Network


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

Hotspot


Dive into the research topics where Stephan Siegel is active.

Publication


Featured researches published by Stephan Siegel.


Epigenomics | 2015

Targeting BET bromodomains for cancer treatment

Marie Jung; Kathy A. Gelato; Amaury Ernesto Fernandez-Montalvan; Stephan Siegel; Bernard Haendler

The bromodomain and extraterminal (BET) subfamily of bromodomain-containing proteins has emerged in the last few years as an exciting, novel target group. BRD4, the best studied BET protein, is implicated in a number of hematological and solid tumors. This is linked to its role in modulating transcription elongation of essential genes involved in cell cycle and apoptosis such as c-Myc and BCL2. Potent BET inhibitors with promising antitumor efficacy in a number of preclinical cancer models have been identified in recent years. This led to clinical studies focusing mostly on the treatment of leukemia and lymphoma, and first encouraging signs of efficacy have already been reported. Here we discuss the biology of BRD4, its known interaction partners and implication in different tumor types. Further, we summarize the current knowledge on BET bromodomain inhibitors.


Antimicrobial Agents and Chemotherapy | 2004

New Class of Bacterial Phenylalanyl-tRNA Synthetase Inhibitors with High Potency and Broad-Spectrum Activity

Dieter Beyer; Hein-Peter Kroll; Rainer Endermann; Guido Schiffer; Stephan Siegel; Marcus Bauser; Jens Pohlmann; Michael Brands; Karl Ziegelbauer; Dieter Dr Haebich; Christine Eymann; Heike Brötz-Oesterhelt

ABSTRACT Phenylalanyl (Phe)-tRNA synthetase (Phe-RS) is an essential enzyme which catalyzes the transfer of phenylalanine to the Phe-specific transfer RNA (tRNAPhe), a key step in protein biosynthesis. Phenyl-thiazolylurea-sulfonamides were identified as a novel class of potent inhibitors of bacterial Phe-RS by high-throughput screening and chemical variation of the screening hit. The compounds inhibit Phe-RS of Escherichia coli, Haemophilus influenzae, Streptococcus pneumoniae, and Staphylococcus aureus, with 50% inhibitory concentrations in the nanomolar range. Enzyme kinetic measurements demonstrated that the compounds bind competitively with respect to the natural substrate Phe. All derivatives are highly selective for the bacterial Phe-RS versus the corresponding mammalian cytoplasmic and human mitochondrial enzymes. Phenyl-thiazolylurea-sulfonamides displayed good in vitro activity against Staphylococcus, Streptococcus, Haemophilus, and Moraxella strains, reaching MICs below 1 μg/ml. The antibacterial activity was partly antagonized by increasing concentrations of Phe in the culture broth in accordance with the competitive binding mode. Further evidence that inhibition of tRNAPhe charging is the antibacterial principle of this compound class was obtained by proteome analysis of Bacillus subtilis. Here, the phenyl-thiazolylurea-sulfonamides induced a protein pattern indicative of the stringent response. In addition, an E. coli strain carrying a relA mutation and defective in stringent response was more susceptible than its isogenic relA+ parent strain. In vivo efficacy was investigated in a murine S. aureus sepsis model and a S. pneumoniae sepsis model in rats. Treatment with the phenyl-thiazolylurea-sulfonamides reduced the bacterial titer in various organs by up to 3 log units, supporting the potential value of Phe-RS as a target in antibacterial therapy.


Journal of Medicinal Chemistry | 2017

Benzoisoquinolinediones as Potent and Selective Inhibitors of BRPF2 and TAF1/TAF1L Bromodomains

Léa Bouché; Clara D. Christ; Stephan Siegel; Amaury Ernesto Fernandez-Montalvan; Simon Holton; Oleg Fedorov; Antonius ter Laak; Tatsuo Sugawara; Detlef Stöckigt; Cynthia Tallant; Jim Bennett; Octovia P. Monteiro; Laura Díaz-Sáez; Paulina Siejka; Julia Meier; Vera Pütter; Jörg Weiske; Susanne Müller; Kilian Huber; Ingo V. Hartung; Bernard Haendler

Bromodomains (BD) are readers of lysine acetylation marks present in numerous proteins associated with chromatin. Here we describe a dual inhibitor of the bromodomain and PHD finger (BRPF) family member BRPF2 and the TATA box binding protein-associated factors TAF1 and TAF1L. These proteins are found in large chromatin complexes and play important roles in transcription regulation. The substituted benzoisoquinolinedione series was identified by high-throughput screening, and subsequent structure–activity relationship optimization allowed generation of low nanomolar BRPF2 BD inhibitors with strong selectivity against BRPF1 and BRPF3 BDs. In addition, a strong inhibition of TAF1/TAF1L BD2 was measured for most derivatives. The best compound of the series was BAY-299, which is a very potent, dual inhibitor with an IC50 of 67 nM for BRPF2 BD, 8 nM for TAF1 BD2, and 106 nM for TAF1L BD2. Importantly, no activity was measured for BRD4 BDs. Furthermore, cellular activity was evidenced using a BRPF2– or TAF1–histone H3.3 or H4 interaction assay.


British Journal of Haematology | 2017

Preclinical evaluation of the BET bromodomain inhibitor BAY 1238097 for the treatment of lymphoma.

Elena Bernasconi; Eugenio Gaudio; Pascale Lejeune; Chiara Tarantelli; Luciano Cascione; Ivo Kwee; Filippo Spriano; Andrea Rinaldi; Afua A. Mensah; Elaine Yee Lin Chung; Anastasios Stathis; Stephan Siegel; Norbert Schmees; Matthias Ocker; Emanuele Zucca; Bernard Haendler; Francesco Bertoni

The epigenome is often deregulated in cancer and treatment with inhibitors of bromodomain and extra‐terminal proteins, the readers of epigenetic acetylation marks, represents a novel therapeutic approach. Here, we have characterized the anti‐tumour activity of the novel bromodomain and extra‐terminal (BET) inhibitor BAY 1238097 in preclinical lymphoma models. BAY 1238097 showed anti‐proliferative activity in a large panel of lymphoma‐derived cell lines, with a median 50% inhibitory concentration between 70 and 208 nmol/l. The compound showed strong anti‐tumour efficacy in vivo as a single agent in two diffuse large B cell lymphoma models. Gene expression profiling showed BAY 1238097 targeted the NFKB/TLR/JAK/STAT signalling pathways, MYC and E2F1‐regulated genes, cell cycle regulation and chromatin structure. The gene expression profiling signatures also highly overlapped with the signatures obtained with other BET Bromodomain inhibitors and partially overlapped with HDAC‐inhibitors, mTOR inhibitors and demethylating agents. Notably, BAY 1238097 presented in vitro synergism with EZH2, mTOR and BTK inhibitors. In conclusion, the BET inhibitor BAY 1238097 presented promising anti‐lymphoma preclinical activity in vitro and in vivo, mediated by the interference with biological processes driving the lymphoma cells. Our data also indicate the use of combination schemes targeting EZH2, mTOR and BTK alongside BET bromodomains.


Oncotarget | 2018

BET inhibition is an effective approach against KRAS-driven PDAC and NSCLC

Toni Jauset; Daniel Massó-Vallés; Sandra Martínez-Martín; Marie-Eve Beaulieu; Laia Foradada; Francesco Paolo Fiorentino; Jun Yokota; Bernard Haendler; Stephan Siegel; Jonathan R. Whitfield; Laura Soucek

Effectively treating KRAS-driven tumors remains an unsolved challenge. The inhibition of downstream signaling effectors is a way of overcoming the issue of direct targeting of mutant KRAS, which has shown limited efficacy so far. Bromodomain and Extra-Terminal (BET) protein inhibition has displayed anti-tumor activity in a wide range of cancers, including KRAS-driven malignancies. Here, we preclinically evaluate the effect of BET inhibition making use of a new BET inhibitor, BAY 1238097, against Pancreatic Ductal Adenocarcinoma (PDAC) and Non-Small Cell Lung Cancer (NSCLC) models harboring RAS mutations both in vivo and in vitro. Our results demonstrate that BET inhibition displays significant therapeutic impact in genetic mouse models of KRAS-driven PDAC and NSCLC, reducing both tumor area and tumor grade. The same approach also causes a significant reduction in cell number of a panel of RAS-mutated human cancer cell lines (8 PDAC and 6 NSCLC). In this context, we demonstrate that while BET inhibition by BAY 1238097 decreases MYC expression in some cell lines, at least in PDAC cells its anti-tumorigenic effect is independent of MYC regulation. Together, these studies reinforce the use of BET inhibition and prompt the optimization of more efficient and less toxic BET inhibitors for the treatment of KRAS-driven malignancies, which are in urgent therapeutic need.


Cancer Research | 2018

Abstract 1646: Discovery and characterization of BAY-6035, a novel benzodiazepine-based SMYD3 inhibitor

Stefan Gradl; Holger Steuber; Jörg Weiske; Norbert Schmees; Stephan Siegel; Detlef Stoeckigt; Clara D. Christ; Fengling Li; Shawna Organ; Dalia Barsyte-Lovejoy; Magdalena M. Szewczyk; Steven Kennedy; Viacheslav Trush; Masoud Vedadi; C.H. Arrowsmith; Peter J. Brown; Manfred Husemann; Amaury Ernesto Fernández-Montalván; Volker Badock; Marcus Bauser; Andrea Haegebarth; Ingo Hartung; Carlo Stresemann

SMYD3 (SET and MYND domain-containing protein 3) is a protein lysine methyltransferase (PKMT) which was initially described as H3K4 methyltransferase involved in transcriptional regulation. SMYD3 has recently been reported to methylate and regulate several non-histone cancer relevant proteins such as mitogen-activated protein kinase kinase kinase 2 (MAP3K2), vascular endothelial growth factor receptor 1 (VEGFR1), and the human epidermal growth factor receptor 2 (HER2). In addition overexpression of SMYD3 has been linked to poor prognosis in certain cancers, thus supporting a possible oncogenic role for SMYD3 and making it an attractive target for anticancer drug development. Here we report the discovery of a novel potent and selective SMYD3 inhibitor series. We performed a thermal shift assay based (TSA) high throughput screening followed by extensive biophysical validation resulting in identification of a benzodiazepine-based SMYD3 inhibitor series. The co-crystallization structures revealed that this series binds to the substrate binding site and occupies the hydrophobic pocket for lysine binding using an unprecedented hydrogen bond pattern. The competitive behavior of the inhibitor in biochemical assays was consistent with the binding mode observed in the crystal structure. Further optimization generated BAY-6035, which showed improved nanomolar potency and was selective against kinases and other PKMTs. Furthermore, BAY-6035 specifically inhibited methylation of MAP3K2 by SMYD3 in a cellular assay with similar potency. In summary, BAY-6035 is a novel selective and potent SMYD3 inhibitor probe and will foster the exploration of the biologic role of SMYD3 in diseased and non-diseased tissues. Citation Format: Stefan Gradl, Holger Steuber, Jorg Weiske, Norbert Schmees, Stephan Siegel, Detlef Stoeckigt, Clara D. Christ, Fengling Li, Shawna Organ, Dalia Barsyte-Lovejoy, Magdalena M. Szewczyk, Steven Kennedy, Viacheslav Trush, Masoud Vedadi, Cheryl H. Arrowsmith, Peter J. Brown, Manfred Husemann, Amaury E. Fernandez-Montalvan, Volker Badock, Marcus Bauser, Andrea Haegebarth, Ingo V. Hartung, Carlo Stresemann. Discovery and characterization of BAY-6035, a novel benzodiazepine-based SMYD3 inhibitor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1646.


Cancer Research | 2017

Abstract 5239: Probing the cancer epigenome: empowering target validation by open innovation

Ingo V. Hartung; C.H. Arrowsmith; Volker Badock; Naomi Barak; Markus Berger; Peter J. Brown; Clara D. Christ; Erik Eggert; Ursula Egner; Oleg Fedorov; Amaury Ernesto Fernandez-Montalvan; Matyas Gorjanacz; Andrea Haegebarth; Bernard Haendler; Roman Hillig; Simon Holton; Kilian Huber; Seong Joo Koo; Antonius ter Laak; Susanne Mueller; Anke Mueller-Fahrnow; Cora Scholten; Stephan Siegel; Timo Stellfeld; Detlef Stoeckigt; Carlo Stresemann; Masoud Vedadi; Joerg Weiske; Hilmar Weinmann

Low reproducibility of published target validation studies as well as the frequent failure of genetic knock-down effects to phenocopy those of small molecule inhibitors have been recognized as road blocks for cancer drug discovery. Academic and industrial institutions have started to address these issues by providing access to high quality small molecular probes for novel targets of interest. Here we discuss probe discovery challenges and quality criteria based on the generation of three novel inhibitors for epigenetic targets. ATAD2 (ATPase family AAA-domain containing protein 2) is an epigenetic regulator that binds to chromatin through its bromodomain (BD). ATAD2 has been proposed to act as a co-factor for oncogenic transcription factors such as ERα and Myc. A more thorough validation of ATAD2 as a therapeutic target has been hampered by the lack of appropriate ATAD2 inhibitors. Here we disclose a structurally unprecedented series of ATAD2 BD inhibitors identified from a DNA-encoded library screen. Optimization delivered BAY-850, a highly potent and exceptionally selective ATAD2 BD inhibitor, which fully recapitulates effects seen by genetic mutagenesis studies in a cellular assay. The three BD and PHD-finger (BRPF) family members are found in histone acetyltransferase complexes. Whereas bromodomain inhibitors with dual activity against BRPF1 and 2 have been described before, we now disclose BAY-299, the first nanomolar inhibitor of the BRPF2 BD with high selectivity against its paralogs. Isoform selectivity was confirmed in cellular protein-protein interaction assays and rationalized based on X-Ray structures. BAY-598, a highly selective, cellularly active and orally bioavailable inhibitor of the protein lysine methyl transferase SMYD2, had been disclosed previously (Stresemann et al., AACR 2015). Development of BAY-598 allowed the identification of new methylation targets of SMYD2 as well as a proposed role of SMYD2 in pancreatic cancer. These results support further development of small molecule inhibitors as research tools to probe the functional role of novel epigenetic targets and underscore the power of open innovation for advancing our understanding of cancer target biology. Citation Format: Ingo V. Hartung, Cheryl Arrowsmith, Volker Badock, Naomi Barak, Markus Berger, Peter J. Brown, Clara D. Christ, Erik Eggert, Ursula Egner, Oleg Fedorov, Amaury E. Fernandez-Montalvan, Matyas Gorjanacz, Andrea Haegebarth, Bernard Haendler, Roman C. Hillig, Simon H. Holton, Kilian V. Huber, Seong J. Koo, Antonius ter Laak, Susanne Mueller, Anke Mueller-Fahrnow, Cora Scholten, Stephan Siegel, Timo Stellfeld, Detlef Stoeckigt, Carlo Stresemann, Masoud Vedadi, Joerg Weiske, Hilmar Weinmann. Probing the cancer epigenome: empowering target validation by open innovation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5239. doi:10.1158/1538-7445.AM2017-5239


Cancer Research | 2014

Abstract 4749: New benzazepine BET-inhibitors with improved oral bioavailability

Norbert Schmees; Bernard Haendler; Pascale Lejeune; Antje Stresemann; Roland Neuhaus; Stephan Siegel; Amaury Ernesto Fernandez-Montalvan; Hilmar Weinmann; Volker Gekeler

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Bromodomain protein 4 (BRD4), a member of the bromodomain and extra-terminal domain (BET) protein family, binds to acetylated histone tails via its two bromodomains BD1 and BD2. It forms a complex with the positive transcription elongation factor b which controls phosphorylation of RNA polymerase II, ultimately leading to stimulation of transcription elongation. An essential role of BRD4 in cell proliferation and cancer growth has been reported in several recent studies. The benzodiazepine JQ-1 is a strong antagonist of the binding of BRD4 to acetylated histone tails and has been used to elucidate the functions of the BET protein family members. JQ-1 is active in vivo in several therapeutic models but reveals a poor pharmacokinetic profile with high clearance and low oral bioavailability in animal studies. We investigated the benzodiazepine core of JQ-1 with the aim of optimization of oral bioavailability. Several possible core variations were identified that kept overall cellular activity but increased metabolic stability. The benzazepine BAY6356 was selected as a potent BET inhibitor with an improved overall pharmacokinetic profile and oral bioavailability between 60 and 100% in mouse, rat and dog. The strong antiproliferative activity observed in vitro in acute myeloid leukemia (AML) and multiple myeloma (MM) cell lines was confirmed in vivo in the MOLM-13 (AML) and MOLP-8 (MM) tumor models implanted in SCID mice. Daily oral treatment at the Maximal Tolerated Dose of 30 mg/kg led to strong tumor reduction in MOLM-13 (17% T/C on day 13 post tumor implantation) and in MOLP-8 (4% T/C on day 19 post tumor implantation) xenografts (T/C≤ 40% = active, T/C≤10% = highly active, according to NCI criteria). In the MOLM-13 model, c-Myc down-regulation was shown in vivo. Furthermore, for a same total dose, intermittent dosing every other day or twice a week in the MOLP-8 model proved to be as active as daily dosing, demonstrating exposure-driven efficacy. These favorable preclinical data support the evaluation of BAY6356 for further development. Citation Format: Norbert Schmees, Bernard Haendler, Pascale Lejeune, Antje Stresemann, Roland Neuhaus, Stephan Siegel, Amaury Ernesto Fernandez-Montalvan, Hilmar Weinmann, Volker Gekeler. New benzazepine BET-inhibitors with improved oral bioavailability. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4749. doi:10.1158/1538-7445.AM2014-4749


Archive | 2008

Substituted imidazopyrimidines and triazolopyrimidines

Stephan Siegel; Andreas Wilmen; Susanne Röhrig; Niels Svenstrup; Mark Jean Gnoth; Stefan Heitmeier; Ulrich Rester; Dmitry Zubov; Jochen Strayle; Michael Sperzel


Archive | 2012

6H-THIENO[3,2-f][1,2,4]TRIAZOLO[4,3-a][1,4]DIAZEPINE

Norbert Schmees; Joachim Kuhnke; Bernard Haendler; Philip Lienau; Amaury Ernesto Fernandez-Montalvan; Pascale Lejeune; Stephan Siegel; William J. Scott

Collaboration


Dive into the Stephan Siegel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan Heitmeier

Bayer HealthCare Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge