Advances in the application of nanotechnology in the diagnosis and treatment of gastrointestinal tumors (Review)

  • Authors:
    • Bo Sun
    • Yantian Fang
    • Zhengyang Li
    • Zongyou Chen
    • Jianbin Xiang
  • View Affiliations

  • Published online on: December 2, 2014     https://doi.org/10.3892/mco.2014.470
  • Pages: 274-280
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Nanotechnology has broad application prospects in the diagnosis and treatment of cancer. Integrating chemistry, engineering, biology and medicine, nanotechnology is a multidisciplinary research field. Nanoscale imaging technology significantly improves the precision and accuracy of tumor diagnosis. Nanocarriers are able to significantly improve the accuracy of dose and targeted drug delivery and reduce the toxic side effects. This review focuses on the emerging roles of these innovative technologies in gastrointestinal cancer diagnostics and therapeutics. Although several problems and barriers are hampering the development of nanodevices, the potential for nanotechnologies to function as multimodal nanotheranostic agents will likely pave the way for the fight against gastrointestinal cancer.

Introduction

Nanotechnology is a multidisciplinary research field that integrates a broad and diverse array of equipments derived from chemistry, engineering, biology and medicine (1). Nanotechnologies applied to gastrointestinal cancer include nanoparticle-based specific identification of tumors and cancer biomarkers, biologically-targeted contrast agents for magnetic resonance imaging (MRI), detection of sentinel lymph nodes (SLNs), drug delivery systems and novel treatment approaches. Novel nanotechnologies have gained worldwide attention due to their great potential to vastly improve current standards and techniques for the diagnosis and treatment of gastrointestinal cancers.

Devices based on nanotechnology are typically, in at least one dimension, in the 1–100 nm range. The dimensions may be manipulated close to the wavelength for scattering or absorption over a wide spectral range of light, including near-infrared (NIR) light. The nanodevices have a large surface area-to-volume ratio that increases the interaction surface between the target and the nanodevices (2). Nanodevices are able to load drugs at a high concentration, which are then efficiently delivered to specific sites with the advantage of fewer side effects and lower toxicity. The advantages of nanodevices compared to traditional technologies make them attractive modalities for development and they may also help promote the application of personalized therapy based on a patient's genetic content.

Although considerable progress has been achieved over the last few years, certain issues are hampering the development of nanodevices (3). Newly engineered nanoparticles exhibit significantly reduced toxicity; however, the question of tocixity remains a focus of attention (4). The high price of the innovative devices and complex production process currently prevent nanotechnology from being routinely applied clinically for tumor detection. Regarding the future application of nanodrug delivery systems, there is a need for a more complete system of safety pharmacology, drug biotransformation, pharmacokinetic and toxicokinetic studies.

The nanodevices developed for gastrointestinal oncology include nanowires, cantilevers, quantum dots (QDs), nanoshells, gold nanoparticles dendrimers, carbon nanotubes, paramagnetic nanoparticles, liposomes and nanogels. The aim of this review was to summarize the emerging roles of this new technology in gastrointestinal cancer diagnosis and therapy, particularly focusing on nanowires, cantilevers, QDs, nanoshells, dendrimers and nanogels, which may represent exciting opportunities in the fight against gastrointestinal cancer.

Nanowires

A nanowire may be defined as a material consisting of millimeters in length, but achieving a diameter measured in the nanometer range. Nanowire devices are based on field effect transistors (FETs) (5). When biomarkers flow aside, the change in charge density is turned into measurable information in the electric field of the nanowire devices, enabling highly efficacious detection of biological targets (6).

Silicon nanowire (SiNW)-FETs with surface receptors binding into arrays are favorable for selective, highly sensitive, multiplexed and label-free biomarker measurements (7). The integrated control nanowires may further reduce the incidence of false-positive results. Nucleic acid receptors incorporated into arrays may enable real-time assays of the telomerase activity using samples extracted from only 10 tumor cells without using methods such as repeat amplification protocol (8).

However, there remain certain challenges. One of the challenges associated with SiNW-FET sensors is the relatively low analytical signal intensity. Due to the higher ionic strength and possible contamination of the sensors, whole-blood samples cannot be directly examined (9).

The SiNWs device has shown the possibility of highly sensitive label-free and early detection of miRNA as a diagnostic marker for tumors. Zhang et al (10) reported that SiNWs arrays allowed direct hybridization detection of miRNA without the help of any additional biological labelling. The biosensor may identify the concentrations of the miRNA through the resistance change caused by direct hybridization with peptide nucleic acids immobilized on the SiNW device. Biosensors are emerging as promising candidates for detection applications due to their ability to detect target miRNA at concentrations as low as 1 fm (10). Given that the concentrations of cancer biomarkers are extremely low in the tissue or blood samples, a SiNWs-based device is expected to be a reliable and cost-effective sensor with high specificity and sensitivity. Lee et al (11) demonstrated that SiNW-based sensors were ultrasensitive and specific in measuring C-reactive protein (CRP), a marker of inflammation which was recently associated with cancer progression. The new technology is highly efficacious for accurate, rapid and repeatable testing of CRP (11). Zheng et al (12) used nanowires to develop a highly selective assay for ultrasensitive multiplexed detection of prostate-specific antigen (PSA)- α 1-antichymotrypsin complexes, PSA, mucin-1 and carcinoembryonic antigen (CEA). This device is capable of detecting concentrations of at least 0.9 pg/ml in undiluted serum samples (12). Another system reported by Stern et al (13) uses a two-step approach, incorporating microfluidic purification chips that capture multiple biomarkers from whole blood, concentrating the biomarkers of interest and releasing the biomarkers for quantitative detection with silicon nanoribbon detectors. This technique reduces the minimum required sensitivity of the system (13).

Well-designed SiNWs passively penetrate and transfect cells, providing new opportunities for gene therapy of gastrointestinal cancers.

Cantilevers

Nanoscale cantilevers are microscopic flexible beams that are usually arrayed in a row. Cantilevers are able to conduct biosensing through the principle that these tiny probes naturally vibrate at a certain frequency dictated by mechanical and mass properties. When a biological molecule binds to this nanoscale probe, it alters the baseline probe frequency, which is typically measured by a difference in the characteristics of the light deflection pattern of the probe or through electrical means (14). Cantilever vibrations are mainly deflected in atomic force microscopy (AFM) force feedback mode to obtain the real-time imaging (15).

The reported benefits of using nanocantilever systems in cancer detection include that there are no requirements for fluorescent or radioactive labeling; detection may take place in liquid samples and this technology may be easily translated to lab-on-a-chip techniques, providing point-of-care diagnostics (16). They have also been described as a simple replacement to polymerase chain reaction reactions and detection methods, as they are more cost-effective regarding sample preparation, in terms of time and costly materials.

Nanoscale cantilever devices, in which cantilevers are coated with specific receptors, may provide highly sensitive and rapid detection of disease-specific molecules, such as DNA or protein (17). Hansen et al (18) incorporated molecules capable of binding to the specific gene sequence of single-nucleotide polymorphisms (SNPs) into nanocantilevers to inspect the SNPs in DNA target oligonucleotides. Based on the good spatial resolution and good contrast of cancer cells on mica in water, AFM has been combined with the astigmatic detection system for imaging soft DNA molecules. Liao et al (19) suppressed the spurious peaks of the cantilever holder down to 26.0% of the real resonance peak to achieve excellent sensitivity. Methodology composed of the AFM tip-cantilever assembly and silica beads has been fabricated to measure specific ligand-receptor interactions and to adjust receptor positions. Gunning et al (20) adopted this novel approach to detect the force between wheat germ agglutinin and the glycosylated extracellular domain III of the epidermal growth factor receptor (EGFR) on the surface of living human intestinal epithelial cells. The values for single-molecule interactions are typically expected (20). A patent approach incorporating AFM with confocal microscopy was developed to detect bulk nuclear stiffness and to simultaneously visualize the cantilever-nucleus contact. Krause et al (21) used this method to identify nuclear compressibility prior to and following nuclear softening induced by the chromatin-decondensating agent trichostatin A, with the results indicating that this approach may be practical. The proposed tool may be extremely useful for the comprehension of the concept of limiting nuclear deformation in carcinoma invasion (21). Wu et al (22) used a microplate measurement system approach based on the deflection of a flexible microcantilever to measure cell stiffness (in Pa) and adhesion force (in nN) between cells prior to and following transforming growth factor-β1-induced epithelial-to-mesenchymal transition.

Quantum dots (QDs)

QDs are nanocrystals that are composed of semiconductor particles, consisting of an inorganic element in its core with a surrounding metal shell. The diameters of QDs range between 2 and 10 nm. The size and composition of QDs may be adjusted to give the QDs a unique fluorescence emission that may vary between 400 and 2,000 nm (1). Varying wavelengths allow for tuning QDs to any color, which enables recognition and tracking of differently labeled biomarkers using only a single light source (23). Fluorescent-labeled QDs make multicolor imaging in living tissue a reality in the context of multiphoton microscopy (24).

QDs possess promising characteristics, such as stable fluorescence with simple excitation, multispectral tunability, high sensitivity and no requirement for lasers. The red/infrared colors of QDs enable whole-blood assays. However, one problem that traditionally exists with imaging normal healthy tissue is that it often exhibits autofluorescence, which interferes with the signal from cancerous tissue. QDs have been engineered to display fluorescence properties in the NIR spectra and, thus, are able to eliminate this interference to a great extent (25). Another potential problem with using QDs in vivo is whether injection poses a toxic risk. Modifications have been made to decrease potential toxicity; however, further research is required to determine appropriate clinical adaptability (26). As cadmium ions released from the QDs are associated with cytotoxicity, polyethylene glycol (PEG) was developed to reduced the toxicity of uncoated QDs. Indeed, the QDs coated with the PEG polymer induced no obvious immune response, no cytotoxicity and no cell cycle changes, even at high dosage. No more than 0.2% of the human genome was affected according to the genome-wide expression array analysis including 18,400 known gene probe sets (27). Another type of improved nanoparticles are graphene QDs, which may be promising substitutes for QDs based on rigorous research including cellular internalization, distribution and cytotoxicity studies (28).

QDs labeled with specific DNA tags is a practical testing tool for DNA sequences associated with cancer; based on the unique fluorescence emission, they may identify oncogene fragments in the DNA sample. Zhang et al (29) successfully synthesized a microfluidic bead-based nucleic acid sensor labeled with QDs to detect CEA gene fragments with a discrimination limit as low as 5 fm. QDs conjugated with colorectal cancer surface antigens may detect circulating tumor cells which have been associated with metastasis and prognostic significance. Yu et al (30) have created glutathione-thioglycolic acid co-capped cadmium telluride QDs exhibiting high fluorescence intensity and good biological compatibility. When incorporated to the monoclonal antibody ND-1, the novel detection device resoundingly labeled colorectal cancer cells in vitro (30). A microfluidic bead-based nucleic acid sensor with QD labels and multienzyme-nanoparticle amplification was found to be highly effective in detecting circulating cancer cells in blood samples. This device is capable of discriminating one cancer cell in 1 ml of blood (29). Gazouli et al (31) labeled QDs with two antibodies that attach to specific proteins on the surface of circulating tumour cells to perform diagnostic procedures on blood specimens. The detection limit was as low as 10 colorectal cancer cells per ml (31). Apart from circulating tumor cell detection, Bodo et al (32) demonstrated that QD immunofluorescence had the possibility of in situ quantitation of phosphoproteins in fixed samples, providing a promising method for highly cell type-specific detection application in the future.

QDs are considered to be a ideal method for cancer targeting and imaging due to their unique properties, including enhanced permeability and retention effect and nanoscale-vehicle properties with high imaging-agent capacity. PEG-coated QDs may be used for identifying SLNs in mouse tumor models (33).

NIR light possesses promising characteristics, such as high signal-to-background ratio, high photon penetration into living tissues and low tissue autofluorescence, which have attracted significant attention for use in biomedical imaging (34). The NIR QDs have been reported to be a practical imaging tool for simultaneous visualization of SLNs (35, 36). Soltesz et al (37) intra-parenchymally injected NIR fluorescent QDs in different parts of the gastrointestinal tract of pigs and identified lymph node drainage and SLNs during surgery using the NIR fluorescence imaging system. This development was a major breakthrough, since it provided real-time imaging of SLNs without using any radiolabels (37). Hikage et al (38) investigated the possibility of detecting SLNs with NIR QDs under a confocal microscope. In that study, the QDs were introduced into the gastrointestinal wall of pigs. The SLNs were quickly and accurately mapped, demonstrating the possibility of highly sensitive label-free SLN mapping (38). QDs conjugated with bioprobe labels are emerging as promising candidates for more specific imaging applications. Zhang et al (39) created gastric tumor-specific QDs (CC49-QDs), which are QDs labelled with the tumor-associated glycoprotein 72 monoclonal antibody CC49, and found that antibody grafting significantly improved the specifity of QDs in mapping target gastric tumor cells without any difference in optical properties. Geraldo et al (40) produced a new QDs/polyamidoamine (PAMAM)-folate derivative. The supramolecular complexes appear to hold promise for highly efficacious and highly selective imaging of gastric tumor cells. Paramagnetic QDs (cNGR-pQDs) labeled with cyclic Asn-Gly-Arg (cNGR) were created by Oostendorp et al (41) to determine angiogenic tumor vasculature, which is significant for tumor invasion and metastasis. The results demonstrated that cNGR-pQDs were a highly specific tool for in vivo detection and quantification of tumor angiogenic activity using MRI (41). An optical imaging nanoprobe using EGF-conjugated QDs for repetitive and quantifiable imaging of EGFR expression within tumor parenchyma was created by Diagaradjane et al (42). These results indicated that QDs conjugated with antigens enhanced the specificity of imaging application.

The use of multicolor QD probes is increasingly being investigated in the field of immunohistochemistry, which is widely used in the diagnosis of malignant tumors. This technique was applied to assess the expression of Cav-1 and LC3B, which have been found to be independent predictors of gastric cancer prognosis in humans (43, 44). QDs-based multiplexed biomarker detection has attracted significant attention as a reliable predictor of disease progression and treatment response (45). Bostick et al (46) created a panel of bioconjugated QDs to simultaneously detect and quantify well-recognized prognosticators for colorectal cancer on the same histological sections. QDs conjugated with multiple biomarkers may also be used for simultaneous prognosticator detection to predict medical outcomes in gastric cancer through studying the major components of the tumor stroma, including tumor infiltrating macrophages, tumor microvascular density and neovessel maturity, type IV collagen and matrix metalloproteinase 9 (4749).

Dendrimers

Dendrimers are synthetic complex nanostructures with branched concentric layers surrounding an inner core. The shape, size, surface functionalities and branching length of a dendrimer may be manipulated to perform different functions (50, 51). The diameters of dendrimers range between 1 and 10 nm. Dendrimers are proving to be particularly adept at serving as a versatile modularity capable of detecting a number of proteins, which are currently detected by individual ELISA testing. In addition, dendrimer nanoparticles have been created, which may be dually utilized for imaging using MRI or NIR fluorescent modalities in a single probe. This has also been shown to be effective in mouse SLN mapping (52).

Dendrimers were developed to be used as non-viral delivery vectors by exploiting their unique and superior property of enhanced permeability (53). Teow et al (54) demonstrated that the third-generation dendrimer is an ideal carrier system for paclitaxel (PTX) following conjugation with lauryl chains and labeling with fluorescein isothiocyanate, based on its increased permeability compared to PTX alone. The most commonly used dendrimer application in genetic transmission is PAMAM. Dufes et al (55) demonstrated that polypropylenimine dendrimers are able to effectively deliver the tumor necrosis factor α gene into colorectal adenocarcinoma cells to inhibit the growth of colorectal cancer without evident toxicity on the animals, demonstating that dendrimers are a promising carrier for the delivery of targeted antitumor genes for cancer therapy. Li et al (56) investigated a synthetic vector system based on PAMAM dendrimers for effective delivery of survivin antisense oligonucleotide into transplanted colorectal tumor cells to reduce the expression of survivin and inhibit the growth of subcutaneously transplanted colorectal cancer in nude mice. PAMAMs have also been utilized in synthetic drug delivery vehicles due to their properties, such as high loading content, simple synthesis, excellent biodegradability and remarkable versatility (57). Drugs covalently bound to the periphery or loaded into the inner core of a dendrimer are efficiently delivered to the site of action (58, 59). Goldberg et al (58) found that SN38 conjugated with G3.5 dendrimers exhibited enhanced transepithelial transport and reduced gastrointestinal toxicity compared to free SN38 in colorectal cancer cells, demonstrating that dendrimers have enormous potential as carriers in the transport of anticancer drugs. Further research evaluated the anticancer effect of the G3.5-conjugated SN38 via a glycine or β-alanine spacer and found that PAMAM dendrimers significantly enhanced the oral bioavailability as targeted antitumor drug delivery systems (60). PAMAM dendrimers complexed with c-Src antisense effectively decreased c-Src, a member of the non-receptor tyrosine kinase protein family that is overexpressed and activated in a number of human cancer cells and EGFR-dependent downstream genes (61). Morgan et al (62) created a biocompatible polyester dendrimer, able to load three different antitumor drugs, as a novel pharmaceutical carrier that significantly increased cellular uptake and drug retention, while maximizing cytotoxicity toward cancer cells in vitro. PEGylated dendrimers have also shown great promise as drug carriers; they are manufactured in an easy and cost-effective manner, but exhibit higher targeting and anticancer abilities, as van der Poll et al (57) demonstrated in their in vivo study.

Studies in vitro as well as in vivo show promise for the targeted delivery of drugs currently used in clinical practice. Thiagarajan et al (63) synthesized a conjugated macromolecule composed of camptothecin and a polydendrimer, which efficiently inhibited colorectal cancer growth. Nuclear fragmentation was also observed in vitro (63). Lee et al (64) conjugated doxorubicin (DOX) with dendrimers to investigate the pharmacokinetic profiles of attached DOX. The conjugated polymer molecules were then injected into BALB/c mice with subcutaneously transplanted C-26 tumors. The results demonstrated that tumor growth was efficiently inhibited and the life span of the mice was significantly extended (64). Due to the unique properties of PAMAM dendrimers, they have been successfully employed as new MRI contrast agents (65). Khosroshahi et al (66) developed 99mTc-dendrimer G-methionine and found that the newly synthesized complex exhibited better cancer molecular imaging and anticancer properties compared to DTPA-methionine conjugates. Kobayashi et al (67) visualized the vasculature in transplanted tumors using 3D MR angiography. G6-(1B4M-Gd)192 with a generation-6 PAMAM dendrimer core as a newly synthesized MR contrast agent proved to be significantly more useful for intratumoral vasculature imaging compared to Gd-DTPA. The vascular visualization achieved higher scores at all time points during contrast enhancement, without severe side effects (67).

Nanogels

Nano-size hydrogels (often called nanogels) are swollen nano-sized networks formed by non-covalent interactions or covalent cross-linking of polymer chains (68). Given that nanogels have the properties of large surface area-to-volume ratio, size tunability, controlled drug release profile, excellent drug loading capacity and responsiveness to environmental stimuli, they have attracted significant attention in medicine as imaging labels and targeted drug delivery, while reducing systemic side effects (69).

NIR polynagogel (NIR-PNG) was recently considered to be an optimal tracer for SLN navigation surgery in gastric cancer. Kong et al (70) found that IRDye900-conjugated pullulan-cholesterol nanoprobe NIR-PNG exhibited the favorable characteristics of lower dispersion and longer retention in the SLN compared to diluted indocyanine green (ICG) and ICG/poly- γ-glutamic acid complex following injection into the stomachs of dogs and pigs (70). Nanogels may be controlled for various applications in drug delivery and may be tailored to exhibit exceptional stability, low cytotoxicity and higher blood compatibility (71, 72). Nanogels containing 5-fluorouracil (5-FU) were developed to be utilized as a new colon-targeting drug carrier systems, owing to their excellent pH-sensitive release property and effectively reduced toxicity (73). Yim et al (74) found no specific manifestations in organ sections from experimental mice injected with PTX-loaded degradable cationic nanogels in conventional histopathological examinations demonstrating considerable toxicity of the nanoparticles. Nanogels also represent a set of biodegradable nanoparticles that may be utilized as non-viral gene carriers. Heparin-polyethyleneimine (HPEI) nanogels were reported to successfully transfect the plasmid expressing the vesicular stomatitis virus matrix protein (VSVMP) into colon carcinoma cells in vitro and in vivo, exhibiting higher transfection efficiency, lower cytotoxicity and better blood compatibility compared to PEI25K. The development of abdominal and pulmonary metastases was significantly inhibited by intraperitoneal or intravenous injection of the HPEI nanogels loaded with pVSVMP. Yim et al (74) synthesized a biodegradable drug delivery vehicle based on nanogels, which effectively deliver PTX into human cancer xenograft in BALB/c mice. The new complex DpNG-PTX achieved a higher efficacy in inhibiting tumor growth compared to PA-PTX (75). Based on thse findings, nanogels may provide novel opportunities for further advances in drug delivery.

Nanogels have been demonstrated to be potential oral drug delivery systems. The 5-FU nucleoside floxuridine has reported been prepared into polymeric nanogels as a novel oral drug form, which exerts a good antitumor effect on drug-resistant tumors (76). Senanayake et al created an innovative nanoencapsulated drug congjugated with gemcitabine for oral administration in cancer chemotherapy (76). This new medical technology enabled gemcitabine to successfully cross the normally impermeable gastrointestinal barrier and reach the site of interest, according to the results of permeability studies. A good antitumor effect was also observed in vivo in nude mice against several drug-resistant human tumors.

Conclusion and future directions

This review provides an summary of the currently available nanotechnologies developed for the diagnosis and treatment of gastrointestinal tumors. Although several problems and barriers are hampering the development of nanodevices, the potential of nanotechnologies to function as multimodal nanotheranostic agents will likely pave the way for the fight against gastrointestinal cancer and promote the development of personalized therapy for early diagnosis and treatment of cancer.

Acknowledgements

This study was supported by a grants from the Shanghai Science and Technology Department of Medicine Guiding Biomedical Projects (no. 134119a1400) and the Research Fund for the Doctoral Program of Higher Education (New Teacher) (no. 20130071120048).

References

1 

Cuenca AG, Jiang H, Hochwald SN, Delano M, Cance WG and Grobmyer SR: Emerging implications of nanotechnology on cancer diagnostics and therapeutics. Cancer. 107:459–466. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Gdowski A, Ranjan AP, Mukerjee A and Vishwanatha JK: Nanobiosensors: role in cancer detection and diagnosis. Adv Exp Med Biol. 807:33–58. 2014.PubMed/NCBI

3 

Laroui H, Rakhya P, Xiao B, Viennois E and Merlin D: Nanotechnology in diagnostics and therapeutics for gastrointestinal disorders. Dig Liver Dis. 45:995–1002. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Service RF: Nanotoxicology.Nanotechnology grows up. Science. 304:1732–1734. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Zhang GJ and Ning Y: Silicon nanowire biosensor and its applications in disease diagnostics: a review. Anal Chim Acta. 749:1–15. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Jabir NR, Tabrez S, Ashraf GM, Shakil S, Damanhouri GA and Kamal MA: Nanotechnology-based approaches in anticancer research. Int J Nanomedicine. 7:4391–4408. 2012.PubMed/NCBI

7 

Shen MY, Li BR and Li YK: Silicon nanowire field-effect-transistor based biosensors: from sensitive to ultra-sensitive. Biosens Bioelectron. 60:101–111. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Wang WU, Chen C, Lin KH, Fang Y and Lieber CM: Label-free detection of small-molecule-protein interactions by using nanowire nanosensors. Proc Natl Acad Sci USA. 102:3208–3212. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Elnathan R, Kwiat M, Pevzner A, et al: Biorecognition layer engineering: overcoming screening limitations of nanowire-based FET devices. Nano Lett. 12:5245–5254. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Zhang GJ, Chua JH, Chee RE, Agarwal A and Wong SM: Label-free direct detection of m iRNAs with silicon nanowire biosensors. Biosens Bioelectron. 24:2504–2508. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Lee MH, Lee DH, Jung SW, Lee KN, Park YS and Seong WK: Measurements of serum C-reactive protein levels in patients with gastric cancer and quantification using silicon nanowire arrays. Nanomedicine. 6:78–83. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Zheng G, Patolsky F, Cui Y, Wang WU and Lieber CM: Multiplexed electrical detection of cancer markers with nanowire sensor arrays. Nat Biotechnol. 23:1294–1301. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Stern E, Vacic A, Rajan NK, et al: Label-free biomarker detection from whole blood. Nat Nanotechnol. 5:138–142. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Gupta AK, Nair PR, Akin D, et al: Anomalous resonance in a nanomechanical biosensor. Proc Natl Acad Sci USA. 103:13362–13367. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Meckes B, Arce FT, Connelly LS and Lal R: Insulated conducting cantilevered nanotips and two-chamber recording system for high resolution ion sensing AFM. Sci Rep. 4:44542014. View Article : Google Scholar : PubMed/NCBI

16 

Lee JH, Hwang KS, Park J, Yoon KH, Yoon DS and Kim TS: Immunoassay of prostate-specific antigen (PSA) using resonant frequency shift of piezoelectric nanomechanical microcantilever. Biosens Bioelectron. 20:2157–2162. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Savran CA, Knudsen SM, Ellington AD and Manalis SR: Micromechanical detection of proteins using aptamer-based receptor molecules. Anal Chem. 76:3194–3198. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Hansen KM, Ji HF, Wu G, et al: Cantilever-based optical deflection assay for discrimination of DNA single-nucleotide mismatches. Anal Chem. 73:1567–1571. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Liao HS, Huang KY, Hwang IS, Chang TJ, Hsiao WW, Lin HH, Hwu ET and Chang CS: Operation of astigmatic-detection atomic force microscopy in liquid environments. Rev Sci Instrum. 84:1037092013. View Article : Google Scholar : PubMed/NCBI

20 

Gunning AP, Chambers S, Pin C, Man AL, Morris VJ and Nicoletti C: Mapping specific adhesive interactions on living human intestinal epithelial cells with atomic force microscopy. FASEB J. 22:2331–2339. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Krause M, Te Riet J and Wolf K: Probing the compressibility of tumor cell nuclei by combined atomic force-confocal microscopy. Phys Biol. 10:0650022013. View Article : Google Scholar : PubMed/NCBI

22 

Wu TH, Chiou YW, Chiu WT, Tang MJ, Chen CH and Yeh ML: The F-actin and adherence-dependent mechanical differentiation of normal epithelial cells after TGF-beta1-induced EMT (tEMT) using a microplate measurement system. Biomed Microdevices. 16:465–478. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Zrazhevskiy P, Sena M and Gao X: Designing multifunctional quantum dots for bioimaging, detection, and drug delivery. Chem Soc Rev. 39:4326–4354. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Larson DR, Zipfel WR, Williams RM, et al: Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science. 300:1434–1436. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Morgan NY, English S, Chen W, et al: Real time in vivo non-invasive optical imaging using near-infrared fluorescent quantum dots. Acad Radiol. 12:313–323. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Rizvi SB, Ghaderi S, Keshtgar M and Seifalian AM: Semiconductor quantum dots as fluorescent probes for in vitro and in vivo bio-molecular and cellular imaging. Nano Rev. 1:Aug 16–2010. View Article : Google Scholar : PubMed/NCBI

27 

Zhang T, Stilwell JL, Gerion D, et al: Cellular effect of high doses of silica-coated quantum dot profiled with high throughput gene expression analysis and high content cellomics measurements. Nano Lett. 6:800–808. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Wu C, Wang C, Han T, Zhou X, Guo S and Zhang J: Insight into the cellular internalization and cytotoxicity of graphene quantum dots. Adv Healthc Mater. 2:1613–1619. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Zhang H, Fu X, Hu J and Zhu Z: Microfluidic bead-based multienzyme-nanoparticle amplification for detection of circulating tumor cells in the blood using quantum dots labels. Anal Chim Acta. 779:64–71. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Yu Y, Xu L, Chen J, et al: Hydrothermal synthesis of GSH-TGA co-capped CdTe quantum dots and their application in labeling colorectal cancer cells. Colloids Surf B Biointerfaces. 95:247–253. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Gazouli M, Lyberopoulou A, Pericleous P, et al: Development of a quantum-dot-labelled magnetic immunoassay method for circulating colorectal cancer cell detection. World J Gastroenterol. 18:4419–4426. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Bodo J, Durkin L and Hsi ED: Quantitative in situ detection of phosphoproteins in fixed tissues using quantum dot technology. J Histochem Cytochem. 57:701–708. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Ballou B, Ernst LA, Andreko S, et al: Sentinel lymph node imaging using quantum dots in mouse tumor models. Bioconjug Chem. 18:389–396. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Khullar O, Frangioni JV, Grinstaff M and Colson YL: Image-guided sentinel lymph node mapping and nanotechnology-based nodal treatment in lung cancer using invisible near-infrared fluorescent light. Semin Thorac Cardiovasc Surg. 21:309–315. 2009. View Article : Google Scholar : PubMed/NCBI

35 

He Y, Xu H, Chen C, et al: In situ spectral imaging of marker proteins in gastric cancer with near-infrared and visible quantum dots probes. Talanta. 85:136–141. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Jain R, Dandekar P and Patravale V: Diagnostic nanocarriers for sentinel lymph node imaging. J Control Release. 138:90–102. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Soltesz EG, Kim S, Kim SW, et al: Sentinel lymph node mapping of the gastrointestinal tract by using invisible light. Ann Surg Oncol. 13:386–396. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Hikage M, Gonda K, Takeda M, et al: Nano-imaging of the lymph network structure with quantum dots. Nanotechnology. 21:1851032010. View Article : Google Scholar : PubMed/NCBI

