Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 294
Filter
1.
Biochim Biophys Acta Gen Subj ; 1867(9): 130396, 2023 09.
Article in English | MEDLINE | ID: mdl-37271407

ABSTRACT

BACKGROUND: Liposomes are predominantly used sorts of nanocarriers for active a targeted delivery through surface functionalization using targeting ligand. The folate receptors are overexpressed in various cancers including breast cancer and because of its binding aptitude specifically to folate receptors, folic acid became the attractive ligand. METHODS: In this research, we have developed a folate and Poly-l-Lysine conjugate and coated this conjugate onto the liposomes. The prepared liposomes were characterized using DLS, FTIR, NMR, SEM, TEM, XRD, AFM, stability and drug release studies. Furthermore, in vitro studies were carried out on FR overexpressed breast cancer cell line. RESULTS: The FA-LUT-ABC-Lip have diameter of 183 ± 3.17 nm with positive surface charge +33.65 ± 3 mV and the drug release studies confirm the NIR responsive payload cleavage. The coated formulation (in presence of NIR light) effectively reduced the IC50 values and kills breast cancer cells through FR mediated internalization and accelerated drug release. Moreover, LUT Formulation shows anticancer effect due to significant inhibition of cell migration and proliferation by regulating VEGF expression and induced apoptosis through the caspase-3 up-regulation. CONCLUSION: It is evident from the in vitro studies that the formulation was found to be very effective and can be explored for triggered and targeted delivery of the substances through active targeting. GENERAL SIGNIFICANCE: Combining receptor mediated drug delivery with triggered release aid in more amounts of drug reaching the target site and achieving enhanced therapeutic activity.


Subject(s)
Breast Neoplasms , Liposomes , Humans , Female , Liposomes/chemistry , Breast Neoplasms/drug therapy , Ligands , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism
2.
Bioorg Med Chem Lett ; 86: 129240, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36931350

ABSTRACT

Folate receptors (FRs) are known to be over-expressed in several human malignancies and therefore serve as an important target for small radiolabeled folate derivatives for non-invasive imaging of tumor, which is an important tool for future treatment recourse. In the present article, we report the synthesis of a new 99mTc-labeled radiotracer for the aforementioned application following the well-established 99mTc-'4+1' chemistry. Formation of the desired [99mTc]Tc-complex with >95% radiochemical purity was confirmed by radio-HPLC and its structure was ascertained by characterizing a natural rhenium analogue of the said complex. Although the ligand exhibited a weaker affinity towards FRs compared to native folic acid (IC50 8.09 µM vs 29.46 nM), the 99mTc-labeled complex was found to bind folate receptor-positive KB cells with high specificity (∼90%). Similar studies in a folate receptor negative cell line viz. A549 further corroborated the receptor-specificity of the synthesized complex. In vivo studies in KB tumor xenograft showed moderate uptake of ∼2.6% upto 3 h post-injection with high specificity (∼80%). The favorable features observed warrant further screening of the current design towards achieving an improved molecular probe for the said application.


Subject(s)
Folic Acid , Neoplasms , Humans , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Radiopharmaceuticals , Carrier Proteins/metabolism , Technetium/chemistry
3.
Chem Biol Interact ; 351: 109731, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34728188

