Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharm Biopharm ; 102: 191-201, 2016 May.
Article in English | MEDLINE | ID: mdl-26955751

ABSTRACT

The purpose of the study was to experimentally deduce pH-dependent critical volumes to dissolve applied dose (VDAD) that determine whether a drug candidate can be developed as immediate release (IR) tablet containing crystalline API, or if solubilization technology is needed to allow for sufficient oral bioavailability. pH-dependent VDADs of 22 and 83 compounds were plotted vs. the relative oral bioavailability (AUC solid vs. AUC solution formulation, Frel) in humans and rats, respectively. Furthermore, in order to investigate to what extent Frel rat may predict issues with solubility limited absorption in human, Frel rat was plotted vs. Frel human. Additionally, the impact of bile salts and lecithin on in vitro dissolution of poorly soluble compounds was tested and data compared to Frel rat and human. Respective in vitro - in vivo and in vivo - in vivo correlations were generated and used to build developability criteria. As a result, based on pH-dependent VDAD, Frel rat and in vitro dissolution in simulated intestinal fluid the IR formulation strategy within Pharmaceutical Research and Development organizations can be already set at late stage of drug discovery.


Subject(s)
Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Administration, Oral , Animals , Bile Acids and Salts/chemistry , Biological Availability , Biopharmaceutics/methods , Caco-2 Cells , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Humans , Lecithins/chemistry , Permeability , Pharmaceutical Preparations/metabolism , Rats , Solubility , Tablets/administration & dosage , Tablets/chemistry , Tablets/pharmacokinetics
2.
Cancer Med ; 2(5): 611-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24403227

ABSTRACT

The activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development, tumor progression, and resistance to chemo- and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase-driven HIF-1 reporter cell line under hypoxia. The high-throughput screening led to the identification of a class of aminoalkyl-substituted compounds that inhibited hypoxia-induced HIF-1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87-2243 was found to inhibit HIF-1α and HIF-2α protein accumulation under hypoxic conditions in non-small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF-1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87-2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel-Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase-2. Antitumor activity of BAY 87-2243, suppression of HIF-1α protein levels, and reduction of HIF-1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87-2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87-2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia-induced HIF-1 activity in tumors might be an interesting therapeutic approach to overcome chemo- and radiotherapy-resistance of hypoxic tumors.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Lung Neoplasms/metabolism , Oxadiazoles/pharmacology , Pyrazoles/pharmacology , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Carbonic Anhydrases/genetics , Cell Hypoxia/genetics , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Electron Transport Complex I/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1/biosynthesis , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Sequence Data , Molecular Targeted Therapy/methods , Oxadiazoles/administration & dosage , Oxadiazoles/blood , Oxadiazoles/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/therapeutic use , RNA, Small Interfering/genetics , Small Molecule Libraries , Tumor Burden/drug effects , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Xenograft Model Antitumor Assays/methods
3.
Br J Pharmacol ; 136(5): 773-83, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12086987

ABSTRACT

1. Soluble guanylyl cyclase (sGC) is the only proven receptor for the ubiquitous biological messenger nitric oxide (NO) and is intimately involved in many signal transduction pathways, most notably in regulating vascular tone and platelet function. sGC is a heterodimeric (alpha/ss) protein that converts GTP to cyclic GMP; NO binds to its prosthetic haem group. Here, we report the discovery of a novel sGC activating compound, its interaction with a previously unrecognized regulatory site and its therapeutic implications. 2. Through a high-throughput screen we identified BAY 58-2667, an amino dicarboxylic acid which potently activates sGC in an NO-independent manner. In contrast to NO, YC-1 and BAY 41-2272, the sGC stimulators described recently, BAY 58-2667 activates the enzyme even after it has been oxidized by the sGC inhibitor ODQ or rendered haem deficient. 3. Binding studies with radiolabelled BAY 58-2667 show a high affinity site on the enzyme. 4. Using photoaffinity labelling studies we identified the amino acids 371 (alpha-subunit) and 231 - 310 (ss-subunit) as target regions for BAY 58-2667. 5. sGC activation by BAY 58-2667 results in an antiplatelet activity both in vitro and in vivo and a potent vasorelaxation which is not influenced by nitrate tolerance. 6. BAY 58-2667 shows a potent antihypertensive effect in conscious spontaneously hypertensive rats. In anaesthetized dogs the hemodynamic effects of BAY 58-2667 and GTN are very similar on the arterial and venous system. 7. This novel type of sGC activator is a valuable research tool and may offer a new approach for treating cardiovascular diseases.


Subject(s)
Cardiovascular System/metabolism , Heme/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cardiovascular System/drug effects , Dogs , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Female , Guanylate Cyclase , In Vitro Techniques , Male , Rabbits , Rats , Rats, Inbred SHR , Rats, Wistar , Soluble Guanylyl Cyclase
SELECTION OF CITATIONS
SEARCH DETAIL
...