Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Biol Chem ; 292(44): 18203-18216, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28916726

ABSTRACT

Many cancer treatments, such as those for managing recalcitrant tumors like pancreatic ductal adenocarcinoma, cause off-target toxicities in normal, healthy tissue, highlighting the need for more tumor-selective chemotherapies. ß-Lapachone is bioactivated by NAD(P)H:quinone oxidoreductase 1 (NQO1). This enzyme exhibits elevated expression in most solid cancers and therefore is a potential cancer-specific target. ß-Lapachone's therapeutic efficacy partially stems from the drug's induction of a futile NQO1-mediated redox cycle that causes high levels of superoxide and then peroxide formation, which damages DNA and causes hyperactivation of poly(ADP-ribose) polymerase, resulting in extensive NAD+/ATP depletion. However, the effects of this drug on energy metabolism due to NAD+ depletion were never described. The futile redox cycle rapidly consumes O2, rendering standard assays of Krebs cycle turnover unusable. In this study, a multimodal analysis, including metabolic imaging using hyperpolarized pyruvate, points to reduced oxidative flux due to NAD+ depletion after ß-lapachone treatment of NQO1+ human pancreatic cancer cells. NAD+-sensitive pathways, such as glycolysis, flux through lactate dehydrogenase, and the citric acid cycle (as inferred by flux through pyruvate dehydrogenase), were down-regulated by ß-lapachone treatment. Changes in flux through these pathways should generate biomarkers useful for in vivo dose responses of ß-lapachone treatment in humans, avoiding toxic side effects. Targeting the enzymes in these pathways for therapeutic treatment may have the potential to synergize with ß-lapachone treatment, creating unique NQO1-selective combinatorial therapies for specific cancers. These findings warrant future studies of intermediary metabolism in patients treated with ß-lapachone.


Subject(s)
Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Naphthoquinones/pharmacology , Pancreatic Neoplasms/drug therapy , Prodrugs/pharmacology , Activation, Metabolic , Antineoplastic Agents/metabolism , Biomarkers/metabolism , Carbon Isotopes , Cell Line, Tumor , Cell Survival/drug effects , Citric Acid Cycle/drug effects , DNA Damage , Enzyme Inhibitors/metabolism , Glycolysis/drug effects , Humans , Metabolomics/methods , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Principal Component Analysis , Prodrugs/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
2.
J Surg Oncol ; 116(1): 83-88, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28346693

ABSTRACT

Novel, tumor-selective therapies are needed to increase the survival rate of pancreatic cancer patients. K-Ras-mutant-driven NAD(P)H:quinone oxidoreductase 1 (NQO1) is over-expressed in pancreatic tumor versus associated normal tissue, while catalase expression is lowered compared to levels in associated normal pancreas tissue. ARQ761 undergoes a robust, futile redox cycle in NQO1+ cancer cells, producing massive hydrogen peroxide (H2 O2 ) levels; normal tissues are spared by low NQO1 and high catalase expression. DNA damage created by ARQ761 in pancreatic cancer cells "hyperactivates" PARP1, causing metabolic catastrophe and NAD ± keresis cell death. NQO1: catalase levels (high in tumor, low in normal tissue) are an attractive therapeutic window to treat pancreatic cancer. Based on a growing body of literature, we are leading a clinical trial to evaluate the combination of ARQ761 and chemotherapy in patients with pancreatic cancer.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Naphthoquinones/pharmacology , Pancreatic Neoplasms/drug therapy , Albumins/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Clinical Trials, Phase I as Topic , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , NAD(P)H Dehydrogenase (Quinone)/metabolism , Paclitaxel/pharmacology , Pancreatic Neoplasms/metabolism , Gemcitabine
3.
Cancer Cell ; 30(6): 940-952, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27960087

ABSTRACT

Therapeutic drugs that block DNA repair, including poly(ADP-ribose) polymerase (PARP) inhibitors, fail due to lack of tumor-selectivity. When PARP inhibitors and ß-lapachone are combined, synergistic antitumor activity results from sustained NAD(P)H levels that refuel NQO1-dependent futile redox drug recycling. Significant oxygen-consumption-rate/reactive oxygen species cause dramatic DNA lesion increases that are not repaired due to PARP inhibition. In NQO1+ cancers, such as non-small-cell lung, pancreatic, and breast cancers, cell death mechanism switches from PARP1 hyperactivation-mediated programmed necrosis with ß-lapachone monotherapy to synergistic tumor-selective, caspase-dependent apoptosis with PARP inhibitors and ß-lapachone. Synergistic antitumor efficacy and prolonged survival were noted in human orthotopic pancreatic and non-small-cell lung xenograft models, expanding use and efficacy of PARP inhibitors for human cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , NAD(P)H Dehydrogenase (Quinone)/genetics , Naphthoquinones/administration & dosage , Pancreatic Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Drug Synergism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice , Naphthoquinones/pharmacology , Pancreatic Neoplasms/genetics , Reactive Oxygen Species/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
4.
Biochemistry ; 55(33): 4666-74, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27471863

ABSTRACT

The dramatic increase in the prevalence of antibiotic-resistant bacteria has necessitated a search for new antibacterial agents against novel targets. Moiramide B is a natural product, broad-spectrum antibiotic that inhibits the carboxyltransferase component of acetyl-CoA carboxylase, which catalyzes the first committed step in fatty acid synthesis. Herein, we report the 2.6 Å resolution crystal structure of moiramide B bound to carboxyltransferase. An unanticipated but significant finding was that moiramide B bound as the enol/enolate. Crystallographic studies demonstrate that the (4S)-methyl succinimide moiety interacts with the oxyanion holes of the enzyme, supporting the notion that an anionic enolate is the active form of the antibacterial agent. Structure-activity studies demonstrate that the unsaturated fatty acid tail of moiramide B is needed only for entry into the bacterial cell. These results will allow the design of new antibacterial agents against the bacterial form of carboxyltransferase.


Subject(s)
Amides/metabolism , Anti-Bacterial Agents/metabolism , Carboxyl and Carbamoyl Transferases/chemistry , Staphylococcus aureus/enzymology , Succinimides/metabolism , Carboxyl and Carbamoyl Transferases/metabolism , Crystallography, X-Ray , Protein Conformation
5.
Eur J Med Chem ; 122: 1-16, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27341379

ABSTRACT

Selenium-containing quinone-based 1,2,3-triazoles were synthesized using click chemistry, the copper catalyzed azide-alkyne 1,3-dipolar cycloaddition, and evaluated against six types of cancer cell lines: HL-60 (human promyelocytic leukemia cells), HCT-116 (human colon carcinoma cells), PC3 (human prostate cells), SF295 (human glioblastoma cells), MDA-MB-435 (melanoma cells) and OVCAR-8 (human ovarian carcinoma cells). Some compounds showed IC50 values < 0.3 µM. The cytotoxic potential of the quinones evaluated was also assayed using non-tumor cells, exemplified by peripheral blood mononuclear (PBMC), V79 and L929 cells. Mechanistic role for NAD(P)H: Quinone Oxidoreductase 1 (NQO1) was also elucidated. These compounds could provide promising new lead derivatives for more potent anticancer drug development and delivery, and represent one of the most active classes of lapachones reported.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzoquinones/chemistry , Selenium/chemistry , Triazoles/chemistry , Triazoles/chemical synthesis , Triazoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Death/drug effects , Cell Line, Tumor , Chemistry Techniques, Synthetic , Drug Design , Humans , Leukocytes, Mononuclear/drug effects , Oxidation-Reduction , Structure-Activity Relationship , Triazoles/toxicity
6.
New Phytol ; 209(2): 762-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26010225

ABSTRACT

Pteris vittata exhibits enhanced arsenic uptake, but the corresponding mechanisms are not well known. The prevalent form of arsenic in most soils is arsenate, which is a phosphate analog and a substrate for Phosphate transporter 1 (Pht1) transporters. Herein we identify and characterize three P. vittata Pht1 transporters. Pteris vittata Pht1 cDNAs were isolated and characterized via heterologous expression in Saccharomyces cerevisiae (yeast) and Nicotiana benthamiana leaves. Expression of the PvPht1 loci in P. vittata gametophytes was also examined in response to phosphate deficiency and arsenate exposure. Expression of each of the PvPht1 cDNAs complemented the phosphate uptake defect of a yeast mutant. Compared with yeast cells expressing Arabidopsis thaliana Pht1;5, cells expressing PvPht1;3 were more sensitive to arsenate, and accumulated more arsenic. Uptake assays with yeast cells and radiolabeled (32)P revealed that PvPht1;3 and AtPht1;5 have similar affinities for phosphate, but the affinity of PvPht1;3 for arsenate is much greater. In P. vittata gametophytes, PvPht1;3 transcript levels increased in response to phosphate (Pi) deficiency and arsenate exposure. PvPht1;3 is induced by Pi deficiency and arsenate, and encodes a phosphate transporter that has a high affinity for arsenate. PvPht1;3 probably contributes to the enhanced arsenate uptake capacity and affinity exhibited by P. vittata.


Subject(s)
Arsenates/metabolism , Plant Proteins/metabolism , Pteris/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arsenates/pharmacokinetics , Gene Expression Regulation, Plant , Mutation , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Phosphates/metabolism , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Proteins/genetics , Plants, Genetically Modified , Pteris/drug effects , Pteris/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Nicotiana/genetics
7.
Sci Rep ; 5: 17066, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26602448

ABSTRACT

Base excision repair (BER) is an essential pathway for pancreatic ductal adenocarcinoma (PDA) survival. Attempts to target this repair pathway have failed due to lack of tumor-selectivity and very limited efficacy. The NAD(P)H: Quinone Oxidoreductase 1 (NQO1) bioactivatable drug, ß-lapachone (ARQ761 in clinical form), can provide tumor-selective and enhanced synergy with BER inhibition. ß-Lapachone undergoes NQO1-dependent futile redox cycling, generating massive intracellular hydrogen peroxide levels and oxidative DNA lesions that stimulate poly(ADP-ribose) polymerase 1 (PARP1) hyperactivation. Rapid NAD(+)/ATP depletion and programmed necrosis results. To identify BER modulators essential for repair of ß-lapachone-induced DNA base damage, a focused synthetic lethal RNAi screen demonstrated that silencing the BER scaffolding protein, XRCC1, sensitized PDA cells. In contrast, depleting OGG1 N-glycosylase spared cells from ß-lap-induced lethality and blunted PARP1 hyperactivation. Combining ß-lapachone with XRCC1 knockdown or methoxyamine (MeOX), an apyrimidinic/apurinic (AP)-modifying agent, led to NQO1-dependent synergistic killing in PDA, NSCLC, breast and head and neck cancers. OGG1 knockdown, dicoumarol-treatment or NQO1- cancer cells were spared. MeOX + ß-lapachone exposure resulted in elevated DNA double-strand breaks, PARP1 hyperactivation and TUNEL+ programmed necrosis. Combination treatment caused dramatic antitumor activity, enhanced PARP1-hyperactivation in tumor tissue, and improved survival of mice bearing MiaPaca2-derived xenografts, with 33% apparent cures. SIGNIFICANCE: Targeting base excision repair (BER) alone has limited therapeutic potential for pancreatic or other cancers due to a general lack of tumor-selectivity. Here, we present a treatment strategy that makes BER inhibition tumor-selective and NQO1-dependent for therapy of most solid neoplasms, particularly for pancreatic cancer.


Subject(s)
DNA Repair/drug effects , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Animals , Autophagy/drug effects , Catalase/genetics , Catalase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Glycosylases/antagonists & inhibitors , DNA Glycosylases/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dicumarol/pharmacology , Female , Humans , Hydroxylamines/pharmacology , Hydroxylamines/therapeutic use , Mice , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , X-ray Repair Cross Complementing Protein 1
8.
J Med Chem ; 57(21): 8947-59, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25280369

ABSTRACT

There is an urgent demand for the development of new antibiotics due to the increase in drug-resistant pathogenic bacteria. A novel target is the multifunctional enzyme acetyl-CoA carboxylase (ACC), which catalyzes the first committed step in fatty acid synthesis and consists of two enzymes: biotin carboxylase and carboxyltransferase. Covalently attaching known inhibitors against these enzymes with saturated hydrocarbon linkers of different lengths generated dual-ligand inhibitors. Kinetic results revealed that the dual-ligands inhibited the ACC complex in the nanomolar range. Microbiology assays showed that the dual-ligand with a 15-carbon linker did not exhibit any antibacterial activity, while the dual-ligand with a 7-carbon linker displayed broad-spectrum antibacterial activity as well as a decreased susceptibility in the development of bacterial resistance. These results suggest that the properties of the linker are vital for antibacterial activity and show how inhibiting two different enzymes with the same compound increases the overall potency while also impeding the development of resistance.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Oxazoles/chemical synthesis , Anti-Bacterial Agents/pharmacology , Carboxyl and Carbamoyl Transferases , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests , Oxazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL