Network


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

Hotspot


Dive into the research topics where Jens Wuerthner is active.

Publication


Featured researches published by Jens Wuerthner.


Cancer Cell | 2016

The Public Repository of Xenografts Enables Discovery and Randomized Phase II-like Trials in Mice

Elizabeth Townsend; Mark A. Murakami; Alexandra N. Christodoulou; Amanda L. Christie; Johannes Köster; Tiffany DeSouza; Elizabeth A. Morgan; Scott P. Kallgren; Huiyun Liu; Shuo-Chieh Wu; Olivia Plana; Joan Montero; Kristen E. Stevenson; Prakash Rao; Raga Vadhi; Michael Andreeff; Philippe Armand; Karen K. Ballen; Patrizia Barzaghi-Rinaudo; Sarah Cahill; Rachael A. Clark; Vesselina G. Cooke; Matthew S. Davids; Daniel J. DeAngelo; David M. Dorfman; Hilary Eaton; Benjamin L. Ebert; Julia Etchin; Brant Firestone; David C. Fisher

More than 90% of drugs with preclinical activity fail in human trials, largely due to insufficient efficacy. We hypothesized that adequately powered trials of patient-derived xenografts (PDX) in mice could efficiently define therapeutic activity across heterogeneous tumors. To address this hypothesis, we established a large, publicly available repository of well-characterized leukemia and lymphoma PDXs that undergo orthotopic engraftment, called the Public Repository of Xenografts (PRoXe). PRoXe includes all de-identified information relevant to the primary specimens and the PDXs derived from them. Using this repository, we demonstrate that large studies of acute leukemia PDXs that mimic human randomized clinical trials can characterize drug efficacy and generate transcriptional, functional, and proteomic biomarkers in both treatment-naive and relapsed/refractory disease.


Cancer Cell | 2016

Erratum: The Public Repository of Xenografts Enables Discovery and Randomized Phase II-like Trials in Mice (Cancer Cell (2016) 29 (574–586))

Elizabeth Townsend; Mark A. Murakami; Alexandra N. Christodoulou; Amanda L. Christie; Johannes Köster; Tiffany DeSouza; Elizabeth A. Morgan; Scott P. Kallgren; Huiyun Liu; Shuo Chieh Wu; Olivia Plana; Joan Montero; Kristen E. Stevenson; Prakash Rao; Raga Vadhi; Michael Andreeff; Philippe Armand; Karen K. Ballen; Patrizia Barzaghi-Rinaudo; Sarah Cahill; Rachael A. Clark; Vesselina G. Cooke; Matthew S. Davids; Daniel J. DeAngelo; David M. Dorfman; Hilary Eaton; Benjamin L. Ebert; Julia Etchin; Brant Firestone; David C. Fisher

Elizabeth C. Townsend, Mark A. Murakami, Alexandra Christodoulou, Amanda L. Christie, Johannes Köster, Tiffany A. DeSouza, Elizabeth A. Morgan, Scott P. Kallgren, Huiyun Liu, Shuo-Chieh Wu, Olivia Plana, Joan Montero, Kristen E. Stevenson, Prakash Rao, Raga Vadhi, Michael Andreeff, Philippe Armand, Karen K. Ballen, Patrizia Barzaghi-Rinaudo, Sarah Cahill, Rachael A. Clark, Vesselina G. Cooke, Matthew S. Davids, Daniel J. DeAngelo, David M. Dorfman, Hilary Eaton, Benjamin L. Ebert, Julia Etchin, Brant Firestone, David C. Fisher, Arnold S. Freedman, Ilene A. Galinsky, Hui Gao, Jacqueline S. Garcia, Francine Garnache-Ottou, Timothy A. Graubert, Alejandro Gutierrez, Ensar Halilovic, Marian H. Harris, Zachary T. Herbert, Steven M. Horwitz, Giorgio Inghirami, Andrew M. Intlekofer, Moriko Ito, Shai Izraeli, Eric D. Jacobsen, Caron A. Jacobson, Sébastien Jeay, Irmela Jeremias, Michelle A. Kelliher, Raphael Koch, Marina Konopleva, Nadja Kopp, Steven M. Kornblau, Andrew L. Kung, Thomas S. Kupper, Nicole R. LeBoeuf, Ann S. LaCasce, Emma Lees, Loretta S. Li, A. Thomas Look, Masato Murakami, Markus Muschen, Donna Neuberg, Samuel Y. Ng, Oreofe O. Odejide, Stuart H. Orkin, Rachel R. Paquette, Andrew E. Place, Justine E. Roderick, Jeremy A. Ryan, Stephen E. Sallan, Brent Shoji, Lewis B. Silverman, Robert J. Soiffer, David P. Steensma, Kimberly Stegmaier, Richard M. Stone, Jerome Tamburini, Aaron R. Thorner, Paul van Hummelen, Martha Wadleigh, Marion Wiesmann, Andrew P. Weng, Jens U. Wuerthner, David A. Williams, Bruce M. Wollison, Andrew A. Lane, Anthony Letai, Monica M. Bertagnolli, Jerome Ritz, Myles Brown, Henry Long, Jon C. Aster, Margaret A. Shipp, James D. Griffin, and David M. Weinstock* *Correspondence: [email protected] http://dx.doi.org/10.1016/j.ccell.2016.06.008


Molecular Cancer Therapeutics | 2015

Inhibition of Wild-Type p53-Expressing AML by the Novel Small Molecule HDM2 Inhibitor CGM097

Ellen Weisberg; Ensar Halilovic; Vesselina G. Cooke; Atsushi Nonami; Tao Ren; Takaomi Sanda; Irene Simkin; Jing Yuan; Brandon Antonakos; Louise Barys; Moriko Ito; Richard Stone; Ilene Galinsky; Kristen Cowens; Erik Nelson; Martin Sattler; Sébastien Jeay; Jens Wuerthner; Sean McDonough; Marion Wiesmann; James D. Griffin

The tumor suppressor p53 is a key regulator of apoptosis and functions upstream in the apoptotic cascade by both indirectly and directly regulating Bcl-2 family proteins. In cells expressing wild-type (WT) p53, the HDM2 protein binds to p53 and blocks its activity. Inhibition of HDM2:p53 interaction activates p53 and causes apoptosis or cell-cycle arrest. Here, we investigated the ability of the novel HDM2 inhibitor CGM097 to potently and selectively kill WT p53-expressing AML cells. The antileukemic effects of CGM097 were studied using cell-based proliferation assays (human AML cell lines, primary AML patient cells, and normal bone marrow samples), apoptosis, and cell-cycle assays, ELISA, immunoblotting, and an AML patient–derived in vivo mouse model. CGM097 potently and selectively inhibited the proliferation of human AML cell lines and the majority of primary AML cells expressing WT p53, but not mutant p53, in a target-specific manner. Several patient samples that harbored mutant p53 were comparatively unresponsive to CGM097. Synergy was observed when CGM097 was combined with FLT3 inhibition against oncogenic FLT3-expressing cells cultured both in the absence as well as the presence of cytoprotective stromal-secreted cytokines, as well as when combined with MEK inhibition in cells with activated MAPK signaling. Finally, CGM097 was effective in reducing leukemia burden in vivo. These data suggest that CGM097 is a promising treatment for AML characterized as harboring WT p53 as a single agent, as well as in combination with other therapies targeting oncogene-activated pathways that drive AML. Mol Cancer Ther; 14(10); 2249–59. ©2015 AACR.


Cancer Research | 2016

High order drug combinations are required to effectively kill colorectal cancer cells

Thomas Horn; Stephane Ferretti; Nicolas Ebel; Angela Tam; Samuel Ho; Fred Harbinski; Ali Farsidjani; Matthew Zubrowski; William R. Sellers; Robert Schlegel; Dale Porter; Erick Morris; Jens Wuerthner; Sébastien Jeay; Joel Greshock; Ensar Halilovic; Levi A. Garraway; Giordano Caponigro; Joseph Lehar

Like classical chemotherapy regimens used to treat cancer, targeted therapies will also rely upon polypharmacology, but tools are still lacking to predict which combinations of molecularly targeted drugs may be most efficacious. In this study, we used image-based proliferation and apoptosis assays in colorectal cancer cell lines to systematically investigate the efficacy of combinations of two to six drugs that target critical oncogenic pathways. Drug pairs targeting key signaling pathways resulted in synergies across a broad spectrum of genetic backgrounds but often yielded only cytostatic responses. Enhanced cytotoxicity was observed when additional processes including apoptosis and cell cycle were targeted as part of the combination. In some cases, where cell lines were resistant to paired and tripled drugs, increased expression of antiapoptotic proteins was observed, requiring a fourth-order combination to induce cytotoxicity. Our results illustrate how high-order drug combinations are needed to kill drug-resistant cancer cells, and they also show how systematic drug combination screening together with a molecular understanding of drug responses may help define optimal cocktails to overcome aggressive cancers. Cancer Res; 76(23); 6950-63. ©2016 AACR.


eLife | 2017

Combined ALK and MDM2 inhibition increases antitumor activity and overcomes resistance in human ALK mutant neuroblastoma cell lines and xenograft models

Hui Qin Wang; Ensar Halilovic; Xiaoyan Li; Jinsheng Liang; Yichen Cao; Daniel Rakiec; David A. Ruddy; Sébastien Jeay; Jens Wuerthner; Noelito Timple; Shailaja Kasibhatla; Nanxin Li; Juliet Williams; William R. Sellers; Alan Huang; Fang Li

The efficacy of ALK inhibitors in patients with ALK-mutant neuroblastoma is limited, highlighting the need to improve their effectiveness in these patients. To this end, we sought to develop a combination strategy to enhance the antitumor activity of ALK inhibitor monotherapy in human neuroblastoma cell lines and xenograft models expressing activated ALK. Herein, we report that combined inhibition of ALK and MDM2 induced a complementary set of anti-proliferative and pro-apoptotic proteins. Consequently, this combination treatment synergistically inhibited proliferation of TP53 wild-type neuroblastoma cells harboring ALK amplification or mutations in vitro, and resulted in complete and durable responses in neuroblastoma xenografts derived from these cells. We further demonstrate that concurrent inhibition of MDM2 and ALK was able to overcome ceritinib resistance conferred by MYCN upregulation in vitro and in vivo. Together, combined inhibition of ALK and MDM2 may provide an effective treatment for TP53 wild-type neuroblastoma with ALK aberrations. DOI: http://dx.doi.org/10.7554/eLife.17137.001


Neuroendocrinology | 2018

The HDM2 (MDM2) Inhibitor NVP-CGM097 Inhibits Tumor Cell Proliferation and Shows Additive Effects with 5-Fluorouracil on the p53-p21-Rb-E2F1 Cascade in the p53wild type Neuroendocrine Tumor Cell Line GOT1

Clemens Reuther; Vera Heinzle; Svenja Nölting; Sabine Herterich; Stefanie Hahner; Ensar Halilovic; Sébastien Jeay; Jens Wuerthner; Elke Tatjana Aristizabal Prada; Gerald Spöttl; Julian Maurer; Christoph J. Auernhammer

Background/Aims: The tumor suppressor p53 is depleted in many tumor cells by the E3 ubiquitin ligase mouse double minute 2 homolog (MDM2) through MDM2/p53 interaction. A novel target for inhibiting p53 degradation and for causing reexpression of p53wild type is inhibition of MDM2. The small molecule NVP-CGM097 is a novel MDM2 inhibitor. We investigated MDM2 inhibition as a target in neuroendocrine tumor cells in vitro. Methods: Human neuroendocrine tumor cell lines from the pancreas (BON1), lung (NCI-H727), and midgut (GOT1) were incubated with the MDM2 inhibitor NVP-CGM097 (Novartis) at concentrations from 4 to 2,500 nM. Results: While p53wild type GOT1 cells were sensitive to NVP-CGM097, p53mutated BON1 and p53mutated NCI-H727 cells were resistant to NVP-CGM097. Incubation of GOT1 cells with NVP-CGM097 at 100, 500, and 2,500 nM for 96 h caused a significant decline in cell viability to 84.9 ± 9.2% (p < 0.05), 77.4 ± 6.6% (p < 0.01), and 47.7 ± 9.2% (p < 0.01). In a Western blot analysis of GOT1 cells, NVP-CGM097 caused a dose-dependent increase in the expression of p53 and p21 tumor suppressor proteins and a decrease in phospho-Rb and E2F1. Experiments of co-incubation of NVP-CGM097 with 5-fluorouracil, temozolomide, or everolimus each showed additive antiproliferative effects in GOT1 cells. NVP-CGM097 and 5-fluorouracil increased p53 and p21 expression in an additive manner. Conclusions: MDM2 inhibition seems a promising novel therapeutic target in neuroendocrine tumors harboring p53wild type. Further investigations should examine the potential role of MDM2 inhibitors in neuroendocrine tumor treatment.


Cancer Research | 2014

Abstract 5466: The Mdm2 inhibitor, NVP-CGM097, in combination with the BRAF inhibitor NVP-LGX818 elicits synergistic antitumor effects in melanoma

Hui Qin Wang; Matthew Zubrowski; Erling Emerson; Elina Pradhan; Sébastien Jeay; Marion Wiesmann; Giordano Caponigro; Jens Wuerthner; Robert Schlegel; Z. Alexander Cao; Alan Huang; Ensar Halilovic

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA RAF kinase inhibitors have shown substantial therapeutic effects in patients with BRAF-mutant melanoma. However, despite impressive initial responses, therapeutic effects are often temporary due to acquired resistance. This resistance highlights the need to identify therapeutic approaches to improve the durability of responses to RAF inhibitors. In this study, we demonstrate that a novel inhibitor of p53-Mdm2 interaction, NVP-CGM097, causes marked reduction of melanoma cell viability. The activation of p53 signaling and consequent reduction in cell viability required wild-type p53, but was independent of BRAF status. Moreover, combined inhibition of Mdm2, using NVP-CGM097, and BRAF using NVP-LGX818, synergistically inhibited the viability of BRAF mutant melanoma cells in vitro and tumor growth in vivo. NVP-CGM097 caused induction of p21 and Bax, while NVP-LGX818 did not. Instead, NVP-LGX818 caused induction of p27 and Bim. Therefore, together they induced a complementary set of anti-proliferative and apoptosis stimulating molecules, which resulted in strong synergistic antitumor effects. In vivo, the combination treatment resulted in sustained tumor regressions and markedly prolonged survival relative to either single agent. Collectively, these data show that activation of p53, through inhibition of Mdm2, leads to antitumor effects in melanoma. Furthermore, the data show that concomitant activation of tumor suppressor p53 and inhibition of BRAF synergistically suppresses melanoma growth. Therefore, combined inhibition of Mdm2 and BRAF in melanoma may provide an effective therapeutic modality capable of overcoming the resistance observed with the BRAF inhibitor monotherapy and thus lead to more durable responses in the clinic. Citation Format: Hui Qin Wang, Matthew Zubrowski, Erling Emerson, Elina Pradhan, Sebastien Jeay, Marion Wiesmann, Giordano Caponigro, Jens Wuerthner, Robert Schlegel, Z. Alexander Cao, Alan Huang, Ensar Halilovic. The Mdm2 inhibitor, NVP-CGM097, in combination with the BRAF inhibitor NVP-LGX818 elicits synergistic antitumor effects in melanoma. [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 5466. doi:10.1158/1538-7445.AM2014-5466


Cancer Research | 2018

Dose and schedule determine distinct molecular mechanisms underlying the efficacy of the p53-MDM2 inhibitor HDM201.

Sébastien Jeay; Stephane Ferretti; Philipp Holzer; Jeannette Fuchs; Emilie Chapeau; Markus Wartmann; Dario Sterker; Vincent Romanet; Masato Murakami; Grainne Kerr; Eric Y. Durand; Swann Gaulis; Marta Cortes-Cros; Stephan Ruetz; therese-marie stachyra; Joerg Kallen; Pascal Furet; Jens Wuerthner; Nelson Guerreiro; Ensar Halilovic; Astrid Jullion; Audrey Kauffmann; Emil T. Kuriakose; Marion Wiesmann; Michael Rugaard Jensen; Francesco Hofmann; William R. Sellers

Activation of p53 by inhibitors of the p53-MDM2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. Here, we report distinct mechanisms by which the novel, potent, and selective inhibitor of the p53-MDM2 interaction HDM201 elicits therapeutic efficacy when applied at various doses and schedules. Continuous exposure of HDM201 led to induction of p21 and delayed accumulation of apoptotic cells. By comparison, high-dose pulses of HDM201 were associated with marked induction of PUMA and a rapid onset of apoptosis. shRNA screens identified PUMA as a mediator of the p53 response specifically in the pulsed regimen. Consistent with this, the single high-dose HDM201 regimen resulted in rapid and marked induction of PUMA expression and apoptosis together with downregulation of Bcl-xL in vivo Knockdown of Bcl-xL was identified as the top sensitizer to HDM201 in vitro, and Bcl-xL was enriched in relapsing tumors from mice treated with intermittent high doses of HDM201. These findings define a regimen-dependent mechanism by which disruption of MDM2-p53 elicits therapeutic efficacy when given with infrequent dosing. In an ongoing HDM201 trial, the observed exposure-response relationship indicates that the molecular mechanism elicited by pulse dosing is likely reproducible in patients. These data support the clinical comparison of daily and intermittent regimens of p53-MDM2 inhibitors.Significance: Pulsed high doses versus sustained low doses of the p53-MDM2 inhibitor HDM201 elicit a proapoptotic response from wild-type p53 cancer cells, offering guidance to current clinical trials with this and other drugs that exploit the activity of p53. Cancer Res; 78(21); 6257-67. ©2018 AACR.


Cancer Research | 2016

Abstract 1311: High order drug combinations are required to effectively kill colorectal cancer cells

Thomas Horn; Stephane Ferretti; Nicolas Ebel; Angela Tam; Samuel Ho; Fred Harbinski; Ali Farsidjani; Matt Zubrowski; William R. Sellers; Robert Schlegel; Dale Porter; Erick Morris; Jens Wuerthner; Sébastien Jeay; Joel Greshock; Ensar Halilovic; Levi A. Garraway; Giordano Caponigro; Joseph Lehár

Tumors are complex biological systems that often retain proliferative capacity even when challenged with drug treatment. Given this resiliency, drug combinations may provide greater therapeutic benefit, however, which molecules to combine and how many to include in combinations for effective responses is not clear yet. Using image-based proliferation and apoptosis assays in colorectal cancer cell lines we systematically investigated combinations that ranged in number from two to six drugs and targeted critical oncogenic pathways. Drug pairs targeting key signaling pathways resulted in synergies across a broad spectrum of genetic backgrounds, but often yielded only cytostatic responses. Enhanced cytotoxicity was observed when additional processes including apoptosis and cell cycle were targeted as part of the combination. In many cases, where cell lines were resistant to two- and three-way drug combinations, increased expression of anti-apoptotic proteins was observed and induction of cytotoxic responses required up to fourth-order combinations. Our results demonstrate that high-order drug combinations might be needed to kill cancers and show how systematic drug combination screening together with a molecular understanding of drug responses can guide their identification. Citation Format: Thomas Horn, Stephane Ferretti, Nicolas Ebel, Angela Tam, Samuel Ho, Fred Harbinski, Ali Farsidjani, Matt Zubrowski, William R. Sellers, Robert Schlegel, Dale Porter, Erick Morris, Jens Wuerthner, Sebastien Jeay, Joel Greshock, Ensar Halilovic, Levi A. Garraway, Giordano Caponigro, Joseph Lehar. High order drug combinations are required to effectively kill colorectal cancer cells. [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 1311.


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.

Collaboration


Dive into the Jens Wuerthner's collaboration.

Researchain Logo
Decentralizing Knowledge