Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carmen B. Rödl is active.

Publication


Featured researches published by Carmen B. Rödl.


Bioorganic & Medicinal Chemistry Letters | 2012

SAR-study on a new class of imidazo[1,2-a]pyridine-based inhibitors of 5-lipoxygenase.

Martina Hieke; Carmen B. Rödl; Joanna M. Wisniewska; Estel.la Buscató; Holger Stark; Manfred Schubert-Zsilavecz; Dieter Steinhilber; Bettina Hofmann; Ewgenij Proschak

A novel class of 5-lipoxygenase (5-LO) inhibitors characterized by a central imidazo[1,2-a]pyridine scaffold, a cyclohexyl moiety and an aromatic system, is presented. This scaffold was identified in a virtual screening study and exhibits promising inhibitory potential on the 5-LO. Here, we investigate the structure-activity relationships of this compound class. With N-cyclohexyl-6-methyl-2-(4-morpholinophenyl)imidazo[1,2-a]pyridine-3-amine (14), we identified a potent 5-LO inhibitor (IC(50)=0.16μM (intact cells) and 0.1μM (cell-free)), which may possess potential as an effective lead compound intervening with inflammatory diseases and certain types of cancer.


Journal of Medicinal Chemistry | 2011

A Class of 5-Benzylidene-2-phenylthiazolinones with High Potency as Direct 5-Lipoxygenase Inhibitors

Bettina Hofmann; Sebastian Barzen; Carmen B. Rödl; Andreas Kiehl; Julia Borig; Aleksandra Zivkovic; Holger Stark; Gisbert Schneider; Dieter Steinhilber

A novel class of potent direct 5-lipoxygenase (5-LO) inhibitors bearing a thiazolinone-scaffold identified by virtual screening is presented. A range of substitutions and the importance of the 2-phenyl moiety were evaluated. This series is characterized by high potency in intact polymorphonuclear leukocytes and a cell-free system, exemplified by (Z)-2-(4-chlorophenyl)-5-(4-methoxybenzylidene)-5H-thiazol-4-one (18, IC(50) = 0.28 and 0.09 μM). These disubstituted thiazolinones may possess potential for intervention with inflammatory and allergic diseases and certain cancer types.


Biochemical Pharmacology | 2012

Molecular characterization of EP6—A novel imidazo[1,2-a]pyridine based direct 5-lipoxygenase inhibitor

Joanna M. Wisniewska; Carmen B. Rödl; Astrid S. Kahnt; Estel.la Buscató; Sandra Ulrich; Yusuf Tanrikulu; Janosch Achenbach; Florian Rörsch; Sabine Grösch; Gisbert Schneider; Jindrich Cinatl; Ewgenij Proschak; Dieter Steinhilber; Bettina Hofmann

5-Lipoxygenase (5-LO) is a crucial enzyme of the arachidonic acid (AA) cascade and catalyzes the formation of bioactive leukotrienes (LTs) which are involved in inflammatory diseases and allergic reactions. The pathophysiological effects of LTs are considered to be prevented by 5-LO inhibitors. In this study we present cyclohexyl-[6-methyl-2-(4-morpholin-4-yl-phenyl)-imidazo[1,2-a]pyridin-3-yl]-amine (EP6), a novel imidazo[1,2-a]pyridine based compound and its characterization in several in vitro assays. EP6 suppresses 5-LO activity in intact polymorphonuclear leukocytes with an IC(50) value of 0.16μM and exhibits full inhibitory potency in cell free assays (IC(50) value of 0.05μM for purified 5-LO). The efficacy of EP6 was not affected by the redox tone or the concentration of exogenous AA, characteristic drawbacks known for the class of nonredox-type 5-LO inhibitors. Furthermore, EP6 suppressed 5-LO activity independently of the cell stimulus or the activation pathway of 5-LO contrary to what is known for some nonredox-type inhibitors. Using molecular modeling and site-directed mutagenesis studies, we were able to derive a feasible binding region within the C2-like domain of 5-LO that can serve as a new starting point for optimization and development of new 5-LO inhibitors targeting this site. EP6 has promising effects on cell viability of tumor cells without mutagenic activity. Hence the drug may possess potential for intervention with inflammatory and allergic diseases and certain types of cancer including leukemia.


Journal of Medicinal Chemistry | 2013

Synthesis and structure-activity relationship studies of novel dual inhibitors of soluble epoxide hydrolase and 5-lipoxygenase.

Karin Meirer; Carmen B. Rödl; Joanna M. Wisniewska; Sven George; Ann-Kathrin Häfner; Estel.la Buscató; Franca-Maria Klingler; Steffen Hahn; Dirk Berressem; Sandra K. Wittmann; Dieter Steinhilber; Bettina Hofmann; Ewgenij Proschak

Current research leads to the assumption that drugs affecting more than one target could result in a more efficient treatment of diseases and fewer safety concerns. Administration of drugs inhibiting only one branch of the arachidonic acid cascade is usually accompanied by side effects. We therefore designed and synthesized a library of hybrid molecules incorporating an imidazo[1,2-a]pyridine and an urea moiety as novel soluble epoxide hydrolase (sEH)/5-lipoxygenase (5-LO) dual inhibitors. Evaluation of the compounds was accomplished by in vitro testing using recombinant enzyme assays.


ACS Medicinal Chemistry Letters | 2013

Exploring the Chemical Space of Multitarget Ligands Using Aligned Self-Organizing Maps

Janosch Achenbach; Franca-Maria Klingler; René Blöcher; Daniel Moser; Ann-Kathrin Häfner; Carmen B. Rödl; Simon B.M. Kretschmer; Björn Krüger; Frank Löhr; Holger Stark; Bettina Hofmann; Dieter Steinhilber; Ewgenij Proschak

Design of multitarget drugs and polypharmacological compounds has become popular during the past decade. However, the main approach to design such compounds is to link two selective ligands via a flexible linker. Although such chimeric ligands often have reasonable potency in vitro, the in vivo efficacy is low due to high molecular weight, low ligand efficiency, and poor pharmacokinetic profile. We developed an unprecedented in silico approach for fragment-based design of multitarget ligands. It relies on superposition of the chemical spaces related to the affinity on single targets represented by self-organizing maps. We used this approach for screening of molecular fragments, which bind to the enzymes 5-lipoxygenase (5-LO) and soluble epoxide hydrolase (sEH). Using STD-NMR and activity-based assays, we were able to identify fragments binding to both targets. Furthermore, we were able to expand one of the fragments to a potent dual inhibitor bearing a reasonable molecular weight (MW = 446) and high affinity to both targets (IC50 of 0.03 μM toward 5-LO and 0.17 μM toward sEH).


European Journal of Medicinal Chemistry | 2014

Multi-dimensional target profiling of N,4-diaryl-1,3-thiazole-2-amines as potent inhibitors of eicosanoid metabolism.

Carmen B. Rödl; Dominik Vogt; Simon B.M. Kretschmer; Katja Ihlefeld; Sebastian Barzen; Astrid Brüggerhoff; Janosch Achenbach; Ewgenij Proschak; Dieter Steinhilber; Holger Stark; Bettina Hofmann

Eicosanoids like leukotrienes and prostaglandins play a considerable role in inflammation. Produced within the arachidonic acid (AA) cascade, these lipid mediators are involved in the pathogenesis of pain as well as acute and chronic inflammatory diseases like rheumatoid arthritis and asthma. With regard to the lipid cross-talk within the AA pathway, a promising approach for an effective anti-inflammatory therapy is the development of inhibitors targeting more than one enzyme of this cascade. Within this study, thirty N-4-diaryl-1,3-thiazole-2-amine based compounds with different substitution patterns were synthesized and tested in various cell-based assays to investigate their activity and selectivity profile concerning five key enzymes involved in eicosanoid metabolism (5-, 12-, 15-lipoxygenase (LO), cyclooxygenase-1 and -2 (COX-1/-2)). With compound 7, 2-(4-phenyl)thiazol-2-ylamino)phenol (ST-1355), a multi-target ligand targeting all tested enzymes is presented, whereas compound 9, 2-(4-(4-chlorophenyl)thiazol-2-ylamino)phenol (ST-1705), represents a potent and selective 5-LO and COX-2 inhibitor with an IC50 value of 0.9 ± 0.2 μM (5-LO) and a residual activity of 9.1 ± 1.1% at 10 μM (COX-2 product formation). The promising characteristics and the additional non-cytotoxic profile of both compounds reveal new lead structures for the treatment of eicosanoid-mediated diseases.


Bioorganic & Medicinal Chemistry | 2012

Synthesis and biological evaluation of a class of 5-benzylidene-2-phenyl-thiazolinones as potent 5-lipoxygenase inhibitors

Sebastian Barzen; Carmen B. Rödl; Andreas Lill; Dieter Steinhilber; Holger Stark; Bettina Hofmann

A class of 5-lipoxygenase (5-LO) inhibitors characterized by a central 5-benzylidene-2-phenyl-thiazolinone scaffold was synthesized as a new series of molecular modifications and extensions of a previously reported series. Compounds were tested in a cell-based and a cell-free assay and furthermore evaluated for their influence on cell viability. The presented substituted thiazolinone scaffold turned out to be essential for both the 5-LO inhibitory activity and the non-cytotoxic profile. With (Z)-5-(4-methoxybenzylidene)-2-(naphthalen-2-yl)-5H-thiazol-4-one (2k, ST1237), a potent, direct, non-cytotoxic 5-LO inhibitor with IC(50) of 0.08 μM and 0.12 μM (cell-free assay and intact cells), we present a promising lead optimization and development for further investigations as novel anti-inflammatory drug.


British Journal of Pharmacology | 2012

Molecular pharmacological profile of a novel thiazolinone‐based direct and selective 5‐lipoxygenase inhibitor

Bettina Hofmann; Carmen B. Rödl; Astrid S. Kahnt; Tj Maier; Aa Michel; M Hoffmann; O Rau; K Awwad; M Pellowska; Mario Wurglics; M Wacker; A Živković; Ingrid Fleming; Manfred Schubert-Zsilavecz; Holger Stark; Gisbert Schneider; Dieter Steinhilber

BACKGROUND AND PURPOSE The potency of many 5‐lipoxygenase (5‐LOX) inhibitors depends on the cellular peroxide tone and the mechanism of 5‐LOX enzyme activation. Therefore, new inhibitors that act regardless of the mode of enzyme activation need to be developed. Recently, we identified a novel class of thiazolinone‐based compounds as potent 5‐LOX inhibitors. Here, we present the molecular pharmacological profile of (Z)‐5‐(4‐methoxybenzylidene)‐2‐(p‐tolyl)‐5H‐thiazol‐4‐one, compound C06.


Biochimica et Biophysica Acta | 2012

Inhibition of 5-lipoxygenase by U73122 is due to covalent binding to cysteine 416.

Michael Hörnig; Stavroula Markoutsa; Ann-Kathrin Häfner; Sven George; Joanna M. Wisniewska; Carmen B. Rödl; Bettina Hofmann; Thorsten J. Maier; Michael Karas; Oliver Werz; Dieter Steinhilber

U73122 which was originally identified as a phospholipase C inhibitor represents a potent direct inhibitor of purified 5-lipoxygenase (5-LO) with an IC50 value of 30 nM. 5-LO catalyzes the conversion of arachidonic acid (AA) into leukotrienes which represent mediators involved in inflammatory and allergic reactions and in host defense reactions against microorganisms. Since the efficient inhibition of the human 5-LO enzyme depended on the thiol reactivity of the maleinimide group of U73122, we used this property to identify cysteine residues in the 5-LO protein that are important for 5-LO inhibition by U73122. We found by MALDI-MS that U73122 covalently binds to cysteine residues 99, 159, 248, 264, 416 and 449. Mutation of Cys416 to serine strongly reduces inhibition of 5-LO by U73122 and the additional mutation of three cysteines close to Cys416 further impairs 5-LO inhibition by the compound. Wash out experiments with U73122 and 5-LO indicated an irreversible binding of U73122. Together, our data suggest that the area around Cys416 which is close to the proposed AA entry channel to the active site is an interesting target for the development of new 5-LO inhibitors.


ChemMedChem | 2011

Potent Inhibitors of 5‐Lipoxygenase Identified using Pseudoreceptors

Carmen B. Rödl; Yusuf Tanrikulu; Joanna M. Wisniewska; Ewgenij Proschak; Gisbert Schneider; Dieter Steinhilber; Bettina Hofmann

Virtual screening has become a routine method in pharmaceutical drug discovery. Two typical strategies in rational drug design are ligandand receptor-based screening approaches. Receptor-based screening methods depend on information about the three-dimensional (3D) structure of the receptor. However, a suitable 3D structure might not always be directly available, in which case ligand-based strategies are the method of choice for virtual screening. Ligand-based techniques can be applied without any information about the structure of the receptor. Pseudoreceptor models combine these two strategies in rational drug design. The only information they need are at least one active reference ligand, or a 3D alignment of a reference ligand selection. By capturing shape and important interaction points, a virtual binding pocket is constructed around one or more reference ligands. So called pseudoatoms are positioned around the ligand(s) with valid interaction distances and angles (potential pharmacophore points). These pseudoatoms mimic putative receptor atom positions and the noncovalent interaction properties that need to be occupied for successful ligand binding. Here, we show the successful application of a pseudoreceptor model to the identification of novel inhibitors of the 5-lipoxygenase (5-LO) pathway. 5-LO catalyzes the first step in the biosynthesis of leukotrienes (LTs) from arachidonic acid. Based on the multiple pathophysiological actions of LTs, there exist increasing therapeutic indications for antileukotriene therapy, including inflammation, allergic rhinitis, cardiovascular diseases, cancer, and osteoporosis. To date, the only direct 5-LO inhibitor in clinical use is the hydrolytic stable N-hydroxyurea derivative A-64077 (zileuton). Pseudoreceptor models could be explicitly useful in the identification of novel 5-LO inhibitors, as there is only limited protein structural information available that is suitable for direct use in virtual screening. Recently, a crystal structure of the so called “stable 5-LO” was solved. It represents an apoprotein structure without a bound ligand. Apoprotein structures have their limitations for virtual screening, though several workarounds like molecular dynamics (MD) simulations exist. In the case of 5-LO, due to the intrinsic structural flexibility and observed induced fit upon substrate/ inhibitor binding shown for other LOs, special attention must be paid when working with a static crystal structure. Therefore, a receptor-structure-independent approach might be a promising workaround. This encouraged us to apply a pseudoreceptor model for the identification of novel scaffolds inhibiting LT formation using known active ligands. We previously reported the use of ligand-based virtual screening to identify new inhibitors of 5-LO product formation using two similarity search methods, “Charge3D” 15] and “TripleCharge3D”. 16] Within this study, a series of imidazo-[1,2a]-pyridines was identified as highly potent 5-LO inhibitors with IC50 values of approximately 1 mm (Table 1). [13] Starting

Collaboration


Dive into the Carmen B. Rödl's collaboration.

Top Co-Authors

Avatar

Bettina Hofmann

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Dieter Steinhilber

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Holger Stark

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Ewgenij Proschak

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Estel.la Buscató

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Gisbert Schneider

École Polytechnique Fédérale de Lausanne

View shared research outputs
Top Co-Authors

Avatar

Janosch Achenbach

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge