Network


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

Hotspot


Dive into the research topics where Rita Andraos is active.

Publication


Featured researches published by Rita Andraos.


Cancer Cell | 2012

JAK2/STAT5 Inhibition Circumvents Resistance to PI3K/mTOR Blockade: A Rationale for Cotargeting These Pathways in Metastatic Breast Cancer

Adrian Britschgi; Rita Andraos; Heike Brinkhaus; Ina Klebba; Vincent Romanet; Urs Müller; Masato Murakami; Thomas Radimerski; Mohamed Bentires-Alj

Hyperactive PI3K/mTOR signaling is prevalent in human malignancies and its inhibition has potent antitumor consequences. Unfortunately, single-agent targeted cancer therapy is usually short-lived. We have discovered a JAK2/STAT5-evoked positive feedback loop that dampens the efficacy of PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this feedback loop and combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell number and tumor growth, decreased tumor seeding and metastasis, and also increased overall survival of the animals. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Enhanced sensitivity to IGF-II signaling links loss of imprinting of IGF2 to increased cell proliferation and tumor risk.

Atsushi Kaneda; Chiaochun J. Wang; Raymond Cheong; Winston Timp; Patrick Onyango; Bo Wen; Christine A. Iacobuzio-Donahue; Rolf Ohlsson; Rita Andraos; Mark Pearson; Alexei A. Sharov; Dan L. Longo; Minoru S.H. Ko; Andre Levchenko; Andrew P. Feinberg

Loss of imprinting (LOI) of the insulin-like growth factor-II gene (IGF2), leading to abnormal activation of the normally silent maternal allele, is a common human epigenetic population variant associated with a 5-fold increased frequency of colorectal neoplasia. Here, we show first that LOI leads specifically to increased expression of proliferation-related genes in mouse intestinal crypts. Surprisingly, LOI(+) mice also have enhanced sensitivity to IGF-II signaling, not simply increased IGF-II levels, because in vivo blockade with NVP-AEW541, a specific inhibitor of the IGF-II signaling receptor, showed reduction of proliferation-related gene expression to levels half that seen in LOI(−) mice. Signal transduction assays in microfluidic chips confirmed this enhanced sensitivity with marked augmentation of Akt/PKB signaling in LOI(+) cells at low doses of IGF-II, which was reduced in the presence of the inhibitor to levels below those found in LOI(−) cells, and was associated with increased expression of the IGF1 and insulin receptor genes. We exploited this increased IGF-II sensitivity to develop an in vivo chemopreventive strategy using the azoxymethane (AOM) mutagenesis model. LOI(+) mice treated with AOM showed a 60% increase in premalignant aberrant crypt foci (ACF) formation over LOI(−) mice. In vivo IGF-II blockade with NVP-AEW541 abrogated this effect, reducing ACF to a level 30% lower even than found in exposed LOI(−) mice. Thus, LOI increases cancer risk in a counterintuitive way, by increasing the sensitivity of the IGF-II signaling pathway itself, providing a previously undescribed epigenetic chemoprevention strategy in which cells with LOI are “IGF-II addicted” and undergo reduced tumorigenesis in the colon upon IGF-II pathway blockade.


Cancer Discovery | 2012

Modulation of Activation-Loop Phosphorylation by JAK Inhibitors Is Binding Mode Dependent

Rita Andraos; Zhiyan Qian; Débora Bonenfant; Joëlle Rubert; Eric Vangrevelinghe; Clemens Scheufler; Fanny Marque; Catherine H. Regnier; Alain De Pover; Hugues Ryckelynck; Neha Bhagwat; Priya Koppikar; Aviva Goel; Lorenza Wyder; Gisele Tavares; Fabienne Baffert; Carole Pissot-Soldermann; Paul W. Manley; Christoph Gaul; Hans Voshol; Ross L. Levine; William R. Sellers; Francesco Hofmann; Thomas Radimerski

Janus kinase (JAK) inhibitors are being developed for the treatment of rheumatoid arthritis, psoriasis, myeloproliferative neoplasms, and leukemias. Most of these drugs target the ATP-binding pocket and stabilize the active conformation of the JAK kinases. This type I binding mode can lead to an increase in JAK activation loop phosphorylation, despite blockade of kinase function. Here we report that stabilizing the inactive state via type II inhibition acts in the opposite manner, leading to a loss of activation loop phosphorylation. We used X-ray crystallography to corroborate the binding mode and report for the first time the crystal structure of the JAK2 kinase domain in an inactive conformation. Importantly, JAK inhibitor-induced activation loop phosphorylation requires receptor interaction, as well as intact kinase and pseudokinase domains. Hence, depending on the respective conformation stabilized by a JAK inhibitor, hyperphosphorylation of the activation loop may or may not be elicited.


Clinical Cancer Research | 2013

JAK1/2 and Pan-Deacetylase Inhibitor Combination Therapy Yields Improved Efficacy in Preclinical Mouse Models of JAK2V617F-Driven Disease

Emeline Evrot; Nicolas Ebel; Vincent Romanet; Claudia Roelli; Rita Andraos; Zhiyan Qian; Arno Dölemeyer; Ernesta Dammassa; Dario Sterker; Robert Cozens; Francesco Hofmann; Masato Murakami; Fabienne Baffert; Thomas Radimerski

Purpose: The myeloproliferative neoplasm myelofibrosis is characterized by frequent deregulation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, and JAK inhibitors were shown to reduce splenomegaly and ameliorate disease-related symptoms. However, the mutant clone and bone marrow fibrosis persist in the majority of patients. Using preclinical models, we explored whether JAK and pan-deacetylase inhibitor combination yielded additional benefits. Experimental Design: The combination of the JAK1/2 inhibitor ruxolitinib and panobinostat was investigated using two different mouse models of JAK2V617F-driven disease. A Ba/F3 JAK2V617F cell–driven leukemic disease model was used to identify tolerated and efficacious doses. The drugs were then evaluated alone and in combination in a mouse model of myeloproliferative neoplasm–like disease based on transplantation of bone marrow transduced with a retrovirus expressing JAK2V617F. Exposures were determined in blood and tissues, and phosphorylated STAT5 and acetylated histone H3 pharmacodynamic readouts were assessed in spleen and bone marrow. Histologic analysis was conducted on spleen and bone marrow, including staining of reticulin fibers in the latter organ. Results: The combination of ruxolitinib and panobinostat was found to have a more profound effect on splenomegaly, as well as on bone marrow and spleen histology, compared with either agent alone, and the analysis of pharmacodynamic readouts showed that ruxolitinib and panobinostat have nonoverlapping and complementary effects. Conclusion: Combining JAK1/2 and pan-deacetylase inhibitors was fairly well tolerated and resulted in improved efficacy in mouse models of JAK2V617F-driven disease compared with the single agents. Thus, the combination of ruxolitinib and panobinostat may represent a promising novel therapeutic modality for myeloproliferative neoplasms. Clin Cancer Res; 19(22); 6230–41. ©2013 AACR.


Blood | 2013

Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model

Lucia Kubovcakova; Pontus Lundberg; Jean Grisouard; Hui Hao-Shen; Vincent Romanet; Rita Andraos; Masato Murakami; Stephan Dirnhofer; Kay Uwe Wagner; Thomas Radimerski; Radek C. Skoda

To establish a preclinical animal model for testing drugs with potential effects on myeloproliferative neoplasms (MPNs), we first performed a detailed phenotypic characterization of Cre-inducible transgenic JAK2-V617F mice. Deleting the conditional mouse Jak2-knockout alleles increased erythropoiesis and accentuated the polycythemia vera phenotype, but did not alter platelet or granulocyte levels. In a transplantation assay, JAK2-V617F(+) BM cells had an advantage over wild-type competitor cells. Using this competitive repopulation assay, we compared the effects of INC424 (ruxolitinib), a dual Jak1/Jak2 inhibitor, and hydroxyurea (HU). HU led to weight loss, but did not reduce spleen weight. The hematologic parameters were lowered and a slight decrease of the mutant allele burden was noted. INC424 had little effect on body weight, but strongly decreased spleen size and rapidly normalized RBC and neutrophil parameters. No significant decrease in the mutant allele burden was observed. INC424 reduced the phospho-Stat5 levels, whereas HU strongly increased phospho-Stat5, most likely because of the elevated erythropoietin levels in response to the HU-induced anemia. This compensatory increase in JAK/STAT signaling may counteract the beneficial effects of cytoreduction at higher doses of HU and represents an adverse effect that should be avoided.


BMC Cancer | 2011

Bim and Mcl-1 exert key roles in regulating JAK2V617F cell survival

Joëlle Rubert; Zhiyan Qian; Rita Andraos; Daniel Guthy; Thomas Radimerski

BackgroundThe JAK2V617F mutation plays a major role in the pathogenesis of myeloproliferative neoplasms and is found in the vast majority of patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia or from primary myelofibrosis. The V617F mutation is thought to provide hematopoietic stem cells and myeloid progenitors with a survival and proliferation advantage. It has previously been shown that activated JAK2 promotes cell survival by upregulating the anti-apoptotic STAT5 target gene Bcl-xL. In this study, we have investigated the role of additional apoptotic players, the pro-apoptotic protein Bim as well as the anti-apoptotic protein Mcl-1.MethodsPharmacological inhibition of JAK2/STAT5 signaling in JAK2V617F mutant SET-2 and MB-02 cells was used to study effects on signaling, cell proliferation and apoptosis by Western blot analysis, WST-1 proliferation assays and flow cytometry. Cells were transfected with siRNA oligos to deplete candidate pro- and anti-apoptotic proteins. Co-immunoprecipitation assays were performed to assess the impact of JAK2 inhibition on complexes of pro- and anti-apoptotic proteins.ResultsTreatment of JAK2V617F mutant cell lines with a JAK2 inhibitor was found to trigger Bim activation. Furthermore, Bim depletion by RNAi suppressed JAK2 inhibitor-induced cell death. Bim activation following JAK2 inhibition led to enhanced sequestration of Mcl-1, besides Bcl-xL. Importantly, Mcl-1 depletion by RNAi was sufficient to compromise JAK2V617F mutant cell viability and sensitized the cells to JAK2 inhibition.ConclusionsWe conclude that Bim and Mcl-1 have key opposing roles in regulating JAK2V617F cell survival and propose that inactivation of aberrant JAK2 signaling leads to changes in Bim complexes that trigger cell death. Thus, further preclinical evaluation of combinations of JAK2 inhibitors with Bcl-2 family antagonists that also tackle Mcl-1, besides Bcl-xL, is warranted to assess the therapeutic potential for the treatment of chronic myeloproliferative neoplasms.


Cancer Research | 2013

Abstract LB-36: JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: A rationale for co-targeting these pathways in metastatic breast cancer.

Rita Andraos; Heike Brinkhaus; Inna Klebba; Masato Murakami; Vincent Romanet; Urs Müller; Thomas Radimerski; Mohamed Bentires-Alj

Hyperactive PI3K/mTOR signaling is prevalent in the majority of human malignancies (1) and its inhibition exhibits potent antitumor activity in a wide spectrum of solid cancers. Unfortunately, single-agent targeted cancer therapy is usually short-lived and thwarted by different resistance mechanisms (2). Here, we report the discovery of a JAK2/STAT5-evoked positive feedback loop that causes adaptive resistance to dual PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary triple-negative breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this vicious feedback loop. Combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell viability in vitro as well as tumor growth in vivo, and decreased tumor seeding and metastasis due to its impact on the IL-8 receptor CXCR1+ tumor-initiating subpopulation of cells. We further found that combined PI3K/mTOR and JAK2 inhibition increased event-free as a well as overall survival of tumor bearing animals. This study reveals a new link between growth factor signaling, JAK/STAT activation, cytokine secretion and metastasis. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease. Citation Format: Adrian Britschgi, Rita Andraos, Heike Brinkhaus, Inna Klebba, Masato Murakami, Vincent Romanet, Urs Muller, Thomas Radimerski, Mohamed Bentires-Alj. JAK2/STAT5 inhibition circumvents resistance to PI3K/mTOR blockade: A rationale for co-targeting these pathways in metastatic breast cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr LB-36. doi:10.1158/1538-7445.AM2013-LB-36


Blood | 2011

Frontline Therapy with Brentuximab Vedotin Combined with ABVD or AVD in Patients with Newly Diagnosed Advanced Stage Hodgkin Lymphoma

Fabienne Baffert; Emeline Evrot; Nicolas Ebel; Claudia Roelli; Rita Andraos; Zhiyan Qian; Vincent Romanet; Masato Murakami; Thomas Radimerski


Cancer Cell | 2015

CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms

Sara C. Meyer; Matthew Keller; Sophia Chiu; Priya Koppikar; Olga A. Guryanova; Franck Rapaport; Ke Xu; Katia Manova; Dmitry Pankov; Richard J. O’Reilly; Maria Kleppe; Anna Sophia McKenney; Alan H. Shih; Kaitlyn Shank; Jihae Ahn; Eftymia Papalexi; Barbara Spitzer; Nick Socci; Agnes Viale; Emeline Mandon; Nicolas Ebel; Rita Andraos; Joëlle Rubert; Ernesta Dammassa; Vincent Romanet; Arno Dölemeyer; Michael Zender; Melanie Heinlein; Raajit Rampal; Rona S. Weinberg


Cancer Cell | 2015

Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia

Shuo-Chieh Wu; Loretta S. Li; Nadja Kopp; Joan Montero; Bjoern Chapuy; Akinori Yoda; Amanda L. Christie; Huiyun Liu; Alexandra N. Christodoulou; Diederik van Bodegom; Jordy C. Van der Zwet; Jacob V. Layer; Trevor Tivey; Andrew A. Lane; Jeremy Ryan; Samuel Y. Ng; Daniel J. DeAngelo; Richard Stone; David P. Steensma; Martha Wadleigh; Marian H. Harris; Emeline Mandon; Nicolas Ebel; Rita Andraos; Vincent Romanet; Arno Dölemeyer; Dario Sterker; Michael Zender; Scott J. Rodig; Masato Murakami

Collaboration


Dive into the Rita Andraos's collaboration.

Researchain Logo
Decentralizing Knowledge