ABSTRACT

We designed amine-functionalized nanocrystalline cellulose grafted folic acid/magnetic nanoparticles (AF-NCC/Fe3O4 NPs) against folate receptors for targeted delivery of doxorubicin (DOX). Toxicity is a major side effect of DOX, damaging vital organs such as the heart, kidney, and liver; for example, it causes dilated cardiomyopathy and hepatotoxicity. Accordingly, we aimed to reduce this adverse effect and increase the targeted delivery of DOX to the right point of cancer cells by using the unique features of cancer cells. The characterizations were approved in each step using Fourier transform infrared (FTIR), scanning electron microscope (SEM), X-ray diffraction (XRD), transmission electron microscopy (TEM), energy dispersive X-ray (EDX), zeta potential, and dynamic light scattering (DLS) analysis techniques. Encapsulation efficacy of AF-NCC/Fe3O4 NPs was 99.6%; drug release investigations showed excellent stability in physiological conditions (pH âˆ¼ 7.4) and a high release rate in the low pH condition of cancer environments (pH âˆ¼ 5.0). The hemolysis assay and Masson's trichrome and hematoxylin and eosin (H&E) staining results showed that the nanocarrier was entirely biocompatible. In vitro cell viability study approved that the designed nanocarrier increased the therapeutic effects of DOX on Saos-2 cells. The cellular internalization results displayed a high percentage of uptake within 2 h. Real-time reverse transcriptase-polymerase chain reaction (RT-PCR) was applied for the evaluation of tumor protein p53 (p53), p21, and Bcl-2-associated X protein (Bax). DOX exerted its effects through DNA damage and oxidative stress that led to p53 upregulation, and p53 inhibited cell cycle progression. This arrest initiated apoptosis and inhibited cell migration. In summary, encapsulating DOX in AF-NCC/Fe3O4 NPs dramatically decreases the toxic effects of this chemotherapeutic agent on vital organs, especially on the heart. This smart nanocarrier increases the delivery of DOX using acid folic on its surface and also enhances the DOX release in the acidic environment of cancer cells. DOX exerts its therapeutic effects by the initiation of apoptosis and inhibition of migration.


Subject(s)
Antineoplastic Agents/pharmacology , Cellulose/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Magnetite Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cellulose/metabolism , Cellulose/toxicity , Doxorubicin/chemistry , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Carriers/toxicity , Drug Liberation , Female , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/analogs & derivatives , Folic Acid/metabolism , Folic Acid/toxicity , Humans , Magnetite Nanoparticles/toxicity , Mice, Inbred BALB C
4.
Cells ; 10(9)2021 08 25.
Article in English | MEDLINE | ID: mdl-34571839

ABSTRACT

The aim of this study was to fabricate a reactive oxygen species (ROS)-sensitive and folate-receptor-targeted nanophotosensitizer for the efficient photodynamic therapy (PDT) of cervical carcinoma cells. Chlorin e6 (Ce6) as a model photosensitizer was conjugated with succinyl ß-cyclodextrin via selenocystamine linkages. Folic acid (FA)-poly(ethylene glycol) (PEG) (FA-PEG) conjugates were attached to these conjugates and then FA-PEG-succinyl ß-cyclodextrin-selenocystamine-Ce6 (FAPEGbCDseseCe6) conjugates were synthesized. Nanophotosensitizers of FaPEGbCDseseCe6 conjugates were fabricated using dialysis membrane. Nanophotosensitizers showed spherical shapes with small particle sizes. They were disintegrated in the presence of hydrogen peroxide (H2O2) and particle size distribution changed from monomodal distribution pattern to multimodal pattern. The fluorescence intensity and Ce6 release rate also increased due to the increase in H2O2 concentration, indicating that the nanophotosensitizers displayed ROS sensitivity. The Ce6 uptake ratio, ROS generation and cell cytotoxicity of the nanophotosensitizers were significantly higher than those of the Ce6 itself against HeLa cells in vitro. Furthermore, the nanophotosensitizers showed folate-receptor-specific delivery capacity and phototoxicity. The intracellular delivery of nanophotosensitizers was inhibited by folate receptor blocking, indicating that they have folate-receptor specificity in vitro and in vivo. Nanophotosensitizers showed higher efficiency in inhibition of tumor growth of HeLa cells in vivo compared to Ce6 alone. These results show that nanophotosensitizers of FaPEGbCDseseCe6 conjugates are promising candidates as PDT of cervical cancer.


Subject(s)
Folate Receptors, GPI-Anchored/metabolism , Nanoparticles/administration & dosage , Oxidation-Reduction/drug effects , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Uterine Cervical Neoplasms/drug therapy , beta-Cyclodextrins/pharmacology , Animals , Cell Line , Cell Line, Tumor , Chlorophyllides , Female , Folic Acid/metabolism , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Particle Size , Photochemotherapy/methods , Uterine Cervical Neoplasms/metabolism
5.
Biomed Pharmacother ; 142: 111985, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34352716

ABSTRACT

Gold nanoparticles and their conjugates have significant potential in the field of diagnosis of various diseases due to their SPR, which enhances light scattering and absorption. Conjugates of gold nanoparticles with various ligands can be used for imaging biomolecules or detecting malignant neoplasms at an early stage. This study focuses on the construction of composite (or hybrid) phospholipid-gold nanoparticles using soy phosphatidylcholine and a targeted ligand (folic acid derivative) to attach specific targeting properties. According to the method of dynamic light scattering, the diameter of the obtained nanoparticles was less than 100 nm, the results of the MTT test indicated their moderate cytotoxicity. In vitro and in vivo experiments showed a significant increase in the accumulation of phospholipid-gold nanoparticles with a targeted fragment compared to those without a targeted fragment both in HeLa cells and in a tumor (in BDF mice with an injected LLC tumor). The resulting nanoparticles are suitable for specific delivery into tumor cells and visualization of various malignant neoplasms, including at early stages, due to the increased expression of the folate receptor characteristic of cells of a wide range of tumors.


Subject(s)
Drug Delivery Systems , Folic Acid/pharmacology , Gold , Metal Nanoparticles , Animals , Carcinoma, Lewis Lung/drug therapy , Dynamic Light Scattering , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/administration & dosage , Folic Acid/chemistry , HeLa Cells , Humans , Ligands , Male , Mice , Particle Size , Phosphatidylcholines/chemistry , Phospholipids/chemistry , Glycine max/chemistry , Surface Plasmon Resonance
6.
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
7.
Drug Des Devel Ther ; 15: 2843-2855, 2021.
Article in English | MEDLINE | ID: mdl-34234415

ABSTRACT

PURPOSE: A novel folate receptor-targeted ß-cyclodextrin (ß-CD) drug delivery vehicle was constructed to improve the bioavailability, biosafety, and drug loading capacity of curcumin. Controlled release and targeted delivery was achieved by modifying the nanoparticles with folic acid (FA). METHODS: Folate-conjugated ß-CD-polycaprolactone block copolymers were synthesized and characterized. Curcumin-loaded nanoparticles (FA-Cur-NPs) were structured by self-assembly. The physicochemical properties, stability, release behavior and tumor-targeting ability of the fabricated nanoparticles were studied. RESULTS: The average particle size and drug loading of FA-Cur-NPs was 151.8 nm and 20.27%, respectively. Moreover, the FA-Cur-NPs exhibited good stability in vitro for 72 h. The drug release profiles showed that curcumin from FA-Cur-NPs was released significantly faster in a pH 6.4 phosphate buffered solution (PBS) than in pH 7.4, indicating that curcumin can be enriched around the tumor site compared with normal cells. Additionally, the internalization of FA-Cur-NPs was aided by FA receptor-mediated endocytosis, and its cytotoxicity was proportional to the cellular uptake efficiency. Furthermore, in vivo studies confirmed that FA-Cur-NPs exhibited marked accumulation in the tumor site and excellent antitumor activity. CONCLUSION: These findings suggest that FA-Cur-NPs are a promising approach for improving cancer therapy through active targeting and controllable release.


Subject(s)
Curcumin/administration & dosage , Drug Delivery Systems , Folic Acid/administration & dosage , Nanoparticles , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Curcumin/pharmacokinetics , Curcumin/pharmacology , Drug Carriers/chemistry , Drug Liberation , Female , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/pharmacokinetics , Folic Acid/pharmacology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Particle Size , Polyesters/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays , beta-Cyclodextrins/chemistry
8.
Methods Mol Biol ; 2275: 341-361, 2021.
Article in English | MEDLINE | ID: mdl-34118049

ABSTRACT

The development of nanoparticles has provided a powerful weapon in the fight against cancer due to the discovery of their selective accumulation in tumoral tissues, known as enhanced permeation and retention (EPR) effect (Peer et al, Nat Nanotechnol 2:751-760, 2007). Tumoral tissues require afastformation of blood vessels to sustain this rapid growth.


Subject(s)
Folic Acid/pharmacology , Mitochondria/chemistry , Prostatic Neoplasms/metabolism , Silicon Dioxide/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Nanoparticles , Organophosphorus Compounds/chemistry , Porosity
9.
Bioorg Chem ; 112: 104946, 2021 07.
Article in English | MEDLINE | ID: mdl-33989916

ABSTRACT

Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Folate Receptors, GPI-Anchored/antagonists & inhibitors , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Folate Receptors, GPI-Anchored/metabolism , Humans , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
10.
Int J Med Sci ; 18(11): 2355-2365, 2021.
Article in English | MEDLINE | ID: mdl-33967612

ABSTRACT

The majority of patients diagnosed with nasopharyngeal carcinoma (NPC) present with advanced-stage disease. The main treatment for these patients is concurrent chemoradiotherapy, which has various side effects. To improve the therapeutic effects and reduce the side effects of NPC chemoradiotherapy, we constructed a multifunctional folic acid (FA)-targeted magnetic nanocomposite codelivering tissue factor pathway inhibitor-2 (TFPI-2) and cisplatin (CDDP). This novel nanocomposite (FA-MNP/CDDP/TFPI-2) was obtained by amidation and electrostatic adsorption between FA-methoxypolyethylene glycol-polyethyleneimine (FA-MPEG-PEI) containing the TFPI-2 plasmid and magnetic nanoparticles modified by aldehyde sodium alginate loaded with CDDP. Transmission electron microscopy (TEM) images showed that the size of the individual magnetite particle core was approximately 11.5 nm. The structure and composition of the nanocomposites were identified and examined by 1H nuclear magnetic resonance (NMR) spectroscopy and ultraviolet (UV) spectrophotometry. The fluorescence analysis, Prussian blue iron staining, magnetic resonance (MR) imaging and whole-body fluorescence imaging results demonstrated that FA-MNP/CDDP/TFPI-2 showed high gene transfection efficiency and could target tumor cells via folate receptor (FR)-mediated delivery. The codelivery analysis showed that the obtained FA-MNP/CDDP/TFPI-2 composite could cause significantly more apoptosis than treatment with CDDP or TFPI-2 alone. The results showed that the FA-MNP/CDDP/TFPI-2 composites were successfully synthesized and indicated to be a specific molecular target for the FR with significant inhibitory effects on the growth of HNE-1 cells.


Subject(s)
Cisplatin/administration & dosage , Drug Carriers/chemistry , Glycoproteins/genetics , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/therapy , Animals , Cell Line, Tumor , Drug Carriers/pharmacology , Drug Compounding/methods , Female , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , Magnetite Nanoparticles/chemistry , Mice , Molecular Targeted Therapy/methods , Nanocomposites/chemistry , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Plasmids/administration & dosage , Plasmids/genetics , Xenograft Model Antitumor Assays
11.
J Med Chem ; 64(8): 4612-4622, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33818111

ABSTRACT

The currently used photodynamic therapy (PDT) photosensitizers (PSs) are generally associated with a poor cancer cell selectivity, which is responsible for some undesirable side effects. To overcome these problems, there is an urgent need for a selective drug delivery system for PDT PSs. Herein, the encapsulation of a promising Ru(II) polypyridine complex in a polymer with terminal folate groups to form nanoparticles is presented. While the Ru(II) complex itself has a cytotoxic effect in the dark, the encapsulation is able to overcome this drawback. Upon light exposure, the nanoparticles were found to be highly phototoxic in 2D monolayer cells as well as 3D multicellular tumor spheroids upon 480 or 595 nm irradiation. Importantly, the nanoparticles demonstrated a high selectivity for cancerous cells over noncancerous cells and were found to be active in drug resistant cancer cells lines, indicating that they are able to overcome drug resistances.


Subject(s)
Coordination Complexes/chemistry , Photosensitizing Agents/chemistry , Polymers/chemistry , Ruthenium/chemistry , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Drug Resistance, Neoplasm/drug effects , Folate Receptors, GPI-Anchored/chemistry , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Humans , Light , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Singlet Oxygen/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
12.
Bioorg Med Chem ; 37: 116093, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33773393

ABSTRACT

We discovered 6-substituted thieno[2,3-d]pyrimidine compounds (3-9) with 3-4 bridge carbons and side-chain thiophene or furan rings for dual targeting one-carbon (C1) metabolism in folate receptor- (FR) expressing cancers. Synthesis involved nine steps starting from the bromo-aryl carboxylate. From patterns of growth inhibition toward Chinese hamster ovary cells expressing FRα or FRß, the proton-coupled folate transporter or reduced folate carrier, specificity for uptake by FRs was confirmed. Anti-proliferative activities were demonstrated toward FRα-expressing KB tumor cells and NCI-IGROV1 ovarian cancer cells. Inhibition of de novo purine biosynthesis at both 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase and glycinamide ribonucleotide formyltransferase (GARFTase) was confirmed by metabolite rescue, metabolomics and enzyme assays. X-ray crystallographic structures were obtained with compounds 3-5 and human GARFTase. Our studies identify first-in-class C1 inhibitors with selective uptake by FRs and dual inhibition of enzyme targets in de novo purine biosynthesis, resulting in anti-tumor activity. This series affords an exciting new platform for selective multi-targeted anti-tumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Pyrimidines/pharmacology , Thiophenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetulus , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Folate Receptors, GPI-Anchored/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/metabolism , Protein Binding , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism
13.
Int J Nanomedicine ; 16: 1775-1787, 2021.
Article in English | MEDLINE | ID: mdl-33692622

ABSTRACT

PURPOSE: To avoid undefined metabolic mechanisms and to eliminate potential side effects of traditional nanocarriers, new green carriers are urgently needed in cancer treatment. Carrier-free nanoparticles (NPs) based on ursolic acid (UA) have attracted significant attention, but the UA NPs targeting the folate receptor have never been explored. We designed a novel self-assembled UA-Methotrexate (MTX) NPs targeting the folate-receptor and its synergetic anticancer activity was studied in vitro and in vivo. METHODS: UA-MTX NPs were prepared using the solvent precipitation method. Characterization of the UA-MTX NPs preparation was performed using a size analyzer, transmission electron microscopy, and UV-vis spectrophotometry. The in vitro pH-responsive drug release capability of UA-MTX NPs was tested at different pH values. The UA-MTX NPs targeting of folates was determined by comparing the endocytosis rates of cell lines with low or overexpression of the folate receptor (A549 and MCF-7 cells). The cytotoxicity and cell apoptosis of UA-MTX NPs were also studied to determine the in vitro synergistic effects. Combination chemotherapy of UA-MTX NPs in vivo was evaluated using MCF-7 xenografted tumor models. RESULTS: Compared with free UA or MTX, the water solubility of UA-MTX NPs improved significantly. Drug-release from the UA-MTX NPs was faster at pH 5.0 than pH 7.4, suggesting MTX-UA NPs could rapidly release MTX in the acidic conditions of the tumor microenvironment. Confocal laser scanning microscopy revealed the excellent folate receptor targeting of UA-MTX NPs in MCF-7 cells. Cytotoxicity and cell apoptosis results demonstrated greater antiproliferative capacity of UA-MTX NPs than that of free drug in folate receptor overexpressing MCF-7 cells. Anticancer effects in vivo suggested MTX-UA NPs exhibited good biological safety and could enhance antitumor efficacy due to the combination therapy. CONCLUSION: Our findings indicate that the UA-MTX NPs targeting folate-receptors is an efficient strategy for combination chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Drug Delivery Systems , Folate Receptors, GPI-Anchored/metabolism , Methotrexate/pharmacology , Nanoparticles/chemistry , Triterpenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Death/drug effects , Cell Survival/drug effects , Drug Liberation , Drug Synergism , Endocytosis/drug effects , Female , Folic Acid/chemistry , Humans , MCF-7 Cells , Methotrexate/administration & dosage , Methotrexate/chemistry , Mice, Nude , Nanoparticles/ultrastructure , Rats, Wistar , Triterpenes/administration & dosage , Triterpenes/chemistry , Ursolic Acid
14.
J Med Chem ; 64(6): 2967-2970, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33728917

ABSTRACT

Both nitroreductase and the folate receptor are highly expressed in hypoxic tumors. The folate receptor is a potential target for tumors, and nitroreductase can selectively turn on the fluorescence of probes by reducing the nitro group to an amino group. Karan et al. (Karana, S., Cho, M., Lee, H., Lee, H., Park, H., Sundararajan, M., Sessler, J., and Hong, K. Near-infrared fluorescent probe activated by nitroreductase for in vitro and in vivo hypoxic tumor detection. J. Med. Chem. 2021) developed a near-infrared fluorescent probe for hypoxia tumor imaging analysis in vitro and in vivo using folate as the targeting site. This promising strategy may promote advances in precision diagnosis and treatment models of malignant tumors. This Viewpoint looks ahead to new possibilities rendered by their studies.


Subject(s)
Fluorescent Dyes/metabolism , Neoplasms/diagnostic imaging , Nitroreductases/metabolism , Tumor Hypoxia , Cell Line, Tumor , Fluorescent Dyes/chemistry , Folate Receptors, GPI-Anchored/metabolism , Humans , Neoplasms/metabolism , Optical Imaging/methods
15.
Mol Pharm ; 18(1): 33-43, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33251808

ABSTRACT

Methotrexate is a gold standard among disease modifying antirheumatic drugs and is also extensively used clinically in combination with oncological therapies. Thus, it is not surprising that nuclear medicine found an interest in methotrexate in the search for diagnostic and therapeutic solutions. Numerous folate-related radiopharmaceuticals have been proposed for nuclear medicine purposes; however, methotrexate radioagents represent only a minority. This imbalance results from the fact that methotrexate has significantly weaker affinity for folate receptors than folic acid. Nevertheless, radiolabeled methotrexate agents utilized as a tool for early detection and imaging of inflammation in rheumatoid arthritis patients gave promising results. Similarly, the use of multimodal MTX-release nanosystems may find potential applications in radiosynovectomy and theranostic approaches in folate receptor positive cancers.


Subject(s)
Methotrexate/chemistry , Methotrexate/therapeutic use , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use , Antirheumatic Agents/chemistry , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Humans , Nuclear Medicine/methods
16.
Nucl Med Biol ; 93: 1-10, 2021 02.
Article in English | MEDLINE | ID: mdl-33212346

ABSTRACT

INTRODUCTION: Strategic design and synthesis of nanoparticle based preparations could improve diagnostic screening of several cancer types, thereby facilitating better clinical management of the disease. Towards this, the present work aims to develop and evaluate a radioactive technetium-99m (99mTc) labeled gold nanoparticle (NP) preparation modified with folic acid, so as to diagnose folate receptor positive cancers viz. ovarian, breast, etc. METHODS: 11-Bromoundecanoic acid (UA) was synthetically modified both with folic acid and Hydrazinonicotinic acid (HYNIC) chelate at the carboxylic acid end and subsequently converted to thiol functionality at the bromo terminal to yield folic acid-UA-SH and HYNIC-UA-SH ligands respectively. Gold NPs modified with folic acid and HYNIC chelator were obtained on direct addition of folic acid-UA-SH and HYNIC-UA-SH to chloroauric acid in polysorbate 80 solution under reducing conditions. These NPs were then radiolabeled with 99mTc following HYNIC labeling approach. Both the inactive and 99mTc-labeled gold NPs were then tested for their biological efficacy in folate receptor (FR) positive KB cancer cell lines. Also, biodistribution studies of 99mTc-labeled gold NPs were carried in KB tumor xenografts to ascertain the efficacy towards FR in in vivo system. RESULTS: Polysorbate 80 could stabilize the gold NP preparation with average size <10 nm as determined by TEM. Inhibition of [3H]folic acid with functionalized gold nanoparticle revealed affinity towards FR positive KB cell lines with an IC50 ~ 9 µM. Biodistribution studies of 99mTc-labeled gold NP preparation in SCID mice bearing KB tumor showed an uptake of 1.39 ± 0.18%ID/g in tumor and 5.48 ± 0.72%ID/g in kidneys at 3 h post-injection. In vivo distribution in folic acid pre-treated animals could not establish the specificity towards folate receptors. CONCLUSIONS: Biological evaluation of functionalized gold NP showed affinity towards FR positive cancer cell lines. 99mTc-labeled NP exhibited target uptake in both in vitro and in vivo models, but folic acid inhibition could not establish the target specificity. Nevertheless, in vivo pharmacokinetics envisaged in the present design was achieved using the present gold functionalized NP preparation.


Subject(s)
Folate Receptors, GPI-Anchored/metabolism , Gold/chemistry , Molecular Imaging/methods , Nanostructures/chemistry , Technetium/chemistry , Animals , Cell Line, Tumor , Female , Humans , Isotope Labeling , Mice , Technetium/pharmacokinetics , Tissue Distribution
17.
Nat Chem ; 13(1): 77-88, 2021 01.
Article in English | MEDLINE | ID: mdl-33349694

ABSTRACT

Membrane proteins on the cell surface perform a myriad of biological functions; however, ligand discovery for membrane proteins is highly challenging, because a natural cellular environment is often necessary to maintain protein structure and function. DNA-encoded chemical libraries (DELs) have emerged as a powerful technology for ligand discovery, but they are mainly limited to purified proteins. Here we report a method that can specifically label membrane proteins with a DNA tag, and thereby enable target-specific DEL selections against endogenous membrane proteins on live cells without overexpression or any other genetic manipulation. We demonstrate the generality and performance of this method by screening a 30.42-million-compound DEL against the folate receptor, carbonic anhydrase 12 and the epidermal growth factor receptor on live cells, and identify and validate a series of novel ligands for these targets. Given the high therapeutic significance of membrane proteins and their intractability to traditional high-throughput screening approaches, this method has the potential to facilitate membrane-protein-based drug discovery by harnessing the power of DEL.


Subject(s)
Carbonic Anhydrases/chemistry , DNA/chemistry , ErbB Receptors/chemistry , Folate Receptors, GPI-Anchored/chemistry , Small Molecule Libraries/chemistry , Antibodies, Immobilized/chemistry , Antibodies, Immobilized/immunology , Carbonic Anhydrases/metabolism , ErbB Receptors/immunology , ErbB Receptors/metabolism , Fluorescein-5-isothiocyanate/chemistry , Folate Receptors, GPI-Anchored/metabolism , HeLa Cells , Humans , Ligands , Microscopy, Fluorescence , Small Molecule Libraries/metabolism
18.
Biomolecules ; 10(12)2020 12 09.
Article in English | MEDLINE | ID: mdl-33317162

ABSTRACT

This report presents the synthesis and folate receptor target-specificity of amino-functionalized polyacrylamide nanoparticles (AFPAA NPs) for near-infrared (NIR) fluorescence imaging of cancer. For the synthesis of desired nano-constructs, the AFPAA NPs (hereafter referred to as NPs) were reacted with a NIR cyanine dye (CD) bearing carboxylic acid functionality by following our previously reported approach, and the resulting conjugate (NP-CD) on further reaction with folic acid (FA) resulted in a new nano-construct, FA-NP-CD, which demonstrated significantly higher uptake in folate receptor-positive breast cancer cells (KB+) and in folate receptor over-expressed tumors in vivo. The target-specificity of these nanoparticles was further confirmed by inhibition assay in folate receptor-positive (KB+) and -negative (HT-1080) cell lines. To show the advantages of polyacrylamide (PAA)-based NPs in folate receptor target-specificity, the CD used in preparing the FA-NP-CD construct was also reacted with folic acid alone and the synthetic conjugate (CD-FA) was also investigated for its target-specificity. Interestingly, in contrast to NPs (FA-NP-CD), the CD-FA conjugate did not show any significant in vitro or in vivo specificity toward folate receptors, showing the advantages of PAA-based nanotechnology in delivering the desired agent to tumor cells.


Subject(s)
Breast Neoplasms/diagnostic imaging , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Nanoparticles/chemistry , Optical Imaging/methods , Acrylic Resins/chemistry , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbocyanines/chemistry , Carbocyanines/metabolism , Cell Line, Tumor , Female , Fibroblasts/metabolism , Fibrosarcoma/pathology , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Folic Acid/metabolism , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Heterografts , Humans , Infrared Rays , KB Cells , Mice , Mice, Nude
19.
Int J Mol Sci ; 21(22)2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33187164

ABSTRACT

Combined treatments which use nanoparticles and drugs could be a synergistic strategy for the treatment of a variety of cancers to overcome drug resistance, low efficacy, and high-dose-induced systemic toxicity. In this study, the effects on human colon adenocarcinoma cells of surface modified Fe3O4 magnetic nanoparticles (MNPs) in combination with sodium butyrate (NaBu), added as a free formulation, were examined demonstrating that the co-delivery produced a cytotoxic effect on malignant cells. Two different MNP coatings were investigated: a simple polyethylene glycol (PEG) layer and a mixed folic acid (FA) and PEG layer. Our results demonstrated that MNPs with FA (FA-PEG@MNPs) have a better cellular uptake than the ones without FA (PEG@MNPs), probably due to the presence of folate that acts as an activator of folate receptors (FRs) expression. However, in the presence of NaBu, the difference between the two types of MNPs was reduced. These similar behaviors for both MNPs likely occurred because of the differentiation induced by butyrate that increases the uptake of ferromagnetic nanoparticles. Moreover, we observed a strong decrease of cell viability in a NaBu dose-dependent manner. Taking into account these results, the cooperation of multifunctional MNPs with NaBu, taking into consideration the particular cancer-cell properties, can be a valuable tool for future cancer treatment.


Subject(s)
Antineoplastic Agents/chemistry , Butyric Acid/chemistry , Ferric Compounds/chemistry , Folic Acid/chemistry , Magnetite Nanoparticles/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Drug Delivery Systems/methods , Folate Receptors, GPI-Anchored/metabolism , Humans , Magnetics/methods , Polyethylene Glycols/chemistry
20.
Curr Drug Metab ; 21(11): 902-909, 2020.
Article in English | MEDLINE | ID: mdl-32851958

ABSTRACT

BACKGROUND: Among all cancers, lung cancer has high mortality among patients in most of the countries in the world. Targeted delivery of anticancer drugs can significantly reduce the side effects and dramatically improve the effects of the treatment. Folate, a suitable ligand, can be modified to the surface of tumor-selective drug delivery systems because it can selectively bind to the folate receptor, which is highly expressed on the surface of lung tumor cells. OBJECTIVE: This study aimed to construct a kind of folate-targeted topotecan liposomes for investigating their efficacy and mechanism of action in the treatment of lung cancer in preclinical models. METHODS: We conjugated topotecan liposomes with folate, and the liposomes were characterized by particle size, entrapment efficiency, cytotoxicity to A549 cells and in vitro release profile. Technical evaluations were performed on lung cancer A549 cells and xenografted A549 cancer cells in female nude mice, and the pharmacokinetics of the drug were evaluated in female SD rats. RESULTS: The folate-targeted topotecan liposomes were proven to show effectiveness in targeting lung tumors. The anti-tumor effects of these liposomes were demonstrated by the decreased tumor volume and improved therapeutic efficacy. The folate-targeted topotecan liposomes also lengthened the topotecan blood circulation time. CONCLUSION: The folate-targeted topotecan liposomes are effective drug delivery systems and can be easily modified with folate, enabling the targeted liposomes to deliver topotecan to lung cancer cells and kill them, which could be used as potential carriers for lung chemotherapy.


Subject(s)
Folic Acid/administration & dosage , Lung Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Topoisomerase I Inhibitors/administration & dosage , Topotecan/administration & dosage , A549 Cells , Administration, Intravenous , Animals , Cell Survival/drug effects , Female , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/pharmacokinetics , Humans , Liposomes , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Polyethylene Glycols/pharmacokinetics , Rats, Sprague-Dawley , Topoisomerase I Inhibitors/blood , Topoisomerase I Inhibitors/pharmacokinetics , Topotecan/blood , Topotecan/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...