Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Theranostics ; 8(8): 2107-2116, 2018.
Article in English | MEDLINE | ID: mdl-29721066

ABSTRACT

Rational: In a subset of cancers, arginine auxotrophy occurs due to the loss of expression of argininosuccinate synthetase 1 (ASS1). This loss of ASS1 expression makes cancers sensitive to arginine starvation that is induced by PEGylated arginine deiminase (ADI-PEG20). Although ADI-PEG20 treatment is effective, it does have important limitations. Arginine starvation is only beneficial in patients with cancers that are ASS1-deficient. Also, these tumors may metabolically reprogram to express ASS1, transforming them from an auxotrophic phenotype to a prototrophic phenotype and thus rendering ADI-PEG20 ineffective. Due to these limitations of ADI-PEG20 treatment and the potential for developing resistance, non-invasive tools to monitor sensitivity to arginine starvation are needed. Methods: Within this study, we assess the utility of a novel positron emission tomography (PET) tracer to determine sarcomas reliant on extracellular arginine for survival by measuring changes in amino acid transport in arginine auxotrophic sarcoma cells treated with ADI-PEG20. The uptake of the 18F-labeled histidine analogue, (S)-2-amino-3-[1-(2-[18F]fluoroethyl)-1H-[1,2,3]triazol-4-yl]propanoic acid (AFETP), was assessed in vitro and in vivo using human-derived sarcoma cell lines. In addition, we examined the expression and localization of cationic amino acid transporters in response to arginine starvation with ADI-PEG20. Results: In vitro studies revealed that in response to ADI-PEG20 treatment, arginine auxotrophs increase the uptake of L-[3H]arginine and [18F]AFETP due to an increase in the expression and localization to the plasma membrane of the cationic amino acid transporter CAT-1. Furthermore, in vivo PET imaging studies in mice with arginine-dependent osteosarcoma xenografts showed increased [18F]AFETP uptake in tumors 4 days after ADI-PEG20 treatment compared to baseline. Conclusion: CAT-1 transporters localizes to the plasma membrane as a result of arginine starvation with ADI-PEG20 in ASS1-deficient tumor cells and provides a mechanism for using cationic amino acid transport substrates such as [18F]AFETP for identifying tumors susceptible to ADI-PEG20 treatment though non-invasive PET imaging techniques. These findings indicate that [18F]AFETP-PET may be suitable for the early detection of tumor response to arginine depletion due to ADI-PEG20 treatment.


Subject(s)
Amino Acids/metabolism , Arginine/deficiency , Argininosuccinate Synthase/deficiency , Fluorine Radioisotopes/chemistry , Sarcoma/enzymology , Amino Acid Transport System y+/metabolism , Argininosuccinate Synthase/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Hydrolases/pharmacology , Models, Biological , Polyethylene Glycols/pharmacology , Positron-Emission Tomography , Sarcoma/pathology
2.
Cell Rep ; 18(4): 991-1004, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28122247

ABSTRACT

Targeting defects in metabolism is an underutilized strategy for the treatment of cancer. Arginine auxotrophy resulting from the silencing of argininosuccinate synthetase 1 (ASS1) is a common metabolic alteration reported in a broad range of aggressive cancers. To assess the metabolic effects that arise from acute and chronic arginine starvation in ASS1-deficient cell lines, we performed metabolite profiling. We found that pharmacologically induced arginine depletion causes increased serine biosynthesis, glutamine anaplerosis, oxidative phosphorylation, and decreased aerobic glycolysis, effectively inhibiting the Warburg effect. The reduction of glycolysis in cells otherwise dependent on aerobic glycolysis is correlated with reduced PKM2 expression and phosphorylation and upregulation of PHGDH. Concurrent arginine deprivation and glutaminase inhibition was found to be synthetic lethal across a spectrum of ASS1-deficient tumor cell lines and is sufficient to cause in vivo tumor regression in mice. These results identify two synthetic lethal therapeutic strategies exploiting metabolic vulnerabilities of ASS1-negative cancers.


Subject(s)
Argininosuccinate Synthase/genetics , Glutamine/metabolism , Serine/biosynthesis , Animals , Arginine/chemistry , Argininosuccinate Synthase/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Glucose/metabolism , Glucose/pharmacology , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/pharmacology , Glycolysis/drug effects , Humans , Hydrolases/pharmacology , Membrane Proteins/metabolism , Metabolomics , Mice , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Phosphorylation/drug effects , Polyethylene Glycols/pharmacology , RNA Interference , Thyroid Hormones/metabolism , Up-Regulation/drug effects , Thyroid Hormone-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL