Network


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

Hotspot


Dive into the research topics where Stephan Ruetz is active.

Publication


Featured researches published by Stephan Ruetz.


Cancer Cell | 2004

In vivo antitumor activity of NVP-AEW541—A novel, potent, and selective inhibitor of the IGF-IR kinase

Carlos Garcia-Echeverria; Mark Pearson; Andreas Marti; Thomas Meyer; Juergen Mestan; Johann Zimmermann; Jiaping Gao; Josef Brueggen; Hans-Georg Capraro; Robert Cozens; Dean B. Evans; Doriano Fabbro; Pascal Furet; Diana Graus Porta; Janis Liebetanz; Georg Martiny-Baron; Stephan Ruetz; Francesco Hofmann

IGF-IR-mediated signaling promotes survival, anchorage-independent growth, and oncogenic transformation, as well as tumor growth and metastasis formation in vivo. NVP-AEW541 is a pyrrolo[2,3-d]pyrimidine derivative small molecular weight kinase inhibitor of the IGF-IR, capable of distinguishing between the IGF-IR (IC50 = 0.086 microM) and the closely related InsR (IC50 = 2.3 microM) in cells. As expected for a specific IGF-IR kinase inhibitor, NVP-AEW541 abrogates IGF-I-mediated survival and colony formation in soft agar at concentrations that are consistent with inhibition of IGF-IR autophosphorylation. In vivo, this orally bioavailable compound inhibits IGF-IR signaling in tumor xenografts and significantly reduces the growth of IGF-IR-driven fibrosarcomas. Thus, NVP-AEW541 represents a class of selective, small molecule IGF-IR kinase inhibitors with proven in vivo antitumor activity and potential therapeutic application.


Cell | 2005

The mTOR Inhibitor RAD001 Sensitizes Tumor Cells to DNA-Damaged Induced Apoptosis through Inhibition of p21 Translation

Iwan Beuvink; Anne Boulay; Stefano Fumagalli; Frederic Zilbermann; Stephan Ruetz; Terence O’Reilly; Francois Natt; Jonathan Hall; Heidi Lane; George Thomas

Although DNA damaging agents have revolutionized chemotherapy against solid tumors, a narrow therapeutic window combined with severe side effects has limited their broader use. Here we show that RAD001 (everolimus), a rapamycin derivative, dramatically enhances cisplatin-induced apoptosis in wild-type p53, but not mutant p53 tumor cells. The use of isogenic tumor cell lines expressing either wild-type mTOR cDNA or a mutant that does not bind RAD001 demonstrates that the effects of RAD001 are through inhibition of mTOR function. We further show that RAD001 sensitizes cells to cisplatin by inhibiting p53-induced p21 expression. Unexpectedly, this effect is attributed to a small but significant inhibition of p21 translation combined with its short half-life. These findings provide the molecular rationale for combining DNA damaging agents with RAD001, showing that a general effect on a major anabolic process may dramatically enhance the efficacy of an established drug protocol in the treatment of cancer patients with solid tumors.


Pharmacology & Therapeutics | 2002

Protein kinases as targets for anticancer agents: from inhibitors to useful drugs

Doriano Fabbro; Stephan Ruetz; Elisabeth Buchdunger; Sandra W. Cowan-Jacob; Gabriele Fendrich; Janis Liebetanz; Terence O'Reilly; Peter Traxler; Bhabatosh Chaudhuri; Heinz Fretz; Jürg Zimmermann; Thomas Meyer; Giorgio Caravatti; Pascal Furet; Paul W. Manley

Many components of mitogenic signaling pathways in normal and neoplastic cells have been identified, including the large family of protein kinases, which function as components of signal transduction pathways, playing a central role in diverse biological processes, such as control of cell growth, metabolism, differentiation, and apoptosis. The development of selective protein kinase inhibitors that can block or modulate diseases caused by abnormalities in these signaling pathways is widely considered a promising approach for drug development. Because of their deregulation in human cancers, protein kinases, such as Bcr-Abl, those in the epidermal growth factor-receptor (HER) family, the cell cycle regulating kinases such as the cyclin-dependent kinases, as well as the vascular endothelial growth factor-receptor kinases involved in the neo-vascularization of tumors, are among the protein kinases considered as prime targets for the development of selective inhibitors. These drug-discovery efforts have generated inhibitors and low-molecular weight therapeutics directed against the ATP-binding site of various protein kinases that are in various stages of development (up to Phase II/III clinical trials). Three examples of inhibitors of protein kinases are reviewed, including low-molecular weight compounds targeting the cell cycle kinases; a potent and selective inhibitor of the HER1/HER2 receptor tyrosine kinase, the pyrollopyrimidine PKI166; and the 2-phenyl-aminopyrimidine STI571 (Glivec(R), Gleevec) a targeted drug therapy directed toward Bcr-Abl, the key player in chronic leukemia (CML). Some members of the HER family of receptor tyrosine kinases, in particular HER1 and HER2, have been found to be overexpressed in a variety of human tumors, suggesting that inhibition of HER signaling would be a viable antiproliferative strategy. The pyrrolo-pyrimidine PKI166 was developed as an HER1/HER2 inhibitor with potent in vitro antiproliferative and in vivo antitumor activity. Based upon its clear association with disease, the Bcr-Abl tyrosine kinase in CML represents the ideal target to validate the clinical utility of protein kinase inhibitors as therapeutic agents. In a preclinical model, STI571 (Glivec(R), Gleevec) showed potent in vitro and in vivo antitumor activity that was selective for Abl, c-Kit, and the platelet-derived growth factor-receptor. Phase I/II studies demonstrated that STI571 is well tolerated, and that it showed promising hematological and cytogenetic responses in CML and clinical responses in the c-Kit-driven gastrointestinal tumors.


Molecular Cancer Therapeutics | 2010

Potent and Selective Inhibition of Polycythemia by the Quinoxaline JAK2 Inhibitor NVP-BSK805

Fabienne Baffert; Catherine H. Regnier; Alain De Pover; Carole Pissot-Soldermann; Gisele A. Tavares; Francesca Blasco; Josef Brueggen; Patrick Chène; Peter Drueckes; Dirk Erdmann; Pascal Furet; Marc Gerspacher; Marc Lang; David Ledieu; Lynda Nolan; Stephan Ruetz; Joerg Trappe; Eric Vangrevelinghe; Markus Wartmann; Lorenza Wyder; Francesco Hofmann; Thomas Radimerski

The recent discovery of an acquired activating point mutation in JAK2, substituting valine at amino acid position 617 for phenylalanine, has greatly improved our understanding of the molecular mechanism underlying chronic myeloproliferative neoplasms. Strikingly, the JAK2V617F mutation is found in nearly all patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia and primary myelofibrosis. Thus, JAK2 represents a promising target for the treatment of myeloproliferative neoplasms and considerable efforts are ongoing to discover and develop inhibitors of the kinase. Here, we report potent inhibition of JAK2V617F and JAK2 wild-type enzymes by a novel substituted quinoxaline, NVP-BSK805, which acts in an ATP-competitive manner. Within the JAK family, NVP-BSK805 displays more than 20-fold selectivity towards JAK2 in vitro, as well as excellent selectivity in broader kinase profiling. The compound blunts constitutive STAT5 phosphorylation in JAK2V617F-bearing cells, with concomitant suppression of cell proliferation and induction of apoptosis. In vivo, NVP-BSK805 exhibited good oral bioavailability and a long half-life. The inhibitor was efficacious in suppressing leukemic cell spreading and splenomegaly in a Ba/F3 JAK2V617F cell-driven mouse mechanistic model. Furthermore, NVP-BSK805 potently suppressed recombinant human erythropoietin-induced polycythemia and extramedullary erythropoiesis in mice and rats. Mol Cancer Ther; 9(7); 1945–55. ©2010 AACR.


Journal of Medicinal Chemistry | 2015

Discovery of a Dihydroisoquinolinone Derivative (NVP-CGM097): A Highly Potent and Selective MDM2 Inhibitor Undergoing Phase 1 Clinical Trials in p53wt Tumors

Philipp Holzer; Keiichi Masuya; Pascal Furet; Joerg Kallen; Therese Valat-Stachyra; Stephane Ferretti; Joerg Berghausen; Michèle Bouisset-Leonard; Nicole Buschmann; Carole Pissot-Soldermann; Caroline Rynn; Stephan Ruetz; Stefan Stutz; Patrick Chène; Sébastien Jeay; François Gessier

As a result of our efforts to discover novel p53:MDM2 protein-protein interaction inhibitors useful for treating cancer, the potent and selective MDM2 inhibitor NVP-CGM097 (1) with an excellent in vivo profile was selected as a clinical candidate and is currently in phase 1 clinical development. This article provides an overview of the discovery of this new clinical p53:MDM2 inhibitor. The following aspects are addressed: mechanism of action, scientific rationale, binding mode, medicinal chemistry, pharmacokinetic and pharmacodynamic properties, and in vivo pharmacology/toxicology in preclinical species.


Molecular Cancer Therapeutics | 2010

Preclinical Antitumor Activity of the Orally Available Heat Shock Protein 90 Inhibitor NVP-BEP800

Andrew Massey; Joseph Schoepfer; Paul Brough; Josef Brueggen; Patrick Chène; Martin J. Drysdale; Ulrike Pfaar; Thomas Radimerski; Stephan Ruetz; Alain Schweitzer; Mike Wood; Carlos Garcia-Echeverria; Michael Rugaard Jensen

Heat shock protein 90 (Hsp90) is a ubiquitously expressed molecular chaperone with ATPase activity involved in the conformational maturation and stability of key signaling molecules involved in cell proliferation, survival, and transformation. Through its ability to modulate multiple pathways involved in oncogenesis, Hsp90 has generated considerable interest as a therapeutic target. NVP-BEP800 is a novel, fully synthetic, orally bioavailable inhibitor that binds to the NH2-terminal ATP-binding pocket of Hsp90. NVP-BEP800 showed activity against a panel of human tumor cell lines and primary human xenografts in vitro at nanomolar concentrations. In A375 melanoma and BT-474 breast cancer cell lines, NVP-BEP800 induced client protein degradation (including ErbB2, B-RafV600E, Raf-1, and Akt) and Hsp70 induction. Oral administration of NVP-BEP800 was well tolerated and induced robust antitumor responses in tumor xenograft models, including regression in the BT-474 breast cancer model. In these tumor models, NVP-BEP800 modulated Hsp90 client proteins and downstream signaling pathways at doses causing antitumor activity. NVP-BEP800 showed in vivo activity in a variety of dosing regimens covering daily to weekly schedules, potentially providing a high degree of flexibility in dose and schedule within the clinical setting. Overall, given the mechanism of action, preclinical activity profile, tolerability, and pharmaceutical properties, NVP-BEP800 is an exciting new oral Hsp90 inhibitor warranting further development. Mol Cancer Ther; 9(4); 906–19. ©2010 AACR.


eLife | 2015

A distinct p53 target gene set predicts for response to the selective p53–HDM2 inhibitor NVP-CGM097

Sébastien Jeay; Swann Gaulis; Stephane Ferretti; Hans Bitter; Moriko Ito; Thérèse Valat; Masato Murakami; Stephan Ruetz; Daniel Guthy; Caroline Rynn; Michael Rugaard Jensen; Marion Wiesmann; Joerg Kallen; Pascal Furet; François Gessier; Philipp Holzer; Keiichi Masuya; Jens Würthner; Ensar Halilovic; Francesco Hofmann; William R. Sellers; Diana Graus Porta

Biomarkers for patient selection are essential for the successful and rapid development of emerging targeted anti-cancer therapeutics. In this study, we report the discovery of a novel patient selection strategy for the p53–HDM2 inhibitor NVP-CGM097, currently under evaluation in clinical trials. By intersecting high-throughput cell line sensitivity data with genomic data, we have identified a gene expression signature consisting of 13 up-regulated genes that predicts for sensitivity to NVP-CGM097 in both cell lines and in patient-derived tumor xenograft models. Interestingly, these 13 genes are known p53 downstream target genes, suggesting that the identified gene signature reflects the presence of at least a partially activated p53 pathway in NVP-CGM097-sensitive tumors. Together, our findings provide evidence for the use of this newly identified predictive gene signature to refine the selection of patients with wild-type p53 tumors and increase the likelihood of response to treatment with p53–HDM2 inhibitors, such as NVP-CGM097. DOI: http://dx.doi.org/10.7554/eLife.06498.001


Current Medicinal Chemistry - Anti-cancer Agents | 2003

Chemical and Biological Profile of Dual Cdk1 and Cdk2 Inhibitors

Stephan Ruetz; Doriano Fabbro; Juerg Zimmermann; Thomas Meyer; Nathanael S. Gray

The importance of Cdks in cell cycle regulation, their interaction with oncogenes and tumor suppressors, and their frequent deregulation in human tumors, has encouraged an active search for agents capable of perturbing the function of Cdks. In our laboratories, a variety of selective and potent low molecular weight inhibitors directed against the ATP binding sites of the Cdk1, Cdk2 have been developed. Extensive biological profiling of two distinct classes of Cdk inhibitors - the phenylamino pyrimidines (PAPs) and trisubstituted purines has revealed distinct differences in their cellular effects in normal cells compared to tumor cells. Due to their intact G1/S checkpoints, normal cells are shown to be reversibly blocked by these Cdk inhibitors in either the G1/S-phase or at the G2/M boarder. In transformed cells these control points are either absent or defective and treatment with the compounds resulted in pronounced proliferation block at the G2/M transition. Furthermore, there is strong evidence that this G2/M arrest is less well tolerated by the cells and consequently, they undergo apoptotic cell death. Finally, these dual Cdk1/ Cdk2 inhibitors are also found to be significantly more active on proliferating cells compared to quiescent cells reflecting their specific activity. Despite these encouraging results demonstrating a distinct outcome after treatment with such dual Cdk inhibitors in normal compared to de-regulated tumor cells, it remains to be determined whether a comparable therapeutic window might be observed in vivo experiments. Furthermore the intracellular kinase selectivity of inhibitors which are putatively selective in vitro remains a complicating feature that is only recently begun to be addressed by affinity chromatography and phosphoproteomics techniques. Once efficacy can be demonstrated in animal models at well-tolerated doses, there will be strong evidence for the development of cell cycle antagonists for cancer therapy.


Clinical Cancer Research | 2010

Quantified tumor t1 is a generic early-response imaging biomarker for chemotherapy reflecting cell viability.

Paul M.J. McSheehy; Claudia Weidensteiner; Catherine Cannet; Stephane Ferretti; Didier Laurent; Stephan Ruetz; Michael Stumm; Peter R. Allegrini

Purpose: Identification of a generic response biomarker by comparison of chemotherapeutics with different action mechanisms on several noninvasive biomarkers in experimental tumor models. Experimental Design: The spin-lattice relaxation time of water protons (T1) was quantified using an inversion recovery-TrueFISP magnetic resonance imaging method in eight different experimental tumor models before and after treatment at several different time points with five different chemotherapeutics. Effects on T1 were compared with other minimally invasive biomarkers including vascular parameters, apparent diffusion coefficient, and interstitial fluid pressure, and were correlated with efficacy at the endpoint and histologic parameters. Results: In all cases, successful chemotherapy significantly lowered tumor T1 compared with vehicle and the fractional change in T1 (ΔT1) correlated with the eventual change in tumor size (range: r2 = 0.21, P < 0.05 to r2 = 0.73, P < 0.0001), except for models specifically resistant to that drug. In RIF-1 tumors, interstitial fluid pressure was decreased, but apparent diffusion coefficient and permeability increased in response to the microtubule stabilizer patupilone and 5-fluorouracil. Although ΔT1 was small (maximum of −20%), the variability was very low (5%) compared with other magnetic resonance imaging methods (24-48%). Analyses ex vivo showed unchanged necrosis, increased apoptosis, and decreased %Ki67 and total choline, but only Ki67 and choline correlated with ΔT1. Correlation of Ki67 and ΔT1 were observed in other models using patupilone, paclitaxel, a VEGF-R inhibitor, and the mammalian target of rapamycin inhibitor everolimus. Conclusions: These results suggest that a decrease in tumor T1 reflects hypocellularity and is a generic marker of response. The speed and robustness of the method should facilitate its use in clinical trials. Clin Cancer Res; 16(1); 212–25


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of dihydroisoquinolinone derivatives as novel inhibitors of the p53-MDM2 interaction with a distinct binding mode.

François Gessier; Joerg Kallen; Edgar Jacoby; Patrick Chène; Thérèse Stachyra-Valat; Stephan Ruetz; Sébastien Jeay; Philipp Holzer; Keiichi Masuya; Pascal Furet

Blocking the interaction between the p53 tumor suppressor and its regulatory protein MDM2 is a promising therapeutic concept under current investigation in oncology drug research. We report here the discovery of the first representatives of a new class of small molecule inhibitors of this protein-protein interaction: the dihydroisoquinolinones. Starting from an initial hit identified by virtual screening, a derivatization program has resulted in compound 11, a low nanomolar inhibitor of the p53-MDM2 interaction showing significant cellular activity. Initially based on a binding mode hypothesis, this effort was then guided by a X-ray co-crystal structure of MDM2 in complex with one of the synthesized analogs. The X-ray structure revealed an unprecedented binding mode for p53-MDM2 inhibitors.

Collaboration


Dive into the Stephan Ruetz's collaboration.

Researchain Logo
Decentralizing Knowledge