Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Drug Metab ; 24(8): 599-610, 2023.
Article in English | MEDLINE | ID: mdl-37592798

ABSTRACT

BACKGROUND: Nifurtimox is an effective treatment for patients with Chagas disease, but knowledge of its biotransformation and excretion is limited. OBJECTIVE: This study aimed to better understand the fate of oral nifurtimox in vivo. METHODS: We investigated the exposure and excretion pathways of [14C]-labeled nifurtimox and its metabolites in rats. We then quantified the prominent metabolites and nifurtimox in the urine and plasma of patients receiving nifurtimox using LC-HRMS with reference standards and quantified these compounds in rat plasma after a single, high dose of nifurtimox. We also investigated potential drug-drug interactions (DDIs) of these compounds in vitro. RESULTS: In rats, orally administered nifurtimox was rapidly absorbed (tmax 0.5 h) and eliminated (t½ 1.4 h). Metabolism of nifurtimox yielded six predominant metabolites (M-1 to M-6) in urine and plasma, and the dose was excreted equally via the renal and fecal routes with only traces of unchanged nifurtimox detectable due to its instability in excreta. In patients with Chagas disease, only M-6 and M-4 achieved relevant exposure levels, and the total amount of excreted metabolites in urine was higher in fed versus fasted patients, consistent with the higher systemic exposure. For nifurtimox, M-6, and M-4, no potential perpetrator pharmacokinetic DDIs with the main cytochrome P- 450 enzymes and drug transporters were identified in vitro. CONCLUSION: This contemporary analysis of the complex metabolite profile and associated exposures emerging after oral dosing of nifurtimox in rats and humans, together with the expected low risk for clinically relevant DDIs, expands the understanding of this important anti-trypanosomal drug.


Subject(s)
Chagas Disease , Nifurtimox , Humans , Rats , Animals , Drug Interactions , Biotransformation , Cytochrome P-450 Enzyme System , Chagas Disease/drug therapy , Administration, Oral
2.
J Med Chem ; 63(20): 11639-11662, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32969660

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a rare and devastating chronic lung disease of unknown etiology. Despite the approved treatment options nintedanib and pirfenidone, the medical need for a safe and well-tolerated antifibrotic treatment of IPF remains high. The human prostaglandin F receptor (hFP-R) is widely expressed in the lung tissue and constitutes an attractive target for the treatment of fibrotic lung diseases. Herein, we present our research toward novel quinoline-based hFP-R antagonists, including synthesis and detailed structure-activity relationship (SAR). Starting from a high-throughput screening (HTS) hit of our corporate compound library, multiple parameter improvements-including increase of the relative oral bioavailability Frel from 3 to ≥100%-led to a highly potent and selective hFP-R antagonist with complete oral absorption from suspension. BAY-6672 (46) represents-to the best of our knowledge-the first reported FP-R antagonist to demonstrate in vivo efficacy in a preclinical animal model of lung fibrosis, thus paving the way for a new treatment option in IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Quinolines/chemical synthesis , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Animals , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Molecular Structure , Quinolines/chemistry , Quinolines/therapeutic use , Rats , Rats, Wistar , Structure-Activity Relationship
3.
Eur J Drug Metab Pharmacokinet ; 45(4): 433-444, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32125665

ABSTRACT

BACKGROUND AND OBJECTIVES: In vivo studies were performed with the novel, selective, non-steroidal mineralocorticoid receptor antagonist finerenone to assess the relevance of inductive and/or inhibitory effects on cytochrome P450 (CYP) enzymes observed in vitro. METHODS: CYP isoenzyme-specific substrates were incubated in vitro with finerenone or its metabolites to investigate reversible and irreversible inhibitory as well as inductive potential. Three crossover studies in healthy male volunteers investigated the effects of finerenone (20 mg orally) on the pharmacokinetics of the index substrates midazolam (CYP3A4, n = 30), repaglinide (CYP2C8, n = 28) and warfarin (CYP2C9, n = 24). RESULTS: Finerenone caused direct inhibitory effects on CYP activities in vitro in the rank order CYP2C8, CYP1A1 > CYP3A4 > CYP2C9 and CYP2C19, but not on other major CYP isoforms. Moreover, irreversible inhibition of CYP3A4 was observed. The major metabolites of finerenone demonstrated minor reversible inhibition of CYP1A1, CYP2C9 and CYP3A4 with no hint of time-dependent inhibition of any CYP isoform. Calculations from in vitro data according to regulatory guidelines suggested likely inhibition of CYP2C8 and CYP3A4 in vivo, whereas this was not the case for CYP1A1, CYP2C9 and CYP2C19. Furthermore, finerenone and three of its metabolites were inducers of CYP3A4 in vitro with predicted weak-to-moderate in vivo relevance. Studies in healthy volunteers, prompted by these results, demonstrated no effect of finerenone on CYP isoenzymes for which in vitro data had indicated potential inhibition or induction. CONCLUSION: Administration of finerenone 20 mg once daily confers no risk of clinically relevant drug-drug interactions with substrates of cytochrome P450 enzymes.


Subject(s)
Carbamates/pharmacokinetics , Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Midazolam/pharmacokinetics , Mineralocorticoid Receptor Antagonists/pharmacology , Naphthyridines/pharmacology , Piperidines/pharmacokinetics , Warfarin/pharmacokinetics , Administration, Oral , Adolescent , Adult , Cross-Over Studies , Cytochrome P-450 CYP2C8/metabolism , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inducers/administration & dosage , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Double-Blind Method , Drug Interactions , Germany , Humans , Isoenzymes , Male , Middle Aged , Mineralocorticoid Receptor Antagonists/administration & dosage , Models, Biological , Naphthyridines/administration & dosage , Substrate Specificity , Young Adult
4.
J Med Chem ; 62(24): 11194-11217, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31746599

ABSTRACT

The P2X4 receptor is a ligand-gated ion channel that is expressed on a variety of cell types, especially those involved in inflammatory and immune processes. High-throughput screening led to a new class of P2X4 inhibitors with substantial CYP 3A4 induction in human hepatocytes. A structure-guided optimization with respect to decreased pregnane X receptor (PXR) binding was started. It was found that the introduction of larger and more polar substituents on the ether linker led to less PXR binding while maintaining the P2X4 inhibitory potency. This translated into significantly reduced CYP 3A4 induction for compounds 71 and 73. Unfortunately, the in vivo pharmacokinetic (PK) profiles of these compounds were insufficient for the desired profile in humans. However, BAY-1797 (10) was identified and characterized as a potent and selective P2X4 antagonist. This compound is suitable for in vivo studies in rodents, and the anti-inflammatory and anti-nociceptive effects of BAY-1797 were demonstrated in a mouse complete Freund's adjuvant (CFA) inflammatory pain model.


Subject(s)
Acetamides/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A/metabolism , Drug Discovery , Inflammation/drug therapy , Pain/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/chemistry , Acetamides/chemistry , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP3A Inducers/chemistry , Enzyme Induction , Female , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation/pathology , Ligands , Male , Mice , Mice, Inbred C57BL , Pain/metabolism , Pain/pathology , Purinergic P2X Receptor Antagonists/chemistry , Rats , Rats, Wistar
5.
Eur J Drug Metab Pharmacokinet ; 44(6): 747-759, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31571146

ABSTRACT

BACKGROUND AND OBJECTIVES: Darolutamide is a novel androgen receptor (AR) antagonist approved for the treatment of nonmetastatic castration-resistant prostate cancer (nmCRPC). Accordingly, the drug-drug interaction (DDI) potential of darolutamide was investigated in both nonclinical and clinical studies. METHODS: In vitro studies were performed to determine the potential for darolutamide to be a substrate, inducer or inhibitor for cytochrome P450 (CYP) isoforms, other metabolizing enzymes and drug transporters. A phase I drug-interaction study in healthy volunteers evaluated the impact of co-administering rifampicin [CYP3A4 and P-glycoprotein (P-gp) inducer] and itraconazole [CYP3A4, P-gp and breast cancer resistance protein (BCRP) inhibitor] on the pharmacokinetics of darolutamide. Two further phase I studies assessed the impact of co-administering oral darolutamide on the pharmacokinetics of midazolam (sensitive CYP3A4 substrate) and dabigatran etexilate (P-gp substrate) and the impact on the pharmacokinetics of co-administered rosuvastatin [a substrate for BCRP, organic anion-transporting polypeptide (OATP)1B1, OATP1B3 and organic anion transporter (OAT)3]. RESULTS: In vitro, darolutamide was predominantly metabolized via oxidative biotransformation catalyzed by CYP3A4 and was identified as a substrate for P-gp and BCRP. The enzymatic activity of nine CYP isoforms was not inhibited or slightly inhibited in vitro with darolutamide, and a rank order and mechanistic static assessment indicated that risk of clinically relevant DDIs via CYP inhibition is very low. In vitro, darolutamide exhibited no relevant induction of CYP1A2 or CYP2B6 activity. Inhibition of BCRP-, P-gp-, OAT3-, MATE1-, MATE2-K-, OATP1B1- and OATP1B3-mediated transport was observed in vitro. Phase I data showed that darolutamide exposure increased 1.75-fold with co-administered itraconazole and decreased by 72% with rifampicin. Co-administration of darolutamide with CYP3A4/P-gp substrates showed no effect or only minor effects. Rosuvastatin exposure increased 5.2-fold with darolutamide because of BCRP and probably also OATPB1/OATPB3 inhibition. CONCLUSIONS: Darolutamide has a low potential for clinically relevant DDIs with drugs that are substrates for CYP or P-gp; increased exposure of BCRP and probably OATP substrates was the main interaction of note.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Drug Interactions , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Aged , Cells, Cultured , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Dabigatran/pharmacokinetics , Enzyme Induction/drug effects , Female , Humans , Itraconazole/pharmacology , Male , Membrane Transport Proteins/drug effects , Microsomes, Liver/drug effects , Midazolam/pharmacokinetics , Middle Aged , Pyrazoles/blood , Pyrazoles/urine , Rifampin/pharmacology , Rosuvastatin Calcium/pharmacokinetics
6.
Chem Res Toxicol ; 32(6): 1115-1122, 2019 06 17.
Article in English | MEDLINE | ID: mdl-30950278

ABSTRACT

Cytochrome P450s (CYPs), a superfamily of enzymes, are involved in the biotransformation of endogenous and xenobiotic chemicals and mainly responsible for the metabolic clearance of widely prescribed drugs. Out of the 57 human isoforms, only a few, most notably CYP3A4, are considered to be important in this process. CYP1A1, one of the three isoforms of the CYP1 family, is widely believed to play an important role in the metabolism and activation of numerous procarcinogens, e.g., polyaromatic hydrocarbons (PAHs) or aromatic amines. It is also known that CYP1A1 is highly inducible by endogenous and exogenous factors, e.g., PAHs. However, CYP1A1 has not been considered to play a significant role in the metabolic clearance of drugs, since this isoform has been detected only in extrahepatic tissues in small amounts. In contrast to conventional wisdom, we herein demonstrate the expression of CYP1A1 protein in human liver microsomal preparations. The expression levels of CYP1A1 were quantified by Western blot and LC/MS analyses and corresponded well with enzymatic activities of highly selective CYP1A1 reactions. In a panel of 29 individual liver microsomal preparations, highly variable and substantial expression levels (up to ∼10 pmol/mg) were measured. Together with the high selectivity and especially the high metabolic efficiency of CYP1A1 shown for granisetron and riociguat, it is demonstrated that CYP1A1 plays an important role in the metabolic clearance of these drugs and is responsible for the clinically observed interindividual variability in their pharmacokinetics. Therefore, the importance of CYP1A1 in drug discovery and development needs to be reconsidered.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Granisetron/pharmacokinetics , Liver/drug effects , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Blotting, Western , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A1/analysis , Humans , Liver/metabolism , Mass Spectrometry , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism
7.
Drug Metab Dispos ; 44(5): 617-23, 2016 May.
Article in English | MEDLINE | ID: mdl-26669328

ABSTRACT

An antibody-drug conjugate (ADC) is a unique therapeutic modality composed of a highly potent drug molecule conjugated to a monoclonal antibody. As the number of ADCs in various stages of nonclinical and clinical development has been increasing, pharmaceutical companies have been exploring diverse approaches to understanding the disposition of ADCs. To identify the key absorption, distribution, metabolism, and excretion (ADME) issues worth examining when developing an ADC and to find optimal scientifically based approaches to evaluate ADC ADME, the International Consortium for Innovation and Quality in Pharmaceutical Development launched an ADC ADME working group in early 2014. This white paper contains observations from the working group and provides an initial framework on issues and approaches to consider when evaluating the ADME of ADCs.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunoconjugates/metabolism , Pharmaceutical Preparations/metabolism , Animals , Drug Industry/methods , Humans
8.
Nat Methods ; 6(10): 741-4, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19749761

ABSTRACT

We report a proteomics strategy to both identify and quantify cellular target protein interactions with externally introduced ligands. We determined dissociation constants for target proteins interacting with the ligand of interest by combining quantitative mass spectrometry with a defined set of affinity purification experiments. We demonstrate the general utility of this methodology in interaction studies involving small-molecule kinase inhibitors, a tyrosine-phosphorylated peptide and an antibody as affinity ligands.


Subject(s)
Cell Extracts/chemistry , Chromatography, Affinity/methods , Mass Spectrometry/methods , Protein Interaction Mapping/methods , Proteome/metabolism , Proteomics/methods
9.
Mol Cell ; 31(3): 438-48, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18691976

ABSTRACT

Protein kinases are pivotal regulators of cell signaling that modulate each other's functions and activities through site-specific phosphorylation events. These key regulatory modifications have not been studied comprehensively, because low cellular abundance of kinases has resulted in their underrepresentation in previous phosphoproteome studies. Here, we combine kinase-selective affinity purification with quantitative mass spectrometry to analyze the cell-cycle regulation of protein kinases. This proteomics approach enabled us to quantify 219 protein kinases from S and M phase-arrested human cancer cells. We identified more than 1000 phosphorylation sites on protein kinases. Intriguingly, half of all kinase phosphopeptides were upregulated in mitosis. Our data reveal numerous unknown M phase-induced phosphorylation sites on kinases with established mitotic functions. We also find potential phosphorylation networks involving many protein kinases not previously implicated in mitotic progression. These results provide a vastly extended knowledge base for functional studies on kinases and their regulation through site-specific phosphorylation.


Subject(s)
Cell Cycle , Phosphoproteins/analysis , Phosphotransferases/metabolism , Proteomics , Amino Acid Sequence , Enzyme Activation , HeLa Cells , Humans , Mitosis , Molecular Sequence Data , Phosphopeptides/analysis , Phosphorylation , Phosphotransferases/chemistry , S Phase , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...