39 

Zhang YP, Sun P, Zhang XR and Yang WL: In vitro gastric cancer cell imaging using near-infrared quantum dot-conjugated CC49. Oncol Lett. 4:996–1002. 2012.PubMed/NCBI

40 

Geraldo DA, Duran-Lara EF, Aguayo D, et al: Supramolecular complexes of quantum dots and a polyamidoamine (PAMAM)-folate derivative for molecular imaging of cancer cells. Anal Bioanal Chem. 400:483–492. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Oostendorp M, Douma K, Hackeng TM, et al: Quantitative molecular magnetic resonance imaging of tumor angiogenesis using cNGR-labeled paramagnetic quantum dots. Cancer Res. 68:7676–7683. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Diagaradjane P, Orenstein-Cardona JM, Colon-Casasnovas NE, et al: Imaging epidermal growth factor receptor expression in vivo: pharmacokinetic and biodistribution characterization of a bioconjugated quantum dot nanoprobe. Clin Cancer Res. 14:731–741. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Zhao X, He Y, Gao J, et al: Caveolin-1 expression level in cancer associated fibroblasts predicts outcome in gastric cancer. PLoS One. 8:e591022013. View Article : Google Scholar : PubMed/NCBI

44 

He Y, Zhao X, Gao J, et al: Quantum dots-based immunofluorescent imaging of stromal fibroblasts caveolin-1 and light chain 3B expression and identification of their clinical significance in human gastric cancer. Int J Mol Sci. 13:13764–13780. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Xing Y, Chaudry Q, Shen C, et al: Bioconjugated quantum dots for multiplexed and quantitative immunohistochemistry. Nat Protoc. 2:1152–1165. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Bostick RM, Kong KY, Ahearn TU, Chaudry Q, Cohen V and Wang MD: Detecting and quantifying biomarkers of risk for colorectal cancer using quantum dots and novel image analysis algorithms. Conf Proc IEEE Eng Med Biol Soc. 1:3313–3316. 2006.PubMed/NCBI

47 

Hu WQ, Fang M, Zhao HL, et al: Tumor invasion unit in gastric cancer revealed by QDs-based in situ molecular imaging and multispectral analysis. Biomaterials. 35:4125–4132. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Peng CW, Tian Q, Yang GF, et al: Quantum-dots based simultaneous detection of multiple biomarkers of tumor stromal features to predict clinical outcomes in gastric cancer. Biomaterials. 33:5742–5752. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Peng CW, Liu XL, Chen C, et al: Patterns of cancer invasion revealed by QDs-based quantitative multiplexed imaging of tumor microenvironment. Biomaterials. 32:2907–2917. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Svenson S and Tomalia DA: Dendrimers in biomedical applications - reflections on the field. Adv Drug Deliv Rev. 57:2106–2129. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Kannan RM, Nance E, Kannan S and Tomalia DA: Emerging concepts in dendrimer-based nanomedicine: from design principles to clinical applications. J Intern Med. Jul 3–2014.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

52 

Talanov VS, Regino CA, Kobayashi H, Bernardo M, Choyke PL and Brechbiel MW: Dendrimer-based nanoprobe for dual modality magnetic resonance and fluorescence imaging. Nano Lett. 6:1459–1463. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Davis SS: Biomedical applications of nanotechnology - implications for drug targeting and gene therapy. Trends Biotechnol. 15:217–224. 1997. View Article : Google Scholar : PubMed/NCBI

54 

Teow HM, Zhou Z, Najlah M, Yusof SR, Abbott NJ and D'Emanuele A: Delivery of paclitaxel across cellular barriers using a dendrimer-based nanocarrier. Int J Pharm. 441:701–711. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Dufes C, Keith WN, Bilsland A, Proutski I, Uchegbu IF and Schatzlein AG: Synthetic anticancer gene medicine exploits intrinsic antitumor activity of cationic vector to cure established tumors. Cancer Res. 65:8079–8084. 2005. View Article : Google Scholar : PubMed/NCBI

56 

Li Z, Huang ZH, Cui DX, Yao H, Yu JL, Li Q, Pan BF and Gao F: Polyamidoamine dendrimer-mediated survivin antisense oligonucleotide inhibits the growth of subcutaneously transplanted colorectal cancer in nude mice. J South Med Univ. 28:1935–1938. 2008.(In Chinese).

57 

van der Poll DG, Kieler-Ferguson HM, Floyd WC, et al: Design, synthesis, and biological evaluation of a robust, biodegradable dendrimer. Bioconjug Chem. 21:764–773. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Goldberg DS, Vijayalakshmi N, Swaan PW and Ghandehari H: G3.5 PAMAM dendrimers enhance transepithelial transport of SN38 while minimizing gastrointestinal toxicity. J Control Release. 150:318–325. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Vijayalakshmi N, Ray A, Malugin A and Ghandehari H: Carboxyl-terminated PAMAM-SN38 conjugates: synthesis, characterization, and in vitro evaluation. Bioconjug Chem. 21:1804–1810. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Pan B, Cui D, Sheng Y, et al: Dendrimer-modified magnetic nanoparticles enhance efficiency of gene delivery system. Cancer Res. 67:8156–8163. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Nourazarian AR, Pashaei-Asl R, Omidi Y and Najar AG: c-Src antisense complexed with PAMAM dendrimers decreases c-Src expression and EGFR-dependent downstream genes in the human HT-29 colon cancer cell line. Asian Pac J Cancer Prev. 13:2235–2240. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Morgan MT, Nakanishi Y, Kroll DJ, et al: Dendrimer-encapsulated camptothecins: increased solubility, cellular uptake, and cellular retention affords enhanced anticancer activity in vitro. Cancer Res. 66:11913–11921. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Thiagarajan G, Ray A, Malugin A and Ghandehari H: PAMAM-camptothecin conjugate inhibits proliferation and induces nuclear fragmentation in colorectal carcinoma cells. Pharm Res. 27:2307–2316. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Lee CC, Gillies ER, Fox ME, Guillaudeu SJ, Fréchet JM, Dy EE and Szoka FC: A single dose of doxorubicin-functionalized bow-tie dendrimer cures mice bearing C-26 colon carcinomas. Proc Natl Acad Sci USA. 103:16649–16654. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Tomalia DA, Reyna LA and Svenson S: Dendrimers as multi-purpose nanodevices for oncology drug delivery and diagnostic imaging. Biochem Soc Trans. 35:61–67. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Khosroshahi AG, Amanlou M, Sabzevari O, et al: A comparative study of two novel nanosized radiolabeled analogues of methionine for SPECT tumor imaging. Curr Med Chem. 20:123–133. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Kobayashi H, Sato N, Kawamoto S, et al: 3D MR angiography of intratumoral vasculature using a novel macromolecular MR contrast agent. Magn Reson Med. 46:579–585. 2001. View Article : Google Scholar : PubMed/NCBI

68 

Kabanov AV and Vinogradov SV: Nanogels as pharmaceutical carriers: finite networks of infinite capabilities. Angew Chem Int Ed Engl. 48:5418–5429. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Arunraj TR, Sanoj Rejinold N, Ashwin Kumar N and Jayakumar R: Bio-responsive chitin-poly(L-lactic acid) composite nanogels for liver cancer. Colloids Surf B Biointerfaces. 113:394–402. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Kong SH, Noh YW, Suh YS, et al: Evaluation of the novel near-infrared fluorescence tracers pullulan polymer nanogel and indocyanine green/gamma-glutamic acid complex for sentinel lymph node navigation surgery in large animal models. Gastric Cancer. Jan 31–2014.(Epub ahead of print).

71 

Chacko RT, Ventura J, Zhuang J and Thayumanavan S: Polymer nanogels: a versatile nanoscopic drug delivery platform. Adv Drug Deliv Rev. 64:836–851. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Gou M, Men K, Zhang J, et al: Efficient inhibition of C-26 colon carcinoma by VSVMP gene delivered by biodegradable cationic nanogel derived from polyethyleneimine. ACS Nano. 4:5573–5584. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Ashwanikumar N, Kumar NA, Nair SA and Kumar GV: Methacrylic-based nanogels for the pH-sensitive delivery of 5-fluorouracil in the colon. Int J Nanomedicine. 7:5769–5779. 2012.PubMed/NCBI

74 

Yim H, Park SJ, Bae YH and Na K: Biodegradable cationic nanoparticles loaded with an anticancer drug for deep penetration of heterogeneous tumours. Biomaterials. 34:7674–7682. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Senanayake TH, Warren G and Vinogradov SV: Novel anticancer polymeric conjugates of activated nucleoside analogues. Bioconjug Chem. 22:1983–1993. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Senanayake TH, Warren G, Wei X and Vinogradov SV: Application of activated nucleoside analogs for the treatment of drug-resistant tumors by oral delivery of nanogel-drug conjugates. J Control Release. 167:200–209. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 3 Issue 2

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun B, Fang Y, Li Z, Chen Z and Xiang J: Advances in the application of nanotechnology in the diagnosis and treatment of gastrointestinal tumors (Review). Mol Clin Oncol 3: 274-280, 2015
APA
Sun, B., Fang, Y., Li, Z., Chen, Z., & Xiang, J. (2015). Advances in the application of nanotechnology in the diagnosis and treatment of gastrointestinal tumors (Review). Molecular and Clinical Oncology, 3, 274-280. https://doi.org/10.3892/mco.2014.470
MLA
Sun, B., Fang, Y., Li, Z., Chen, Z., Xiang, J."Advances in the application of nanotechnology in the diagnosis and treatment of gastrointestinal tumors (Review)". Molecular and Clinical Oncology 3.2 (2015): 274-280.
Chicago
Sun, B., Fang, Y., Li, Z., Chen, Z., Xiang, J."Advances in the application of nanotechnology in the diagnosis and treatment of gastrointestinal tumors (Review)". Molecular and Clinical Oncology 3, no. 2 (2015): 274-280. https://doi.org/10.3892/mco.2014.470