Network


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

Hotspot


Dive into the research topics where Carsten Schultz-Fademrecht is active.

Publication


Featured researches published by Carsten Schultz-Fademrecht.


Nature | 2014

The E3 ligase Cbl-b and TAM receptors regulate cancer metastasis via natural killer cells

Magdalena Paolino; Axel Choidas; Stephanie Wallner; Blanka Pranjic; Iris Uribesalgo; Stefanie Loeser; Amanda M. Jamieson; Wallace Y. Langdon; Fumiyo Ikeda; Juan Pablo Fededa; Shane J. Cronin; Roberto Nitsch; Carsten Schultz-Fademrecht; Jan Eickhoff; Sascha Menninger; Anke Unger; Robert Torka; Thomas Gruber; Reinhard Hinterleitner; Gottfried Baier; Dominik Wolf; Axel Ullrich; Bert Klebl; Josef M. Penninger

Tumour metastasis is the primary cause of mortality in cancer patients and remains the key challenge for cancer therapy. New therapeutic approaches to block inhibitory pathways of the immune system have renewed hopes for the utility of such therapies. Here we show that genetic deletion of the E3 ubiquitin ligase Cbl-b (casitas B-lineage lymphoma-b) or targeted inactivation of its E3 ligase activity licenses natural killer (NK) cells to spontaneously reject metastatic tumours. The TAM tyrosine kinase receptors Tyro3, Axl and Mer (also known as Mertk) were identified as ubiquitylation substrates for Cbl-b. Treatment of wild-type NK cells with a newly developed small molecule TAM kinase inhibitor conferred therapeutic potential, efficiently enhancing anti-metastatic NK cell activity in vivo. Oral or intraperitoneal administration using this TAM inhibitor markedly reduced murine mammary cancer and melanoma metastases dependent on NK cells. We further report that the anticoagulant warfarin exerts anti-metastatic activity in mice via Cbl-b/TAM receptors in NK cells, providing a molecular explanation for a 50-year-old puzzle in cancer biology. This novel TAM/Cbl-b inhibitory pathway shows that it might be possible to develop a ‘pill’ that awakens the innate immune system to kill cancer metastases.


Journal of Medicinal Chemistry | 2009

Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors.

Philip Jones; Sergio Altamura; Julia K. Boueres; Federica Ferrigno; Massimiliano Fonsi; Claudia Giomini; Stefania Lamartina; Edith Monteagudo; Jesus M. Ontoria; Maria Vittoria Orsale; Maria Cecilia Palumbi; Silvia Pesci; Giuseppe Roscilli; Rita Scarpelli; Carsten Schultz-Fademrecht; Carlo Toniatti; Michael Rowley

We disclose the development of a novel series of 2-phenyl-2H-indazole-7-carboxamides as poly(ADP-ribose)polymerase (PARP) 1 and 2 inhibitors. This series was optimized to improve enzyme and cellular activity, and the resulting PARP inhibitors display antiproliferation activities against BRCA-1 and BRCA-2 deficient cancer cells, with high selectivity over BRCA proficient cells. Extrahepatic oxidation by CYP450 1A1 and 1A2 was identified as a metabolic concern, and strategies to improve pharmacokinetic properties are reported. These efforts culminated in the identification of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide 56 (MK-4827), which displays good pharmacokinetic properties and is currently in phase I clinical trials. This compound displays excellent PARP 1 and 2 inhibition with IC(50) = 3.8 and 2.1 nM, respectively, and in a whole cell assay, it inhibited PARP activity with EC(50) = 4 nM and inhibited proliferation of cancer cells with mutant BRCA-1 and BRCA-2 with CC(50) in the 10-100 nM range. Compound 56 was well tolerated in vivo and demonstrated efficacy as a single agent in a xenograft model of BRCA-1 deficient cancer.


Bioorganic & Medicinal Chemistry Letters | 2008

2-Trifluoroacetylthiophenes, a novel series of potent and selective class II histone deacetylase inhibitors.

Philip Jones; Matthew J. Bottomley; Andrea Carfi; Ottavia Cecchetti; Federica Ferrigno; Paola Lo Surdo; Jesus M. Ontoria; Michael Rowley; Rita Scarpelli; Carsten Schultz-Fademrecht; Christian Steinkühler

The identification of class II HDAC inhibitors has been hampered by lack of efficient enzyme assays, in the preceding paper two assays have been developed to improve the efficiency of these enzymes: mutating an active site histidine to tyrosine, or by the use of a trifluoroacetamide lysine substrate, allowing screening to identify class II HDAC inhibitors. Herein, 2-trifluoroacetylthiophenes have been demonstrated to inhibit class II HDACs, resulting in the development of a series of 5-(trifluoroacetyl)thiophene-2-carboxamides as novel, potent and selective class II HDAC inhibitors. X-ray crystal structures of the HDAC 4 catalytic domain with a bound inhibitor demonstrate these compounds are active site inhibitors and bind in their hydrated form.


Journal of Medicinal Chemistry | 2009

Identification of novel, selective, and stable inhibitors of class II histone deacetylases. Validation studies of the inhibition of the enzymatic activity of HDAC4 by small molecules as a novel approach for cancer therapy.

Jesus M. Ontoria; Sergio Altamura; Annalise Di Marco; Federica Ferrigno; Ralph Laufer; Ester Muraglia; Maria Cecilia Palumbi; Michael Rowley; Rita Scarpelli; Carsten Schultz-Fademrecht; Sergio Serafini; Christian Steinkühler; Philip Jones

5-Aryl-2-(trifluoroacetyl)thiophenes were identified as a new series of class II HDAC inhibitors (HDACi). Further development of this new series led to compounds such as 6h, a potent inhibitor of HDAC4 and HDAC6 (HDAC4 WT IC(50) = 310 nM, HDAC6 IC(50) = 70 nM) that displays 40-fold selectivity over HDAC1 and improved stability in HCT116 cancer cells (t(1/2) = 11 h). Compounds 6h and 2 show inhibition of alpha-tubulin deacetylation in HCT116 cells at 1 microM concentration and antiproliferation effects only at concentrations where inhibition of histone H3 deacetylation is observed.


Nature Communications | 2016

Identification of pyrazolopyridazinones as PDEδ inhibitors

Björn Papke; Sandip Murarka; Holger A Vogel; Pablo Martín-Gago; Marija Kovacevic; Dina C Truxius; Eyad K. Fansa; Shehab Ismail; Gunther Zimmermann; Kaatje Heinelt; Carsten Schultz-Fademrecht; Alaa Al Saabi; Matthias Baumann; Peter Nussbaumer; Alfred Wittinghofer; Herbert Waldmann; Philippe I. H. Bastiaens

The prenyl-binding protein PDEδ is crucial for the plasma membrane localization of prenylated Ras. Recently, we have reported that the small-molecule Deltarasin binds to the prenyl-binding pocket of PDEδ, and impairs Ras enrichment at the plasma membrane, thereby affecting the proliferation of KRas-dependent human pancreatic ductal adenocarcinoma cell lines. Here, using structure-based compound design, we have now identified pyrazolopyridazinones as a novel, unrelated chemotype that binds to the prenyl-binding pocket of PDEδ with high affinity, thereby displacing prenylated Ras proteins in cells. Our results show that the new PDEδ inhibitor, named Deltazinone 1, is highly selective, exhibits less unspecific cytotoxicity than the previously reported Deltarasin and demonstrates a high correlation with the phenotypic effect of PDEδ knockdown in a set of human pancreatic cancer cell lines.


Journal of Medicinal Chemistry | 2015

Targeting Drug Resistance in EGFR with Covalent Inhibitors: A Structure-Based Design Approach.

Julian Engel; André Richters; Matthäus Getlik; Stefano Tomassi; Marina Keul; Termathe M; Jonas Lategahn; Christian F. W. Becker; Svenja Mayer-Wrangowski; Christian Grütter; Uhlenbrock N; Krüll J; Schaumann N; Eppmann S; Patrick Kibies; Franziska Hoffgaard; Jochen Heil; Sascha Menninger; Sandra Ortiz-Cuaran; Johannes M. Heuckmann; Tinnefeld; René P. Zahedi; Martin L. Sos; Carsten Schultz-Fademrecht; Roman K. Thomas; Stefan M. Kast; Daniel Rauh

Receptor tyrosine kinases represent one of the prime targets in cancer therapy, as the dysregulation of these elementary transducers of extracellular signals, like the epidermal growth factor receptor (EGFR), contributes to the onset of cancer, such as non-small cell lung cancer (NSCLC). Strong efforts were directed to the development of irreversible inhibitors and led to compound CO-1686, which takes advantage of increased residence time at EGFR by alkylating Cys797 and thereby preventing toxic effects. Here, we present a structure-based approach, rationalized by subsequent computational analysis of conformational ligand ensembles in solution, to design novel and irreversible EGFR inhibitors based on a screening hit that was identified in a phenotype screen of 80 NSCLC cell lines against approximately 1500 compounds. Using protein X-ray crystallography, we deciphered the binding mode in engineered cSrc (T338M/S345C), a validated model system for EGFR-T790M, which constituted the basis for further rational design approaches. Chemical synthesis led to further compound collections that revealed increased biochemical potency and, in part, selectivity toward mutated (L858R and L858R/T790M) vs nonmutated EGFR. Further cell-based and kinetic studies were performed to substantiate our initial findings. Utilizing proteolytic digestion and nano-LC-MS/MS analysis, we confirmed the alkylation of Cys797.


Journal of Medicinal Chemistry | 2014

Structure Guided Design and Kinetic Analysis of Highly Potent Benzimidazole Inhibitors Targeting the PDEδ Prenyl Binding Site

Gunther Zimmermann; Carsten Schultz-Fademrecht; Philipp Küchler; Sandip Murarka; Shehab Ismail; Gemma Triola; Peter Nussbaumer; Alfred Wittinghofer; Herbert Waldmann

K-Ras is one of the most frequently mutated signal transducing human oncogenes. Ras signaling activity requires correct cellular localization of the GTPase. The spatial organization of K-Ras is controlled by the prenyl binding protein PDEδ, which enhances Ras diffusion in the cytosol. Inhibition of the Ras-PDEδ interaction by small molecules impairs Ras localization and signaling. Here we describe in detail the identification and structure guided development of Ras-PDEδ inhibitors targeting the farnesyl binding pocket of PDEδ with nanomolar affinity. We report kinetic data that characterize the binding of the most potent small molecule ligands to PDEδ and prove their binding to endogenous PDEδ in cell lysates. The PDEδ inhibitors provide promising starting points for the establishment of new drug discovery programs aimed at cancers harboring oncogenic K-Ras.


Journal of Medicinal Chemistry | 2009

Discovery of a Potent Class I Selective Ketone Histone Deacetylase Inhibitor with Antitumor Activity in Vivo and Optimized Pharmacokinetic Properties

Olaf Kinzel; Laura Llauger-Bufi; Giovanna Pescatore; Michael Rowley; Carsten Schultz-Fademrecht; Edith Monteagudo; Massimiliano Fonsi; Odalys Gonzalez Paz; Fabrizio Fiore; Christian Steinkühler; Philip Jones

The optimization of a potent, class I selective ketone HDAC inhibitor is shown. It possesses optimized pharmacokinetic properties in preclinical species, has a clean off-target profile, and is negative in a microbial mutagenicity (Ames) test. In a mouse xenograft model it shows efficacy comparable to that of vorinostat at a 10-fold reduced dose.


Bioorganic & Medicinal Chemistry Letters | 2009

Histone deacetylase inhibitors with a primary amide zinc binding group display antitumor activity in xenograft model.

Barbara Attenni; Jesus M. Ontoria; Jonathan C. Cruz; Michael Rowley; Carsten Schultz-Fademrecht; Christian Steinkühler; Philip Jones

Histone deacetylase (HDAC) inhibition causes hyperacetylation of histones leading to differentiation, growth arrest and apoptosis of malignant cells, representing a new strategy in cancer therapy. Many of the known HDAC inhibitors (HDACi) that are in clinical trials possess a hydroxamic acid, that is a strong Zn(2+) binding group, thereby inhibiting some of the class I and class II isoforms. Herein we describe the identification of a selective class I HDAC inhibitor bearing a primary carboxamide moiety as zinc binding group. This HDACi displays good antiproliferative activity against multiple cancer cell lines, and demonstrates efficacy in a xenograft model comparable to vorinostat.


Angewandte Chemie | 2016

Insight into the Inhibition of Drug-Resistant Mutants of the Receptor Tyrosine Kinase EGFR.

Julian Engel; Christian Becker; Jonas Lategahn; Marina Keul; Julia Ketzer; Thomas Mühlenberg; Laxmikanth Kollipara; Carsten Schultz-Fademrecht; René P. Zahedi; Sebastian Bauer; Daniel Rauh

Targeting acquired drug resistance represents the major challenge in the treatment of EGFR-driven non-small-cell lung cancer (NSCLC). Herein, we describe the structure-based design, synthesis, and biological evaluation of a novel class of covalent EGFR inhibitors that exhibit excellent inhibition of EGFR-mutant drug-resistant cells. Protein X-ray crystallography combined with detailed kinetic studies led to a deeper understanding of the mode of inhibition of EGFR-T790M and provided insight into the key principles for effective inhibition of the recently discovered tertiary mutation at EGFR-C797S.

Collaboration


Dive into the Carsten Schultz-Fademrecht's collaboration.

Top Co-Authors

Avatar

Philip Jones

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

Bert Klebl

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rita Scarpelli

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Axel Choidas

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge