Nanochemoprevention: sustained release of bioactive food components for cancer prevention
.
Nutr Cancer
2010
;
62
:
883
90
.
244.
Steinbach
G
,
Lynch
PM
,
Phillips
RK
,
Wallace
MH
,
Hawk
E
,
Gordon
GB
, et al
The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis
.
N Engl J Med
2000
;
342
:
1946
52
.
245.
Barton
MK
. 
Daily aspirin reduces colorectal cancer incidence in patients with Lynch syndrome
.
CA Cancer J Clin
2012
;
62
:
143
4
.
246.
Solomon
SD
,
McMurray
JJ
,
Pfeffer
MA
,
Wittes
J
,
Fowler
R
,
Finn
P
, et al
Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention
.
N Engl J Med
2005
;
352
:
1071
80
.
247.
Morgen
M
,
Bloom
C
,
Beyerinck
R
,
Bello
A
,
Song
W
,
Wilkinson
K
, et al
Polymeric nanoparticles for increased oral bioavailability and rapid absorption using celecoxib as a model of a low-solubility, high-permeability drug
.
Pharm Res
2012
;
29
:
427
40
.
248.
Margulis-Goshen
K
,
Weitman
M
,
Major
DT
,
Magdassi
S
. 
Inhibition of crystallization and growth of celecoxib nanoparticles formed from volatile microemulsions
.
J Pharm Sci
2011 May 31
. [Epub ahead of print].
249.
Tan
A
,
Davey
AK
,
Prestidge
CA
. 
Silica-lipid hybrid (SLH) versus non-lipid formulations for optimising the dose-dependent oral absorption of celecoxib
.
Pharm Res
2011
;
28
:
2273
87
.
250.
Venkatesan
P
,
Puvvada
N
,
Dash
R
,
Prashanth Kumar
BN
,
Sarkar
D
,
Azab
B
, et al
The potential of celecoxib-loaded hydroxyapatite-chitosan nanocomposite for the treatment of colon cancer
.
Biomaterials
2011
;
32
:
3794
806
.
251.
Thakkar
H
,
Kumar Sharma
R
,
Murthy
RS
. 
Enhanced retention of celecoxib-loaded solid lipid nanoparticles after intra-articular administration
.
Drugs R D
2007
;
8
:
275
85
.
252.
Lanza
FL
,
Marathi
UK
,
Anand
BS
,
Lichtenberger
LM
. 
Clinical trial: comparison of ibuprofen-phosphatidylcholine and ibuprofen on the gastrointestinal safety and analgesic efficacy in osteoarthritic patients
.
Aliment Pharmacol Ther
2008
;
28
:
431
42
.
253.
Grandhi
BK
,
Thakkar
A
,
Wang
J
,
Prabhu
S
. 
A novel combinatorial nanotechnology-based oral chemopreventive regimen demonstrates significant suppression of pancreatic cancer neoplastic lesions
.
Cancer Prev Res
2013
;
6
:
1015
25
.
254.
Bisht
S
,
Mizuma
M
,
Feldmann
G
,
Ottenhof
NA
,
Hong
SM
,
Pramanik
D
, et al
Systemic administration of polymeric nanoparticle-encapsulated curcumin (NanoCurc) blocks tumor growth and metastases in preclinical models of pancreatic cancer
.
Mol Cancer Ther
2010
;
9
:
2255
64
.
255.
Aqil
F
,
Jeyabalan
J
,
Kausar
H
,
Bansal
SS
,
Sharma
RJ
,
Singh
IP
, et al
Multi-layer polymeric implants for sustained release of chemopreventives
.
Cancer Lett
2012
;
326
:
33
40
.
256.
Bansal
SS
,
Goel
M
,
Aqil
F
,
Vadhanam
MV
,
Gupta
RC
. 
Advanced drug delivery systems of curcumin for cancer chemoprevention
.
Cancer Prev Res
2011
;
4
:
1158
71
.