Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Molecules ; 26(15)2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34361705

ABSTRACT

In order to seek novel technetium-99m folate receptor-targeting agents, two folate derivatives (CN5FA and CNPFA) were synthesized and radiolabeled to obtain [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA complexes, which exhibited high radiochemical purity (>95%) without purification, hydrophilicity, and good stability in vitro. The KB cell competitive binding experiments indicated that [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA had specificity to folate receptor. Biodistribution studies in KB tumor-bearing mice illustrated that [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA had specific tumor uptake. Compared with [99mTc]Tc-CN5FA, the tumor/muscle ratios of [99mTc]Tc-CNPFA were higher, resulting in a better SPECT/CT imaging background. According to the results, the two 99mTc complexes have potential as tumor imaging agents to target folate receptors.


Subject(s)
Diagnostic Imaging/methods , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Kidney/diagnostic imaging , Nitriles/chemistry , Radiopharmaceuticals/chemical synthesis , Tomography, Emission-Computed, Single-Photon/methods , Animals , Binding, Competitive , Drug Stability , Folate Receptors, GPI-Anchored/genetics , Folic Acid/pharmacokinetics , Gene Expression , Humans , Hydrophobic and Hydrophilic Interactions , KB Cells , Kidney/metabolism , Mice , Protein Binding , Radiopharmaceuticals/pharmacokinetics , Technetium/chemistry , Tissue Distribution
2.
Nucleic Acids Res ; 48(1): 75-85, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31777918

ABSTRACT

One of the major hurdles in RNAi research has been the development of safe and effective delivery systems for siRNAs. Although various chemical modifications have been proposed to improve their pharmacokinetic behaviour, their delivery to target cells and tissues presents many challenges. In this work, we implemented a receptor-targeting strategy to selectively deliver siRNAs to cancer cells using folic acid as a ligand. Folic acid is capable of binding to cell-surface folate receptors with high affinity. These receptors have become important molecular targets for cancer research as they are overexpressed in numerous cancers despite being expressed at low levels in normal tissues. Employing a post-column copper-catalyzed alkyne-azide cycloaddition (CuAAC), we report the synthesis of siRNAs bearing folic acid modifications at different positions within the sense strand. In the absence of a transfection carrier, these siRNAs were selectively taken up by cancer cells expressing folate receptors. We show that centrally modified folic acid-siRNAs display enhanced gene-silencing activity against an exogenous gene target (∼80% knockdown after 0.75 µM treatment) and low cytotoxicity. In addition, these siRNAs achieved potent dose-dependent knockdown of endogenous Bcl-2, an important anti-apoptotic gene.


Subject(s)
Folic Acid/chemistry , Gene Silencing , Gene Targeting/methods , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Carbonates/chemistry , Cell Survival , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Genes, Reporter , HT29 Cells , HeLa Cells , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Pargyline/analogs & derivatives , Pargyline/chemistry , Potassium/chemistry , Protein Binding , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/chemical synthesis , Transfection
3.
Anticancer Agents Med Chem ; 19(12): 1513-1522, 2019.
Article in English | MEDLINE | ID: mdl-31241440

ABSTRACT

BACKGROUND: Glutathione (GSH), which is the predominant low molecular weight intracellular thiol in mammals, has multiple functions, such as those of protecting against oxidative stress and detoxifying endogenous and exogenous electrophiles. High GSH levels, which have been observed in various types of tumors, have been thought to contribute to the resistance of neoplastic cells to apoptotic stimuli triggered by pro-oxidant therapy. Although L-(S,R)-Buthionine Sulfoximine (BSO), a selective irreversible inhibitor of glutamate cysteine ligase, depletes GSH in vitro and in in vivo and sensitizes tumor cells to radiation and some cancer chemotherapeutics, its toxicity and short in vivo half-life have limited its application to combination anticancer therapies. OBJECTIVE: To demonstrate that a folate-targeted PEGylated BSO conjugate can sensitize cancer cells to a Reactive Oxygen Species (ROS)-generating anticancer agent by depleting GSH. METHODS: A novel folate-targeted PEGylated-BSO conjugate was synthesized and tested in combination with gemcitabine in human cell lines that over-express (HeLa) or do not express (A549) the folate receptor. RESULTS: The prepared folate-PEG-GFLG-BSO conjugate proved to be efficacious in reducing GSH levels and, when used in combination with the pro-oxidant drug gemcitabine, it enhanced drug activity in the cell line overexpressing the folate receptor. CONCLUSION: The folate-PEG-GFLG-BSO conjugate studied was found to be effective in sensitizing folatereceptor positive cancer cells to the ROS-generating drug gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Buthionine Sulfoximine/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Folic Acid/pharmacology , Polyethylene Glycols/pharmacology , A549 Cells , Antimetabolites, Antineoplastic/chemistry , Buthionine Sulfoximine/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Folate Receptors, GPI-Anchored/antagonists & inhibitors , Folate Receptors, GPI-Anchored/genetics , Folic Acid/chemistry , HeLa Cells , Humans , Molecular Structure , Polyethylene Glycols/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Gemcitabine
4.
ACS Appl Mater Interfaces ; 11(19): 17193-17203, 2019 May 15.
Article in English | MEDLINE | ID: mdl-31012571

ABSTRACT

Conjugatable nanobimetals exhibiting broadband light absorption for use as phototherapeutic platforms were assembled via a plug-and-play continuous gas flow route. Electrically produced AuCu nanobunches (NBs) under nitrogen gas flow were directly injected into cysteine (cys) solution through gas pressurization to mechanically spray the solution (AuCu into cys droplets). The sprayed droplets were then exposed to 185 nm UV light (higher photon energy [6.2 eV] than the work functions of Au [5.1 eV] and Cu [4.7 eV]) to initiate photoionization of AuCu NBs for subsequent electrostatic reaction with the SH- group of cys to form cys-inserted AuCu (AuCu-cys) platforms in a single-pass gas stream. These platforms exhibited broadband light absorption spectra because of hybridized interparticle plasmonic coupling and could be conjugated to folic acid (FA) when dispersed in FA solution to form highly dispersible, biocompatible, and cancer-targetable AuCu-cys-FA. This material was suitable for use in targeted phototherapy of folate-receptor (FR)-rich cancers via FR-mediated endocytosis, and loading doxorubicin (DOX) into AuCu-cys-FA (i.e., AuCu-cys-DOXFA) facilitated chemo-phototherapy because of photoresponsive anticancer drug release upon induction of hyperthermia.


Subject(s)
Doxorubicin/pharmacology , Drug Delivery Systems , Neoplasms/therapy , Phototherapy , Cell Line, Tumor , Copper/chemistry , Copper/pharmacology , Cysteine/chemistry , Doxorubicin/chemistry , Endocytosis/drug effects , Folate Receptors, GPI-Anchored/chemistry , Folate Receptors, GPI-Anchored/genetics , Folic Acid/chemistry , Gases/chemistry , Gases/pharmacology , Gold/chemistry , Gold/pharmacology , Humans , Light , Neoplasms/pathology , Nitrogen/chemistry , Nitrogen/pharmacology , Solutions/chemistry
5.
EBioMedicine ; 41: 236-243, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30872130

ABSTRACT

BACKGROUND: Non-invasive lung adenocarcinoma could benefit from limited resection, nonetheless, there is a lack of method to determine preoperative tumour invasiveness. We aimed to investigate whether folate receptor-positive circulating tumour cells (FR+-CTCs) in combination with maximum tumour diameter (MTD) determines, before surgery, the invasiveness of small-sized, indeterminate solitary pulmonary nodules (SPNs). METHODS: A total of 382 patients with suspicious lung adenocarcinoma on computed tomography who were expected to undergo lung resection were enrolled in this study at three participating institutes and randomly assigned into training and validation cohorts. Before surgery, 3 mL peripheral blood was collected from all participants. FR+-CTCs were analyzed using immunomagnetic leukocyte depletion and quantitated by ligand-targeted PCR method. After surgery, the resected tissues were diagnosed by pathologists according to IASLC/ATS/ERS classification. FINDINGS: FR+-CTC levels in the peripheral blood can differentiate benign from malignant nodules with a sensitivity of 78·6%-82·7% and a specificity of 68·8%-78·4%. Both FR+-CTC and MTD are independent predictive indices of invasive tumours for lung adenocarcinoma ≤2 cm based on multivariate analyses. Further, FR+-CTC count in combination with MTD can differentiate non-invasive cancers from invasive cancers with a sensitivity of 63·6%-81·8% and a specificity of 71·4%-89·7%. INTERPRETATION: Detection of FR+-CTC is a reliable method to differentiate malignancy of indeterminate SPNs. Combining of FR+-CTC count and MTD could possibly enhance preoperative determination of the invasiveness of lung nodules and guide surgeons to select limited lung resection and avoid overtreatment for patients with non-invasive lesions. FUND: None.


Subject(s)
Adenocarcinoma/blood , Biomarkers, Tumor/blood , Folate Receptors, GPI-Anchored/metabolism , Lung Neoplasms/blood , Neoplastic Cells, Circulating/metabolism , Solitary Pulmonary Nodule/blood , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Female , Folate Receptors, GPI-Anchored/genetics , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Solitary Pulmonary Nodule/pathology
6.
Nanoscale ; 11(11): 5005-5013, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30839969

