Network


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

Hotspot


Dive into the research topics where Wolfgang Schwede is active.

Publication


Featured researches published by Wolfgang Schwede.


Cancer Research | 2008

Improved Cellular Pharmacokinetics and Pharmacodynamics Underlie the Wide Anticancer Activity of Sagopilone

Jens Hoffmann; Ilio Vitale; Bernd Buchmann; Lorenzo Galluzzi; Wolfgang Schwede; Laura Senovilla; Werner Skuballa; Sonia Vivet; Rosemarie Lichtner; Jose Miguel Vicencio; Theocharis Panaretakis; Gerhard Siemeister; Hermann Lage; Lisa Nanty; Stefanie Hammer; Kevin Mittelstaedt; Sebastian Winsel; Julia Eschenbrenner; Maria Castedo; Carine Demarche; Ulrich Klar; Guido Kroemer

Sagopilone (ZK-EPO) is the first fully synthetic epothilone undergoing clinical trials for the treatment of human tumors. Here, we investigate the cellular pathways by which sagopilone blocks tumor cell proliferation and compare the intracellular pharmacokinetics and the in vivo pharmacodynamics of sagopilone with other microtubule-stabilizing (or tubulin-polymerizing) agents. Cellular uptake and fractionation/localization studies revealed that sagopilone enters cells more efficiently, associates more tightly with the cytoskeleton, and polymerizes tubulin more potently than paclitaxel. Moreover, in contrast to paclitaxel and other epothilones [such as the natural product epothilone B (patupilone) or its partially synthetic analogue ixabepilone], sagopilone is not a substrate of the P-glycoprotein efflux pumps. Microtubule stabilization by sagopilone caused mitotic arrest, followed by transient multinucleation and activation of the mitochondrial apoptotic pathway. Profiling of the proapoptotic signal transduction pathway induced by sagopilone with a panel of small interfering RNAs revealed that sagopilone acts similarly to paclitaxel. In HCT 116 colon carcinoma cells, sagopilone-induced apoptosis was partly antagonized by the knockdown of proapoptotic members of the Bcl-2 family, including Bax, Bak, and Puma, whereas knockdown of Bcl-2, Bcl-X(L), or Chk1 sensitized cells to sagopilone-induced cell death. Related to its improved subcellular pharmacokinetics, however, sagopilone is more cytotoxic than other epothilones in a large panel of human cancer cell lines in vitro and in vivo. In particular, sagopilone is highly effective in reducing the growth of paclitaxel-resistant cancer cells. These results underline the processes behind the therapeutic efficacy of sagopilone, which is now evaluated in a broad phase II program.


Cancer Research | 2017

Abstract 4989: 3D spheroid screen yields SCD1 pathway inhibitors for the treatment of cancer

Sylvia Gruenewald; Carolyn Sperl; Patrick Steigemann; Alexander Walter; Sylvia Zacharias; Uwe Eberspaecher; Roland Neuhaus; Ludwig Zorn; Wolfgang Schwede; Kai Thede; Sven Christian

With three-dimensional growth conditions, multicellular tumor spheroids reproduce several parameters of the tumor microenvironment, including oxygen and nutrient gradients, characteristic of poorly vascularized tumor regions. 3D high content screening (HCS) identified compounds that selectively kill tumor cells in the inner core of tumor cell spheroids by targeting the Stearoyl CoA Desaturase 1 (SCD1) pathway. SCD1 catalyzes the rate-limiting step in the production of mono-unsaturated fatty acids (MUFAs). Cancer cells are dependent on higher levels of MUFAs compared to normal cells and SCD1 is highly expressed in multiple tumor types. Changes in the MUFA / SFA (saturated fatty acid) ratio alters lipid biosynthesis and thus triggers cellular (ER) stress and induces the Unfolded Protein Response. Although the lead compound was very effective in vitro, it had unfavorable PK and physical chemistry properties, including low permeability and solubility and very high lipophilicity. This led to insufficient oral bioavailability, which could be overcome by optimization of PK and physical chemistry properties. Here, we report on the in vitro/in vivo effects of our 3D HCS compounds which showed high potency in the 3D spheroid inner core death assay with T47D breast cancer cells. In this in vitro model compound-induced inner core cell death is enhanced by SCD1 substrates palmitic or stearic acid and rescued by the SCD1 products palmitoleic or oleic acid. Furthermore, the effects can be reproduced in 2D cultures, which become increasingly sensitive to inhibition by our 3D HCS compounds with decreasing FBS concentration in the culture medium and this effect can also be rescued by addition of MUFAs but not of palmitic acid. Mode of action analysis showed that our compounds reduced palmitoleoyl- or oleoyl-CoA levels and simultaneously increased saturated fatty acyl-CoAs of palmitate or stearate in several cell lines as well as in vivo. In the sensitive T47D cells, the compounds induced expression of stress response genes and genes related to lipid metabolism. While these results support the SCD1 pathway as target for our 3D HCS compounds, we also observed striking differences to published SCD1 inhibitors suggesting a new cancer target beyond SCD1. Thus, further validation of our inhibitors in vitro and in vivo will be required, but these results suggest that 3D spheroid cultures may be a valuable tool for elucidation of new drug targets for cancer therapy. Citation Format: Sylvia Gruenewald, Carolyn Sperl, Patrick Steigemann, Alexander Walter, Sylvia Zacharias, Uwe Eberspaecher, Roland Neuhaus, Ludwig Zorn, Wolfgang Schwede, Kai Thede, Sven Christian. 3D spheroid screen yields SCD1 pathway inhibitors for the treatment of cancer [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 4989. doi:10.1158/1538-7445.AM2017-4989


Cancer Research | 2017

Abstract 3248: Identification and optimization of a highly active, cross reactive Complex-1 inhibitor

Jeffrey Mowat; Sven Christian; Carolyn Sperl; Alexander Ehrmann; Stephan Menz; Judith Guenther; Roman Hillig; Marcus Bauser; Andrea Haegebarth; Wolfgang Schwede

Mitochondria are key regulators of both energy supply and apoptotic cell death. The mitochondrial electron transport chain (ETC) consists of four enzyme complexes that transfer electrons from NADH to oxygen. During electron transfer, the ETC pumps protons into the inter-membrane space, generating a gradient across the inner mitochondrial membrane that is used by Complex V to drive ATP synthesis. Recent publications have shown that tumor cells harboring specific mutations (LKB1, mIDH and others) are more sensitive to Complex I inhibition, potentially providing an opportunity for selectively targeting tumor cells. Based on a high throughput screen (HTS), we identified new, albeit moderately active, lead structures with cross reactivity between mouse and human Complex 1. SAR elaboration of the lead structure allowed for optimization of the potency, although compounds still suffered from low metabolic stability. Further improvement of the in vitro and in vivo PK properties finally permitted in vivo animal studies. Herein, we report for the first time the preclinical profile and structure of a highly active, optimized, human/mouse cross-reactive Complex I inhibitor that allowed for the further investigation into the therapeutic potential of Complex I inhibition in cancer. Citation Format: Jeffrey Mowat, Sven Christian, Carolyn Sperl, Alexander Ehrmann, Stephan Menz, Judith Guenther, Roman Hillig, Marcus Bauser, Andrea Haegebarth, Wolfgang Schwede. Identification and optimization of a highly active, cross reactive Complex-1 inhibitor [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 3248. doi:10.1158/1538-7445.AM2017-3248


Archive | 1998

16-Halogen-epothilone derivatives, method for producing them and their pharmaceutical use

Ulrich Klar; Wolfgang Schwede; Werner Skuballa; Bernd Buchmann


Archive | 1999

Epothilon derivatives, their preparation process, intermediate products and their pharmaceutical use

Bernd Buchmann; Ulrich Klar; Werner Skuballa; Wolfgang Schwede; Michael Schirner; Andreas Menrad


Archive | 2003

Thiazolidinones and the use therof as polo-like kinase inhibitors

Wolfgang Schwede; Volker Schulze; Knut Eis; Bernd Buchmann; Hans Briem; Gerhard Siemeister; Ulf Boemer; Karsten Parczyk


Archive | 1998

ANTIGESTAGENIC STEROIDS WITH A FLUORINATED 17ALPHA-ALKYL CHAIN

Wolfgang Schwede; Arwed Cleve; Ulrich Klar; Günter Neef; Kristof Chwalisz; Martin Schneider; Ulrike Fuhrmann; Holger Hess-Stumpp


Archive | 2011

Progesterone receptor antagonists

Wolfgang Schwede; Ulrich Klar; Carsten Möller; Andrea Rotgeri; Wilhelm Bone; Christoph Huwe


Archive | 2001

Novel epothilone derivatives, method for the preparation thereof and their pharmaceutical use

Bernd Buchmann; Ulrich Klar; Werner Skuballa; Wolfgang Schwede; Rosemarie Lichtner; Jens Hoffmann


Synthesis | 2005

Efficient Chiral Pool Synthesis of the C1-C6 Fragment of Epothilones

Ulrich Klar; Bodo Röhr; Frank Kuczynski; Wolfgang Schwede; Markus Berger; Werner Skuballa; Bernd Buchmann

Collaboration


Dive into the Wolfgang Schwede's collaboration.

Researchain Logo
Decentralizing Knowledge