Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Enzyme Inhib Med Chem ; 32(1): 648-657, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28262028

ABSTRACT

Specifically targeted drug delivery systems with low immunogenicity and toxicity are deemed to increase efficacy of cancer chemotherapy. Acridine Orange (AO) is an acidophilic dye with a strong tumoricidal action following excitation with a light source at 466 nm. However, to date the clinical use of AO is limited by the potential side effects elicited by systemic administration. The endogenous nanocarrier exosomes have been recently introduced as a natural delivery system for therapeutic molecules. In this article, we show the outcome of the administration to human melanoma cells of AO charged Exosomes (Exo-AO), in both monolayer and spheroid models. The results showed an extended drug delivery time of Exo-AO to melanoma cells as compared to the free AO, improving the cytotoxicity of AO. This study shows that Exo-AO have a great potential for a real exploitation as a new theranostic approach against tumors based on AO delivered through the exosomes.


Subject(s)
Acridine Orange/chemistry , Drug Delivery Systems , Exosomes , Melanoma/drug therapy , Theranostic Nanomedicine , Acridine Orange/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Microscopy, Confocal
2.
J Cell Physiol ; 232(3): 490-495, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27464761

ABSTRACT

Electrochemotherapy (ECT) is a medical strategy that allows an increased efficacy of chemotherapy agents after the application of permeabilizing electric pulses having appropriate characteristics (form, voltage, frequency). In the past 10 years, the clinical efficacy of this therapeutic approach in several spontaneous models of tumors in animals has been shown. Moreover, some of the molecular and cellular mechanisms responsible for this phenomenon have been elucidated. Our group has been deeply involved in the development of new ECT protocols for companion animals, implementing the use of the technique as first line treatment, and evaluating different chemotherapy agents in laboratory animals as well as pets. This article summarizes the most important advances in veterinary ECT, including the development of novel equipment, therapeutic protocols, and their translation to humans. J. Cell. Physiol. 232: 490-495, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Electrochemotherapy/methods , Hospitals, Animal , Translational Research, Biomedical , Animals , Electricity , Electroporation , Humans
3.
PLoS One ; 11(7): e0159763, 2016.
Article in English | MEDLINE | ID: mdl-27447181

ABSTRACT

Tumor extracellular acidity is a hallmark of malignant cancers. Thus, in this study we evaluated the effects of the oral administration of a commercially available water alkalizer (Basenpulver®) (BP) on tumor growth in a syngenic melanoma mouse model. The alkalizer was administered daily by oral gavage starting one week after tumor implantation in CB57/BL mice. Tumors were calipered and their acidity measured by in vivo MRI guided 31P MRS. Furthermore, urine pH was monitored for potential metabolic alkalosis. BP administration significantly reduced melanoma growth in mice; the optimal dose in terms of tolerability and efficacy was 8 g/l (p< 0.05). The in vivo results were supported by in vitro experiments, wherein BP-treated human and murine melanoma cell cultures exhibited a dose-dependent inhibition of tumor cell growth. This investigation provides the first proof of concept that systemic buffering can improve tumor control by itself and that this approach may represent a new strategy in prevention and/or treatment of cancers.


Subject(s)
Animal Feed , Dietary Supplements , Hydrogen-Ion Concentration , Melanoma/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Magnetic Resonance Imaging/methods , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Tumor Burden
4.
J Enzyme Inhib Med Chem ; 31(sup1): 119-125, 2016.
Article in English | MEDLINE | ID: mdl-27142956

ABSTRACT

CONTEXT: Proton Pump Inhibitors (PPIs) reduce tumor acidity and therefore resistance of tumors to drugs. Carbonic Anhydrase IX (CA IX) inhibitors have proven to be effective against tumors, while tumor acidity might impair their full effectiveness. OBJECTIVE: To analyze the effect of PPI/CA IX inhibitors combined treatment against human melanoma cells. METHODS: The combination of Lansoprazole (LAN) and CA IX inhibitors (FC9-399A and S4) has been investigated in terms of cell proliferation inhibition and cell death in human melanoma cells. RESULTS: The combination of these inhibitors was more effective than the single treatments in both inhibiting cell proliferation and in inducing cell death in human melanoma cells. DISCUSSION: These results represent the first successful attempt in combining two different proton exchanger inhibitors. CONCLUSION: This is the first evidence on the effectiveness of a new approach against tumors based on the combination of PPI and CA IX inhibitors, thus providing an alternative strategy against tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Lansoprazole/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Lansoprazole/chemical synthesis , Lansoprazole/chemistry , Molecular Structure , Structure-Activity Relationship
5.
J Enzyme Inhib Med Chem ; 31(4): 538-45, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26018420

ABSTRACT

CONTEXT: Tumor acidity represents a major cause of chemoresistance. Proton pump inhibitors (PPIs) can neutralize tumor acidity, sensitizing cancer cells to chemotherapy. OBJECTIVE: To compare the anti-tumor efficacy of different PPIs in vitro and in vivo. MATERIALS AND METHODS: In vitro experiments PPIs anti-tumor efficacy in terms of cell proliferation and cell death/apoptosis/necrosis evaluation were performed. In vivo PPIs efficacy experiments were carried out using melanoma xenograft model in SCID mice. RESULTS: Lansoprazole showed higher anti-tumor effect when compared to the other PPIs. The lansoprazole effect lasted even upon drug removal from the cell culture medium and it was independent from the lipophilicity of the PPIs formulation. DISCUSSION: These PPIs have shown different anti-tumoral efficacy, and the most effective at low dose was lansoprazole. CONCLUSION: The possibility to contrast tumor acidity by off-label using PPIs opens a new field of oncology investigation.


Subject(s)
Antineoplastic Agents/classification , Antineoplastic Agents/pharmacology , Drugs, Generic/classification , Drugs, Generic/pharmacology , Melanoma, Experimental/drug therapy , Proton Pump Inhibitors/classification , Proton Pump Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Drugs, Generic/chemical synthesis , Drugs, Generic/chemistry , Female , Humans , Melanoma, Experimental/pathology , Mice , Mice, SCID , Proton Pump Inhibitors/chemical synthesis , Proton Pump Inhibitors/chemistry , Structure-Activity Relationship
6.
Drug Resist Updat ; 23: 69-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26341193

ABSTRACT

Despite the major progresses in biomedical research and the development of novel therapeutics and treatment strategies, cancer is still among the dominant causes of death worldwide. One of the crucial challenges in the clinical management of cancer is primary (intrinsic) and secondary (acquired) resistance to both conventional and targeted chemotherapeutics. Multiple mechanisms have been identifiedthat underlie intrinsic and acquired chemoresistance: these include impaired drug uptake, increased drug efflux, deletion of receptors, altered drug metabolism, quantitative and qualitative alterations in drug targets, increased DNA damage repair and various mechanisms of anti-apoptosis. The fast efflux of anticancer drugs mediated by multidrug efflux pumps and the partial or complete reversibility of chemoresistance combined with the absence of genetic mutations suggests a multifactorial process. However, a growing body of recent evidence suggests that chemoresistance is often triggered by the highly acidic microenvironment of tumors. The vast majority of drugs, including conventional chemotherapeutics and more recent biological agents, are weak bases that are quickly protonated and neutralized in acidic environments, such as the extracellular microenvironment and the acidic organelles of tumor cells. It is therefore essential to develop new strategies to overcome the entrapment and neutralization of weak base drugs. One such strategy is the use of proton pump inhibitors which can enhance tumor chemosensitivity by increasing the pH of the tumor microenvironment. Recent clinical trials in animals with spontaneous tumors have indicated that patient alkalization is capable of reversing acquired chemoresistance in a large percentage of tumors that are refractory to chemotherapy. Of particular interest was the benefit of alkalization for patients undergoing metronomic regimens which are becoming more widely used in veterinary medicine. Overall, these results provide substantial new evidence that altering the acidic tumor microenvironment is an effective, well tolerated and low cost strategy for the overcoming of anticancer drug resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Proton Pump Inhibitors/therapeutic use , Tumor Microenvironment/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biological Transport/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Hydrogen-Ion Concentration , Metabolic Networks and Pathways/drug effects , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction
7.
Int J Cardiol ; 197: 333-47, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26159041

ABSTRACT

BACKGROUND: We recently demonstrated that epicardial progenitor cells participate in the regenerative response to myocardial infarction (MI) and factors released in the pericardial fluid (PF) may play a key role in this process. Exosomes are secreted nanovesicles of endocytic origin, identified in most body fluids, which may contain molecules able to modulate a variety of cell functions. Here, we investigated whether exosomes are present in the PF and their potential role in cardiac repair. METHODS AND RESULTS: Early gene expression studies in 3day-infarcted mouse hearts showed that PF induces epithelial-to-mesenchymal transition (EMT) in epicardial cells. Exosomes were identified in PFs from non-infarcted patients (PFC) and patients with acute MI (PFMI). A shotgun proteomics analysis identified clusterin in exosomes isolated from PFMI but not from PFC. Notably, clusterin has a protective effect on cardiomyocytes after acute MI in vivo and is an important mediator of TGFß-induced. Clusterin addition to the pericardial sac determined an increase in epicardial cells expressing the EMT marker α-SMA and, interestingly, an increase in the number of epicardial cells ckit(+)/α-SMA(+), 7days following MI. Importantly, clusterin treatment enhanced arteriolar length density and lowered apoptotic rates in the peri-infarct area. Hemodynamic studies demonstrated an improvement in cardiac function in clusterin-treated compared to untreated infarcted hearts. CONCLUSIONS: Exosomes are present and detectable in the PFs. Clusterin was identified in PFMI-exosomes and might account for an improvement in myocardial performance following MI through a framework including EMT-mediated epicardial activation, arteriogenesis and reduced cardiomyocyte apoptosis.


Subject(s)
Clusterin/metabolism , Coronary Vessels/metabolism , Exosomes/metabolism , Myocardial Infarction/metabolism , Pericardial Fluid/metabolism , Pericardium/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis/physiology , Biomarkers/analysis , Biomarkers/metabolism , Clusterin/analysis , Coronary Vessels/chemistry , Exosomes/chemistry , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Infarction/diagnosis , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , Pericardial Fluid/chemistry , Pericardium/chemistry , Pericardium/pathology
8.
J Gen Virol ; 96(Pt 7): 1969-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25805411

ABSTRACT

In most forms of prion diseases, blood is infectious, but detection by immunochemistry techniques of the only available marker of infection (the misfolded prion protein, PrPTSE) in blood remains elusive. We developed a novel method for the detection of PrPTSE in blood of prion-infected rodents based on the finding that PrPTSE is associated with plasma exosomes. However, further purification of the exosomes on a sucrose gradient was necessary to remove plasma immunoglobulins, which interfere with PrPTSE, masking its detection by immunochemistry. Finally, we report that about 20% of plasma infectivity is associated with exosomes.


Subject(s)
Exosomes/chemistry , Prions/analysis , Animals , Blood Chemical Analysis , Female , Immunochemistry , Mesocricetus , Prion Diseases/diagnosis , Specimen Handling/methods
9.
Cancer Lett ; 356(2 Pt B): 697-703, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25449440

ABSTRACT

Tumor acidity is now considered an important determinant of drug-resistance and tumor progression, and anti-acidic approaches, such as Proton Pump inhibitors (PPIs), have demonstrated promising antitumor and chemo-sensitizing efficacy. The main purpose of the present study was to evaluate the possible PPI-induced sensitization of human melanoma cells to Paclitaxel (PTX). Our results show that PTX and the PPI Lansoprazole (LAN) combination was extremely efficient against metastatic melanoma cells, as compared to the single treatments, both in vitro and in vivo. We also showed that acidity plays an important role on the anti-tumor activity of these drugs, being detrimental for PTX activity, while crucial for the synergistic effect of PTX following pretreatment with LAN, due to its nature of pro-drug needing protonation for a full activation. We obtained straightforward results in a human melanoma xenograft model combining well tolerated LAN doses with suboptimal and poorly toxic doses of PTX. With this study we provide a clear evidence that the PPI LAN may be included in new combined therapy of human melanoma together with low doses of PTX.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/drug effects , Lansoprazole/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Paclitaxel/pharmacology , Proton Pump Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Mice , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
PLoS One ; 9(2): e88193, 2014.
Article in English | MEDLINE | ID: mdl-24516610

ABSTRACT

Intrinsic resistance to cytotoxic drugs has been a main issue in cancer therapy for decades. Microenvironmental acidity is a simple while highly efficient mechanism of chemoresistance, exploited through impairment of drug delivery. The latter is achieved by extracellular protonation and/or sequestration into acidic vesicles. This study investigates the importance of extracellular acidosis and nanovesicle (exosome) release in the resistance of human tumour cell to cisplatin (CisPt); in parallel to proton pump inhibitors (PPI) ability of interfering with these tumour cell features. The results showed that CisPt uptake by human tumour cells was markedly impaired by low pH conditions. Moreover, exosomes purified from supernatants of these cell cultures contained various amounts of CisPt, which correlated to the pH conditions of the culture medium. HPLC-Q-ICP-MS analysis revealed that exosome purified from tumour cell culture supernatants contained CisPt in its native form. PPI pre-treatment increased cellular uptake of CisPt, as compared to untreated cells, in an acidic-depend manner. Furthermore, it induced a clear inhibition of exosome release by tumour cells. Human tumours obtained from xenografts pretreated with PPI contained more CisPt as compared to tumours from xenografts treated with CisPt alone. Further analysis showed that in vivo PPI treatment induced a clear reduction in the plasmatic levels of tumour-derived exosomes which also contained lower level of CisPt. Altogether, these findings point to the identification of a double mechanism that human malignant melanoma use in resisting to a dreadful cellular poison such as cisplatin. This framework of resistance includes both low pH-dependent extracellular sequestration and an exosome-mediated elimination. Both mechanisms are markedly impaired by proton pump inhibition, leading to an increased CisPt-dependent cytotoxicity.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Exosomes/metabolism , Melanoma/pathology , Animals , Buffers , Cell Death/drug effects , Cell Line, Tumor , Cellular Microenvironment/drug effects , Chromatography, High Pressure Liquid , Exosomes/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Humans , Hydrogen-Ion Concentration/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice, SCID , Proton Pump Inhibitors/pharmacology , Reference Standards , Solutions , Spectrophotometry, Atomic , Xenograft Model Antitumor Assays
11.
Biol Chem ; 394(1): 1-15, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23241589

ABSTRACT

Nanomedicine aims to exploit the improved and often novel physical, chemical, and biological properties of materials at the nanometric scale, possibly with the highest level of biomimetism, an approach that simulates what occurs in nature. Although extracellularly released vesicles include both microvesicles (MVs) and exosomes, only exosomes have the size that may be considered suitable for potential use in nanomedicine. In fact, recent reports have shown that exosomes are able to interact with target cells within an organ or at a distance using different mechanisms. Much is yet to be understood about exosomes, and currently, we are looking at the visible top of an iceberg, with most of what we have to understand on these nanovesicles still under the sea. In fact, we know that exosomes released by normal cells always trigger positive effects, whereas those released by cells in pathological condition, such as tumor or infected cells, may induce undesired, dangerous, and mostly unknown effects, but we cannot exclude the possibility that exosomes may also be detrimental for the body in normal conditions. However, whether we consider extracellular vesicles as a whole, thus including MVs, it appears that even in normal conditions, extracellular vesicles may lead to unwanted effects, depending on gender and age. This review aims to critically emphasize existing data in the literature that support the possible roles of exosomes in both diagnostic and therapeutic scopes.


Subject(s)
Drug Delivery Systems , Exosomes/chemistry , Exosomes/metabolism , Nanoparticles/chemistry , Neoplasms/diagnosis , Neoplasms/therapy , Animals , Biomarkers, Tumor/analysis , Humans , Lipids/chemistry , MicroRNAs/chemistry , MicroRNAs/metabolism , Proteins/chemistry , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...