ABSTRACT

It is acknowledged that the targeting ability of multivalent ligand-modified nanoparticles (MLNs) strongly depends on the ligand spatial presentation determined by ligand valency. However, the receptor overexpression level varies between different types or stages of tumors. Thus, it is essential to explore the influence of ligand valency on the targeting ability of MLNs to tumors with different levels of receptor overexpression. In this study, a dual-acting agent raltitrexed was used as a ligand to target the folate receptor (FR). Different copies of the raltitrexed-modified multivalent dendritic polyethyleneimine ligand cluster PRn (n = 2, 4, and 8) were conjugated onto magnetic nanoparticles to form multivalent magnetic NPs (MMNs) with different valences. The in vitro studies demonstrated that Fe-PR4 was the most effective valency in the treatment of high FR overexpressing KB cells with a decentralized receptor distribution, owing to the fact that Fe-PR2 was negative in statistical rebinding and Fe-PR8 could induce steric hindrance in the limited binding area. Instead, in moderate FR overexpressing HeLa cells with clustered receptor display, the extra ligands on Fe-PR8 would facilitate statistical rebinding more beneficially. Furthermore, in in vivo tumor inhibition and targeted magnetic resonance imaging (MRI) of KB tumors and another moderate FR expressing H22 tumor, similar results were obtained with the cell experiments. Overall, the optimizable treatment effect of Fe-PRn by modulating the ligand valency based on the overexpressing tumor receptor distribution behavior supports the potential of Fe-PRn as a nanomedicine for personalized theranostics.


Subject(s)
Contrast Media/chemistry , Drug Carriers/chemistry , Folate Receptors, GPI-Anchored/metabolism , Magnetite Nanoparticles/chemistry , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems , Endocytosis , Folate Receptors, GPI-Anchored/genetics , Gene Expression , Humans , Magnetic Resonance Imaging , Mice , Neoplasms/drug therapy , Polyethyleneimine/chemistry , Quinazolines/administration & dosage , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Thiophenes/administration & dosage , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Xenograft Model Antitumor Assays
7.
Cytokine Growth Factor Rev ; 45: 45-52, 2019 02.
Article in English | MEDLINE | ID: mdl-30770191

ABSTRACT

The glycoprotein FRα is a membrane-attached transport protein that is shielded from the immune system in healthy cells. However, it is upregulated in various malignancies, involved in cancer development and is also immunogenic. Furthermore, FRα is a tumor-associated antigen endowed with unique properties, thus rendering it a suitable target for immunotherapeutic development in cancer. Various anti- FRα immunotherapeutic strategies are thus currently being developed and clinically assessed for the treatment of various solid tumors. These approaches include passive anti-FRα immunotherapies, such as monoclonal antibodies, or active immunotherapies, such as CART, folate haptens and vaccines. In this review, we will explore the advances in the field of FRα-based immune therapies and discuss both their successes and shortcomings in the clinical setting.


Subject(s)
Folate Receptor 1/antagonists & inhibitors , Folate Receptors, GPI-Anchored/immunology , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Female , Folate Receptor 1/immunology , Folate Receptors, GPI-Anchored/genetics , Humans , Mice , Neoplasms/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy
8.
Colloids Surf B Biointerfaces ; 173: 101-108, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30273870

ABSTRACT

Molybdenum disulfide (MoS2) has been extensively explored for biomedical applications due to its excellent photothermal conversion ability. In this paper, we report a nanoplatform based on folic acid (FA) targeted dual-stimuli responsive MoS2 nanosheets and explore this for the treatment of FA-receptor positive human breast cancer. The nanocomposites generated had a uniform diameter (ca. 133 nm), and could be loaded with the anti-cancer drug doxorubicin (DOX) to a high capacity (151.4 mg/g). The release of DOX was greatly accelerated at pH 5.0 as compared to pH 7.4. In addition, it was found that drug release is enhanced under near infrared laser (NIR) irradiation, showing that the composites can be used as dual responsive systems, with DOX release controllable through pH or NIR irradiation. MTT assays and confocal experiments showed that the MoS2-based nanoplatform could selectively target and kill FA-positive MDA-MB-231 cells (a human breast cancer cell line). The platform also allowed the combination of chemotherapy and photothermal therapy, which led to synergistic effects superior to either monotherapy. The functionalized MoS2 nanoplatform developed in this work hence could be a potent system for targeted drug delivery and synergistic chemo-photothermal cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Combined Modality Therapy/methods , Disulfides/chemistry , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Molecular Targeted Therapy/methods , Molybdenum/chemistry , Nanocomposites/chemistry , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Doxorubicin/chemistry , Drug Compounding/methods , Drug Liberation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Humans , Hydrogen-Ion Concentration , Infrared Rays , Kinetics , Lasers , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mammary Glands, Human/radiation effects , Particle Size , Protein Binding
9.
Biomater Sci ; 6(7): 1818-1833, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29785434

ABSTRACT

Combinational cancer therapy has been considered as a promising strategy to achieve synergetic therapeutic effects and suppression of multidrug resistance. Herein, we adopted a combination of methotrexate (MTX), an antimetabolite acting on cytoplasm, and 10-hydroxycamptothecin (HCPT), an alkaloid acting on nuclei, to treat cancer. Given the different solubilities, membrane permeabilities, and anticancer mechanisms of both drugs, we developed a dual-targeting delivery system based on 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-hyaluronic acid (a principal ligand of CD44 receptors)-MTX (a selective ligand of folate receptors) nanoparticles, which was exploited to carry HCPT-MTX conjugate for synergistically boosting dual-drug co-delivery. The HCPT-MTX conjugate was synthesized by a blood-stable yet intracellularly hydrolysable ester bond. The core-shell-corona DSPE-HA-MTX nanoparticles encapsulating HCPT-MTX (HCPT-MTX@DHM) exhibited high drug entrapment efficiency (∼91.8%) and pH/esterase-controlled release behavior. Cellular uptake studies confirmed significant increase in the efficiency of selective internalization of HCPT-MTX@DHM via CD44/folate receptors compared with those of DSPE-HA nanoparticles encapsulating HCPT-MTX (HCPT-MTX@DH), both drugs, or each individual drug. Furthermore, in vivo near-infrared fluorescence and photoacoustic dual-modal imaging indicated that DiR-doped HCPT-MTX@DHM nanoparticles efficiently accumulated at the tumor sites through passive-plus-active targeting. Finally, the synergistic active targeting and synchronous dual-drug release at a synergistic drug-to-drug ratio resulted in highly synergetic tumor cell-killing and tumor growth inhibition in MCF-7 tumor-bearing mice. Therefore, HCPT-MTX@DHM nanoparticles can be an efficient and smart platform for tumor-targeting therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Hyaluronic Acid/chemistry , Methotrexate/pharmacology , Phosphatidylethanolamines/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Camptothecin/chemistry , Camptothecin/pharmacology , Drug Carriers , Drug Compounding/methods , Drug Synergism , Female , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Humans , MCF-7 Cells , Methotrexate/chemistry , Mice , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
Birth Defects Res ; 110(12): 973-981, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29732742

ABSTRACT

BACKGROUND: Folate receptors (FRs) facilitate embryonic uptake of folates and are important for proper early embryonic development. There is accumulating evidence that blocking FR autoantibodies contribute to developmental diseases. However, genetic factors associated with the expression of FR autoantibodies remain unknown. OBJECTIVE: We investigated the effects of genetic polymorphisms in folate pathway genes on FR autoantibody titers in women. METHODS: We recruited 302 pregnant women in China. The FR antigen-down immunoassay was used to measure levels of FR autoantibodies including human immunoglobulin G (IgG) and immunoglobulin M (IgM) in maternal plasma. Genotypes were identified by matrix-assisted laser desorption/ionization time of flight mass spectrometry and polymerase chain reaction methods. General linear model was used to analyze the effects of genetic variants on FR autoantibody levels. RESULTS: Significant associations were observed between genotypic variations and levels of FR autoantibodies. Plasma levels of FR autoantibodies in women with the TT genotype at MTHFR rs1801133 were significantly higher than those of women with the CC genotype (IgG: ß = 0.62, 95% CI 0.21-1.04; IgM: ß = 0.42, 95% CI 0.12-0.72). For DNMT3A rs7560488, the level of FR autoantibody IgG significantly increased in the TT genotype compared with CC genotype (ß = 0.90, 95% CI 0.20-1.59). For MTHFD2 rs828903, genotype GG was associated with elevated levels of FR autoantibody IgM compared to the AA genotype (ß = 0.60, 95% CI 0.10-1.10). No association was detected between genetic variants of the DHFR gene with FR autoantibodies levels. CONCLUSION: Genetic variations in MTHFR, DNMT3A, and MTHFD2 genes were associated with elevated plasma levels of FR autoantibodies.


Subject(s)
Autoantibodies/blood , Folate Receptors, GPI-Anchored/genetics , Folic Acid/metabolism , Genetic Variation , Adult , Case-Control Studies , Female , Homocystinuria/genetics , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity/genetics , Neural Tube Defects/genetics , Polymorphism, Single Nucleotide/genetics , Pregnancy , Psychotic Disorders/genetics
11.
Talanta ; 182: 306-313, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29501157

ABSTRACT

A novel dual-target recognition sandwich strategy for selective capture and detection of MCF-7 breast cancer cells based on core-shell magnetic mesoporous silica (Fe3O4@nSiO2@mSiO2@apt) nanoparticles was developed. Fe3O4@nSiO2@mSiO2@apt nanoparticles, which were prepared by a layer-by-layer method and were used for the first time to capture cancer cells, have large surface areas, particularly accessible mesochannels, and good biocompatibility, enabling aptamers to be compactly anchored onto the surface of the core-shell magnetic nanoparticles. A mucin 1 protein (MUC1)-targeted Fe3O4@nSiO2@mSiO2@apt nanoparticle was used as an affinity magnetic isolate material to capture target MCF-7 cells selectively and to reduce interference through affinity interaction between the anti-MUC1 aptamer and the MUC1 protein over-expressed on the surface of the MCF-7 cells. Meanwhile, a folate receptor (FR)-targeted affinity fluorescent probe (FA-BSA-FITC) was developed by coupling folic acid and FITC to the surface of BSA, enabling high sensitivity, selective fluorescent labeling of FR over-expressed MCF-7 cells. A dual-target recognition sandwich assay was developed based on the MUC1-targeted magnetic nanoparticles and the FR-targeted fluorescent probes. Under optimum conditions, a quantitative assay of MCF-7 cells was achieved with a dynamic range of 102-105 cells/mL (R2 = 0.9991). This assay showed high specificity and sensitivity to the target MCF-7 cells. Finally, the proposed strategy could be extended to detect MCF-7 cells in human plasma and whole blood with a recovery range of 86.1-104.0% and a RSD range of 1.2-8.4%, respectively. This indicates that the dual-target recognition method developed in this research exhibits good selectivity, anti-interference capability, and reliability even in plasma and whole blood samples and is more suitable for complex samples than previous targeted assays. Therefore, the approach proposed here may have great potential for early breast cancer diagnosis.


Subject(s)
Biological Assay , Breast Neoplasms/diagnosis , Cell Separation/methods , Magnetite Nanoparticles/chemistry , Neoplastic Cells, Circulating/metabolism , Aptamers, Peptide/chemical synthesis , Aptamers, Peptide/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Ferrosoferric Oxide/chemistry , Fluorescein-5-isothiocyanate/chemistry , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Gene Expression , Humans , MCF-7 Cells , Magnetite Nanoparticles/ultrastructure , Mucin-1/genetics , Mucin-1/metabolism , Neoplastic Cells, Circulating/immunology , Neoplastic Cells, Circulating/pathology , Particle Size , Protein Binding , Protein Transport , Serum Albumin, Bovine/chemistry , Silicon Dioxide/chemistry
12.
Mikrochim Acta ; 185(3): 205, 2018 03 03.
Article in English | MEDLINE | ID: mdl-29594762

ABSTRACT

Water-soluble and functional copper nanoclusters (CuNCs) were prepared by using folic acid (FA) that serves both as a reducing reagent and a stabilizer. FA also acts as a functional ligand on the surface of the CuNCs, and this can be exploited to target the folate receptor which is over-expressed on the surface of HeLa cells. The FA-modified CuNCs nanoclusters have an average size of ca. 0.9 nm and are stable in aqueous medium for 30 days. Under photoexcitation at λex 270 and 350 nm, the FA-CuNCs display strong blue fluorescence with an emission peak at 440 nm. The FA-CuNCs exhibit low cytotoxicity and favorable biocompatibility as demonstrated by an MTT assay. A cell viability of >80% is found when incubating HeLa cells for 20 h with FA-CuNCs at levels of up to 200 µg mL-1. The targeting capability of the FA-CuNCs is demonstrated by live cell imaging. It is shown that HeLa cells with over-expressed folate receptor are much brighter than A549 cells where the receptor is not over-expressed. This is further corroborated by the fact that the copper content in HeLa cells (1.5 pg/cell) is 6.5-fold higher than that of A549 cells (0.23 pg/cell), both measured after the same incubation time of 3 h. If free FA is introduced into the cell culture medium, the folate receptors will be preoccupied with FA, and this results in a significant decrease in the cellular uptake of the FA-CuNCs by HeLa cells. Graphical Abstract Biocompatible copper nanoclusters (CuNCs) coated with folic acid (FA) were prepared and are shown to be viable probes for the differentiation between FR-positive HeLa cells and FR-negative A549 cells.


Subject(s)
Copper/chemistry , Folate Receptors, GPI-Anchored/genetics , Folic Acid/chemistry , Nanostructures/chemistry , Optical Imaging/methods , A549 Cells , Biological Transport , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Gene Expression , HeLa Cells , Humans , Water/chemistry
13.
Mol Ther ; 26(2): 390-403, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29241970

ABSTRACT

Non-small cell lung cancer (NSCLC) is the number one cancer killer in the United States. Despite attempted curative surgical resection, nearly 40% of patients succumb to recurrent disease. High recurrence rates may be partially explained by data suggesting that 20% of NSCLC patients harbor synchronous disease that is missed during resection. In this report, we describe the use of a novel folate receptor-targeted near-infrared contrast agent (OTL38) to improve the intraoperative localization of NSCLC during pulmonary resection. Using optical phantoms, fluorescent imaging with OTL38 was associated with less autofluorescence and greater depth of detection compared to traditional optical contrast agents. Next, in in vitro and in vivo NSCLC models, OTL38 reliably localized NSCLC models in a folate receptor-dependent manner. Before testing intraoperative molecular imaging with OTL38 in humans, folate receptor-alpha expression was confirmed to be present in 86% of pulmonary adenocarcinomas upon histopathologic review of 100 human pulmonary resection specimens. Lastly, in a human feasibility study, intraoperative molecular imaging with OTL38 accurately identified 100% of pulmonary adenocarcinomas and allowed for identification of additional subcentimeter neoplastic processes in 30% of subjects. This technology may enhance the surgeon's ability to identify NSCLC during oncologic resection and potentially improve long-term outcomes.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Contrast Media , Folate Receptors, GPI-Anchored/metabolism , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Molecular Imaging , Adenocarcinoma/genetics , Adenocarcinoma/surgery , Animals , Disease Models, Animal , Female , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folate Receptors, GPI-Anchored/genetics , Gene Expression , Humans , Immunohistochemistry , Intraoperative Care , Lung Neoplasms/genetics , Lung Neoplasms/surgery , Male , Mice , Molecular Imaging/methods , Xenograft Model Antitumor Assays
14.
Colloids Surf B Biointerfaces ; 162: 76-89, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29154189

ABSTRACT

The novel drug delivery system based on self-assembly of zinc phthalocyanine-soybean phosphatidylcholine (ZnPc-SPC) complex was developed by a co-solvent method followed by a nanoprecipitaion technique. DSPE-PEG-methotrexate (DSPE-PEG-MTX) was introduced on the surface of ZnPc-SPC self-assembled nanoparticles (ZS) to endow them with folate receptor-targeting property. NMR, XRD, FTIR, and UV-vis-NIR analysis demonstrated the weak molecular interaction between ZnPc and SPC. The ZS functionalized with DSPE-PEG-MTX (ZSPM) was successfully constructed with an average particle size of ∼170nm, a narrow size distribution, and could remain physiologically stable for at least 7days. In vitro cellular uptake and cytotoxicity studies demonstrated that ZSPM exhibited stronger cellular uptake efficacy and photodynamic cytotoxicity against HeLa and MCF-7 cells than ZS functionalized with DSPE-mPEG (ZSP) and free ZnPc. More importantly, ZSPM showed the enhanced accumulation effect at the tumor region compared with ZSP by the active-plus-passive targeting via enhanced permeability and retention (EPR) effect and folate receptor-mediated endocytosis. Furthermore, in vivo antitumor effect and histological analysis demonstrated the superior tumor growth inhibition effect of ZSPM. In addition, the needle-shape ZSP (ZSPN) exhibited better in vitro cellular uptake and in vivo tumor accumulation compared with ZSP due to the shape-assisted effect. Moreover, the interesting off-on switch effect of reactive oxygen species (ROS) production of ZnPc-SPC complex-based nanoparticles was discovered to achieve photodynamic treatment in a controllable way. These findings suggested that the ZnPc-SPC complex-based self-assembled nanoparticles could serve as a promising and effective formulation to achieve tumor-targeting fluorescence imaging and enhanced photodynamic treatment.


Subject(s)
Drug Delivery Systems/methods , Indoles/pharmacology , Mammary Neoplasms, Animal/therapy , Methotrexate/pharmacology , Nanoparticles/chemistry , Organometallic Compounds/pharmacology , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/agonists , Animals , Endocytosis , Female , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , HeLa Cells , Humans , Indoles/pharmacokinetics , Injections, Subcutaneous , Isoindoles , MCF-7 Cells , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Methotrexate/pharmacokinetics , Mice , Mice, Nude , Nanoparticles/administration & dosage , Optical Imaging/methods , Organometallic Compounds/pharmacokinetics , Particle Size , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Photochemotherapy/methods , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Reactive Oxygen Species/metabolism , Theranostic Nanomedicine/methods , Zinc Compounds
15.
Mol Ther ; 25(7): 1544-1555, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28109960

ABSTRACT

Targeted inhibition of oncogenic miRNA-21 has been proposed to treat glioblastoma by rescuing tumor suppressors, PTEN and PDCD4. However, systemic delivery of anti-miR-21 sequences requires a robust and efficient delivery platform to successfully inhibit this druggable target. Three-way-junction (3WJ)-based RNA nanoparticles (RNP), artificially derived from pRNA of bacteriophage phi29 DNA packaging motor, was recently shown to target glioblastoma. Here, we report that multi-valent folate (FA)-conjugated 3WJ RNP constructed to harbor anti-miR-21 LNA sequences (FA-3WJ-LNA-miR21) specifically targeted and delivered anti-miR-21 LNA and knocked down miR-21 expression in glioblastoma cells in vitro and in vivo with favorable biodistribution. Systemically injected FA-3WJ-LNA-miR21 RNP efficiently rescued PTEN and PDCD4, resulting in glioblastoma cell apoptosis and tumor growth regression. Overall survival rate was also significantly improved by FA-3WJ-LNA-miR21 RNP. These results are indicative of the clinical benefit of FA-3WJ RNP-based gene therapy for the successful targeted therapy of developing and even recurring glioblastoma.


Subject(s)
Antagomirs/pharmacology , Brain Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Glioblastoma/therapy , MicroRNAs/antagonists & inhibitors , Nanoparticles/administration & dosage , Animals , Antagomirs/chemistry , Antagomirs/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Carriers , Female , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Nanoparticles/chemistry , Oligonucleotides/chemistry , Oligonucleotides/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
16.
Nanotechnology ; 28(8): 085603, 2017 Feb 24.
Article in English | MEDLINE | ID: mdl-28055982

ABSTRACT

Herein, we prepared folate-targeting core crosslinked polymeric micelles (CCL/FA) containing multiple disulfide bonds located at the interface and core of the micelles to co-deliver doxorubicin (DOX) and the P-glycoprotein (P-gp) inhibitor tariquidar (TQR) for reversing drug resistance. The stability and redox-responsive behavior of the CCL/FA micelles was evaluated through the changes in morphology, molecular weight and hydrodynamic size. On the one hand, the micelles possessed good stability, which led to the suppression of drug release from the CCL micelles in the physiological environment. On the other hand, under reductive conditions, the CCL micelles collapsed rapidly and accelerated drug release markedly. In vitro cytotoxicity measurements, combined with confocal laser scanning microscopy (CLSM) and flow cytometry, confirmed that the dual-drug-loaded micelles exhibited obviously higher cytotoxicity to MCF-7/ADR-resistant cells than free DOX · HCl, single-drug loaded CCL micelles and nontargeted CCL micelles. The results imply that co-delivering DOX and TQR by CCL/FA micelles may be a promising way of overcoming multidrug resistance in tumor treatments.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Carriers , Drug Resistance, Neoplasm/drug effects , Polymers/chemistry , Quinolines/pharmacology , Antibiotics, Antineoplastic/chemistry , Biological Transport, Active/drug effects , Doxorubicin/chemistry , Drug Compounding/methods , Drug Liberation , Drug Synergism , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Gene Expression , HeLa Cells , Humans , Kinetics , MCF-7 Cells , Micelles , Molecular Targeted Therapy , Polyethylene Glycols/chemistry , Pyridines/chemistry , Quinolines/chemistry
17.
Zhongguo Fei Ai Za Zhi ; 19(12): 813-820, 2016 Dec 20.
Article in Chinese | MEDLINE | ID: mdl-27978866

ABSTRACT

BACKGROUND: The aim of this study is to determine the efficacy and feasibility of a novel folate receptor (FR)-based circulating tumor cell (CTC) detection method in the diagnosis of lung cancer. CTCs were collected from 3 mL of blood based on negative enrichment by immunomagnetic beads and then labeled by a conjugate of a tumor-specific ligand folate and an oligonucleotide. METHODS: After washing off redundant conjugates, the bound conjugates were removed and analyzed by quantitative polymerase chain reaction. RESULTS: The CTC levels of 97 patients with lung cancer were significantly higher than that of the controls (18 patients with benign lung diseases; P<0.001). With a threshold of 8.7 Folate units, the method showed a sensitivity of 82.5% and a specificity of 72.2% in the diagnosis of lung cancer, especially a sensitivity of 86.8% in stage I disease. Compared with the existing clinical biomarkers such as neuron-specific enolase (NSE), carcinoembryonic antigen (CEA), and CYFRA21-1, the method showed the highest diagnostic efficiency in lung cancer (area under the curve, 0.859; 95%CI: 0.779-0.939) and stage I lung cancer (area under the curve, 0.912; 95%CI: 0.829-0.994). For future work, the CTC levels of 5 lung cancer patients higher than 8.7 Folate units/3 mL in their postoperation. CONCLUSIONS: FR-positive CTCs were feasible diagnostic biomarkers in patients with lung cancer, as well as in early-stage tumors.


Subject(s)
Folate Receptors, GPI-Anchored/blood , Lung Neoplasms/diagnosis , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Biomarkers, Tumor/blood , Carcinoembryonic Antigen/blood , Female , Folate Receptors, GPI-Anchored/genetics , Humans , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplastic Cells, Circulating/pathology , Pilot Projects
18.
J Colloid Interface Sci ; 482: 121-130, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27497233

ABSTRACT

We report that through facile lipid self-assembly, biomimetic membrane-conjugated mesoporous silica-coated graphene oxide is constructed as targeting nanocarrier toward efficient combination of photothermal therapy and chemotherapy. Impressively, the simple surface modification with folate-contained lipid bilayer allows the graphene-based nanoarchitecture above to be selectively internalized by tumor cells overexpressing relevant receptors. Compared to pure drug, 7-fold doxorubicin is delivered into tumor cells by the nanoarchitecture. After cellular internalization, upon near infrared light illumination, graphene oxide in the nanoarchitecture can convert light energy into heat to kill cancer cells partially. Simultaneously, hyperthermia will drive rapid release of doxorubicin from the nanoarchitecture above to further cause the death of more cancer cells. Thus, integrated cancer treatment with higher efficacy is achieved in vitro compared to that of individual therapy.


Subject(s)
Biomimetic Materials/chemical synthesis , Drug Carriers , Nanoconjugates/chemistry , Phototherapy/methods , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Biomimetic Materials/pharmacology , Biomimetic Materials/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Combined Modality Therapy , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Compounding , Endocytosis , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Gene Expression , Graphite/chemistry , HeLa Cells , Humans , Infrared Rays , Lipid Bilayers/chemistry , MCF-7 Cells , Nanoconjugates/radiation effects , Nanoconjugates/ultrastructure , Silicon Dioxide/chemistry
19.
Dalton Trans ; 44(37): 16304-12, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26299897

ABSTRACT

Magnetic fluorescent nanoparticles (NPs) have great potential applications for diagnostics, imaging and therapy. We developed a facile polyol method to synthesize multifunctional Fe3O4@CeF3:Tb@CeF3 NPs with small size (<20 nm), high water solubility and good biocompatibility. The NPs were modified by ligand exchange reactions with citric acid (CA) to obtain carboxyl-functionalized NPs (Fe3O4@CeF3:Tb@CeF3-COOH). Folic acid (FA) as an affinity ligand was then covalently conjugated onto NPs to yield Fe3O4@CeF3:Tb@CeF3-FA NPs. They were then applied as multimodal imaging agents for simultaneous in vitro targeted fluorescence imaging and magnetic resonance imaging (MRI) of HeLa cells with overexpressed folate receptors (FR). The results indicated that these NPs had strong luminescence and enhanced T2-weighted MR contrast and would be promising candidates as multimodal probes for both fluorescence and MRI imaging.


Subject(s)
Cesium/chemistry , Fluorides/chemistry , Folic Acid/chemistry , Nanoparticles/chemistry , Terbium/chemistry , Cell Survival/drug effects , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , HeLa Cells , Humans , MCF-7 Cells , Magnetic Resonance Imaging , Magnetics , Microscopy, Confocal , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Spectroscopy, Fourier Transform Infrared
20.
J Thorac Oncol ; 10(8): 1163-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26200270

ABSTRACT

INTRODUCTION: To investigate the diagnostic performance of folate receptor-positive circulating tumor cells in distinguishing non-small-cell lung cancer (NSCLC) from lung benign disease by using a novel ligand-targeted polymerase chain reaction (PCR) detection technique. METHODS: Circulating tumor cells were enriched from 3-ml peripheral blood by immunomagnetic depletion of leukocytes and then labeled with a conjugate of a tumor-specific ligand folic acid and a synthesized oligonucleotide. After washing off free conjugates, the stripped bound conjugates were analyzed by quantitative PCR. RESULTS: Seven hundred fifty-six participants (473 patients with NSCLC, 227 patients with lung benign disease, and 56 healthy donors) were randomly assigned to a training set and a test set. The circulating tumor cell (CTC) levels in patients with NSCLC were significant higher than those with lung benign disease (p < 0.001) and healthy donors (p < 0.001). Compared with carcinoembryonic antigen, neuron-specific enolase, and Cyfra21-1, CTCs displayed the highest area under the receiver operating characteristic curve (training set, 0.815; validation set, 0.813) in the diagnosis of NSCLC, with a markedly sensitivity (training set, 72.46%; validation set, 76.37%) and specificity (training set, 88.65%; validation set, 82.39%). The model combining CTCs with carcinoembryonic antigen, neuron-specific enolase, and Cyfra21-1 was more effective for the diagnosis of NSCLC than tumor makers alone (sensitivity and specificity in the training set, 84.21% and 83.91%; validation set, 88.78% and 87.36%, respectively). In addition, the CTC levels were higher in patients with stage III/IV NSCLC compared with those with stage I/II disease. CONCLUSION: Ligand-targeted PCR technique was feasible and reliable for detecting folate receptor-positive CTCs in patients with NSCLC, and CTC levels could be used as a useful biomarker for the diagnosis of NSCLC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Folate Receptors, GPI-Anchored/blood , Lung Neoplasms/diagnosis , Neoplastic Cells, Circulating/chemistry , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Area Under Curve , Bronchiectasis/blood , Bronchiectasis/diagnosis , Carcinoembryonic Antigen/blood , Carcinoma, Non-Small-Cell Lung/blood , Case-Control Studies , Diagnosis, Differential , Double-Blind Method , Female , Folate Receptors, GPI-Anchored/genetics , Humans , Keratin-19/blood , Ligands , Lung Neoplasms/blood , Male , Middle Aged , Phosphopyruvate Hydratase/blood , Pneumonia/blood , Pneumonia/diagnosis , Pneumothorax/blood , Pneumothorax/diagnosis , Polymerase Chain Reaction/methods , Prospective Studies , ROC Curve , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/diagnosis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...