Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
1.
Toxicology ; 464: 153019, 2021 12.
Article in English | MEDLINE | ID: mdl-34740671

ABSTRACT

Cassava (Manihotesculenta Crantz) is one of the most important root crops in tropical countries. It is a major source of cyanogenic glycosides viz. linamarin and lotaustralin, and these on breakdown liberate HCN and ketone. Cassava cyanide extract (CCE) from cassava leaves and tuber rinds were formulated as a biopesticide against certain borer insect pests of horticultural crops. Adenocarcinomic human alveolar basal epithelial cells (A549) were treated with three different concentrations (100, 200, 400 ppm) of CCE. The MTT and NRU assays showed dose-dependent cytotoxicity. The DCFH-DA assay does not show any free radical scavenging activity, whereas the NRR assay showed a reduction in the nitrile radicals with an increase in the concentration of the bioactive compound. A negative correlation was found between the concentration of the bioactive principles and mitochondrial and lysosomal functions. Various cellular assays demonstrated the cellular response of the CCE, and it was found that at higher concentration (400 ppm), the CCE exert a significant necrotic cell death rather than apoptosis. The results of the study indicated that the CCE have a remarkable tendency of anti-proliferative ability.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Cyanides/pharmacology , Lung Neoplasms/drug therapy , Manihot/chemistry , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Alveolar Epithelial Cells/drug effects , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Cell Death/drug effects , Cell Proliferation/drug effects , Cyanides/administration & dosage , Cyanides/isolation & purification , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/pathology , Necroptosis/drug effects , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Plant Extracts/pharmacology
2.
BMC Immunol ; 22(1): 44, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253166

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGF-ß) is a typical immuno-inhibitory cytokine and highly secreted by lung cancer cells. It was supposed that its immunosuppressive effects to NK cell might be related with the altered expression of activating and inhibitory molecules in lung cancer cells. In this study, we examined the expression of NKG2DLs, PD-L1 and PD-L2 in lung cancer cells after treatment of TGF-ß and a TGF-ß inhibitor, Galunisertib (LY2157299). RESULTS: TGF-ß reduced the level of surface proteins of five NKG2DLs without altered transcription levels in lung cancer cells. Galunisertib reversed the effect of TGF-ß on the expression of NKG2DLs. Since MMP inhibitors, MMPi III and MMP2 inhibitor I, restored the reduced expression of NKG2DLs after treatment of TGF-ß, it was thought that TGF-ß induced the expression of MMP2 which facilitated the shedding of the NKG2DLs in cancer cells. However, the expression of PD-L1, L2 were not changed by treatment with TGF-ß or Galunisertib. CONCLUSIONS: Therefore, inhibition of TGF-ß might reverse the immunosuppressive status on immune cells and restore NK cell mediated anticancer immune responses by upregulation of NKG2DLs in cancer cells.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Transforming Growth Factor beta/metabolism , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Cytotoxicity, Immunologic , Down-Regulation , GPI-Linked Proteins/metabolism , Humans , Immune Tolerance , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Pyrazoles/pharmacology , Quinolines/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Escape
3.
Chemistry ; 27(1): 362-370, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-32716591

ABSTRACT

During the last decades, photodynamic therapy (PDT), an approved medical technique, has received increasing attention to treat certain types of cancer. Despite recent improvements, the treatment of large tumors remains a major clinical challenge due to the low ability of the photosensitizer (PS) to penetrate a 3D cellular architecture and the low oxygen concentrations present in the tumor center. To mimic the conditions found in clinical tumors, exceptionally large 3D multicellular tumor spheroids (MCTSs) with a diameter of 800 µm were used in this work to test a series of new RuII polypyridine complexes as one-photon and two-photon PSs. These metal complexes were found to fully penetrate the 3D cellular architecture and to generate singlet oxygen in the hypoxic center upon light irradiation. While having no observed dark toxicity, the lead compound of this study showed an impressive phototoxicity upon clinically relevant one-photon (595 nm) or two-photon (800 nm) excitation with a full eradication of the hypoxic center of the MCTSs. Importantly, this efficacy was also demonstrated on mice bearing an adenocarcinomic human alveolar basal epithelial tumor.


Subject(s)
Organometallic Compounds , Photochemotherapy , Photosensitizing Agents , Ruthenium , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Animals , HeLa Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Neoplasms, Basal Cell/drug therapy , Neoplasms, Basal Cell/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Photons/therapeutic use , Photosensitizing Agents/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology , Singlet Oxygen/metabolism , Spheroids, Cellular , Tumor Hypoxia , Xenograft Model Antitumor Assays
4.
Carbohydr Res ; 493: 108032, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32417443

ABSTRACT

Milk exosomes (mExo), similar to cell-derived exosomes, are emerging as promising nanocarriers for delivery of therapeutic molecules such as chemical drugs and siRNA, due to the excellent biocompatibility and low-cost production from bovine milk. However, additional modification remains required to apply milk exosomes for tumor-specific drug delivery. Here, we attempted to develop a novel strategy for directing doxorubicin (Dox)-loaded mExo to CD44-overexpressing tumor cells. Hyaluronan (HA), a CD44-specific ligand, was functionalized with an amphiphilic molecule DSPE-PEG2000, which enabled the spontaneous decoration of Dox-loaded mExo with HA onto the phospholipid bilayer. The obtained nanocarrier HA-mExo-Dox was shown to be able to selectively deliver Dox into cells with over-expressed CD44 instead of control cells and trigger the notable tumor cells death in the in vitro analysis. This study demonstrates the potential use of HA-displaying mExo for tumor cell-specific drug delivery and this strategy should prove to be feasible for targeted cancer therapy.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Delivery Systems , Hyaluronic Acid/chemistry , Lung Neoplasms/drug therapy , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Animals , Antibiotics, Antineoplastic/chemistry , Breast Neoplasms/pathology , Carbohydrate Conformation , Cell Line , Cell Survival/drug effects , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Exosomes/chemistry , Humans , Lung Neoplasms/pathology , MCF-7 Cells , Milk/chemistry
5.
Biomater Sci ; 8(6): 1748-1758, 2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32002530

ABSTRACT

In this work, we developed multi-shelled hollow nanospheres [RGD@am-ZnO@CuO@Au@DOX HNSs] as multifunctional therapeutic agents to achieve effective and targeted Zn2+/Cu2+ therapy, induced drug delivery under low pH/red-light conditions, and enhanced phototherapy under single red-light. The photothermal and photodynamic performance of am-ZnO@CuO@Au HNSs was enhanced relative to that of am-ZnO nanoparticles (NPs) or am-ZnO@CuO HNSs by utilizing the resonance energy transfer process and broad red-light absorption. The pH-sensitive am-ZnO@CuO@Au HNSs were dissolved to Zn2+/Cu2+ in the acidic endosomes/lysosomes of cancer cells, resulting in a cancer cell killing effect. The release performance of doxorubicin (DOX) from RGD@am-ZnO@CuO@Au@DOX HNSs was evaluated under low pH and red-light-irradiated conditions, and targeting of HNSs was confirmed by dual-modal imaging (magnetic resonance/fluorescence) of the tumor area. Moreover, in vivo synergistic therapy using RGD@am-ZnO@CuO@Au@DOX HNSs was further evaluated in mice bearing human pulmonary adenocarcinoma (A549) cells, achieving a remarkable synergistic antitumor effect superior to that obtained by monotherapy. This study validated that RGD@am-ZnO@CuO@Au@DOX HNSs can be a promising candidate for efficient postoperative cancer therapy.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/diagnostic imaging , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Copper/chemistry , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Zinc/chemistry , A549 Cells , Animals , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Delivery Systems , Humans , Magnetic Resonance Imaging , Mice , NIH 3T3 Cells , Nanospheres , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Oligopeptides/pharmacology , Phototherapy , Xenograft Model Antitumor Assays
6.
Biochim Biophys Acta Gen Subj ; 1864(4): 129533, 2020 04.
Article in English | MEDLINE | ID: mdl-31953127

ABSTRACT

BACKGROUND: Epidemiologic studies suggest that diabetes is associated with an increased risk of cancer. Concurrently, clinical trials have shown that metformin, which is a first-line antidiabetic drug, displays anticancer activity. The underlying mechanisms for these effects are, however, still not well recognized. METHODS: Methods based on atomic force microscopy (AFM) were used to directly evaluate the influence of metformin on the nanomechanical and adhesive properties of endothelial and cancer cells in chronic hyperglycemia. AFM single-cell force spectroscopy (SCFS) was used to measure the total adhesion force and the work of detachment between EA.hy926 endothelial cells and A549 lung carcinoma cells. Nanoindentation with a spherical AFM probe provided information about the nanomechanical properties of cells, particularly the length and grafting density of the glycocalyx layer. Fluorescence imaging was used for glycocalyx visualization and monitoring of E-selectin and ICAM-1 expression. RESULTS: SCFS demonstrated that metformin attenuates adhesive interactions between EA.hy926 endothelial cells and A549 lung carcinoma cells in chronic hyperglycemia. Nanoindentation experiments, confirmed by confocal microscopy imaging, revealed metformin-induced recovery of endothelial glycocalyx length and density. The recovery of endothelial glycocalyx was correlated with a decrease in the surface expression of E-selectin and ICAM-1. CONCLUSION: Our results identify metformin-induced endothelial glycocalyx restoration as a key factor responsible for the attenuation of adhesion between EA.hy926 endothelial cells and A549 lung carcinoma cells. GENERAL SIGNIFICANCE: Metformin-induced glycocalyx restoration and the resulting attenuation of adhesive interactions between the endothelium and cancer cells may account for the antimetastatic properties of this drug.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents/pharmacology , Endothelial Cells/drug effects , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Lung Neoplasms/drug therapy , Metformin/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Cell Adhesion/drug effects , Cells, Cultured , Chronic Disease , Endothelial Cells/pathology , Endothelium/drug effects , Endothelium/metabolism , Glycocalyx/drug effects , Glycocalyx/metabolism , Humans , Hyperglycemia/pathology , Lung Neoplasms/pathology , Microscopy, Atomic Force
7.
Bioorg Chem ; 92: 103282, 2019 11.
Article in English | MEDLINE | ID: mdl-31541801

ABSTRACT

Bisphosphonates are widely used for treatment of osteoporosis. Recently, they have been reported to be effective anticancer agents. In this work, we designed some substituted phenyl (azanediyl) bis (methylene phosphonic acid) to be tested for their anticancer effect. Both molecular docking and dynamics studies were used to select the top ranked highly scored compounds. The selected hits showed potential in vitro anticancer effect against some cell lines. Biodistribution pattern and gamma scintigraphy were conducted to the most effective derivative (BMBP) after radiolabeling with 99mTc. Results of biodistribution and scintigraphic imaging of 99mTc-BMBP in tumor bearing mice showed a notable tumor affinity, and confirmed the targeting affinity of BMBP to the tumor tissues. As a conclusion, BMBP could act as potential anticancer agent and imaging probe.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents/pharmacology , Aza Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Optical Imaging , Phosphorous Acids/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/diagnostic imaging , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Gamma Rays , Geranyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/metabolism , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Models, Molecular , Molecular Structure , Phosphorous Acids/chemical synthesis , Phosphorous Acids/chemistry , Structure-Activity Relationship , Tissue Distribution
8.
Mol Pharm ; 16(2): 561-572, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30592418

ABSTRACT

The chimeric peptide HPRP-A1-iRGD, composed of a chemically conjugated tumor-homing/penetration domain (iRGD) and a cationic anticancer peptide domain (HPRP-A1), was used to study the effect of targeted modification to enhance the peptide's specificity, penetration, and tumor accumulation ability. The iRGD domain exhibits tumor-targeting and tumor-penetrating activities by specifically binding to the neuropilin-1 receptor. Acting as a homing/penetration domain, iRGD contributed to enhancing the tumor selectivity, permeability, and targeting of HPRP-A1 by targeted receptor dependence. As the anticancer active domain, HPRP-A1 kills cancer cells by disrupting the cell membrane and inducing apoptosis. The in vitro membrane selectivity toward cancer cells, such as A549 and MDA-MB-23, and human umbilical vein endothelial cells (HUVECs), normal cells, the penetrability assessment in the A549 3D multiple cell sphere model, and the in vivo tumor-tissue accumulation test in the A549 xenograft model indicated that HPRP-A1-iRGD exhibited significant increases in the selectivity toward membranes that highly express NRP-1, the penetration distance in 3D multiple cell spheres, and the accumulation in tumor tissues after intravenous injection, compared with HPRP-A1 alone. The mechanism of the enhanced targeting ability of HPRP-A1-iRGD was demonstrated by the pull-down assay and biolayer interferometry test, which indicated that the chimeric peptide could specifically bind to the neuropilin-1 protein with high affinity. We believe that chemical conjugation with iRGD to increase the specificity, penetration, and tumor-tissue accumulation of HPRP-A1 is an effective and promising approach for the targeted modification of peptides as anticancer therapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Oligopeptides/chemistry , Peptides/chemistry , Peptides/therapeutic use , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Animals , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neuropilin-1/metabolism
9.
Molecules ; 23(12)2018 Dec 04.
Article in English | MEDLINE | ID: mdl-30518152

ABSTRACT

A phytochemical investigation of the roots of Aspilia plurisetaled to the isolation of ent-kaurane-type diterpenoids and additional phytochemicals (1⁻23). The structures of the isolated compounds were elucidated based on Nuclear Magnetic Resonance (NMR) spectroscopic and mass spectrometric analyses. The absolute configurations of seven of the ent-kaurane-type diterpenoids (3⁻6, 6b, 7 and 8) were determined by single crystal X-ray diffraction studies. Eleven of the compounds were also isolated from the roots and the aerial parts of Aspilia mossambicensis. The literature NMR assignments for compounds 1 and 5 were revised. In a cytotoxicity assay, 12α-methoxy-ent-kaur-9(11),16-dien-19-oic acid (1) (IC50 = 27.3 ± 1.9 µM) and 9ß-hydroxy-15α-angeloyloxy-ent-kaur-16-en-19-oic acid (3) (IC50 = 24.7 ± 2.8 µM) were the most cytotoxic against the hepatocellular carcinoma (Hep-G2) cell line, while 15α-angeloyloxy-16ß,17-epoxy-ent-kauran-19-oic acid (5) (IC50 = 30.7 ± 1.7 µM) was the most cytotoxic against adenocarcinomic human alveolar basal epithelial (A549) cells.


Subject(s)
Asteraceae/chemistry , Cell Survival/drug effects , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Diterpenes, Kaurane/isolation & purification , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Molecular Structure , Plant Components, Aerial/chemistry , Plant Roots/chemistry
10.
J Nanobiotechnology ; 16(1): 100, 2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30526603

ABSTRACT

BACKGROUND: Particokinetic models are important to predict the effective cellular dose, which is key to understanding the interactions of particles with biological systems. For the reliable establishment of dose-response curves in, e.g., the field of pharmacology and toxicology, mostly the In vitro Sedimentation, Diffusion and Dosimetry (ISDD) and Distorted Grid (DG) models have been employed. Here, we used high resolution scanning electron microscopy to quantify deposited numbers of particles on cellular and intercellular surfaces and compare experimental findings with results predicted by the ISDD and DG models. RESULTS: Exposure of human lung epithelial A549 cells to various concentrations of differently sized silica particles (100, 200 and 500 nm) revealed a remarkably higher dose deposited on intercellular regions compared to cellular surfaces. The ISDD and DG models correctly predicted the areal densities of particles in the intercellular space when a high adsorption ("stickiness") to the surface was emulated. In contrast, the lower dose on cells was accurately inferred by the DG model in the case of "non-sticky" boundary conditions. Finally, the presence of cells seemed to enhance particle deposition, as aerial densities on cell-free substrates were clearly reduced. CONCLUSIONS: Our results further validate the use of particokinetic models but also demonstrate their limitations, specifically, with respect to the spatial distribution of particles on heterogeneous surfaces. Consideration of surface properties with respect to adhesion and desorption should advance modelling approaches to ultimately predict the cellular dose with higher precision.


Subject(s)
Nanoparticles/chemistry , Nanoparticles/ultrastructure , Single-Cell Analysis , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/ultrastructure , Adsorption , Dose-Response Relationship, Drug , Humans , Microscopy, Electron, Scanning , Models, Biological , Nanoparticles/metabolism , Particle Size , Silicon Dioxide/chemistry , Surface Properties
11.
Cell Death Dis ; 9(11): 1048, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323180

ABSTRACT

Dihydroartemisinin (DHA) and its analogs are reported to possess selective anticancer activity. Here, we reported a novel DHA derivative DHA-37 that exhibited more potent anticancer activity on the cells tested. Distinct from DHA-induced apoptosis, DHA-37 triggered excessive autophagic cell death, and became the main contributor to DHA-37-induced A549 cell death. Incubation of the cells with DHA-37 but not DHA produced increased dots distribution of GFP-LC3 and expression ratio of LC3-II/LC3-I, and enhanced the formation of autophagic vacuoles as revealed by TEM. Treatment with the autophagy inhibitor 3-MA, LY294002, or chloroquine could reverse DHA-37-induced cell death. In addition, DHA-37-induced cell death was associated significantly with the increased expression of HMGB1, and knockdown of HMGB1 could reverse DHA-37-induced cell death. More importantly, the elevated HMGB1 expression induced autophagy through the activation of the MAPK signal but not PI3K-AKT-mTOR pathway. In addition, DHA-37 also showed a wonderful performance in A549 xenograft mice model. These findings suggest that HMGB1 as a target candidate for apoptosis-resistant cancer treatment and artemisinin-based drugs could be used in inducing autophagic cell death.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Artemisinins/pharmacology , Autophagy/drug effects , Gene Expression Regulation, Neoplastic , HMGB1 Protein/genetics , Lung Neoplasms/drug therapy , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Artemisinins/chemical synthesis , Autophagy/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Burden/drug effects , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure , Xenograft Model Antitumor Assays
12.
Adv Healthc Mater ; 7(16): e1800380, 2018 08.
Article in English | MEDLINE | ID: mdl-29931753

ABSTRACT

Melittin (MEL) is recognized as a highly potent therapeutic peptide for treating various human diseases including cancer. However, the clinical applications of MEL are largely restricted by its severe hemolytic activity and nonspecific cytotoxicity. Here, it is found that MEL can form a stable supramolecular nanocomplex of ≈60 nm with the photosensitizer chlorin e6 (Ce6), which after hyaluronic acid (HA) coating can achieve robust, safe, and imaging-guided tumor ablation. The as-designed nanocomplex (denoted as MEL/Ce6@HA) shows largely reduced hemolysis and selective cytolytic activity toward cancer cells. Upon laser irradiation, the loaded photosensitive Ce6 can synergistically facilitate the membrane-lytic efficiency of melittin and greatly increase the tumor penetration depth of the complexes in multicellular tumor spheroids. In vivo experiments reveal that MEL/Ce6@HA can realize enhanced tumor accumulation, reduced liver deposition, and rapid body clearance, which are beneficial for highly efficient and safe chemo-photodynamic dual therapy. This work develops a unique supramolecular strategy for optimized in vivo delivery of melittin and may have implications for the development of peptide-based theranostics.


Subject(s)
Melitten/chemistry , Nanoparticles/chemistry , Peptides/chemistry , Photosensitizing Agents/chemistry , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Animals , Cell Line, Tumor , Female , Hemolysis/drug effects , Humans , Melitten/pharmacology , Melitten/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Photochemotherapy
13.
J Med Chem ; 61(8): 3478-3490, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29606001

ABSTRACT

Agents inducing both apoptosis and autophagic death can be effective chemotherapeutic drugs. In our present work, we synthesized two organometallic gold(III) complexes harboring C^N ligands that structurally resemble tetrahydroisoquinoline (THIQ): Cyc-Au-1 (AuL1Cl2, L1 = 3,4-dimethoxyphenethylamine) and Cyc-Au-2 (AuL2Cl2, L2 = methylenedioxyphenethylamine). In screening their in vitro activity, we found both gold complexes exhibited lower toxicity, lower resistance factors, and better anticancer activity than those of cisplatin. The organometallic gold(III) complexes accumulate in mitochondria and induce elevated ROS and an ER stress response through mitochondrial dysfunction. These effects ultimately result in simultaneous apoptosis and autophagy. Importantly, compared to cisplatin, Cyc-Au-2 exhibits lower toxicity and better anticancer activity in a murine tumor model. To the best of our knowledge, Cyc-Au-2 is the first organometallic Au(III) compound that induces apoptosis and autophagic death. On the basis of our results, we believe Cyc-Au-2 to be a promising anticancer agent or lead compound for further anticancer drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Coordination Complexes/pharmacology , Endoplasmic Reticulum Stress/drug effects , Tetrahydroisoquinolines/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Coordination Complexes/chemical synthesis , Coordination Complexes/therapeutic use , Coordination Complexes/toxicity , Drug Screening Assays, Antitumor , Humans , Ligands , Male , Mice, Nude , Mitochondria/drug effects , Molecular Structure , Reactive Oxygen Species/metabolism , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/therapeutic use , Tetrahydroisoquinolines/toxicity , Xenograft Model Antitumor Assays
14.
Sci Rep ; 8(1): 2274, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396568

ABSTRACT

To improve the specificity and penetration of anticancer peptides against tumors, in this study, we examined the effects of co-administration of the membrane-active peptide HPRP-A1 and the tumor homing/penetrating peptide iRGD. iRGD peptide is widely recognized as an efficient cell membrane penetration peptide targeting to αvß3 integrins and neuropilin-1 (NRP-1) receptors, which show high expression in many tumor cells. The anticancer activity, cancer specificity and penetration activity in vitro and in vivo of the co-administered peptides were examined on 2D monolayer cells, 3D multi-cellular spheroids (MCS) and xenograft nude mice. Co-administration of iRGD and HPRP-A1 exhibited stronger anticancer activity and tumor specificity against A549 non-small cell lung cancer cells with NRP-1 receptor overexpression compared with HPRP-A1 alone. A549 cells showed uptake of the peptide combination and destruction of the integrity of the cell membrane, as well as adherence to the mitochondrial net, resulting in induction of apoptosis by a caspase-dependent pathway. The iRGD peptide dramatically increased the penetration depth of HPRP-A1 on A549 MCS and anticancer efficacy in an A549 xenograft mouse model. Our results suggest that the co-administration strategy of anticancer and penetrating peptides could be a potential therapeutic approach for cancer treatment in clinical practice.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents/pharmacokinetics , Drug Carriers/pharmacokinetics , Membranes/metabolism , Oligopeptides/pharmacokinetics , A549 Cells , Animals , Apoptosis , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Heterografts , Humans , Mice, Nude , Neoplasm Transplantation , Spheroids, Cellular , Treatment Outcome
15.
Clin Lung Cancer ; 19(1): 84-92, 2018 01.
Article in English | MEDLINE | ID: mdl-28801183

ABSTRACT

BACKGROUND: Before mutation testing of the epidermal growth factor receptor (EGFR) gene was recognized as highly associated with the activity of EGFR tyrosine kinase inhibitors (TKIs), clinically defined patient populations with bronchioloalveolar carcinoma (BAC) and never smokers were identified as likely to benefit from EGFR TKIs. From preclinical and clinical data suggesting potentially improved efficacy with a combination of an EGFR TKI and the antiangiogenic agent bevacizumab, the Southwestern Oncology Group (SWOG) initiated paired phase II trials to evaluate the combination of erlotinib/bevacizumab in patients with advanced BAC (SWOG S0635) or never smokers with advanced lung adenocarcinoma (SWOG S0636). MATERIALS AND METHODS: Eligible patients with BAC or adenocarcinoma with BAC features (SWOG S0635) or never smokers with advanced lung adenocarcinoma (SWOG S0636) received erlotinib 150 mg/day with bevacizumab 15 mg/kg until progression or prohibitive toxicity. Never smokers with BAC were preferentially enrolled to SWOG S0636. The primary endpoint for both trials was overall survival. RESULTS: A total of 84 patients were enrolled in the SWOG S0635 trial and 85 in the SWOG S0636 trial. The objective response rate was 22% (3% complete response) in the SWOG S0635 trial and 50% (38% confirmed; 3% complete response) in the SWOG S0636 trial. The median progression-free survival was 5 and 7.4 months in the S0635 and S0636 trials, respectively. The median overall survival was 21 and 29.8 months, respectively. Toxicity consisted mainly of rash and diarrhea in both trials. CONCLUSION: Although the field has moved toward molecular, rather than clinical, selection of patients as optimal candidates for EGFR TKI therapy, these results support the hypothesis that a subset of patients in whom erlotinib is particularly active could receive an incremental benefit from the addition of bevacizumab.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Diarrhea/epidemiology , Drug-Related Side Effects and Adverse Reactions/epidemiology , Erlotinib Hydrochloride/therapeutic use , Exanthema/epidemiology , Lung Neoplasms/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/epidemiology , Adenocarcinoma, Bronchiolo-Alveolar/mortality , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Cigarette Smoking/adverse effects , Diarrhea/etiology , Diarrhea/mortality , Drug-Related Side Effects and Adverse Reactions/mortality , Erlotinib Hydrochloride/adverse effects , Exanthema/etiology , Exanthema/mortality , Female , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Survival Analysis , Treatment Outcome
16.
Sci Rep ; 7(1): 10071, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28855534

ABSTRACT

Barriers presented by the tumor microenvironment including the abnormal tumor vasculature and interstitial matrix invariably lead to heterogeneous distribution of nanotherapeutics. Inspired by the close association between cyclooxygenase-2 (COX-2) and tumor-associated angiogenesis, as well as tumor matrix formation, we proposed that tumor microenvironment normalization by COX-2 inhibitors might improve the distribution and efficacy of nanotherapeutics for solid tumors. The present study represents the first time that celecoxib, a special COX-2 inhibitor widely used in clinics, was explored to normalize the tumor microenvironment and to improve tumor nanotherapeutics delivery using a human-derived A549 tumor xenograft as the solid tumor model. Immunofluorescence staining of tumor slices demonstrated that oral celecoxib treatment at a dose of 200 mg/kg for two weeks successfully normalized the tumor microenvironment, including tumor-associated fibroblast reduction, fibronectin bundle disruption, tumor vessel normalization, and tumor perfusion improvement. Furthermore, it also significantly enhanced the in vivo accumulation and deep penetration of 22-nm micelles rather than 100-nm nanoparticles in tumor tissues by in vivo imaging and distribution experiments and improved the therapeutic efficacy of paclitaxel-loaded micelles in tumor xenograft-bearing mouse models in the pharmacodynamics experiment. As celecoxib is widely and safely used in clinics, our findings may have great potential in clinics to improve solid tumor treatment.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/genetics , Lung Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/drug effects , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/blood supply , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Cyclooxygenase 2/metabolism , Drug Administration Schedule , Drug Delivery Systems/methods , Drug Synergism , Drug Therapy, Combination , Gene Expression , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Micelles , Nanoparticles/chemistry , Nanoparticles/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
J Antibiot (Tokyo) ; 70(9): 923-928, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28676717

ABSTRACT

Trichopeptides A (1) and B (2), new linear tetrapeptide and tripeptide, respectively, and three new diketopiperazines trichocyclodipeptides A-C (3-5) were isolated from the fermentation of the ascomycete fungus Stagonospora trichophoricola, a fungus isolated from the soil sample surrounding the fruiting body of Ophiocordyceps sinensis in Maqin Country, Qinghai Province, People's Republic of China. Their structures were primarily elucidated by interpretation of NMR and MS experiments. The absolute configurations of 1-5 were assigned through Marfey's method on their acid hydrolyzates. Compound 3 showed antifungal activity against Candida albicans with the IC50 and MIC values of 22 and 90 µg ml-1, respectively.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Antibiotics, Antineoplastic/isolation & purification , Antifungal Agents/isolation & purification , Antimicrobial Cationic Peptides/isolation & purification , Ascomycota/metabolism , Drug Discovery , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Candida albicans/drug effects , Candida albicans/growth & development , Cell Survival/drug effects , Escherichia coli/drug effects , Escherichia coli/growth & development , Fermentation , Humans , Inhibitory Concentration 50 , K562 Cells , Leukemia, Erythroblastic, Acute/drug therapy , Microbial Sensitivity Tests , Oligopeptides/chemistry , Oligopeptides/isolation & purification , Oligopeptides/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Stereoisomerism
18.
Biomacromolecules ; 18(8): 2552-2563, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28700211

ABSTRACT

We report a versatile synthesis for polyphenolic polymersomes of controlled submicron (<500 nm) size for intracellular delivery of high and low molecular weight compounds. The nanoparticles are synthesized by stabilizing the vesicular morphology of thermally responsive poly(N-vinylcaprolactam)n-b-poly(N-vinylpyrrolidone)m (PVCLn-PVPONm) diblock copolymers with tannic acid (TA), a hydrolyzable polyphenol, via hydrogen bonding at a temperature above the copolymer's lower critical solution temperature (LCST). The PVCL179-PVPONm diblock copolymers are produced by controlled reversible addition-fragmentation chain transfer (RAFT) polymerization of PVPON using PVCL as a macro-chain transfer agent. The size of the TA-locked (PVCL179-PVPONm) polymersomes at room temperature and upon temperature variations are controlled by the PVPON chain length and TA:PVPON molar unit ratio. The particle diameter decreases from 1000 to 950, 770, and 250 nm with increasing PVPON chain length (m = 107, 166, 205, 234), and it further decreases to 710, 460, 290, and 190 nm, respectively, upon hydrogen bonding with TA at 50 °C. Lowering the solution temperature to 25 °C results in a slight size increase for vesicles with longer PVPON. We also show that TA-locked polymersomes can encapsulate and store the anticancer drug doxorubicin (DOX) and higher molecular weight fluorescein isothiocyanate (FITC)-dextran in a physiologically relevant pH and temperature range. Encapsulated DOX is released in the nuclei of human alveolar adenocarcinoma tumor cells after 6 h incubation via biodegradation of the TA shell with the cytotoxicity of DOX-loaded polymersomes being concentration-dependent. Our approach offers biocompatible and intracellular degradable nanovesicles of controllable size for delivery of a variety of encapsulated materials. Considering the particle monodispersity, high loading capacity, and a facile two-step aqueous assembly based on the reversible temperature-responsiveness of PVCL, these polymeric vesicles have significant potential as novel drug nanocarriers and provide a new perspective for fundamental studies on thermo-triggered polymer assemblies in solutions.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents , Caprolactam/analogs & derivatives , Doxorubicin , Drug Carriers , Lung Neoplasms/drug therapy , Polymers , Polyphenols , Povidone , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Caprolactam/chemistry , Caprolactam/pharmacokinetics , Caprolactam/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Hot Temperature , Humans , Lung Neoplasms/metabolism , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology , Polyphenols/chemistry , Polyphenols/pharmacokinetics , Polyphenols/pharmacology , Povidone/chemistry , Povidone/pharmacokinetics , Povidone/pharmacology
19.
Med Sci Monit ; 23: 223-237, 2017 Jan 14.
Article in English | MEDLINE | ID: mdl-28087861

ABSTRACT

BACKGROUND Jolkinolide A (JA) and Jolkinolide B (JB) are diterpenoids extracted from the roots of Euphorbia fischeriana Steud and have been shown to have anti-tumor activity. However, their effects on the ability of tumor cells to invade blood vessels and metastasize remain largely unknown. Investigations into the effects of JA and JB on the angiogenesis of tumor tissues may facilitate the identification of new natural drugs with anti-tumor growth and metastasis activities. MATERIAL AND METHODS We used different concentrations of JA and JB (20 µg/ml, 40 µg/ml, 60 µg/ml, 80 µg/ml, and 100 µg/ml) to stimulate A549 cells and then studied the effects on the growth and metastasis of lung cancers. In addition, we used conditional media from A549 cells (A549-CM) stimulated by either JA or JB in different concentrations to culture human umbilical vein endothelial cells (HUVECs). RESULTS We found that both JA and JB significantly inhibited the Akt-STAT3-mTOR signaling pathway and reduced the expression of VEGF in A549 cells, but JB exhibited more significant inhibitory effects than JA. The JB-stimulated A549 cell conditional media had a greater inhibitory effect on the proliferation and migration of HUVECs than did the conditional media of JA-stimulated A549 cells. This effect gradually increased with increasing concentrations of either type of Jolkinolide. CONCLUSIONS Our results suggest that JA and JB inhibited VEGF expression in A549 cells through the inhibition of the Akt-STAT3-mTOR signaling pathway, and directly inhibited the proliferation and migration of HUVECs. These findings are of great significance for the development of new plant-derived chemotherapy agents for the treatment of cancer.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Diterpenes/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Lung Neoplasms/drug therapy , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drugs, Chinese Herbal/pharmacology , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/drug effects , Xenograft Model Antitumor Assays
20.
J Thorac Oncol ; 12(1): 145-151, 2017 01.
Article in English | MEDLINE | ID: mdl-27697581

ABSTRACT

INTRODUCTION: This phase I trial was conducted to determine the safety, maximum tolerated dose (MTD)/recommended phase II dose, and efficacy of crizotinib plus erlotinib in patients with advanced NSCLC. METHODS: Patients with NSCLC and an Eastern Cooperative Oncology Group performance status of 0 to 2 after failure of one or two prior chemotherapy regimens were eligible. Erlotinib, 100 mg, was given continuously once daily starting between day -14 and -7; crizotinib, 200 mg twice daily (dose level 1) or 150 mg twice daily (dose level -1), was added continuously beginning on day 1 of treatment cycle 1. Potential pharmacokinetic interactions between crizotinib and erlotinib were evaluated. RESULTS: Twenty-seven patients received treatment; 26 received crizotinib plus erlotinib. Frequent adverse events were diarrhea, rash, decreased appetite, and fatigue. Dose-limiting toxicities were dehydration, diarrhea, dry eye, dysphagia, dyspepsia, esophagitis and vomiting. The MTD was crizotinib, 150 mg twice daily, with erlotinib, 100 mg once daily. Crizotinib increased the erlotinib area under the concentration-time curve 1.5-fold (dose level -1) and 1.8-fold (dose level 1). The plasma level of crizotinib appeared to be unaffected by coadministration of erlotinib. Two patients whose tumors harbored activating EGFR mutations achieved confirmed partial responses, one at each crizotinib dose level. CONCLUSIONS: The MTD of the combination of crizotinib and erlotinib in patients with advanced NSCLC was crizotinib, 150 mg twice daily, with erlotinib, 100 mg once daily, which is less than the approved dose of either agent. The phase II portion of the study was not initiated.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Papillary/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/secondary , Adenocarcinoma, Bronchiolo-Alveolar/secondary , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Papillary/secondary , Crizotinib , Erlotinib Hydrochloride/administration & dosage , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Prognosis , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Survival Rate , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...