Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Cancer Med ; 8(18): 7705-7719, 2019 12.
Article in English | MEDLINE | ID: mdl-31568691

ABSTRACT

Despite advances in cancer therapeutics, pancreatic cancer remains difficult to treat and often develops resistance to chemotherapies. We have evaluated a bioavailable genistein analogue, AXP107-11 which has completed phase Ib clinical trial, as an approach to sensitize tumor cells to chemotherapy. Using organotypic cultures of 14 patient-derived xenografts (PDX) of pancreatic ductal adenocarcinoma, we found that addition of AXP107-11 indeed sensitized 57% of cases to gemcitabine treatment. Results were validated using PDX models in vivo. Further, RNA-Seq from responsive and unresponsive tumors proposed a 41-gene treatment-predictive signature. Functional and molecular assays were performed in cell lines and demonstrated that the effect was synergistic. Transcriptome analysis indicated activation of G-protein-coupled estrogen receptor (GPER1) as the main underlying mechanism of action, which was corroborated using GPER1-selective agonists and antagonists. GPER1 expression in pancreatic tumors was indicative of survival, and our study proposes that activation of GPER1 may constitute a new avenue for pancreatic cancer therapeutics.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Genistein/pharmacology , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Drug Synergism , Female , Genistein/analogs & derivatives , Genistein/therapeutic use , Humans , Mice , Models, Biological , Mucin-1/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays , Gemcitabine
2.
Pancreatology ; 16(4): 640-5, 2016.
Article in English | MEDLINE | ID: mdl-27234064

ABSTRACT

BACKGROUND: AXP107-11 is a novel, multi-component crystalline form of the naturally occurring compound genistein. AXP107-11 has improved physiochemical properties and oral bioavailability compared to the natural form of genistein, and it is possible that combining AXP107-11 with chemotherapy may increase the effect and reduce chemoresistance. The purpose of this dose escalation phase Ib study was to assess the safety, maximum tolerated dose (MTD) and pharmacokinetics (PK) of AXP107-11 in combination with gemcitabine in treatment-naïve patients with inoperable pancreatic carcinoma. PATIENTS AND METHODS: AXP107-11 was given orally in escalating doses (400 mg-1600 mg daily) in combination with standard gemcitabine treatment (1000 mg/m(2)/week) for the first seven of eight weeks and thereafter for a maximum of four × four-week treatment cycles. PK, safety, MTD and efficacy of AXP107-11 in combination with gemcitabine were evaluated. RESULTS: Sixteen patients were enrolled and received AXP107-11. The maximum concentration in serum of unconjugated (free) genistein was 1 µM. Neither dose-limiting toxicities (DLTs) nor signs of hematological or non-hematological toxicities related to AXP107-11 were observed over a period ranging from 0.7 to 13.2 months. The median overall survival time was 4.9 months (range 1.5-19.5 months). Seven patients (44%) survived longer than six months and 19% were alive at the one-year follow-up. CONCLUSION: Treatment of pancreatic cancer patients with AXP107-11 in combination with gemcitabine resulted in a favorable PK-profile with high serum levels without signs of either hematological or non-hematological toxicity. Accordingly, we suggest further studies with AXP107-11 in pancreatic cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Genistein/administration & dosage , Genistein/pharmacokinetics , Humans , Karnofsky Performance Status , Liver Neoplasms/secondary , Male , Maximum Tolerated Dose , Middle Aged , Survival Analysis , Treatment Outcome , Gemcitabine
3.
Nucl Recept Signal ; 12: e003, 2014.
Article in English | MEDLINE | ID: mdl-25422593

ABSTRACT

The establishment of effective high throughput screening cascades to identify nuclear receptor (NR) ligands that will trigger defined, therapeutically useful sets of NR activities is of considerable importance. Repositioning of existing approved drugs with known side effect profiles can provide advantages because de novo drug design suffers from high developmental failure rates and undesirable side effects which have dramatically increased costs. Ligands that target estrogen receptor ß (ERß) could be useful in a variety of diseases ranging from cancer to neurological to cardiovascular disorders. In this context, it is important to minimize cross-reactivity with ERα, which has been shown to trigger increased rates of several types of cancer. Because of high sequence similarities between the ligand binding domains of ERα and ERß, preferentially targeting one subtype can prove challenging. Here, we describe a sequential ligand screening approach comprised of complementary in-house assays to identify small molecules that are selective for ERß. Methods include differential scanning fluorimetry, fluorescence polarization and a GAL4 transactivation assay. We used this strategy to screen several commercially-available chemical libraries, identifying thirty ERß binders that were examined for their selectivity for ERß versus ERα, and tested the effects of selected ligands in a prostate cancer cell proliferation assay. We suggest that this approach could be used to rapidly identify candidates for drug repurposing.


Subject(s)
Drug Evaluation, Preclinical/methods , Estrogen Receptor beta/metabolism , Cell Line, Tumor , Estrogen Receptor beta/genetics , Humans , Ligands , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Substrate Specificity , Transcriptional Activation/drug effects
4.
Proc Natl Acad Sci U S A ; 105(2): 663-7, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18160532

ABSTRACT

Atherosclerotic cardiovascular disease is a major problem despite the availability of drugs that influence major risk factors. New treatments are needed, and there is growing interest in therapies that may have multiple actions. Thyroid hormone modulates several cardiovascular risk factors and delays atherosclerosis progression in humans. However, use of thyroid hormone is limited by side effects, especially in the heart. To overcome this limitation, pharmacologically selective thyromimetics that mimic metabolic effects of thyroid hormone and bypass side effects are under development. In animal models, such thyromimetics have been shown to stimulate cholesterol elimination through LDL and HDL pathways and decrease body weight without eliciting side effects. We report here studies on a selective thyromimetic [KB2115; (3-[[3,5-dibromo-4-[4-hydroxy-3-(1-methylethyl)-phenoxy]-phenyl]-amino]-3-oxopropanoic acid)] in humans. In moderately overweight and hypercholesterolemic subjects KB2115 was found to be safe and well tolerated and elicited up to a 40% lowering of total and LDL cholesterol after 14 days of treatment. Bile acid synthesis was stimulated without evidence of increased cholesterol production, indicating that KB2115 induced net cholesterol excretion. KB2115 did not provoke detectable effects on the heart, suggesting that the pharmacological selectivity observed in animal models translates to humans. Thus, selective thyromimetics deserve further study as agents to treat dyslipidemia and other risk factors for atherosclerosis.


Subject(s)
Anilides/pharmacology , Bile Acids and Salts/metabolism , Cholesterol, LDL/metabolism , Heart/drug effects , Thyroid Gland/drug effects , Thyroid Hormones/metabolism , Adolescent , Adult , Anilides/chemistry , Double-Blind Method , Female , Humans , Lipid Metabolism , Male , Middle Aged , Placebos , Risk Factors , Thiazolidinediones/pharmacology
5.
Curr Opin Pharmacol ; 5(2): 171-6, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780827

ABSTRACT

Drugs that target the nuclear hormone receptor family constitute one of the largest and most potent groups of pharmaceuticals currently in use. However, although many of these human nuclear receptors have been clearly demonstrated to be key sensors and regulators of lipid metabolism, the full pharmacological potential of this drug target class has not been fully explored. There are two main reasons for this. First, a rationale approach is needed to identify pharmacologically selective drug candidates to nuclear receptors that have a large therapeutic window between the beneficial effects and the unwanted side effects. This appears to apply to all ligand-regulated nuclear receptors, including those nuclear receptors more recently proposed as novel targets for diseases related to lipid metabolism such as the peroxisome proliferator-activated receptors, liver X receptors and farnesoid X-activated receptor. The second reason is that any sub-group of nuclear receptors important for the regulation of lipid metabolism might be pharmacologically inaccessible by conventional low molecular weight compounds, owing to the lack of a classical ligand-binding-pocket, as recently revealed by X-ray crystallography. Accordingly, targeting of classical nuclear receptor family members with better characterized endocrinology and roles in lipid metabolism, such as the thyroid and steroid hormone receptors, could become of renewed pharmacological interest, as these targets provide well-characterized alternatives to the more recently discovered nuclear receptors.


Subject(s)
Lipid Metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Humans
6.
Mech Ageing Dev ; 125(10-11): 707-17, 2004.
Article in English | MEDLINE | ID: mdl-15541766

ABSTRACT

Members of the nuclear receptor gene family act as biological rheostats to maintain metabolic homeostasis in response to endocrine and nutritional changes. The liver X (LXR) and thyroid hormone (TR) receptors have been shown to regulate overlapping but distinct metabolic pathways important for overall lipid homeostasis. Dyslipidemia is one out of four key determinants for cardiovascular risk and both LXRs and TRs may provide attractive targets for intervention of cardiovascular disease. In this review we will compare the two receptor systems to highlight similarities and differences in structure and function with implications for development of novel treatments for dyslipidemia and atherosclerosis.


Subject(s)
DNA-Binding Proteins/physiology , Lipid Metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Thyroid Hormone/physiology , Animals , Arteriosclerosis/physiopathology , Homeostasis/physiology , Humans , Hyperlipidemias/physiopathology , Liver X Receptors , Orphan Nuclear Receptors
7.
Eur J Biochem ; 269(19): 4896-904, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12354121

ABSTRACT

The cytochrome P450 family of enzymes has long been known to metabolize a wide range of compounds, including many of today's most common drugs. A novel nuclear receptor called PXR has been established as an activator of several of the cytochrome P450 genes, including CYP3A4. This enzyme is believed to account for the metabolism of more than 50% of all prescription drugs. PXR is therefore used as a negative selector target and discriminatory filter in preclinical drug development. In this paper we describe the design, construction and characterization by transient transfection of mutant receptors of the human and mouse PXR ligand binding domains. By modeling the human PXR ligand binding domain we have identified and mutated two polar residues in the putative ligand binding pocket which differ between the human and the mouse receptor. The first residue (Q285 in human/I282 in mouse) was mutated between the two species with the corresponding amino acids. These mutants showed that this residue is important for the species specific activation of PXR by the ligand pregnenolone-16alpha-carbonitrile (PCN), while having a less pronounced role in receptor activation by rifampicin. The second residue to be mutated (H407 in human/Q404 in mouse) unexpectedly proved to be important for the basal level of activation of PXR. The H407A mutant of the human receptor showed a high level of constitutive activity, while the Q404H mutant of the mouse receptor demonstrated a sharply decreased basal activity compared to wild-type.


Subject(s)
Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/chemistry , Receptors, Steroid/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Line , Cytochrome P-450 CYP3A , DNA/genetics , Enzyme Activation , Humans , In Vitro Techniques , Ligands , Mice , Models, Molecular , Oxidoreductases, N-Demethylating/metabolism , Point Mutation , Pregnane X Receptor , Protein Conformation , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...