Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
PLoS One ; 16(6): e0252590, 2021.
Article in English | MEDLINE | ID: mdl-34086763

ABSTRACT

Conditions that resemble osteoarthritis (OA) were produced by injection of sodium monoiodoacetate (MIA) into the knee joints of mice. Bone marrow derived mast cells (BMMCs) injected into the OA knee joints enhanced spontaneous pain. Since no spontaneous pain was observed when BMMCs were injected into the knee joints of control mice that had not been treated with MIA, BMMCs should be activated within the OA knee joints and release some pain-inducible factors. Protease activated receptor-2 (PAR2) antagonist (FSLLRY-NH2) almost abolished the pain-enhancing effects of BMMCs injected into the OA knee joints, suggesting that tryptase, a mast cell protease that is capable of activating PAR2, should be released from the injected BMMCs and enhance pain through activation of PAR2. When PAR2 agonist (SLIGKV-NH2) instead of BMMCs was injected into the OA knee joints, it was also enhanced pain. Apyrase, an ATP degrading enzyme, injected into the OA knee joints before BMMCs suppressed the pain enhanced by BMMCs. We showed that purinoceptors (P2X4 and P2X7) were expressed in BMMCs and that extracellular ATP stimulated the release of tryptase from BMMCs. These observations suggest that ATP may stimulate degranulation of BMMCs and thereby enhanced pain. BMMCs injected into the OA knee joints stimulated expression of IL-1ß, IL-6, TNF-α, CCL2, and MMP9 genes in the infrapatellar fat pads, and PAR2 antagonist suppressed the stimulatory effects of BMMCs. Our study suggests that intermittent pain frequently observed in OA knee joints may be due, at least partly, to mast cells through activation of PAR2 and action of ATP, and that intraarticular injection of BMMCs into the OA knee joints may provide a useful experimental system for investigating molecular mechanisms by which pain is induced in OA knee joints.


Subject(s)
Adenosine Triphosphate/metabolism , Arthritis, Experimental/therapy , Chronic Pain/pathology , Knee Joint/pathology , Mast Cells/transplantation , Receptor, PAR-2/metabolism , Adenosine Triphosphate/analysis , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Bone Marrow Cells/cytology , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/toxicity , Chronic Pain/etiology , Disease Models, Animal , Knee Joint/metabolism , Male , Mast Cells/cytology , Mast Cells/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/administration & dosage , Receptor, PAR-2/agonists , Receptor, PAR-2/antagonists & inhibitors , Receptors, Purinergic/metabolism , Synovial Fluid/metabolism
2.
Article in English | MEDLINE | ID: mdl-34023535

ABSTRACT

Neuropeptide AF (NPAF) decreases food and water intake in birds and food intake in mammals. In this study, the objective was to determine the effects of centrally administered NPAF on food and water intake, hypothalamic c-Fos immunoreactivity and hypothalamic mRNA abundance of appetite-regulating factors in Japanese quail (Coturnix japonica). Seven days post hatch, 6 h fasted quail were intracerebroventricularly (ICV) injected with 0 (vehicle), 4, 8, or 16 nmol of NPAF and food and water intake were measured at 30 min intervals for 180 min. In Experiment 1, chicks which received 4, 8, and 16 nmol ICV NPAF had reduced food intake for 120, 60 and 180 min following injection, respectively, and reduced water intake during the entire 180 min observation. In Experiment 2, there was increased c-Fos immunoreactivity in the paraventricular nucleus, the ventromedial nucleus of the hypothalamus, and the dorsomedial hypothalamic nucleus in NPAF-injected quail. In Experiment 3, ICV NPAF was associated with decreased corticotropin-releasing factor mRNA, and an increase in hypothalamic proopiomelanocortin and melanocortin receptor 4 mRNA. These results demonstrate that central NPAF suppresses food and water intake in quail, effects that are likely mediated via the melanocortin system in the hypothalamus.


Subject(s)
Appetite/drug effects , Drinking/drug effects , Feeding Behavior/drug effects , Hypothalamus/drug effects , Melanocortins/metabolism , Oligopeptides/administration & dosage , Animals , Anorexia/chemically induced , Corticotropin-Releasing Hormone/metabolism , Coturnix/metabolism , Disease Models, Animal , Hypothalamus/metabolism , Infusions, Intraventricular , Paraventricular Hypothalamic Nucleus , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Signal Transduction
3.
Sci Rep ; 11(1): 9304, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33927292

ABSTRACT

The novel procedure of few-layer black phosphorus (FLBP) stabilization and functionalisation was here proposed. The cationic polymer PLL and non-ionic PEG have been involved into encapsulation of FLBP to allow sufficient time for further nanofabrication process and overcome environmental degradation. Two different spacer chemistry was designed to bind polymers to tumor-homing peptides. The efficiency of functionalisation was examined by RP-HPLC, microscopic (TEM and SEM) and spectroscopic (FT-IR and Raman) techniques as well supported by ab-initio modelling. The cell and dose dependent cytotoxicity of FLBP and its bioconjugates was evaluated against HB2, MCF-7 and MDA-MB-231 cell lines. Functionalisation allowed not only for improvement of environmental stability, but also enhances therapeutic effect by abolished the cytotoxicity of FLBP against HB2 cell line. Moreover, modification of FLBP with PLL caused increase of selectivity against highly aggressive breast cancer cell lines. Results indicate the future prospect application of black phosphorus nanosheets as nanocarrier, considering its unique features synergistically with conjugated polymeric micelles.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Carriers , Nanostructures , Oligopeptides/pharmacology , Phosphorus , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Survival , Female , Humans , MCF-7 Cells , Micelles , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Phosphorus/pharmacology , Polyethylene Glycols , Polylysine
4.
J Am Heart Assoc ; 10(9): e020006, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33870715

ABSTRACT

Background Survivors of myocardial infarction are at increased risk of late ventricular arrhythmias, with infarct size and scar heterogeneity being key determinants of arrhythmic risk. Gap junctions facilitate the passage of small ions and morphogenic cell signaling between myocytes. We hypothesized that gap junctions enhancement during infarction-reperfusion modulates structural and electrophysiological remodeling and reduces late arrhythmogenesis. Methods and Results Infarction-reperfusion surgery was carried out in male Sprague-Dawley rats followed by 7 days of rotigaptide or saline administration. The in vivo and ex vivo arrhythmogenicity was characterized by programmed electrical stimulation 3 weeks later, followed by diffusion-weighted magnetic resonance imaging and Masson's trichrome histology. Three weeks after 7-day postinfarction administration of rotigaptide, ventricular tachycardia/ventricular fibrillation was induced on programmed electrical stimulation in 20% and 53% of rats, respectively (rotigaptide versus control), resulting in reduction of arrhythmia score (3.2 versus 1.4, P=0.018), associated with the reduced magnetic resonance imaging parameters fractional anisotropy (fractional anisotropy: -5% versus -15%; P=0.062) and mean diffusivity (mean diffusivity: 2% versus 6%, P=0.042), and remodeling of the 3-dimensional laminar structure of the infarct border zone with reduction of the mean (16° versus 19°, P=0.013) and the dispersion (9° versus 12°, P=0.015) of the myofiber transverse angle. There was no change in ECG features, spontaneous arrhythmias, or mortality. Conclusions Enhancement of gap junctions function by rotigaptide administered during the early healing phase in reperfused infarction reduces later complexity of infarct scar morphology and programmed electrical stimulation-induced arrhythmias, and merits further exploration as a feasible and practicable intervention in the acute myocardial infarction management to reduce late arrhythmic risk.


Subject(s)
Arrhythmias, Cardiac/etiology , Electrophysiologic Techniques, Cardiac/methods , Magnetic Resonance Imaging, Cine/methods , Myocardial Infarction/drug therapy , Myocardium/pathology , Oligopeptides/administration & dosage , Ventricular Remodeling/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Infusions, Intravenous , Male , Myocardial Infarction/complications , Myocardial Infarction/diagnosis , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Time Factors
5.
Int J Nanomedicine ; 16: 1037-1050, 2021.
Article in English | MEDLINE | ID: mdl-33603367

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is one of the most common fatal cancers, with no curative therapy available. The concept of ferroptosis is attracting increasing attention in cancer research. Herein, we describe the use of a nanodevice as an effective strategy for inducing ferroptosis to manage HCC. METHODS: To improve ferroptosis-induced treatment of HCC, we constructed sorafenib (sor)-loaded MIL-101(Fe) nanoparticles (NPs) [MIL-101(Fe)@sor] and evaluated the efficacy of ferroptosis-based HCC therapy after co-administration with the iRGD peptide both in vitro and in vivo. RESULTS: The prepared MIL-101(Fe) NPs have several promising characteristics including drug-loading, controllable release, peroxidase activity, biocompatibility, and T2 magnetic resonance imaging ability. MIL-101(Fe)@sor NPs significantly induced ferroptosis in HepG2 cells, increased the levels of lipid peroxidation and malondialdehyde, and reduced those of glutathione and glutathione peroxidase 4 (GPX-4). The in vivo results showed that the MIL-101(Fe)@sor NPs significantly inhibited tumor progression and decreased GPX-4 expression levels, with negligible long-term toxicity. Meanwhile, co-administration of MIL-101(Fe)@sor NPs with iRGD significantly accelerated ferroptosis. CONCLUSION: Our findings suggest that MIL-101(Fe)@sor NPs co-administered with iRGD are a promising strategy for inducing HCC ferroptosis.


Subject(s)
Ferroptosis , Iron/chemistry , Liver Neoplasms/drug therapy , Metal-Organic Frameworks/chemistry , Oligopeptides/administration & dosage , Oligopeptides/therapeutic use , Sorafenib/administration & dosage , Sorafenib/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Endocytosis/drug effects , Ferroptosis/drug effects , Hep G2 Cells , Humans , Lipid Peroxidation/drug effects , Liver Neoplasms/pathology , Metal-Organic Frameworks/ultrastructure , Mice , Nanoparticles/chemistry , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Peroxidase/metabolism , Rabbits , Sorafenib/pharmacology , Toxicity Tests
6.
Biomater Sci ; 8(12): 3443-3453, 2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32412569

ABSTRACT

Reliable diagnosis and efficient targeted therapy are important and may lead to the effective treatment of laryngeal carcinoma. Multifunctional nano-theranostic agents demonstrate great potential in tumor theranostic applications. Thus, herein, we report novel targeting multifunctional theranostic nanoparticles, internalized RGD (iRGD)-modified indocyanine green (ICG) encapsulated liposomes (iLIPICG), for imaging-guided photothermal therapy (PTT) and photodynamic therapy (PDT) for the treatment of laryngeal carcinoma. The iRGD-PEG-DSPE lipid endowed iLIPICG with high affinity for tumor vascular targeting, tumor-penetration and tumor cell targeting. The in vivo results showed that iLIPICG exhibited excellent blood circulation and tumor accumulation. iLIPICG could be spatially and temporally controlled, simultaneously producing hyperthermia and reactive oxygen species as well as a fluorescence-guided effect through ICG to ablate laryngeal carcinoma cells under irradiation from an 808 nm laser. iLIPICG generated synergistic photodynamic-photothermal cytotoxicity against Hep-2 cells, resulting in the efficient ablation of laryngeal carcinoma. Thus, the iLIPICG system provides a promising strategy to improve the precision imaging and effective phototherapy for the treatment of laryngeal carcinoma.


Subject(s)
Coloring Agents/administration & dosage , Indocyanine Green/administration & dosage , Laryngeal Neoplasms/therapy , Oligopeptides/administration & dosage , Phototherapy , Animals , Cell Line, Tumor , Coloring Agents/chemistry , Coloring Agents/pharmacokinetics , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Lasers , Liposomes , Male , Mice, SCID , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Optical Imaging , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacokinetics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Reactive Oxygen Species/metabolism
7.
FASEB J ; 34(4): 5061-5076, 2020 04.
Article in English | MEDLINE | ID: mdl-32043638

ABSTRACT

Recently, interest in using whole food-derived mixtures to alleviate chronic metabolic syndrome through potential synergistic interactions among different components is increasing. In this study, the effects and mechanisms of tuna meat oligopeptides (TMOP) on hyperuricemia and associated renal inflammation were investigated in mice. Dietary administration of TMOP alleviated hyperuricemia and renal inflammation phenotypes, reprogramed uric acid metabolism pathways, inhibited the activation of NLRP3 inflammasome and TLR4/MyD88/NF-κB signaling pathways, and suppressed the phosphorylation of p65-NF-κB. In addition, TMOP treatments repaired the intestinal epithelial barrier, reversed the gut microbiota dysbiosis and increased the production of short-chain fatty acids. Moreover, the antihyperuricemia effects of TMOP were transmissible by transplanting the fecal microbiota from TMOP-treated mice, indicating that the protective effects were at least partially mediated by the gut microbiota. Thus, for the first time, we clarify the potential effects of TMOP as a whole food derived ingredient on alleviating hyperuricemia and renal inflammation in mice, and additional efforts are needed to confirm the beneficial effects of TMOP on humans.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Fish Proteins, Dietary/therapeutic use , Gastrointestinal Microbiome , Hyperuricemia/drug therapy , Nephritis/drug therapy , Oligopeptides/therapeutic use , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Dietary Supplements , Fish Proteins, Dietary/administration & dosage , Fish Proteins, Dietary/chemistry , Hyperuricemia/microbiology , Intestinal Mucosa/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred ICR , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephritis/microbiology , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Toll-Like Receptor 4/metabolism , Tuna , Uric Acid/metabolism
8.
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
9.
Int J Pharm ; 577: 119034, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31991183

ABSTRACT

Relapse in Allergic Rhinitis (AR) is triggered by various unclear mechanisms. Xanthium strumarium L. as a traditional folk medicine can inhibit inflammatory responses through multiple mechanisms. Xanthatin (XT) is a bioactive compound derived from Xanthium strumarium L, and we developed a polymeric micelle (PM) that is dendritic cells (DCs)-specific targeting delivery system loading XT (NGR-XT-PM) based on a cyclic peptide moiety (NGR) to render DCs maturation-resistant for therapy of refractory AR. A murine model of AR was employed to investigate the in vivo therapeutic efficiency and relapse rate compared with the commercial product Budesonide. The results showed intranasal administration of NGR-XT-PM presented significant anti-allergy effect with no recurrence, in contrast, all mice treatment with Budesonide relapsed. NGR-XT-PM could effectively reverse the Th1/Th2 imbalance by depleting the serum inflammatory levels (IgE, histamine and IL-4) and DCs surface costimulatory molecules (CD80, CD86 and I-A/I-E), and promote immune tolerance by upregulating the level of Treg cells and reducing the levels of Th2, Th9 and Th17 cells. Furthermore, we appealed to virtual screening of inflammatory targets and found XT blocking the COX-2/PGE2 signaling pathway, which is a key effector in immune responses. These indicated CD13-specific NGR could facilitate XT selectively targeting DCs for efficiently ameliorating refractory rhinitis, and NGR-XT-PM should be a potential anti-AR drug.


Subject(s)
CD13 Antigens/chemistry , Dendritic Cells/drug effects , Drug Delivery Systems/methods , Furans/chemistry , Furans/pharmacology , Oligopeptides/chemistry , Rhinitis, Allergic/prevention & control , Administration, Intranasal , Animals , Budesonide/pharmacology , CD13 Antigens/administration & dosage , Dendritic Cells/immunology , Furans/administration & dosage , Inflammation Mediators/blood , Male , Mice , Micelles , Nanomedicine/methods , Oligopeptides/administration & dosage , Rhinitis, Allergic/blood , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects
10.
J Cancer Res Ther ; 16(7): 1703-1709, 2020.
Article in English | MEDLINE | ID: mdl-33565520

ABSTRACT

PURPOSE: Transcatheter arterial chemoembolization (TACE) is the first-line therapy for unresectable hepatocellular carcinoma (HCC). However, its therapeutic effects are hampered by the poor distribution of anticancer drugs in tumors. iRGD, a novel tumor-penetrating peptide, enhances the penetration distance and therapeutic efficacy of anticancer drugs. Herein, we evaluated the therapeutic effects of iRGD coupled with TACE in the rabbit VX2 liver tumor model. SUBJECTS AND METHODS: This study had two stages: tumor permeability assay and anticancer efficacy evaluation. In the tumor permeability assay, we coadministered TACE with either iRGD + lipiodol-doxorubicin emulsion (LDE) or LDE in the rabbit VX2 liver tumor model. We evaluated the doxorubicin (DOX) distribution at predetermined times by immunofluorescence microscopy. To evaluate anticancer efficacy, we administered saline, LDE, or iRGD + LDE to tumor-grafted rabbits. We measured tumor volume using magnetic resonance scanning. We quantified the expression levels of Bax, Bcl-2, and cleaved caspase-3 using Western blot (WB) analysis and determined the apoptosis rate in tumor cells using transferase-mediated dUTP nick-end labeling assay. RESULTS: The iRGD + LDE infusion significantly increased the DOX concentration and DOX penetration in tumors compared with the LDE infusion (P < 0.05). The antitumor efficacy of the iRGD + LDE in tumor inhibition was higher than that of the other treatments (P < 0.05). Besides, iRGD + LDE induced more apoptosis (P < 0.05). CONCLUSIONS: We demonstrated that iRGD coadministered with TACE is effective against HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Liver Neoplasms/therapy , Oligopeptides/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis/drug effects , Arteries/surgery , Carcinoma, Hepatocellular/pathology , Catheters , Cell Line, Tumor , Chemoembolization, Therapeutic/instrumentation , Disease Models, Animal , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Ethiodized Oil/administration & dosage , Ethiodized Oil/pharmacokinetics , Humans , Liver/diagnostic imaging , Liver/pathology , Liver Neoplasms/pathology , Magnetic Resonance Imaging , Male , Oligopeptides/pharmacokinetics , Permeability , Rabbits , Tumor Burden
11.
Drug Deliv ; 27(1): 15-25, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31830840

ABSTRACT

Small interfering RNA (siRNA) exhibits great potential as a novel therapeutic option due to its highly sequence-specific ability to silence genes. However, efficient and safe delivery carriers are required for developing novel therapeutic paradigms. Thus, the successful development of efficient delivery platforms for siRNA is a crucial issue for the development of siRNA-based drugs in cancer treatments. In this study, biocompatible selenium nanoparticles (SeNPs) were loaded with RGDfC peptide to fabricate tumor-targeting gene delivery vehicle RGDfC-SeNPs. Subsequently, RGDfC-SeNPs were loaded with Derlin1-siRNA to fabricate RGDfC-Se@siRNA, which are functionalized selenium nanoparticles. RGDfC-Se@siRNA showed greater uptake in HeLa cervical cancer cells in comparison with that in human umbilical vein endothelial cells (HUVECs), verifying the RGDfC-mediated specific uptake of RGDfC-Se@siRNA. RGDfC-Se@siRNA was capable of entering HeLa cells via clathrin-associated endocytosis, and showed faster siRNA release in a cancer cell microenvironment in comparison with a normal physiological environment. qPCR and western blotting assays both indicated that RGDfC-Se@siRNA exhibited an obvious gene silencing efficacy in HeLa cells. RGDfC-Se@siRNA suppressed the invasion, migration and the proliferation of HeLa cells, and triggered HeLa cell apoptosis. Moreover, RGDfC-Se@siRNA induced the disruption of mitochondrial membrane potentials. Meanwhile, RGDfC-Se@siRNA enhanced the generation of reactive oxygen species (ROS) in HeLa cell, suggesting that mitochondrial dysfunction mediated by ROS might play a significant role in RGDfC-Se@siRNA-induced apoptosis. Interestingly, RGDfC-SeNPs@siRNA exhibited significant antitumor activity in a HeLa tumor-bearing mouse model. Additionally, RGDfC-SeNPs@siRNA is nontoxic to main organ of mouse. The above results indicate that RGDfC-Se@siRNA provides a promising potential for cervical cancer therapy.


Subject(s)
Membrane Proteins/drug effects , Nanoparticles/chemistry , Oligopeptides/pharmacology , RNA, Small Interfering/administration & dosage , Selenium/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Migration Inhibition , Cell Proliferation/drug effects , Female , Gene Silencing/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Reactive Oxygen Species/metabolism , Tumor Microenvironment , Uterine Cervical Neoplasms/drug therapy
12.
J Vasc Interv Radiol ; 30(12): 2026-2035.e2, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31590966

ABSTRACT

PURPOSE: To evaluate the effect of transarterial infusion of iRGD-modified and doxorubicin-loaded zirconia-composite nanoparticles (R-DZCNs) with lipiodol in the improvement of the distribution of doxorubicin (DOX) in liver tumors and its antitumor efficacy. MATERIALS AND METHODS: The effect of R-DZCNs was evaluated in vitro by tumor cellular uptake and cytotoxicity assays. For the in vivo study, DOX distribution and antitumor efficiency were assessed. In the DOX distribution study, VX2 tumor-bearing rabbits received transarterial infusion of lipiodol with DOX, doxorubicin-loaded zirconia-composite nanoparticles (DZCNs), or R-DZCNs, respectively. DOX distribution was assessed by immunofluorescence. In the antitumor study, tumor-bearing rabbits received transarterial infusions of lipiodol with DOX, DZCNs, R-DZCNs, or saline respectively. Tumor volume was measured using magnetic resonance imaging, and the expression of apoptosis-related factors (caspase-3, Bax, Bcl-2) was analyzed by immunohistochemistry and Western blotting. RESULTS: R-DZCNs increased cellular uptake and caused stronger cytotoxicity. Compared with the DOX + lipiodol or DZCNs + lipiodol group, the R-DZCNs + lipiodol group showed more DOX fluorescence spots (2,449.15 ± 444.14 vs. 3,464.73 ± 632.75 or 5,062.25 ± 585.62, respectively; P < .001) and longer penetration distance (117.58 ± 19.36 vs 52.64 ± 8.53 or 83.37 ± 13.76 µm, respectively; P < .001). In the antitumor study, the R-DZCNs + lipiodol group showed smaller tumor volumes than the DOX + lipiodol or DZCNs + lipiodol group (1,223.87 ± 223.58 vs. 3,695.26 ± 666.25 or 2281.06 ± 457.21 mm3, respectively; P = .005).The greatest extent of tumor cell apoptosis was observed in R-DZCNs + lipiodol group immunohistochemistry and Western blotting results. CONCLUSIONS: Transarterial infusion of R-DZCNs with lipiodol improved the distribution of DOX and enhanced its antitumor efficacy.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Chemoembolization, Therapeutic , Doxorubicin/administration & dosage , Drug Carriers , Ethiodized Oil/administration & dosage , Liver Neoplasms, Experimental/drug therapy , Metal Nanoparticles , Oligopeptides/administration & dosage , Zirconium/administration & dosage , Animals , Antibiotics, Antineoplastic/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Hep G2 Cells , Humans , Infusions, Intra-Arterial , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Magnetic Resonance Imaging , Male , Oligopeptides/metabolism , Rabbits , Tissue Distribution , Tumor Burden/drug effects , Zirconium/metabolism
13.
Nano Lett ; 19(11): 8333-8341, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31657935

ABSTRACT

A group of chemotherapeutic drugs has gained increasing interest in cancer immunotherapy due to the potential to induce immunogenic cell death (ICD). A critical challenge in using the ICD inducers in cancer immunotherapy is the immunotoxicity accompanying their antiproliferative effects. To alleviate this, a nanocapsule formulation of carfilzomib (CFZ), an ICD-inducing proteasome inhibitor, was developed using interfacial supramolecular assembly of tannic acid (TA) and iron, supplemented with albumin coating. The albumin-coated CFZ nanocapsules (CFZ-pTA-alb) attenuated CFZ release, reducing toxicity to immune cells. Moreover, due to the adhesive nature of the TA assembly, CFZ-pTA-alb served as a reservoir of damage-associated molecular patterns released from dying tumor cells to activate dendritic cells. Upon intratumoral administration, CFZ-pTA-alb prolonged tumor retention of CFZ and showed consistently greater antitumor effects than cyclodextrin-solubilized CFZ (CFZ-CD) in B16F10 and CT26 tumor models. Unlike CFZ-CD, the locally injected CFZ-pTA-alb protected or enhanced CD8+ T cell population in tumors, helped develop splenocytes with tumor-specific interferon-γ response, and delayed tumor development on the contralateral side in immunocompetent mice (but not in athymic nude mice), supporting that CFZ-pTA-alb contributed to activating antitumor immunity. This study demonstrates that sustained delivery of ICD inducers by TA-based nanocapsules is an effective way of translating local ICD induction to systemic antitumor immunity.


Subject(s)
Antineoplastic Agents/administration & dosage , Nanocapsules/chemistry , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Tannins/chemistry , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Humans , Immunity/drug effects , Immunogenic Cell Death/drug effects , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Oligopeptides/therapeutic use
14.
Lancet Oncol ; 20(9): 1286-1294, 2019 09.
Article in English | MEDLINE | ID: mdl-31375469

ABSTRACT

BACKGROUND: National Comprehensive Cancer Network guidelines consider 18F-fluciclovine PET-CT for prostate cancer biochemical recurrence localisation after radical prostatectomy, whereas European Association of Urology guidelines recommend prostate-specific membrane antigen (PSMA) PET-CT. To the best of our knowledge, no prospective head-to-head comparison between these tests has been done so far. The aim of this study was to compare prospectively paired 18F-fluciclovine and PSMA PET-CT scans for localising biochemical recurrence of prostate cancer after radical prostatectomy in patients with low prostate-specific antigen (PSA) concentrations (<2·0 ng/mL). METHODS: This was a prospective, single-centre, open-label, single-arm comparative study done at University of California Los Angeles (Los Angeles, CA, USA). Patients older than 18 years of age with prostate cancer biochemical recurrence after radical prostatectomy and PSA levels ranging from 0·2 to 2·0 ng/mL without any prior salvage therapy and with a Karnofsky performance status of at least 50 were eligible. Patients underwent 18F-fluciclovine (reference test) and PSMA (index test) PET-CT scans within 15 days. Detection rate of biochemical recurrence at the patient level and by anatomical region was the primary endpoint. A statistical power analysis demonstrated that a sample size of 50 patients was needed to show a 22% difference in detection rates in favour of PSMA (test for superiority). Each PET scan was interpreted by three independent masked readers and a consensus majority interpretation was generated (two vs one) to determine positive findings. This study is registered with ClinicalTrials.gov, number NCT02940262, and is complete. FINDINGS: Between Feb 26, 2018, and Sept 20, 2018, 143 patients were screened for eligibility, of whom 50 patients were enrolled into the study. Median follow-up was 8 months (IQR 7-9). The primary endpoint was met; detection rates were significantly lower with 18F-fluciclovine PET-CT (13 [26%; 95% CI 15-40] of 50) than with PSMA PET-CT (28 [56%; 41-70] of 50), with an odds ratio (OR) of 4·8 (95% CI 1·6-19·2; p=0·0026) at the patient level; in the subanalysis of the pelvic nodes region (four [8%; 2-19] with 18F-fluciclovine vs 15 [30%; 18-45] with PSMA PET-CT; OR 12·0 [1·8-513·0], p=0·0034); and in the subanalysis of any extrapelvic lesions (none [0%; 0-6] vs eight [16%; 7-29]; OR non-estimable [95% CI non-estimable], p=0·0078). INTERPRETATION: With higher detection rates, PSMA should be the PET tracer of choice when PET-CT imaging is considered for subsequent treatment management decisions in patients with prostate cancer and biochemical recurrence after radical prostatectomy and low PSA concentrations (≤2·0 ng/mL). Further research is needed to investigate whether higher detection rates translate into improved oncological outcomes. FUNDING: None.


Subject(s)
Carboxylic Acids/administration & dosage , Cyclobutanes/administration & dosage , Edetic Acid/analogs & derivatives , Neoplasm Recurrence, Local/diagnostic imaging , Oligopeptides/administration & dosage , Positron Emission Tomography Computed Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Aged , Contrast Media/administration & dosage , Edetic Acid/administration & dosage , Gallium Isotopes , Gallium Radioisotopes , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Prospective Studies , Prostate-Specific Antigen/blood , Prostatectomy/methods , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
15.
Pediatr Blood Cancer ; 66(8): e27765, 2019 08.
Article in English | MEDLINE | ID: mdl-31012549

ABSTRACT

BACKGROUND: Denintuzumab mafodotin (SGN-CD19A) is a CD19-targeting antibody-drug conjugate, comprising a monoclonal antibody conjugated to the potent cytotoxin monomethyl auristatin F. Since denintuzumab mafodotin has previously shown activity against B-cell malignancies in early-stage clinical trials, it was of interest to test it against the Pediatric Preclinical Testing Program preclinical models of CD19+ pediatric acute lymphoblastic leukemia (ALL). PROCEDURES: Denintuzumab mafodotin was evaluated against eight B-cell lineage ALL patient-derived xenografts (PDXs), representing B-cell precursor ALL, Ph-like ALL, and mixed-lineage leukemia rearranged infant ALL. Denintuzumab mafodotin was administered weekly for 3 weeks at 3 mg/kg. It was also tested in combination with an induction-type chemotherapy regimen of vincristine, dexamethasone, and l-asparaginase (VXL) against three PDXs. The relationship between cell surface and gene expression of CD19 and drug activity was also assessed. RESULTS: Denintuzumab mafodotin significantly delayed the progression of seven of eight PDXs tested and achieved objective responses in five of eight. There was no apparent subtype specificity of denintuzumab mafodotin activity. No correlations were observed between CD19 mRNA or cell surface expression and denintuzumab mafodotin activity, perhaps due to small sample size, and denintuzumab mafodotin treatment did not select for reduced CD19 expression. Combining denintuzumab mafodotin with VXL achieved therapeutic enhancement compared to either treatment alone. CONCLUSIONS: Denintuzumab mafodotin showed single-agent activity against selected B-lineage ALL PDXs, although leukemia growth was evident in most models at 28 days from treatment initiation. This level of activity for denintuzumab mafodotin is consistent with that observed in adults with ALL.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD19/immunology , Immunoconjugates/administration & dosage , Oligopeptides/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Animals , Antigens, CD19/metabolism , Child , Child, Preschool , Drug Evaluation, Preclinical , Female , Humans , Infant , Infant, Newborn , Male , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
J Neuroimmunol ; 332: 8-15, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30925295

ABSTRACT

CD40/CD154-interaction is critical in the development of Experimental Autoimmune Encephalomyelitis (EAE; mouse model of Multiple Sclerosis). Culprit CD4+CD40+ T cells drive a more severe form of EAE than conventional CD4 T cells. Blocking CD40/CD154-interaction with CD154-antibody prevents or ameliorates disease but had thrombotic complications in clinical trials. We targeted CD40 using a CD154-sequence based peptide. Peptides in human therapeutics demonstrate good safety. A small peptide, KGYY6, ameliorates EAE when given as pretreatment or at first symptoms. KGYY6 binds Th40 and memory T cells, affecting expression of CD69 and IL-10 in the CD4 T cell compartment, ultimately hampering disease development.


Subject(s)
CD40 Antigens/antagonists & inhibitors , CD40 Ligand/chemistry , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Oligopeptides/therapeutic use , T-Lymphocyte Subsets/drug effects , Animals , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Immunologic Memory , Mice , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Oligopeptides/chemistry , T-Lymphocyte Subsets/immunology
17.
J Agric Food Chem ; 67(5): 1437-1442, 2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30609899

ABSTRACT

We recently identified a novel, potent antihypertensive peptide, Leu-Arg-Ala (LRA; minimum effective dose = 0.25 mg/kg), from rice bran protein. In this study, we found that LRA potently relaxed mesenteric arteries isolated from spontaneously hypertensive rats (SHRs) (EC50 = 0.1 µM). In contrast, the vasorelaxant activity of each amino acid that constitutes the LRA tripeptide was remarkably attenuated. The LRA-induced vasorelaxant activity was inhibited by N(G)-nitro-l-arginine methyl ester (L-NAME; NO synthase [NOS] inhibitor) but not by an antagonist of bradykinin B2 and Mas receptors or by a phosphoinositide 3-kinase inhibitor. The antihypertensive effect induced after the oral administration of LRA was inhibited by L-NAME. LRA also induced the phosphorylation of endothelial NOS in human umbilical vein endothelial cells. Taken together, LRA may exhibit antihypertensive effects via NO-mediated vasorelaxation. LRA is the first example of a NO-dependent vasorelaxant peptide identified from rice bran protein.


Subject(s)
Antihypertensive Agents/administration & dosage , Human Umbilical Vein Endothelial Cells/drug effects , Hypertension/drug therapy , Nitric Oxide/metabolism , Oligopeptides/administration & dosage , Oryza/chemistry , Plant Extracts/administration & dosage , Vasodilator Agents/administration & dosage , Animals , Antihypertensive Agents/isolation & purification , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/metabolism , Hypertension/physiopathology , Male , Oligopeptides/isolation & purification , Plant Extracts/isolation & purification , Rats , Rats, Inbred SHR , Seeds/chemistry , Vasodilation/drug effects , Vasodilator Agents/isolation & purification
18.
Eur Urol ; 75(4): 548-551, 2019 04.
Article in English | MEDLINE | ID: mdl-30578119

ABSTRACT

In prostate cancer, disease progression after primary treatment and subsequent androgen deprivation therapy is common. Intensification of systemic treatment is the standard of care. Recently, 68Ga prostate-specific membrane antigen positron emission tomography (PSMA-PET) imaging was introduced to identify oligometastatic prostate cancer patients. In this retrospective, exploratory study, we report on the efficacy of PSMA-PET-guided local ablative radiotherapy (aRT) in 15 oligometastatic castration-resistant prostate cancer (CRPC) patients, selected from our prospective institutional database and treated between 2013 and 2016. After multidisciplinary discussion, aRT was delivered with two different schedules. Androgen deprivation therapy remained unchanged. Prostate-specific antigen (PSA) response and time to PSA progression were analysed. For comparison, individual time to PSA progression without aRT was estimated by individual PSA doubling time (PSADT). PSA response was observed in 11 patients (73%). Mean time to PSA progression or last follow-up was 17.9mo, as opposed to 2.9mo estimated from the PSADT without aRT (p<0.001). A relevant subset of CRPC patients had a PSA response with aRT to PET-positive lead metastases. A prospective trial is in preparation. PATIENT SUMMARY: In selected patients with prostate-specific antigen (PSA) increase during androgen deprivation, metastases were detected with prostate-specific membrane antigen positron emission tomography imaging. Fifteen patients with three or fewer metastases were treated with high-dose radiotherapy. Subsequently, PSA values dropped in 11 patients and in six patients no PSA progression was detected for >12mo.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiosurgery , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Databases, Factual , Disease Progression , Dose Fractionation, Radiation , Edetic Acid/administration & dosage , Edetic Acid/analogs & derivatives , Gallium Isotopes , Gallium Radioisotopes , Humans , Kallikreins/blood , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Oligopeptides/administration & dosage , Positron-Emission Tomography , Progression-Free Survival , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/diagnostic imaging , Prostatic Neoplasms, Castration-Resistant/pathology , Radiopharmaceuticals/administration & dosage , Radiosurgery/adverse effects , Retrospective Studies , Time Factors
19.
J Mater Chem B ; 7(12): 2001-2008, 2019 03 28.
Article in English | MEDLINE | ID: mdl-32254804

ABSTRACT

Cancer cell-targeted imaging and efficient therapy are vital for tumor diagnosis and treatments. However, the development of multifunctional plasmonic nanoparticles with high-performance SERS-imaging and NIR light-triggered plasmonic photothermal therapy (PPTT) of cancer cells in both the first (NIR-I) and second (NIR-II) biological windows is still a big challenge. In the present work, gold nanostars which possess a broad NIR absorption band covering the NIR-I and NIR-II windows with good NIR SERS activity and photothermal effects were synthesized by a seed-mediated growth method, using gold chloride (HAuCl4), ascorbic acid (AA) and (1-hexadecyl) trimethylammonium chloride (CTAC) as growth solutions. The gold nanostars were further designed to be multifunctional nanoagents by labeling Raman molecules and then conjugating arginine-glycine-aspartic acid (RGD), which can serve as cancer cell-targeted SERS-imaging tags and photothermal nanoagents in both the NIR-I and NIR-II windows. The investigation of in vitro SERS-mapping and PPTT of the A549 human lung adenocarcinoma cells indicates that the proposed multifunctional gold nanostars have great potential for a wide spectrum of light-mediated applications, such as optical imaging and image-guided phototherapy in both the NIR-I and NIR-II biological windows.


Subject(s)
Gold/chemistry , Nanostructures/chemistry , A549 Cells , Cell Survival/drug effects , Gold/administration & dosage , Humans , Light , Nanostructures/administration & dosage , Neoplasms , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Phototherapy , Spectrum Analysis, Raman
20.
Sci Rep ; 8(1): 16171, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385885

ABSTRACT

Radio Guided Surgery is a technique helping the surgeon in the resection of tumors: a radiolabeled tracer is administered to the patient before surgery and then the surgeon evaluates the completeness of the resection with a handheld detector sensitive to emitted radiation. Established methods rely on γ emitting tracers coupled with γ detecting probes. The efficacy of this technique is however hindered by the high penetration of γ radiation, limiting its applicability to low background conditions. To overtake such limitations, a novel approach to RGS has been proposed, relying on ß- emitting isotopes together with a dedicated ß probe. This technique has been proved to be effective in first ex-vivo trials. We discuss in this paper the possibility to extend its application cases to 68Ga, a ß+ emitting isotope widely used today in nuclear medicine. To this aim, a retrospective study on 45 prostatic cancer patients was performed, analysing their 68Ga-PSMA PET images to asses if the molecule uptake is enough to apply this technique. Despite the expected variability both in terms of SUV (median 4.1, IQR 3.0-6.1) and TNR (median 9.4, IQR 5.2-14.6), the majority of cases have been found to be compatible with ß-RGS with reasonable injected activity and probing time (5 s).


Subject(s)
Beta Particles/therapeutic use , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/surgery , Surgery, Computer-Assisted , Edetic Acid/administration & dosage , Edetic Acid/analogs & derivatives , Gallium Isotopes , Gallium Radioisotopes , Humans , Male , Oligopeptides/administration & dosage , Positron-Emission Tomography , Prostatic Neoplasms/pathology , Radiopharmaceuticals/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL