Network


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

Hotspot


Dive into the research topics where Andrea Vaupel is active.

Publication


Featured researches published by Andrea Vaupel.


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery of a novel class of highly potent inhibitors of the p53-MDM2 interaction by structure-based design starting from a conformational argument.

Pascal Furet; Keiichi Masuya; Joerg Kallen; Thérèse Stachyra-Valat; Stephan Ruetz; Vito Guagnano; Philipp Holzer; Robert Mah; Stefan Stutz; Andrea Vaupel; Patrick Chène; Sébastien Jeay; Achim Schlapbach

The p53-MDM2 interaction is an anticancer drug target under investigation in the clinic. Our compound NVP-CGM097 is one of the small molecule inhibitors of this protein-protein interaction currently evaluated in cancer patients. As part of our effort to identify new classes of p53-MDM2 inhibitors that could lead to additional clinical candidates, we report here the design of highly potent inhibitors having a pyrazolopyrrolidinone core structure. The conception of these new inhibitors originated in a consideration on the MDM2 bound conformation of the dihydroisoquinolinone class of inhibitors to which NVP-CGM097 belongs. This work forms the foundation of the discovery of HDM201, a second generation p53-MDM2 inhibitor that recently entered phase I clinical trial.


Bioorganic & Medicinal Chemistry Letters | 2014

Tetra-substituted imidazoles as a new class of inhibitors of the p53–MDM2 interaction

Andrea Vaupel; Guido Bold; Alain De Pover; Thérèse Stachyra-Valat; Joanna Lisztwan; Joerg Kallen; Keiichi Masuya; Pascal Furet

Capitalizing on crystal structure information obtained from a previous effort in the search for non peptide inhibitors of the p53-MDM2 interaction, we have discovered another new class of compounds able to disrupt this protein-protein interaction, an important target in oncology drug research. The new inhibitors, based on a tetra-substituted imidazole scaffold, have been optimized to low nanomolar potency in a biochemical assay following a structure-guided approach. An appropriate strategy has allowed us to translate the high biochemical potency in significant anti-proliferative activity on a p53-dependent MDM2 amplified cell line.


Tetrahedron-asymmetry | 2000

A practical synthesis of protected β-homolysine

Markus Baenziger; Luca Gobbi; Bernard Riss; Frank Schaefer; Andrea Vaupel

Abstract Protected β-homolysine of high enantiomeric purity (ee>99.5%) is prepared utilizing the stereoselective conjugate addition of lithiated ( S )-(α-methylbenzyl)benzylamide to ( E )-7-(tosyloxy)hept-2-enoic acid tert -butyl ester, followed by subsequent ammonia substitution, Boc protection and removal of the auxiliary.


ACS Medicinal Chemistry Letters | 2017

Discovery of Potent, Selective, and Structurally Novel Dot1L Inhibitors by a Fragment Linking Approach

Henrik Möbitz; Rainer Machauer; Philipp Holzer; Andrea Vaupel; Frédéric Stauffer; Christian Ragot; Giorgio Caravatti; Clemens Scheufler; César Fernández; Ulrich Hommel; Ralph Tiedt; Kim S. Beyer; Chao Chen; Hugh Zhu; Christoph Gaul

Misdirected catalytic activity of histone methyltransferase Dot1L is believed to be causative for a subset of highly aggressive acute leukemias. Targeting the catalytic domain of Dot1L represents a potential therapeutic approach for these leukemias. In the context of a comprehensive Dot1L hit finding strategy, a knowledge-based virtual screen of the Dot1L SAM binding pocket led to the discovery of 2, a non-nucleoside fragment mimicking key interactions of SAM bound to Dot1L. Fragment linking of 2 and 3, an induced back pocket binder identified in earlier studies, followed by careful ligand optimization led to the identification of 7, a highly potent, selective and structurally novel Dot1L inhibitor.


Bioorganic & Medicinal Chemistry Letters | 2016

Optimisation of a 5-[3-phenyl-(2-cyclic-ether)-methyl-ether]-4-aminopyrrolopyrimidine series of IGF-1R inhibitors

Robin Alec Fairhurst; Thomas H. Marsilje; Stefan Stutz; Andreas Boos; Michel Niklaus; Bei Chen; Songchun Jiang; Wenshuo Lu; Pascal Furet; Clive Mccarthy; Frédéric Stauffer; Vito Guagnano; Andrea Vaupel; Pierre-Yves Michellys; Christian Schnell; Sébastien Jeay

Taking the pyrrolopyrimidine derived IGF-1R inhibitor NVP-AEW541 as the starting point, the benzyl ether back-pocket binding moiety was replaced with a series of 2-cyclic ether methyl ethers leading to the identification of novel achiral [2.2.1]-bicyclic ether methyl ether containing analogues with improved IGF-1R activities and kinase selectivities. Further exploration of the series, including a fluorine scan of the 5-phenyl substituent, and optimisation of the sugar-pocket binding moiety identified compound 33 containing (S)-2-tetrahydrofuran methyl ether 6-fluorophenyl ether back-pocket, and cis-N-Ac-Pip sugar-pocket binding groups. Compound 33 showed improved selectivity and pharmacokinetics compared to NVP-AEW541, and produced comparable in vivo efficacy to linsitinib in inhibiting the growth of an IGF-1R dependent tumour xenograft model in the mouse.


Journal of Medicinal Chemistry | 2017

Development of Selective, Orally Active GPR4 Antagonists with Modulatory Effects on Nociception, Inflammation, and Angiogenesis

Juraj Velcicky; Wolfgang Miltz; Berndt Oberhauser; David Orain; Andrea Vaupel; Klaus Weigand; Janet Dawson King; Amanda Littlewood-Evans; Mark S. Nash; Roland Feifel; Pius Loetscher

A novel, selective, and efficacious GPR4 antagonist 13 was developed starting from lead compound 1a. While compound 1a showed promising efficacy in several disease models, its binding to a H3 receptor as well as a hERG channel prevented it from further development. Therefore, a new round of optimization addressing the key liabilities was performed and led to discovery of compound 13 with an improved profile. Compound 13 showed significant efficacy in the rat antigen induced arthritis as well as in the hyperalgesia and angiogenesis model at a well-tolerated dose of 30 mg/kg.


Cancer Research | 2016

Abstract 1239: NVP-HDM201: Biochemical and biophysical profile of a novel highly potent and selective PPI inhibitor of p53-Mdm2

Thérèse Stachyra-Valat; Frédéric Baysang; Anne-Cécile D’Alessandro; Erdmann Dirk; Pascal Furet; Vito Guagnano; Joerg Kallen; Lukas Leder; Robert Mah; Keiichi Masuya; Stefan Stutz; Andrea Vaupel; Francesco Hofmann; Patrick Chène; Sébastien Jeay; Philipp Holzer

An effective strategy to restore p53 activity in cancer cells containing wild type p53 is to inhibit the Mdm2-p53 protein-protein interaction (PPI). NVP-HDM201 is a novel PPI inhibitor currently under evaluation in a Phase I clinical trial. It binds to the p53 binding-site of the Mdm2 protein, disrupting the interaction of the two proteins and leading to the activation of the p53 pathway. NVP-HDM201 belongs to a novel chemical series with a distinct biophysical and biochemical profile. Affinity constant of NVP-HDM201 for Mdm2 is in the picomolar range, with a selectivity ratio greater than a 10000-fold vs. Mdm4. Analysis of its binding mode provides evidence for a distinct set of critical interactions between the small molecule and its target, as compared with our other Mdm2 inhibitor NVP-CGM097, and explains as to why NVP-HDM201 binds equally to human, mouse, rat and dog Mdm2. Characterization of its binding kinetics indicates that the optimized interactions of NVP-HDM201 with Mdm2 protein are responsible for the increased stabilization of the complex resulting in high potency against Mdm2. This feature, together with favorable physicochemical and drug-like properties, supported the selection of NVP-HDM201 for clinical development. Citation Format: Therese Stachyra-Valat, Frederic Baysang, Anne-Cecile D’Alessandro, Erdmann Dirk, Pascal Furet, Vito Guagnano, Joerg Kallen, Lukas Leder, Robert Mah, Keiichi Masuya, Stefan Stutz, Andrea Vaupel, Francesco Hofmann, Patrick Chene, Sebastien Jeay, Philipp Holzer. NVP-HDM201: Biochemical and biophysical profile of a novel highly potent and selective PPI inhibitor of p53-Mdm2. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1239.


Cancer Research | 2016

Abstract 4855: Discovery of NVP-HDM201 - First disclosure of a Next-Generation Mdm2 inhibitor with superior characteristics

Philipp Holzer; Patrick Chène; Stephane Ferretti; Pascal Furet; Tobias Gabriel; Bjoern Gruenenfelder; Vito Guagnano; Francesco Hofmann; Joerg Kallen; Robert Mah; Keiichi Masuya; Rita Ramos; Stephan Ruetz; Caroline Rynn; Thérèse Stachyra-Valat; Stefan Stutz; Andrea Vaupel; Sébastien Jeay

Activation of p53 by blocking the p53-Mdm2 interaction using non-peptidic small-molecule inhibitors has been pursued for many years as a promising cancer therapeutic strategy. We disclose the identity of NVP-HDM201, a novel, highly optimized and selective inhibitor of the p53-Mdm2 interaction. NVP-HDM201 binds to human Mdm2 protein with a sub-nanomolar Ki value, activates p53 and induces robust p53-dependent cell cycle arrest and apoptosis in human p53 wild-type tumor cells. The activity and selectivity of NVP-HDM201 have been tested and confirmed across a panel of cancer cell lines and the molecule displays desirable pharmacokinetic and pharmacodynamic profiles in animals together with excellent oral bioavailability. Application of NVP-HDM201 using various dosing schedules triggers rapid and sustained activation of p53-dependent pharmacodynamic biomarkers resulting in tumor regression in multiple xenografted models of p53 wild-type human cancers. We report here how a promising lead series was discovered and how innovative medicinal chemistry efforts led to further optimization of the potency and physico-chemical properties, culminating in the discovery of NVP-HDM201. The superior characteristics of the compound allowed the fast progression of the compound into the clinic where NVP-HDM201 is currently in Phase 1 clinical trials both as a single agent and as a combination partner in patients pre-selected for p53 wild-type tumors. Citation Format: Philipp Holzer, Patrick Chene, Stephane Ferretti, Pascal Furet, Tobias Gabriel, Bjoern Gruenenfelder, Vito Guagnano, Francesco Hofmann, Joerg Kallen, Robert Mah, Keiichi Masuya, Rita Ramos, Stephan Ruetz, Caroline Rynn, Therese Stachyra-Valat, Stefan Stutz, Andrea Vaupel, Sebastien Jeay. Discovery of NVP-HDM201 - First disclosure of a Next-Generation Mdm2 inhibitor with superior characteristics. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4855.


Cancer Research | 2016

Abstract 1225: NVP-HDM201: cellular and in vivo profile of a novel highly potent and selective PPI inhibitor of p53-Mdm2

Sébastien Jeay; Patrick Chène; Stephane Ferretti; Pascal Furet; Bjoern Gruenenfelder; Vito Guagnano; Nelson Guerreiro; Ensar Halilovic; Francesco Hofmann; Joerg Kallen; Michelle Léonard; Robert Mah; Keiichi Masuya; Rita Ramos; Caroline Rynn; Stephan Ruetz; Thérèse Stachyra-Valat; Stefan Stutz; Andrea Vaupel; Jens Wuerthner; Philipp Holzer

Stabilization of p53 protein by preventing its interaction with the negative regulator Mdm2 leads to selective induction of the p53 pathway, thus offering a promising cancer therapeutic strategy in p53 wild-type tumors. In the present study, we show the identification of NVP-HDM201, a novel, highly optimized, and selective inhibitor of the p53-Mdm2 interaction. NVP-HDM201 activates p53 in human cells and induces robust p53-dependent cell cycle arrest and apoptosis, selectively in p53 wild-type tumor cells. Its activity and selectivity has been tested and confirmed across a large panel of cancer cell lines from the Cancer Cell Line Encyclopedia. In vivo, NVP-HDM201 shows a dose-proportional pharmacokinetic (PK) profile and a clear PK/PD relationship, resulting in tumor growth inhibition and regression in SJSA-1 tumor-bearing rats at well-tolerated oral (p.o.) doses. The validation and understanding of its mechanism of action, the overall favorable drug-like properties and the characterization of its on-target toxicological profile in preclinical species strongly supported the initiation of Phase I clinical trials with NVP-HDM201 in pre-selected patients with p53 wild-type tumors. Citation Format: Sebastien Jeay, Patrick Chene, Stephane Ferretti, Pascal Furet, Bjoern Gruenenfelder, Vito Guagnano, Nelson Guerreiro, Ensar Halilovic, Francesco Hofmann, Joerg Kallen, Michelle Leonard, Robert Mah, Keiichi Masuya, Rita Ramos, Caroline Rynn, Stephan Ruetz, Therese Stachyra-Valat, Stefan Stutz, Andrea Vaupel, Jens Wuerthner, Philipp Holzer. NVP-HDM201: cellular and in vivo profile of a novel highly potent and selective PPI inhibitor of p53-Mdm2. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1225.


Archive | 2003

Diaryl urea derivatives in the treatment of protein kinase dependent diseases

Guido Bold; Giorgio Caravatti; Andreas Floersheimer; Vito Guagnano; Patricia Imbach; Keiichi Masuya; Johannes Roesel; Andrea Vaupel; Carlos Garcia-Echeverria

Collaboration


Dive into the Andrea Vaupel's collaboration.

Researchain Logo
Decentralizing Knowledge