Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Hepatology ; 74(5): 2605-2621, 2021 11.
Article in English | MEDLINE | ID: mdl-34101869

ABSTRACT

BACKGROUND AND AIMS: HCC is the most predominant type of liver cancer affecting 800,000 people globally each year. Various small-molecule compounds targeting diverse oncogenic signaling pathways have been tested for patients with HCC, and clinical outcomes were not satisfactory. In this study, we investigated molecular signaling that determines the efficiency of drug delivery into HCC. APPROACH AND RESULTS: Hydrodynamics-based transfection (HT) was performed to develop mouse models for HCC induced by various oncogenes. Mice bearing liver cancer were treated with verteporfin at 5 weeks after HT. Multicellular HCC organoid (MCHO) models were established that contained various types of stromal cells, such as hepatic stellate cells, fibroblasts, and endothelial cells together with HCC cells. Tumor organoids were treated with verteporfin, and distributions of the drug in the organoids were assessed using fluorescence microscopy. Murine HCC models developed by HT methods showed that a high Yes-associated protein/Transcriptional co-activator with PDZ-binding motif (YAP/TAZ) activity in HCC cells impaired verteporfin penetration into the cancer. Activation of tumor stroma was observed in HCC with a high YAP/TAZ activity. Consistent with the findings in the in vivo models of HCC, MCHOs with activated YAP/TAZ signaling showed stromal activation and impaired penetration of verteporfin into the tumor organoids. Inhibition of YAP/TAZ transcriptional activity in HCC cells significantly increased drug penetration into the MCHO. CONCLUSIONS: Drug delivery into liver cancer is impaired by YAP/TAZ signaling in tumor cells and subsequent activation of stroma by the signaling. Disrupting or targeting activated tumor stroma might improve drug delivery into HCC with an elevated YAP/TAZ activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacokinetics , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , YAP-Signaling Proteins/metabolism , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Endothelial Cells , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Oncogenes/genetics , Organoids , Permeability , Tissue Distribution , Tumor Cells, Cultured , Verteporfin/administration & dosage , Verteporfin/pharmacokinetics
2.
J Mater Chem B ; 8(40): 9351-9361, 2020 10 21.
Article in English | MEDLINE | ID: mdl-32996552

ABSTRACT

Combined X-ray-induced photodynamic therapy (X-PDT) and chemotherapy are of great interest for tumor treatment, but their outcome is still hindered by insufficient drug delivery without tumor specificity and the difficulty of switching to chemotherapy during the X-PDT process. Herein, we report an efficient strategy for preparing a nanocarrier, DANPVP&DOX, with slight-acidity-induced charge conversion and hypoxia-motivated doxorubicin (DOX) release properties to achieve a more precise and synchronous therapeutic effect. Upon a change in the extracellular pH (pHe) in the tumor matrix, the surface charge of DANPVP&DOX converted from negative to positive via dimethyl maleate degradation. Following the increased internalization by tumoral cells, exposure of verteporfin (VP) in DANPVP&DOX to low-dose X-ray radiation resulted in O2 consumption in the cytoplasm to produce cytotoxic reactive oxygen species (ROS), which caused cell killing. Moreover, the hypoxic conditions formed in the tumor area specifically promoted DANPVP&DOX dissociation and on-demand DOX release. Consequently, DANPVP&DOX significantly increased the therapeutic efficacy through X-PDT and cascade chemotherapy. More importantly, this strategy could potentially be extended to various therapeutic agents other than anticancer drugs for precise drug delivery and cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemical synthesis , Drug Carriers/pharmacokinetics , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Organophosphates/chemical synthesis , Organophosphates/chemistry , Organophosphates/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/radiation effects , Photosensitizing Agents/therapeutic use , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Singlet Oxygen/metabolism , Verteporfin/chemistry , Verteporfin/pharmacokinetics , Verteporfin/radiation effects , Verteporfin/therapeutic use , X-Rays , Xenograft Model Antitumor Assays
3.
Int J Nanomedicine ; 14: 10047-10060, 2019.
Article in English | MEDLINE | ID: mdl-31920302

ABSTRACT

BACKGROUND: Nanomedicine can improve traditional therapies by enhancing the controlled release of drugs at targeted tissues in the body. However, there still exists disease- and therapy-specific barriers that limit the efficacy of such treatments. A major challenge in developing effective therapies for one of the most aggressive brain tumors, glioblastoma (GBM), is affecting brain cancer cells while avoiding damage to the surrounding healthy brain parenchyma. Here, we developed poly(ethylene glycol) (PEG)-poly(beta-amino ester) (PBAE) (PEG-PBAE)-based micelles encapsulating verteporfin (VP) to increase tumor-specific targeting. METHODS: Biodegradable, pH-sensitive micelles of different shapes were synthesized via nanoprecipitation using two different triblock PEG-PBAE-PEG copolymers varying in their relative hydrophobicity. The anti-tumor efficacy of verteporfin loaded in these anisotropic and spherical micelles was evaluated in vitro using patient-derived primary GBM cells. RESULTS: For anisotropic micelles, uptake efficiency was ~100% in GBM cells (GBM1A and JHGBM612) while only 46% in normal human astrocytes (NHA) at 15.6 nM VP (p ≤ 0.0001). Cell killing of GBM1A and JHGBM612 vs NHA was 52% and 77% vs 29%, respectively, at 24 hrs post-treatment of 125 nM VP-encapsulated in anisotropic micelles (p ≤ 0.0001), demonstrating the tumor cell-specific selectivity of VP. Moreover, anisotropic micelles showed an approximately fivefold longer half-life in blood circulation than the analogous spherical micelles in a GBM xenograft model in mice. In this model, micelle accumulation to tumors was significantly greater for anisotropic micelle-treated mice compared to spherical micelle-treated mice at both 8 hrs (~1.8-fold greater, p ≤ 0.001) and 24 hrs (~2.1-fold greater, p ≤ 0.0001). CONCLUSION: Overall, this work highlights the promise of a biodegradable anisotropic micelle system to overcome multiple drug delivery challenges and enhance efficacy and safety for the treatment of brain cancer.


Subject(s)
Brain Neoplasms/pathology , Micelles , Polymers/chemistry , Verteporfin/pharmacology , Verteporfin/pharmacokinetics , Animals , Anisotropy , Astrocytes/drug effects , Astrocytes/metabolism , Brain Neoplasms/drug therapy , Cell Death/drug effects , Cell Line, Tumor , Drug Carriers , Drug Liberation , Endocytosis/drug effects , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Hydrogen-Ion Concentration , Mice, Nude , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polymers/chemical synthesis , Solubility , Tissue Distribution/drug effects , Verteporfin/therapeutic use , Xenograft Model Antitumor Assays
4.
Sci Rep ; 7: 46581, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28429726

ABSTRACT

Verteporfin (VP) was first used in Photodynamic therapy, where a non-thermal laser light (689 nm) in the presence of oxygen activates the drug to produce highly reactive oxygen radicals, resulting in local cell and tissue damage. However, it has also been shown that Verteporfin can have non-photoactivated effects such as interference with the YAP-TEAD complex of the HIPPO pathway, resulting in growth inhibition of several neoplasias. More recently, it was proposed that, another non-light mediated effect of VP is the formation of cross-linked oligomers and high molecular weight protein complexes (HMWC) that are hypothesized to interfere with autophagy and cell growth. Here, in a series of experiments, using human uveal melanoma cells (MEL 270), human embryonic kidney cells (HEK) and breast cancer cells (MCF7) we showed that Verteporfin-induced HMWC require the presence of light. Furthermore, we showed that the mechanism of this cross-linking, which involves both singlet oxygen and radical generation, can occur very efficiently even after lysis of the cells, if the lysate is not protected from ambient light. This work offers a better understanding regarding VP's mechanisms of action and suggests caution when one studies the non-light mediated actions of this drug.


Subject(s)
Light , Multiprotein Complexes/metabolism , Neoplasms , Photochemotherapy , Photosensitizing Agents , Verteporfin , Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , Verteporfin/pharmacokinetics , Verteporfin/pharmacology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL