Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 172, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31932600

ABSTRACT

Despite recent advances in drug delivery, the targeted treatment of unhealthy cells or tissues continues to remain a priority. In cancer (much like other pathologies), delivery vectors are designed to exploit physical and biological features of unhealthy tissues that are not always homogenous across the disease. In some cases, shifting the target from unhealthy tissues to the whole organ can represent an advantage. Specifically, the natural organ-specific retention of nanotherapeutics following intravenous administration as seen in the lung, liver, and spleen can be strategically exploited to enhance drug delivery. Herein, we outline the development of a cell-based delivery system using macrophages as a delivery vehicle. When loaded with a chemotherapeutic payload (i.e., doxorubicin), these cellular vectors (CELVEC) were shown to provide continued release within the lung. This study provides proof-of-concept evidence of an alternative class of biomimetic delivery vectors that capitalize on cell size to provide therapeutic advantages for pulmonary treatments.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Biomimetics , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Lung/metabolism , Macrophages/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Liberation , Liposomes , Lung/cytology , Male , Mice , Mice, Nude , Tissue Distribution
2.
Clin Cancer Res ; 26(8): 1932-1943, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31831557

ABSTRACT

PURPOSE: Optimal head and neck squamous cell carcinoma (HNSCC) patient selection for anti-EGFR-based therapy remains an unmet need since only a minority of patients derive long-term benefit from cetuximab treatment. We assessed the ability of state-of-the-art noninvasive in vivo metabolic imaging to probe metabolic shift in cetuximab-sensitive and -resistant HNSCC patient-derived tumor xenografts (PDTXs). EXPERIMENTAL DESIGN: Three models selected based on their known sensitivity to cetuximab in patients (cetuximab-sensitive or acquired-resistant HNC007 PDTXs, cetuximab-naïve UCLHN4 PDTXs, and cetuximab-resistant HNC010 PDTXs) were inoculated in athymic nude mice. RESULTS: Cetuximab induced tumor size stabilization in mice for 4 weeks in cetuximab-sensitive and -naïve models treated with weekly injections (30 mg/kg) of cetuximab. Hyperpolarized 13C-pyruvate-13C-lactate exchange was significantly decreased in vivo in cetuximab-sensitive xenograft models 8 days after treatment initiation, whereas it was not modified in cetuximab-resistant xenografts. Ex vivo analysis of sensitive tumors resected at day 8 after treatment highlighted specific metabolic changes, likely to participate in the decrease in the lactate to pyruvate ratio in vivo. Diffusion MRI showed a decrease in tumor cellularity in the HNC007-sensitive tumors, but failed to show sensitivity to cetuximab in the UCLHN4 model. CONCLUSIONS: This study constitutes the first in vivo demonstration of cetuximab-induced metabolic changes in cetuximab-sensitive HNSCC PDTXs that were not present in resistant tumors. Using metabolic imaging, we were able to identify hyperpolarized 13C-pyruvate as a potential marker for response and resistance to the EGFR inhibitor in HNSCC.


Subject(s)
Carbon Isotopes/analysis , Carcinoma, Squamous Cell/pathology , Cetuximab/pharmacology , Drug Resistance, Neoplasm , Head and Neck Neoplasms/pathology , Lactates/metabolism , Pyruvates/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/antagonists & inhibitors , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
3.
J Cell Mol Med ; 24(2): 1934-1944, 2020 01.
Article in English | MEDLINE | ID: mdl-31833658

ABSTRACT

Nearly all melanoma patients with a BRAF-activating mutation will develop resistance after an initial clinical benefit from BRAF inhibition (BRAFi). The aim of this work is to evaluate whether metabolic imaging using hyperpolarized (HP) 13 C pyruvate can serve as a metabolic marker of early response to BRAFi in melanoma, by exploiting the metabolic effects of BRAFi. Mice bearing human melanoma xenografts were treated with the BRAFi vemurafenib or vehicle. In vivo HP 13 C magnetic resonance spectroscopy was performed at baseline and 24 hours after treatment to evaluate changes in pyruvate-to-lactate conversion. Oxygen partial pressure was measured via electron paramagnetic resonance oximetry. Ex vivo qRT-PCR, immunohistochemistry and WB analysis were performed on tumour samples collected at the same time-points selected for in vivo experiments. Similar approaches were applied to evaluate the effect of BRAFi on sensitive and resistant melanoma cells in vitro, excluding the role of tumour microenvironment. BRAF inhibition induced a significant increase in the HP pyruvate-to-lactate conversion in vivo, followed by a reduction of hypoxia. Conversely, the conversion was inhibited in vitro, which was consistent with BRAFi-mediated impairment of glycolysis. The paradoxical increase of pyruvate-to-lactate conversion in vivo suggests that such conversion is highly influenced by the tumour microenvironment.


Subject(s)
Carbon Isotopes/metabolism , Melanoma/diagnostic imaging , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyruvic Acid/metabolism , Vemurafenib/pharmacology , Xenograft Model Antitumor Assays , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Electron Spin Resonance Spectroscopy , Female , Glycolysis/drug effects , Glycolysis/genetics , Humans , Melanoma/pathology , Mice, Nude , Oximetry , Oxygen Consumption/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Transcription, Genetic/drug effects
4.
Free Radic Res ; 53(4): 405-410, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30864859

ABSTRACT

Melanoma is the most aggressive skin tumour type. Although complete cure can be achieved when the whole tumour is resected, prognostic dramatically drops when melanoma cells reach deeper tissues and lymph nodes. Hence, there is an urgent need to develop accurate tools allowing (i) discriminating benign naevi from malignant tumours and (ii) being able to characterise melanoma infiltration. For that purpose, we exploited the paramagnetic properties of melanin by using electron paramagnetic resonance (EPR) spectroscopy to measure the melanin content in pigmented (B16F10 cancer cells) and non-pigmented melanomas (WM2664 cancer cells) inoculated intradermally in nude mice. Specifically, we took advantage of a new clinical EPR device (1 GHz), which provides sensitive measurements of radical species in vivo. Results showed that the melanin-specific EPR signal increased with tumour growth in pigmented tumours, whereas no EPR signal could be detected in achromic melanomas. These data plead for the development of new EPR spectrometers/imagers with an improved in-depth resolution for the detection of invasive melanomas.


Subject(s)
Electron Spin Resonance Spectroscopy/methods , Melanins/metabolism , Melanoma/metabolism , Melanoma/pathology , Animals , Female , Humans , Mice , Mice, Nude , Proof of Concept Study , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Oncotarget ; 9(24): 16832-16846, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29682188

ABSTRACT

A majority of patients with a V600x melanoma respond quickly to BRAF/MEK inhibition (BRAFi/MEKi) and have an obvious clinical benefit. Nearly all the patients after this initial phase will develop resistance. Therefore, non-invasive early markers of response/non-response are needed in order to identify those patients who, due to intrinsic or acquired resistance, do not respond to treatment and would be eligible for alternative treatments. The aim of this study was to investigate the value of magnetic resonance spectroscopy (1H-MRS) of choline and diffusion-weighted magnetic resonance imaging (DW-MRI) as early markers of response to BRAF inhibition (BRAFi) with vemurafenib alone or in combination with MEK inhibition (MEKi) with trametinib, in BRAFi-sensitive and BRAFi-resistant melanoma xenografts. Tumor response was significantly improved by the combination of BRAFi and MEKi, compared to BRAFi alone, only in sensitive xenografts; thus indicating that vemurafenib-resistant A375R xenografts were cross-resistant to the inhibition of MEK, as confirmed by immunohistochemistry analysis for phosphorylated ERK. In vivo1H-MRS showed that in sensitive melanoma xenografts, a significant blockage of ERK phosphorylation, but not a decrease in cell proliferation, was required to affect total choline (tCho) levels, thus suggesting that tCho could serve as a pharmacodynamic (PD) marker for agents targeting the MAPK cascade. In addition, early effects of the combination therapy on tumor cellularity could be detected via DW-MRI. In particular, skewness and kurtosis of the apparent diffusion coefficient (ADC) distribution may be useful to detect changes in the diffusional heterogeneity that might not affect the global ADC value.

6.
Curr Drug Targets ; 16(14): 1582-90, 2015.
Article in English | MEDLINE | ID: mdl-25316273

ABSTRACT

Nanoparticles have considerable potential for cancer imaging and therapy due to their small size and prolonged circulation. However, biological barriers can impede the delivery of a sufficient dose of a drug to the target site, thereby also resulting in the accumulation of toxic compounds within healthy tissues, and systemic toxicity. Multistage nanovectors (MSV) preferentially accumulate on inflamed endothelium, and can thus serve as carriers for drugs and nanoparticles. Herein, we describe the loading of free (i.e., melittin) and nano-encapsulated (i.e., doxorubicin-loaded micelles) drugs into MSV, and report the impact of surface charge and pore size on drug loading. For both drug formulations, negatively charged MSV (i.e., oxidized) with larger pores were shown to retain higher concentrations of payloads compared to positively charged (i.e., APTES-modified) MSV with small pores. Treatment of human umbilical vein endothelial cells (HUVEC) with melittin-loaded MSV (MEL@MSV) resulted in an 80% reduction in cell viability after 3 days. Furthermore, MEL@MSV conjugated with antivascular endothelial growth factor receptor 2 (VEGFR2) antibodies displayed preferential targeting and delivery of MEL to activated HUVEC expressing VEGFR2. Treatment of HUVEC and MCF7 cells with doxorubicin-loaded micelles (DOXNP@MSV) resulted in a 23% and 47% reduction in cell viability, respectively. Taken together, these results demonstrate increased loading of a payload in oxidized, large pore MSV, and effective delivery of free and nano-encapsulated drugs to endothelial and cancer cells.


Subject(s)
Doxorubicin/pharmacology , Endothelial Cells/drug effects , Melitten/pharmacology , Nanoparticles/chemistry , Antibodies/pharmacology , Capsules , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Micelles , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...