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.
Gynecol Oncol ; 138(2): 363-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26080289

ABSTRACT

OBJECTIVE: The purpose of this study was to test PENAO, a promising new organoarsenical that is in phase 1 testing in patients with solid tumours, on a range of ovarian cancer cell lines with different histotypes, and to understand the molecular basis of drug resistance exhibited by the endometrioid ovarian cancer cell line, SKOV-3. METHODS: Proliferation arrest and cell death induced by PENAO in serous (OVCAR-3), endometrioid (SKOV-3, TOV112D), clear cell (TOV21G) and mucinous (EFO27) ovarian cancer cells in culture, and anti-tumour efficacy in a murine model of SKOV-3 and OVCAR-3 tumours, were measured. Cells were analysed for cell cycle arrest, cell death mechanisms, reactive oxygen species production, mitochondrial depolarisation, oxygen consumption and acid production. RESULTS: PENAO demonstrated promising anti-proliferative activity on the most common (serous, endometrioid) as well as on rare (clear cell, mucinous) subtypes of ovarian cancer cell lines. No cross-resistance with platinum-based drugs was evident. Endometrioid SKOV-3 cells were, however, shown to be resistant to PENAO in vitro and in a xenograft mouse model. This resistance was due to an ability to cope with PENAO-induced oxidative stress, notably through heme oxygenase-1 induction, and a shift in metabolism towards glycolysis. The adaptive glycolytic shift in SKOV-3 was targeted using a mTORC1 inhibitor in combination with PENAO. This strategy was successful with the two drugs acting synergistically to inhibit cell proliferation and to induce cell death via apoptosis and autophagy. CONCLUSION: Mitochondria/mTOR dual-targeting therapy may constitute a new approach for the treatment of recurrent/resistant forms of epithelial ovarian cancer.


Subject(s)
Arsenicals/pharmacology , Mitochondria/drug effects , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Biomimetic Materials/pharmacology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxidative Stress/drug effects , Xenograft Model Antitumor Assays
2.
J Exp Clin Cancer Res ; 34: 14, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25652202

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and malignant primary brain tumor. In contrast to some other tumor types, aberrant glucose metabolism is an important component of GBM growth and chemoresistance. Recent studies of human orthotopic GBM in mice and in situ demonstrated GBM cells rely on both glycolysis and mitochondrial oxidation for glucose catabolism. These observations suggest that the homeostasis of energy metabolism of GBM cells might be further disturbed by dual-inhibition of glucose metabolism. The present study aimed to evaluate the efficacy and the mechanisms of dual-targeting therapy in GBM cells. METHODS: Representative GBM cells (immortalized GBM cell lines and patient-derived GBM cells) and non-cancerous cells were treated with 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid (PENAO), an in-house designed novel arsenic-based mitochondrial toxin, in combination with dichloroacetate (DCA), a pyruvate dehydrogenase kinase inhibitor. The efficacy of this combinatorial therapy was evaluated by MTS assay, clonogenic surviving assay and apoptotic assays. The underlying mechanisms of this dual-targeting treatment were unraveled by using mitochondrial membrane potential measurements, cytosol/mitochondrial ROS detection, western blotting, extracellular flux assay and mass spectrometry. RESULTS: As monotherapies, both PENAO and DCA induced proliferation arrest in a panel of GBM cell lines and primary isolates. PENAO inhibited oxygen consumption, induced oxidative stress and depolarized mitochondrial membrane potential, which in turn activated mitochondria-mediated apoptosis. By combining DCA with PENAO, the two drugs worked synergistically to inhibit cell proliferation (but had no significant effect on non-cancerous cells), impair the clonogenicity, and induce mitochondria-mediated apoptosis. An oxidative stress of mitochondrial origin takes a prominent place in the mechanism by which the combination of PENAO and DCA induces cell death. Additionally, PENAO-induced oxidative damage was enhanced by DCA through glycolytic inhibition which in turn diminished acid production induced by PENAO. Moreover, DCA treatment also led to an alteration in the multidrug resistance (MDR) phenotype of GBM cells, thereby leading to an increased cytosolic accumulation of PENAO. CONCLUSIONS: The findings of this study shed a new light with respect to the dual-targeting of glucose metabolism in GBM cells and the innovative combination of PENAO and DCA shows promise in expanding GBM therapies.


Subject(s)
Glioblastoma/metabolism , Glucose/metabolism , Arsenicals/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dichloroacetic Acid/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression , Glioblastoma/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
3.
Mol Pharm ; 11(5): 1500-11, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24654974

ABSTRACT

γ-Glutamyltransferase (γGT) is a cell surface enzyme that catalyzes hydrolysis of the bond linking the glutamate and cysteine residues of glutathione and glutathione-S-conjugates. We have observed that human pancreatic tumor cells and tumor-associated stellate cells express high levels of this enzyme when compared to normal pancreatic epithelial and stellate cells. Detection of the protein in tumor sections correlated with γGT activity on the surface of the cultured tumor and stellate cells. We tested whether the tumor γGT could be employed to deliver a therapeutic to the tumor endothelial cells. GSAO is a glutathione-S-conjugate of a trivalent arsenical that is activated to enter endothelial cells by γGT cleavage of the γ-glutamyl residue. The arsenical moiety triggers proliferation arrest and death of the endothelial cells by targeting the mitochondria. Human pancreatic tumor and stellate cell γGT activated GSAO in culture and γGT activity positively correlated with GSAO-mediated proliferation arrest and death of endothelial cells in Transwell and coculture systems. A soluble form of γGT is found in blood, and we measured the rate of activation of GSAO by this enzyme. We calculated that systemically administered GSAO would circulate through the pancreatic blood supply several times before appreciable activation by normal blood levels of γGT. In support of this finding, tumor γGT activity positively correlated with GSAO-mediated inhibition of pancreatic tumor angiogenesis and tumor growth in mice. Our findings indicate that pancreatic tumor γGT can be used to deliver a therapeutic to the tumor.


Subject(s)
gamma-Glutamyltransferase/blood , gamma-Glutamyltransferase/metabolism , Animals , Arsenicals/chemistry , Arsenicals/metabolism , Cell Line , Drug Carriers/metabolism , Female , Glutathione/chemistry , Glutathione/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Prodrugs/administration & dosage , Prodrugs/metabolism , Prodrugs/therapeutic use
4.
J Med Chem ; 52(20): 6209-16, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19788237

ABSTRACT

Plasma membrane drug efflux pumps of the multidrug resistance associated protein (MRP) family blunt the effectiveness of anticancer drugs and are often associated with drug resistance. GSAO, a tripeptide trivalent arsenical that targets a key mitochondrial transporter in angiogenic endothelial cells, is an example of a compound whose efficacy is limited by tumor cell expression of MRP isoforms 1 and 2. A cysteine mimetic analogue of GSAO was made, PENAO, which accumulates in cells 85 times faster than GSAO due to increased rate of entry and decreased rate of export via MRP1/2. The faster rate of accumulation of PENAO corresponds to a 44-fold increase in antiproliferative activity in vitro and approximately 20-fold better antitumor efficacy in vivo. This information could be used to improve the efficacy of other small molecule cancer therapeutics.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Arsenicals/chemistry , Cytosol/metabolism , Mitochondria/drug effects , Oligopeptides/metabolism , Oligopeptides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biomimetics , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Endothelial Cells/cytology , Extracellular Space/metabolism , Female , Humans , Mice , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Pancreatic Neoplasms/pathology , Time Factors , Xenograft Model Antitumor Assays
5.
Cancer Res ; 65(24): 11729-34, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357185

ABSTRACT

The synthetic tripeptide arsenical 4-(N-(S-glutathionylacetyl)amino)p-phenylarsenoxide (p-GSAO) is an angiogenesis inhibitor that inactivates mitochondrial adenine nucleotide translocase (ANT) by cross-linking a pair of matrix-facing cysteine residues. This causes an increase in superoxide levels and proliferation arrest of endothelial cells followed by mitochondrial depolarization and apoptosis. p-GSAO induces proliferation arrest in endothelial cells and is a selective inhibitor of endothelial cells compared with tumor cells. An analogue of p-GSAO has been made in which the arsenical moiety is at the ortho instead of the para position on the phenyl ring. o-GSAO, like p-GSAO, bound to ANT in a dithiol-dependent manner but was approximately 8-fold more efficient than p-GSAO at triggering the mitochondria permeability transition in isolated mitochondria. o-GSAO was an approximately 50-fold more potent inhibitor of endothelial and tumor cell proliferation than p-GSAO. The mechanism of this effect was a consequence of approximately 300-fold faster rate of accumulation of o-GSAO in the cells, which is due, at least in part, to impaired export by the multidrug resistance-associated protein 1. Administration of o-GSAO to tumor-bearing mice delayed tumor growth by inhibiting tumor angiogenesis but there were side effects not observed with p-GSAO administration.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Arsenicals/therapeutic use , Cell Proliferation/drug effects , Endothelium, Vascular/drug effects , Glutathione/analogs & derivatives , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/drug therapy , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Cattle , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Glutathione/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/enzymology , Mitochondrial ADP, ATP Translocases/antagonists & inhibitors , Mitochondrial ADP, ATP Translocases/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Pancreatic Neoplasms/blood supply , Stereoisomerism , Superoxides/metabolism , Toluene/analogs & derivatives , Toluene/metabolism
6.
Cancer Cell ; 3(5): 497-509, 2003 May.
Article in English | MEDLINE | ID: mdl-12781367

ABSTRACT

Mitochondria are the powerhouse of the cell and their disruption leads to cell death. We have used a peptide trivalent arsenical, 4-(N-(S-glutathionylacetyl)amino) phenylarsenoxide (GSAO), to inactivate the adenine nucleotide translocator (ANT) that exchanges matrix ATP for cytosolic ADP across the inner mitochondrial membrane and is the key component of the mitochondrial permeability transition pore (MPTP). GSAO triggered Ca(2+)-dependent MPTP opening by crosslinking Cys(160) and Cys(257) of ANT. GSAO treatment caused a concentration-dependent increase in superoxide levels, ATP depletion, mitochondrial depolarization, and apoptosis in proliferating, but not growth-quiescent, endothelial cells. Endothelial cell proliferation drives new blood vessel formation, or angiogenesis. GSAO inhibited angiogenesis in the chick chorioallantoic membrane and in solid tumors in mice. Consequently, GSAO inhibited tumor growth in mice with no apparent toxicity at efficacious doses.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Arsenicals/therapeutic use , Endothelial Cells/metabolism , Eosine Yellowish-(YS)/analogs & derivatives , Mitochondria/drug effects , Neovascularization, Pathologic , Adenine Nucleotide Translocator 1/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Aorta/cytology , Apoptosis , Biotin/pharmacology , Calcium/metabolism , Calcium/pharmacology , Cattle , Cell Division , Cell Survival , Chick Embryo , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Eosine Yellowish-(YS)/pharmacology , Female , Immunohistochemistry , Ion Channels/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Models, Chemical , Peptides/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...