Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Eur J Med Chem ; 227: 113923, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34688013

ABSTRACT

Sphingosine-1-phosphate receptor 2 (S1PR2) has been identified as a brand-new GPCR target for designing antagonists to reverse 5-FU resistance. We herein report the structural optimization and structure-activity relationship of JTE-013 derivatives as S1PR2 antagonists. Compound 9d was the most potent S1PR2 antagonist (KD = 34.8 nM) among developed compounds. Here, compound 9d could significantly inhibit the expression of dihydropyrimidine dehydrogenase (DPD) to reverse 5-FU-resistance in HCT116DPD and SW620/5-FU cells. Further mechanism studies demonstrated that compound 9d not only inhibited S1PR2 but also affected the transcription of S1PR2. In addition, compound 9d also showed acceptable selectivity to normal cells (NCM460). Importantly, compound 9d with suitable pharmacokinetic properties could significantly reverse 5-FU-resistance in the HCT116DPD and SW620/5-FU xenograft models without obvious toxicity, in which the inhibition rates of 5-FU were increased from 23.97% to 65.29% and 27.23% to 60.81%, respectively. Further immunohistochemistry and western blotting analysis also demonstrated that compound 9d significantly decreases the expression of DPD in tumor and liver tissues. These results indicated that compound 9d is a promising lead compound to reverse 5-FU-resistance for colorectal cancer therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/drug therapy , Drug Design , Fluorouracil/pharmacology , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fluorouracil/chemical synthesis , Fluorouracil/chemistry , Humans , Male , Mice , Mice, Nude , Molecular Docking Simulation , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Rats , Rats, Sprague-Dawley , Sphingosine-1-Phosphate Receptors/metabolism , Structure-Activity Relationship
2.
Carbohydr Polym ; 277: 118858, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893265

ABSTRACT

We report herein the development of the novel nanohybrids of gold nanoparticles reduced/stabilized/coated with collagen (AuNPs@collagen) in the first layer and subsequently modified with biotin-quat188-chitosan (Bi-QCS) in the outer layer for 5-fluorouracil (5-FU) delivery to improve cellular uptake and promote specific cell targeting of the nanocarrier. The fabrication of the layer-by-layer technique on the surface of gold nanoparticles (AuNPs) can overcome the limitation of poor drug loading capacity of the classic AuNPs from 64.67% to 87.46%. The AuNPs@collagen coated by the Bi-QCS exhibits strong electrostatic interactions between drug anion (5-FU) and amine groups of the modified chitosan as well as hydrogen bonding. Furthermore, the Bi-QCS-AuNPs@collagen demonstrated a significantly higher anti-inflammatory activity in RAW264.7 macrophage cell line. The Bi-QCS-AuNPs@collagen enhanced the activity of 5-FU approximately 3.3-fold (HeLa) and 6.2-fold (A549), compared to the free 5-Fluorouracil. According to these results, it is very promising that Bi-QCS-AuNPs@collagen can be used as an effective drug delivery carrier in the future.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Drug Delivery Systems , Fluorouracil/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chitosan/chemistry , Collagen/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Fluorouracil/chemical synthesis , Fluorouracil/chemistry , Gold/chemistry , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Metal Nanoparticles/chemistry , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Particle Size , RAW 264.7 Cells
3.
Pharm Res ; 38(6): 1093-1106, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33961188

ABSTRACT

PURPOSE: The clinical application of gemcitabine (GEM) is limited by its pharmacokinetic properties. The aim of this study was to characterize the stability in circulating plasma, tumor targeting, and payload release of liposome-encapsulated GEM, FF-10832. METHODS: Antitumor activity was assessed in xenograft mouse models of human pancreatic cancer. The pharmacokinetics of GEM and its active metabolite dFdCTP were also evaluated. RESULTS: In mice with Capan-1 tumors, the dose-normalized areas under the curve (AUCs) after FF-10832 administration in plasma and tumor were 672 and 1047 times higher, respectively, than after using unencapsulated GEM. The tumor-to-bone marrow AUC ratio of dFdCTP was approximately eight times higher after FF-10832 administration than after GEM administration. These results indicated that liposomal encapsulation produced long-term stability in circulating plasma and tumor-selective targeting of GEM. In mice with Capan-1, SUIT-2, and BxPC-3 tumors, FF-10832 had better antitumor activity and tolerability than GEM. Internalization of FF-10832 in tumor-associated macrophages (TAMs) was revealed by flow cytometry and confocal laser scanning microscopy, and GEM was efficiently released from isolated macrophages of mice treated with FF-10832. These results suggest that TAMs are one of the potential reservoirs of GEM in tumors. CONCLUSION: This study found that FF-10832 had favorable pharmacokinetic properties. The liposomal formulation was more effective and tolerable than unencapsulated GEM in mouse xenograft tumor models. Hence, FF-10832 is a promising candidate for the treatment of pancreatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/blood , Deoxycytidine/analogs & derivatives , Drug Compounding/methods , Drug Delivery Systems/methods , Pancreatic Neoplasms/blood , Xenograft Model Antitumor Assays/methods , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemical synthesis , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/chemical synthesis , Drug Stability , Female , Humans , Liposomes , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Pancreatic Neoplasms/drug therapy , Treatment Outcome , Gemcitabine
4.
J Mater Chem B ; 8(26): 5645-5654, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32538389

ABSTRACT

Nanocarriers have been an important strategy for enhancing the combination therapy of chemotherapy and photodynamic therapy (PDT) (Chem-PDT). However, conventional nanocarriers suffer from the problems of drug leakage in blood and insufficient drug release at target sites. Herein, we have designed a chlorin e6 (Ce6)-loaded GEM prodrug polymer micelle with a singlet oxygen cleavable linker and a pH responsive switch to avoid drug leakage in blood. These Ce6-loaded prodrug micelles possessed a uniform size distribution with a particle size of 78 nm. Meanwhile, the release of Ce6 and GEM was well controlled by acidic pH and laser irradiation. In addition, these micelles showed great acid triggered particle size shrinkage and charge-conversion properties, promoting micelle penetration at tumors and cellular uptake of micelles, which were confirmed by using CLSM of in vitro cell spheres and flow cytometry. Moreover, the in vitro and in vivo1O2 generation ability and antitumor ability of these micelles were impressive. This novel nanocarrier is a potential candidate for efficient Chem-PDT therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Singlet Oxygen/metabolism , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorophyllides , Combined Modality Therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Hydrogen-Ion Concentration , Lasers , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Micelles , Molecular Structure , Optical Imaging , Particle Size , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Polymers/pharmacology , Porphyrins/chemical synthesis , Porphyrins/chemistry , Porphyrins/pharmacology , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Singlet Oxygen/chemistry , Surface Properties , Gemcitabine
5.
J Mater Chem B ; 8(26): 5667-5681, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32500886

ABSTRACT

Carrier-free nanotheranostics directly assembled by using clinically used photosensitizers and chemotherapeutic drugs are a promising alternative to tumor theranostics. However, the weak interaction-driven assembly still suffers from low structural stability against disintegration, lack of targeting specificity, and poor stimulus-responsive property. Moreover, almost all exogenous ligands possess no therapeutic effect. Enlightened by the concept of metal-organic frameworks, we developed a novel self-recognizing metal-coordinated nanotheranostic agent by the coordination-driven co-assembly of photosensitizer indocyanine green (ICG) and chemo-drug methotrexate (MTX, also served as a specific "targeting ligand" towards folate receptors), in which ferric (FeIII) ions acted as a bridge to tightly associate ICG with MTX. Such carrier-free metal-coordinated nanotheranostics with high dual-drug payload (∼94 wt%) not only possessed excellent structural and physiological stability, but also exhibited prolonged blood circulation. In addition, the nanotheranostics could achieve the targeted on-demand drug release by both stimuli of internal lysosomal acidity and external near-infrared laser. More importantly, the nanotheranostics could self-recognize the cancer cells and selectively target the tumors, and therefore they decreased toxicity to normal tissues and organs. Consequently, the nanotheranostics showed strongly synergistic potency for tumor photo-chemotherapy under the precise guidance of magnetic resonance/photoacoustic/fluorescence imaging, thereby achieving highly effective tumor curing efficiency. Considering that ICG and bi-functional MTX are approved by the Food and Drug Administration, and FeIII ions have high biosafety, the self-recognizing and stimulus-responsive carrier-free metal-coordinated nanotheranostics may hold potential applications in tumor theranostics.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Indocyanine Green/pharmacology , Methotrexate/pharmacology , Photosensitizing Agents/pharmacology , Phototherapy , Theranostic Nanomedicine , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Indocyanine Green/chemical synthesis , Indocyanine Green/chemistry , Magnetic Resonance Imaging , Methotrexate/chemical synthesis , Methotrexate/chemistry , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Optical Imaging , Particle Size , Photoacoustic Techniques , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Surface Properties
6.
J Mater Chem B ; 8(12): 2410-2417, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32100811

ABSTRACT

Gemcitabine (GEM) has been the recommended first-line drug for patients with pancreatic ductal adenocarcinoma cancer (PDAC) for the last twenty years. However, GEM-based treatment has failed in many patients because of the drug resistance acquired during tumorigenesis and development. To override resistance to GEM in pancreatic cancer, we developed a visualisable, photothermally controlled, drug release nanosystem (VPNS). This nanosystem has NaLuF4:Nd@NaLuF4 nanoparticles as the luminescent core, octabutoxyphthalocyanine palladium(ii) (PdPc) as the photothermal agent, and phosphorylated gemcitabine (pGEM) as the chemodrug. pGEM, one of the active forms of GEM, can circumvent the insufficient activation of GEM in cancer cell metabolism. The NaLuF4:Nd@NaLuF4 nanoparticles were employed to visualise the tumor lesion in vivo by their near-infrared luminescence. The near-infrared light-triggered photothermal effect from PdPc could trigger the release of pGEM loaded in a thermally responsive ligand and simultaneously enable photothermal cancer treatment. This work presents an effective method that suppresses the growth of tumour cells with dual-mode treatment and enables the improved treatment of orthotopic nude mice afflicted with pancreatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Theranostic Nanomedicine , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Optical Imaging , Pancreatic Neoplasms/diagnostic imaging , Particle Size , Photothermal Therapy , Surface Properties , Tumor Cells, Cultured , Gemcitabine
7.
Drug Des Devel Ther ; 12: 3301-3309, 2018.
Article in English | MEDLINE | ID: mdl-30323562

ABSTRACT

BACKGROUND: Gliomas are one of the most common types of primary brain tumors. It is usually evaluated by gadolinium(III)-based contrast agents by magnetic resonance imaging (MRI) in the clinic. Methotrexate (MTX), as a type of folate analog that inhibits the enzyme dihydrofolate reductase, is widely used as a chemotherapeutic agent to treat gliomas in the experiment. PURPOSE: In this study, a novel theranostic agent MTX-DOTA-Gd (MTX-Gd) was synthesized, which integrates magnetic resonance imaging (MRI) with anticancer treatment. METHODS: MTX-Gd was synthesized by connecting MTX and Gd through 1,4,7,10-tetraazacy-clododecane-1,4,7,10-tetraacetic acid (DOTA). The characterization of MTX-Gd was detected by ultraviolet (UV) and infrared spectroscopy (IR). To confirm the antitumor effect of MTX-Gd, the cytotoxicity of MTX-Gd was examined by the MTT assay. The contrast enhancement of the MTX-Gd was measured through MRI in vitro. Then, nude mice bearing C6 tumor xenografts were used to study in vivo imaging capabilities. RESULTS: The ultraviolet-visible-near infrared radiation (UV-NIR) absorption curve indicated that MTX-Gd had a broad absorption in the region of 500-700 nm. The formation of MTX-Gd was confirmed from the characteristic bands of MTX-DOTA-Gd in the 1413 cm-1 (C-N), 1577 cm-1 (-NH2), and 3429 cm-1 (N-H), in the fourier-transform infrared (FTIR) spectra. MTX-Gd showed little difference in the cell viability compared with MTX, except for the highest concentration (270 µM). In vitro, the imaging of MTX-Gd was significantly brighter than Gd-DOTA at the same concentration, and the brightness and signal intensity of MRI were increased followed by the increased concentration of MTX-Gd. And it also showed that MTX was not visualized on MRI. The other images revealed that the concentration of 4 mM MTX-Gd had the same imaging effect with the concentration of 10 mM Gd-DOTA. Then, MTX-Gd was injected in nude mice bearing C6 tumor xenografts through the tail vein. Significant contrast enhancement was observed at the tumor site from 0.5 h to 3 h. The signal of tumor area was strongest at 3 h due to accumulation by size effect of macromolecules. CONCLUSION: A novel stable and unique theranostic agent (MTX-Gd) was successfully synthe-sized, and it has good stability, strong anticancer ability and excellent magnetic capacity. The methotrexate component of MTX-Gd, as a chemotherapeutic agent, played an important role in targeted therapies of cancer. The DOTA-Gd component of MTX-Gd performed as the MRI contrast agent. The superior MRI imaging performance and synergetic chemical antineoplastic ability of MTX-Gd was revealed, and it has great potential in the diagnosis and treatment of glioma and potentially other cancers, with prospects of clinical application in the near future.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Contrast Media/pharmacology , Gadolinium/pharmacology , Glioma/diagnosis , Glioma/drug therapy , Methotrexate/pharmacology , Organometallic Compounds/pharmacology , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Contrast Media/chemical synthesis , Contrast Media/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Gadolinium/chemistry , Glioma/diagnostic imaging , Glioma/pathology , Humans , Magnetic Resonance Imaging , Methotrexate/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship
8.
J Med Chem ; 60(21): 8758-8780, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29016119

ABSTRACT

P-Glycoprotein (P-gp, ABCB1), multidrug resistance-associated protein 1 (MRP1, ABCC1), and breast cancer resistance protein (BCRP, ABCG2) are the three major ABC transport proteins conferring resistance to many structurally diverse anticancer agents, leading to the phenomenon called multidrug resistance (MDR). Much effort has been put into the development of clinically useful compounds to reverse MDR. Broad-spectrum inhibitors of ABC transport proteins can be of great use in cancers that simultaneously coexpress two or three transporters. In this work, we continued our effort to generate new, potent, nontoxic, and multiply effective inhibitors of the three major ABC transporters. The best compound was active in a very low micromolar concentration range against all three transporters and restored sensitivity toward daunorubicin (P-gp and MRP1) and SN-38 (BCRP) in A2780/ADR (P-gp), H69AR (MRP1), and MDCK II BCRP (BCRP) cells. Additionally, the compound is a noncompetitive inhibitor of daunorubicin (MRP1), calcein AM (P-gp), and pheophorbide A (BCRP) transport.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Purine Nucleosides/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/pharmacology , Humans , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors
9.
RNA ; 23(4): 567-577, 2017 04.
Article in English | MEDLINE | ID: mdl-28096517

ABSTRACT

Nonsense mutations resulting in a premature stop codon in an open reading frame occur in critical tumor suppressor genes in a large number of the most common forms of cancers and are known to cause or contribute to the progression of disease. Low molecular weight compounds that induce readthrough of nonsense mutations offer a new means of treating patients with genetic disorders or cancers resulting from nonsense mutations. We have identified the nucleoside analog clitocine as a potent and efficacious suppressor of nonsense mutations. We determined that incorporation of clitocine into RNA during transcription is a prerequisite for its readthrough activity; the presence of clitocine in the third position of a premature stop codon directly induces readthrough. We demonstrate that clitocine can induce the production of p53 protein in cells harboring p53 nonsense-mutated alleles. In these cells, clitocine restored production of full-length and functional p53 as evidenced by induced transcriptional activation of downstream p53 target genes, progression of cells into apoptosis, and impeded growth of nonsense-containing human ovarian cancer tumors in xenograft tumor models. Thus, clitocine induces readthrough of nonsense mutations by a previously undescribed mechanism and represents a novel therapeutic modality to treat cancers and genetic diseases caused by nonsense mutations.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Biomimetic Materials/pharmacology , Codon, Nonsense/drug effects , Furans/pharmacology , Nucleosides/pharmacology , Ovarian Neoplasms/drug therapy , Pyrimidine Nucleosides/pharmacology , Tumor Suppressor Protein p53/agonists , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/metabolism , Apoptosis/drug effects , Biomimetic Materials/chemical synthesis , Biomimetic Materials/metabolism , Cell Line, Tumor , Female , Furans/chemical synthesis , Furans/metabolism , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Nude , Nucleosides/chemical synthesis , Nucleosides/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Biosynthesis , Pyrimidine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/metabolism , Signal Transduction , Transcriptional Activation , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
10.
Molecules ; 22(1)2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28067759

ABSTRACT

An effective synthesis of nucleosides using glycosyl chlorides as glycosyl donors in the absence of Lewis acid has been developed. Glycosyl chlorides have been shown to be pivotal intermediates in the classical silyl-Hilbert-Johnson reaction. A possible mechanism that differs from the currently accepted mechanism advanced by Vorbrueggen has been proposed and verified by experiments. In practice, this catalyst-free method provides easy access to Capecitabine in high yield.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Capecitabine/chemical synthesis , Chlorides/chemistry , Nucleosides/chemical synthesis , Catalysis , Glycosylation
11.
J Colloid Interface Sci ; 487: 239-249, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27776282

ABSTRACT

The anti-leukemia effect of cytarabine (Ara-C) is severely restricted by its high hydrophilic properties and rapid plasma degradation. Herein, a novel amphiphilic small molecular prodrug of Ara-C was developed by coupling a short aliphatic chain, hexanoic acid (HA) to 4-NH2 of the parent drug. Based on the amphiphilic nature, the resulting bioconjugate (HA-Ara) could spontaneously self-assemble into stable spherical nanoassemblies (NAs) with an extremely high drug loading (∼71wt%). Moreover, folate receptor (FR)-targeting NAs with high grafting efficient folic acid - bovine serum albumin (FA-BSA) conjugate immobilized on the surface (NAs/FA-BSA) was prepared. The results of MTT assays on FR-positive K562 cells and FR-negative A549 cells demonstrated higher cytotoxicity of HA-Ara NAs than the native drug. Especially, the IC50 values revealed that NAs/FA-BSA was 3 and 2-fold effective than non-targeted NAs after 24 and 48h treatment with K562 cells, respectively indicating FR-mediated enhanced anti-tumor efficacy. In vitro cellular uptake, larger accumulation of HA-Ara NAs were observed in comparative with the free FITC and the results further confirmed the selective uptake of NAs/FA-BSA in folate receptor enriched cancer cells. Above all, self-assembled HA-Ara NAs exhibited potential superiority for Ara-C delivery and FA-modified NAs would be an excellent candidate for targeting leukemia therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Caproates/chemistry , Cytarabine/pharmacology , Drug Carriers , Nanoconjugates/chemistry , Prodrugs/pharmacology , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/metabolism , Biological Transport , Cattle , Cytarabine/chemical synthesis , Cytarabine/metabolism , Drug Compounding , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Gene Expression , Humans , Inhibitory Concentration 50 , K562 Cells , Molecular Targeted Therapy , Nanoconjugates/ultrastructure , Prodrugs/chemical synthesis , Prodrugs/metabolism , Protein Binding , Serum Albumin, Bovine/chemistry
12.
Molecules ; 21(12)2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27898006

ABSTRACT

This paper describes the immobilization of the neutral protease from Bacillus subtilis and its application in the regioselective hydrolysis of acetylated nucleosides, including building blocks useful for the preparation of anticancer products. Regarding the immobilization study, different results have been obtained depending on the immobilization procedure. Epoxy hydrophobic carriers gave a poorly stable derivative that released almost 50% of the immobilized protein under the required reaction conditions. On the contrary, covalent immobilization on a differently activated hydrophilic carrier (agarose) resulted in very stable enzyme derivatives. In an attempt to explain the obtained enzyme immobilization results, the hypothetical localization of lysines on the enzyme surface was predicted in a 3D structure model of B. subtilis protease N built in silico by using the structure of Staphylococcus aureus metalloproteinase as the template. The immobilized enzyme shown a high regioselectivity in the hydrolysis of different peracetylated nucleosides. A stable enzyme derivative was obtained and successfully used in the development of efficient preparative bioprocesses for the hydrolysis of acetylated nucleosides, giving new intermediates for the synthesis of capecitabine in high yield.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Bacillus subtilis/enzymology , Capecitabine/chemical synthesis , Enzymes, Immobilized/chemistry , Nucleosides/chemistry , Peptide Hydrolases/chemistry , Acetylation , Hydrolysis
13.
Neoplasia ; 18(1): 33-48, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26806350

ABSTRACT

In this study, a new compound, 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine (DHA-dFdC), was synthesized and characterized. Its antitumor activity was evaluated in cell culture and in mouse models of pancreatic cancer. DHA-dFdC is a poorly soluble, pale yellow waxy solid, with a molecular mass of 573.3Da and a melting point of about 96°C. The activation energy for the degradation of DHA-dFdC in an aqueous Tween 80-based solution is 12.86kcal/mol, whereas its stability is significantly higher in the presence of vitamin E. NCI-60 DTP Human Tumor Cell Line Screening revealed that DHA-dFdC has potent and broad-spectrum antitumor activity, especially in leukemia, renal, and central nervous system cancer cell lines. In human and murine pancreatic cancer cell lines, the IC50 value of DHA-dFdC was up to 10(5)-fold lower than that of dFdC. The elimination of DHA-dFdC in mouse plasma appeared to follow a biexponential model, with a terminal phase t1/2 of about 58minutes. DHA-dFdC significantly extended the survival of genetically engineered mice that spontaneously develop pancreatic ductal adenocarcinoma. In nude mice with subcutaneously implanted human Panc-1 pancreatic tumors, the antitumor activity of DHA-dFdC was significantly stronger than the molar equivalent of dFdC alone, DHA alone, or the physical mixture of them (1:1, molar ratio). DHA-dFdC also significantly inhibited the growth of Panc-1 tumors orthotopically implanted in the pancreas of nude mice, whereas the molar equivalent dose of dFdC alone did not show any significant activity. DHA-dFdC is a promising compound for the potential treatment of cancers in organs such as the pancreas.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Calorimetry, Differential Scanning , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Stability , Female , Humans , Mice , Mice, Transgenic , Solubility , X-Ray Diffraction , Xenograft Model Antitumor Assays , Gemcitabine
14.
Pak J Pharm Sci ; 29(6): 1997-2004, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28375116

ABSTRACT

The objective of this work is to synthesize indolacin-5-fluorouracil-1-ylmethyl ester and the structure was confirmed by means of UV, IR, 1H-NMR, 13C-NMR and mass spectrometry. The physicochemical parameters of melting point, solubility, apparent partition coefficient were investigated. S180 sarcoma, H22 hapatitic cancer and Lewis-transplanted mice were used to evaluate the anti-tumor activity of indolacini-5-fluorouracil-1-ylmethyl ester compared with 5-fluorouracil in vivo. Anti-inflammatory and analgesic activities were evaluated in mice. The inhibitory ratio of indolacini- 5-fluorouracil-1-ylmethyl ester is comparative to that of 5-fluorouracil. This study indicates that 5-fluorouracil-1-ylmethyl ester may represent a new anticancer predrug of 5-fluorouracil to produce a combined effect of indolacin and 5-fluorouracil for cancer therapy.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Lewis Lung/drug therapy , Fluorouracil/analogs & derivatives , Fluorouracil/chemical synthesis , Fluorouracil/pharmacology , Indoleacetic Acids/chemical synthesis , Indoleacetic Acids/pharmacology , Sarcoma 180/drug therapy , Analgesics/chemical synthesis , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Antimetabolites, Antineoplastic/toxicity , Carbon-13 Magnetic Resonance Spectroscopy , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Fluorouracil/toxicity , Indoleacetic Acids/toxicity , Lethal Dose 50 , Mass Spectrometry , Mice , Molecular Structure , Proton Magnetic Resonance Spectroscopy , Sarcoma 180/pathology , Solubility , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Transition Temperature , Tumor Burden/drug effects
15.
Bioorg Med Chem ; 23(13): 3287-96, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25960323

ABSTRACT

Phospholipid derivatives of anticancer nucleosides cladribine and fludarabine (F-ara-A) bearing 1,2- and 1,3-diacylglycerol moieties have been prepared by the H-phosphonate approach using 1,1,3,3-tetraisopropyldisiloxane-1,3-diyl protecting group for cladribine and a combination of tert-butyldimethylsilyl and levulinyl protecting groups for 2-fluoroadenine nucleosides. The synthesized conjugates exhibited lower in vitro antiproliferative activity against human tumor cell lines in comparison with the same concentrations of the parent cladribine and fludarabine phosphate. In the course of biokinetic study, it was found that intragastric administration of phospholipid F-ara-A derivatives to Wistar rats and ICR outbred male mice led to a slow release of F-ara-A into the bloodstream, a smooth increase in nucleoside concentration, and prolonged serum circulation of liberated nucleoside. The oral bioavailability of F-ara-A from 1,2-dimyristoylglycerophosphate derivative 29 was similar to its oral bioavailability from fludarabine phosphate.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Cladribine/pharmacokinetics , Diglycerides/chemistry , Phospholipids/chemistry , Prodrugs , Vidarabine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/chemical synthesis , Biological Availability , Biotransformation , Cell Line, Tumor , Cell Survival/drug effects , Cladribine/analogs & derivatives , Cladribine/blood , Cladribine/chemical synthesis , Diglycerides/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Molecular Structure , Organophosphonates/chemistry , Phospholipids/metabolism , Rats , Rats, Wistar , Vidarabine/blood , Vidarabine/chemical synthesis , Vidarabine/pharmacokinetics
16.
Carbohydr Res ; 406: 71-5, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25681996

ABSTRACT

Gemcitabine, 2'-deoxy-2',2'-difluorocytidine, is currently prescribed against a number of cancers. Here we report a linear synthesis of gemcitabine with a high-yielding direct conversion of 3,5-di-O-benzoyl-2-deoxy-2,2-difluororibose into the corresponding glycosyl urea as the key step, followed by conventional conversion to the cytosine base via the uracil derivative. The process proceeded with modest anomeric selectivity.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Deoxycytidine/analogs & derivatives , Acylation , Catalysis , Cyclization , Deoxycytidine/chemical synthesis , Urea/chemistry , Gemcitabine
17.
Pharmazie ; 69(4): 271-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24791590

ABSTRACT

The broad-spectrum antitumor agent 5-fluorouracil (5-FU), has been used to treat various solid malignant tumors. However, its short life-time in vivo and poor ability to cross the blood-brain barrier has limited its application to brain tumor therapy. In order to develop a 5-FU derivative that localizes efficiently to the brain while retaining potent antitumor activity, we conjugated 5-FU with N,N-dimethylethylenediamine via an amide bond. The stability of the resulting 5-FU derivative (D-FU) was tested in vitro in phosphate buffer, rat plasma and brain homogenate. The pharmacokinetic and biodistribution studies in brains of the rats showed a higher C(max) (the maximal concentration) and an increased AUC(0-t) (the area under the concentration-time curve) which was 6-fold that of 5-FU. In addition, compared to 5-FU, D-FU exhibited lower toxicity in an acute toxicity assay and similar antitumor activity in the C6 cell line. In conclusion, D-FU has the potential to be developed into an efficient brain delivery drug.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Fluorouracil/analogs & derivatives , Fluorouracil/pharmacology , Algorithms , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/toxicity , Area Under Curve , Brain/metabolism , Buffers , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Drug Stability , Fluorouracil/chemical synthesis , Hydrogen-Ion Concentration , Indicators and Reagents , Magnetic Resonance Spectroscopy , Male , Mice , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Tissue Distribution
18.
J Med Chem ; 57(10): 4023-34, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24784222

ABSTRACT

The motuporamines isolated from the sea sponge Xestospongia exigua are of biological interest because of their unique antimigration and antiangiogenic properties. Key bioactive features were found to be a saturated 15-membered heterocycle and a norspermidine motif. This paper describes new analogues that modulate the cytotoxicity of this compound class and have enhanced antimigration properties. By movement of the polyamine chain outside the ring, new carbocycles were discovered that doubled the antimigration potency and reduced compound toxicity by 133-fold. Mice injected with metastatic human L3.6pl pancreatic cancer cells demonstrated significant reduction in liver metastases when treated with N(1)-(3-aminopropyl)-N(3)-(cyclopentadecylmethyl)propane-1,3-diamine compared with dihydromotuporamine C. Significant changes in specific ceramide populations (N16:0 and N22:1) were noted in L3.6pl cells treated with dihydromotuporamine C but not for the cyclopentadecylmethylnorspermidine derivative, which had lower toxicity. Both compounds gave increased levels of specific low molecular weight sphingomyelins, suggesting that they may act upon sphingomyelin processing enzymes.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Heterocyclic Compounds, 1-Ring/pharmacology , Neoplasm Metastasis/prevention & control , Propylamines/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , CHO Cells , Cell Movement/drug effects , Cricetinae , Cricetulus , Humans , Mice, Nude , Sphingomyelins/metabolism , Xenograft Model Antitumor Assays
19.
Int J Pharm ; 468(1-2): 142-51, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24731731

ABSTRACT

Although gemcitabine (Gem) constitutes first-line therapy for pancreatic cancer, its clinical outcome suffers from rapid metabolism and acquired drug resistance. To overcome its limitations, several lipophilic prodrugs including 4-(N)-stearoyl Gem (GemC18) have been studied for their efficacy over Gem. Herein, we aimed to prepare and characterize the GemC18-loaded poly(ethylene glycol)-poly(d,l-lactide) (PEG-PLA) polymeric micelles (PMs) as well as its self-assembled nanoparticles (NPs). A D-optimal design was also utilized to investigate the effects of formulation variables, namely initial drug/polymer ratio, total solid content, and the type of organic solvent on properties of GemC18-loaded PMs. The optimized formulation showed a particle size of about 120 nm, encapsulation efficiency >90%, and a sustained release behavior of the drug. Alternatively, the prodrug NPs were harvested in larger size (∼300 nm) and more negative zeta potential, but less chemical stability compared to the optimized PMs. In Panc-1 and AsPC-1 cell lines, both GemC18-loaded PMs and NPs were significantly more cytotoxic than GemC18 solution. Chiefly, they could effectively reduce the viability of Gem high-resistant AsPC-1 cells in culture, whereas the molar equivalent doses of Gem did not show any acceptable cytotoxicity. Thus, these results suggest a promising direction for alternative Gem delivery systems for future therapeutic applications.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Deoxycytidine/analogs & derivatives , Drug Carriers , Nanoparticles , Polymers/chemistry , Prodrugs/chemical synthesis , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Deoxycytidine/chemical synthesis , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Stability , Humans , Kinetics , Micelles , Nanotechnology , Pancreatic Neoplasms/pathology , Particle Size , Polyesters/chemistry , Polyethylene Glycols/chemistry , Prodrugs/pharmacology , Solubility , Solvents/chemistry , Technology, Pharmaceutical/methods
20.
Bioconjug Chem ; 25(3): 501-9, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24506698

ABSTRACT

The present study reports the synthesis, characterization, and biological evaluation of a novel macromolecular bipill, synthesized by appending two different anticancer agents, viz., gemcitabine (GEM) and methotrexate (MTX), to the distal ends of a long-circulating poly(ethylene glycol) (PEG) spacer. Covalent conjugation of GEM and MTX via PEG linker not only transformed the solubility profiles of constituent drug molecules, but significantly improved their stability in the presence of plasma. In vitro cytotoxicity studies confirmed that GEM-PEG-MTX exerts higher cytotoxicity (IC50 0.181 µM at 24 h) in human breast adenocarcinoma MCF-7 cell lines, when compared to free drug congeners, i.e., free GEM (IC50 0.294 µM at 24 h) and free MTX (IC50 0.591 µM at 24 h). Tumor growth inhibition studies in chemically induced breast cancer bearing rats established the superiority of GEM-PEG-MTX conjugate over all other pharmaceutical preparations including free drugs, physical mixture of GEM and MTX, and PEGylated GEM/MTX. Toxicity studies in tumor bearing rats as well as healthy mice corroborated that dual drug conjugation is an effective means to synergize the therapeutic indices of potential drug candidates while alleviating drug-associated side effects.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Mammary Neoplasms, Experimental/drug therapy , Methotrexate/pharmacology , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/chemical synthesis , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , MCF-7 Cells , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Mammary Neoplasms, Experimental/pathology , Methotrexate/chemical synthesis , Methotrexate/chemistry , Mice , Molecular Structure , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Risk Factors , Structure-Activity Relationship , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...