Network


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

Hotspot


Dive into the research topics where Carlos Bais is active.

Publication


Featured researches published by Carlos Bais.


Nature | 2007

Bv8 regulates myeloid-cell-dependent tumour angiogenesis

Farbod Shojaei; Xiumin Wu; Cuiling Zhong; Lanlan Yu; Xiaohuan Liang; Jenny Yao; Dominique Blanchard; Carlos Bais; Franklin Peale; Nicholas van Bruggen; Calvin Ho; Jed Ross; Martha Tan; Richard A. D. Carano; Y. Gloria Meng; Napoleone Ferrara

Bone-marrow-derived cells facilitate tumour angiogenesis, but the molecular mechanisms of this facilitation are incompletely understood. We have previously shown that the related EG-VEGF and Bv8 proteins, also known as prokineticin 1 (Prok1) and prokineticin 2 (Prok2), promote both tissue-specific angiogenesis and haematopoietic cell mobilization. Unlike EG-VEGF, Bv8 is expressed in the bone marrow. Here we show that implantation of tumour cells in mice resulted in upregulation of Bv8 in CD11b+Gr1+ myeloid cells. We identified granulocyte colony-stimulating factor as a major positive regulator of Bv8 expression. Anti-Bv8 antibodies reduced CD11b+Gr1+ cell mobilization elicited by granulocyte colony-stimulating factor. Adenoviral delivery of Bv8 into tumours was shown to promote angiogenesis. Anti-Bv8 antibodies inhibited growth of several tumours in mice and suppressed angiogenesis. Anti-Bv8 treatment also reduced CD11b+Gr1+ cells, both in peripheral blood and in tumours. The effects of anti-Bv8 antibodies were additive to those of anti-Vegf antibodies or cytotoxic chemotherapy. Thus, Bv8 modulates mobilization of CD11b+Gr1+ cells from the bone marrow during tumour development and also promotes angiogenesis locally.


The EMBO Journal | 2012

Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway

Guanglei Zhuang; Xiumin Wu; Zhaoshi Jiang; Ian Kasman; Jenny Yao; Yinghui Guan; Jason Oeh; Zora Modrusan; Carlos Bais; Deepak Sampath; Napoleone Ferrara

Angiogenesis plays a crucial role during tumorigenesis and much progress has been recently made in elucidating the role of VEGF and other growth factors in the regulation of angiogenesis. Recently, microRNAs (miRNAs) have been shown to modulate a variety of physiogical and pathological processes. We identified a set of differentially expressed miRNAs in microvascular endothelial cells co‐cultured with tumour cells. Unexpectedly, most miRNAs were derived from tumour cells, packaged into microvesicles (MVs), and then directly delivered to endothelial cells. Among these miRNAs, we focused on miR‐9 due to the strong morphological changes induced in cultured endothelial cells. We found that exogenous miR‐9 effectively reduced SOCS5 levels, leading to activated JAK‐STAT pathway. This signalling cascade promoted endothelial cell migration and tumour angiogenesis. Remarkably, administration of anti‐miR‐9 or JAK inhibitors suppressed MV‐induced cell migration in vitro and decreased tumour burden in vivo. Collectively, these observations suggest that tumour‐secreted miRNAs participate in intercellular communication and function as a novel pro‐angiogenic mechanism.


Cell | 2014

Glycosylation-Dependent Lectin-Receptor Interactions Preserve Angiogenesis in Anti-VEGF Refractory Tumors

Diego O. Croci; Juan P. Cerliani; Tomas Dalotto-Moreno; Santiago P. Méndez-Huergo; Ivan D. Mascanfroni; Sebastián Dergan-Dylon; Marta A. Toscano; Julio J. Caramelo; Juan J. Garcia-Vallejo; Jing Ouyang; Enrique A. Mesri; Melissa R. Junttila; Carlos Bais; Margaret A. Shipp; Mariana Salatino; Gabriel A. Rabinovich

The clinical benefit conferred by vascular endothelial growth factors (VEGF)-targeted therapies is variable, and tumors from treated patients eventually reinitiate growth. Here, we identify a glycosylation-dependent pathway that compensates for the absence of cognate ligand and preserves angiogenesis in response to VEGF blockade. Remodeling of the endothelial cell (EC) surface glycome selectively regulated binding of galectin-1 (Gal1), which upon recognition of complex N-glycans on VEGFR2, activated VEGF-like signaling. Vessels within anti-VEGF-sensitive tumors exhibited high levels of α2-6-linked sialic acid, which prevented Gal1 binding. In contrast, anti-VEGF refractory tumors secreted increased Gal1 and their associated vasculature displayed glycosylation patterns that facilitated Gal1-EC interactions. Interruption of β1-6GlcNAc branching in ECs or silencing of tumor-derived Gal1 converted refractory into anti-VEGF-sensitive tumors, whereas elimination of α2-6-linked sialic acid conferred resistance to anti-VEGF. Disruption of the Gal1-N-glycan axis promoted vascular remodeling, immune cell influx and tumor growth inhibition. Thus, targeting glycosylation-dependent lectin-receptor interactions may increase the efficacy of anti-VEGF treatment.


Cell | 2013

Targeting placental growth factor/neuropilin 1 pathway inhibits growth and spread of medulloblastoma

Matija Snuderl; Ana Batista; Nathaniel D. Kirkpatrick; Carmen Ruiz de Almodovar; Lars Riedemann; Elisa C. Walsh; Rachel Anolik; Yuhui Huang; John D. Martin; Walid S. Kamoun; Ellen Knevels; Thomas Schmidt; Christian T. Farrar; Benjamin J. Vakoc; Nishant Mohan; Euiheon Chung; Sylvie Roberge; Teresa Peterson; Carlos Bais; Boryana Zhelyazkova; Stephen Yip; Martin Hasselblatt; Claudia Rossig; Elisabeth Niemeyer; Napoleone Ferrara; Michael Klagsbrun; Dan G. Duda; Dai Fukumura; Lei Xu; Peter Carmeliet

Medulloblastoma is the most common pediatric malignant brain tumor. Although current therapies improve survival, these regimens are highly toxic and are associated with significant morbidity. Here, we report that placental growth factor (PlGF) is expressed in the majority of medulloblastomas, independent of their subtype. Moreover, high expression of PlGF receptor neuropilin 1 (Nrp1) correlates with poor overall survival in patients. We demonstrate that PlGF and Nrp1 are required for the growth and spread of medulloblastoma: PlGF/Nrp1 blockade results in direct antitumor effects in vivo, resulting in medulloblastoma regression, decreased metastasis, and increased mouse survival. We reveal that PlGF is produced in the cerebellar stroma via tumor-derived Sonic hedgehog (Shh) and show that PlGF acts through Nrp1-and not vascular endothelial growth factor receptor 1-to promote tumor cell survival. This critical tumor-stroma interaction-mediated by Shh, PlGF, and Nrp1 across medulloblastoma subtypes-supports the development of therapies targeting PlGF/Nrp1 pathway.


Cell | 2010

PlGF Blockade Does Not Inhibit Angiogenesis during Primary Tumor Growth

Carlos Bais; Xiumin Wu; Jenny Yao; Suya Yang; Yongping Crawford; Krista McCutcheon; Christine Tan; Ganesh Kolumam; Jean-Michel Vernes; Jeffrey Eastham-Anderson; Peter Haughney; Marcin Kowanetz; Thijs J. Hagenbeek; Ian Kasman; Hani Bou Reslan; Jed Ross; Nick van Bruggen; Richard A. D. Carano; Yu-Ju Gloria Meng; Jo-Anne Hongo; Jean Philippe Stephan; Masabumi Shibuya; Napoleone Ferrara

It has been recently reported that treatment with an anti-placenta growth factor (PlGF) antibody inhibits metastasis and primary tumor growth. Here we show that, although anti-PlGF treatment inhibited wound healing, extravasation of B16F10 cells, and growth of a tumor engineered to overexpress the PlGF receptor (VEGFR-1), neutralization of PlGF using four novel blocking antibodies had no significant effect on tumor angiogenesis in 15 models. Also, genetic ablation of the tyrosine kinase domain of VEGFR-1 in the host did not result in growth inhibition of the anti-VEGF-A sensitive or resistant tumors tested. Furthermore, combination of anti-PlGF with anti-VEGF-A antibodies did not result in greater antitumor efficacy than anti-VEGF-A monotherapy. In conclusion, our data argue against an important role of PlGF during primary tumor growth in most models and suggest that clinical evaluation of anti-PlGF antibodies may be challenging.


Journal of Clinical Oncology | 2015

Patients With Proneural Glioblastoma May Derive Overall Survival Benefit From the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial

Thomas Sandmann; Richard Bourgon; Josep Garcia; Congfen Li; Timothy F. Cloughesy; Olivier Chinot; Wolfgang Wick; Ryo Nishikawa; Warren P. Mason; Roger Henriksson; Frank Saran; Albert Lai; Nicola Moore; Samir Kharbanda; Franklin Peale; Priti Hegde; Lauren E. Abrey; Heidi S. Phillips; Carlos Bais

PURPOSE The AVAglio (Avastin in Glioblastoma) and RTOG-0825 randomized, placebo-controlled phase III trials in newly diagnosed glioblastoma reported prolonged progression-free survival (PFS), but not overall survival (OS), with the addition of bevacizumab to radiotherapy plus temozolomide. To establish whether certain patient subgroups derived an OS benefit from the addition of bevacizumab to first-line standard-of-care therapy, AVAglio patients were retrospectively evaluated for molecular subtype, and bevacizumab efficacy was assessed for each patient subgroup. PATIENTS AND METHODS A total of 349 pretreatment specimens (bevacizumab arm, n = 171; placebo arm, n = 178) from AVAglio patients (total, N = 921) were available for biomarker analysis. Samples were profiled for gene expression and isocitrate dehydrogenase 1 (IDH1) mutation status and classified into previously identified molecular subtypes. PFS and OS were assessed within each subtype. RESULTS A multivariable analysis accounting for prognostic covariates revealed that bevacizumab conferred a significant OS advantage versus placebo for patients with proneural IDH1 wild-type tumors (17.1 v 12.8 months, respectively; hazard ratio, 0.43; 95% CI, 0.26 to 0.73; P = .002). This analysis also revealed an interaction between the proneural subtype biomarker and treatment arm (P = .023). The group of patients with mesenchymal and proneural tumors derived a PFS benefit from bevacizumab compared with placebo; however, this translated to an OS benefit in the proneural subset only. CONCLUSION Retrospective analysis of AVAglio data suggests that patients with IDH1 wild-type proneural glioblastoma may derive an OS benefit from first-line bevacizumab treatment. The predictive value of the proneural subtype observed in AVAglio should be validated in an independent data set.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Expression of a functional VEGFR-1 in tumor cells is a major determinant of anti-PlGF antibodies efficacy

Jenny Yao; Xiumin Wu; Guanglei Zhuang; Ian Kasman; Tobias K. Vogt; Vernon Phan; Masabumi Shibuya; Napoleone Ferrara; Carlos Bais

PlGF, one of the ligands for VEGFR-1, has been implicated in tumor angiogenesis. However, more recent studies indicate that genetic or pharmacological inhibition of PlGF signaling does not result in reduction of microvascular density in a variety of tumor models. Here we screened 12 human tumor cell lines and identified 3 that are growth inhibited by anti-PlGF antibodies in vivo. We found that efficacy of anti-PlGF treatment strongly correlates with VEGFR-1 expression in tumor cells, but not with antiangiogenesis. In addition, PlGF induced VEGFR-1 signaling and biological responses in tumor cell lines sensitive to anti-PlGF, but not in refractory tumor cell lines or in endothelial cells. Also, genetic ablation of VEGFR-1 signaling in the host did not affect the efficacy of PlGF blockade. Collectively, these findings suggest that the role of PlGF in tumorigenesis largely consists of promoting autocrine/paracrine growth of tumor cells expressing a functional VEGFR-1 rather than stimulation of angiogenesis.


Clinical Cancer Research | 2013

Identification and Analysis of In Vivo VEGF Downstream Markers Link VEGF Pathway Activity with Efficacy of Anti- VEGF Therapies

Matthew J. Brauer; Guanglei Zhuang; Maike Schmidt; Jenny Yao; Xiumin Wu; Joshua S. Kaminker; Stefanie S. Jurinka; Ganesh Kolumam; Alicia S. Chung; Adrian M. Jubb; Zora Modrusan; Tomoko Ozawa; C. David James; Heidi S. Phillips; Benjamin Haley; Rachel Tam; Anne C Clermont; Jason H. Cheng; Sherry X. Yang; Sandra M. Swain; Daniel Chen; Stefan J. Scherer; Hartmut Koeppen; Ru Fang Yeh; Peng Yue; Jean Philippe Stephan; Priti Hegde; Napoleone Ferrara; Mallika Singh; Carlos Bais

Purpose: The aim of this study was to identify conserved pharmacodynamic and potential predictive biomarkers of response to anti-VEGF therapy using gene expression profiling in preclinical tumor models and in patients. Experimental Design: Surrogate markers of VEGF inhibition [VEGF-dependent genes or VEGF-dependent vasculature (VDV)] were identified by profiling gene expression changes induced in response to VEGF blockade in preclinical tumor models and in human biopsies from patients treated with anti-VEGF monoclonal antibodies. The potential value of VDV genes as candidate predictive biomarkers was tested by correlating high or low VDV gene expression levels in pretreatment clinical samples with the subsequent clinical efficacy of bevacizumab (anti-VEGF)-containing therapy. Results: We show that VDV genes, including direct and more distal VEGF downstream endothelial targets, enable detection of VEGF signaling inhibition in mouse tumor models and human tumor biopsies. Retrospective analyses of clinical trial data indicate that patients with higher VDV expression in pretreatment tumor samples exhibited improved clinical outcome when treated with bevacizumab-containing therapies. Conclusions: In this work, we identified surrogate markers (VDV genes) for in vivo VEGF signaling in tumors and showed clinical data supporting a correlation between pretreatment VEGF bioactivity and the subsequent efficacy of anti-VEGF therapy. We propose that VDV genes are candidate biomarkers with the potential to aid the selection of novel indications as well as patients likely to respond to anti-VEGF therapy. The data presented here define a diagnostic biomarker hypothesis based on translational research that warrants further evaluation in additional retrospective and prospective trials. Clin Cancer Res; 19(13); 3681–92. ©2013 AACR.


Science Signaling | 2013

Phosphoproteomic Analysis Implicates the mTORC2-FoxO1 Axis in VEGF Signaling and Feedback Activation of Receptor Tyrosine Kinases

Guanglei Zhuang; Kebing Yu; Zhaoshi Jiang; Alicia Chung; Jenny Yao; Connie Ha; Karen Toy; Robert Soriano; Benjamin Haley; Elizabeth Blackwood; Deepak Sampath; Carlos Bais; Jennie R. Lill; Napoleone Ferrara

Pathological angiogenesis mediated by the VEGF receptor could be treated by combining mTOR and PI3K inhibitors. Combining Forces Against Angiogenesis Pathological angiogenesis contributes to the growth of solid tumors, which has led to the development of several antiangiogenic therapies that target the VEGF (vascular endothelial growth factor) signaling pathway, which promotes angiogenesis. Zhuang et al. mapped the signaling networks activated in endothelial cells by VEGF through its receptor tyrosine kinases and found that VEGF promoted the survival of endothelial cells by stimulating the kinase activity of PI3K (phosphatidylinositol 3-kinase) and of mTORC2 (mammalian target of rapamycin complex 2). Because VEGF-activated endothelial cells exposed to mTOR kinase inhibitors exhibited increased activity of multiple receptor tyrosine kinases and PI3K, the cells were less susceptible to the toxic effects of mTOR inhibition. However, combining mTOR and PI3K inhibitors effectively reduced endothelial cell survival. Thus, understanding the signaling pathways that are activated by VEGF can identify potential therapeutic strategies to treat pathological angiogenesis. The vascular endothelial growth factor (VEGF) signaling pathway plays a pivotal role in normal development and also represents a major therapeutic target for tumors and intraocular neovascular disorders. The VEGF receptor tyrosine kinases promote angiogenesis by phosphorylating downstream proteins in endothelial cells. We applied a large-scale proteomic approach to define the VEGF-regulated phosphoproteome and its temporal dynamics in human umbilical vein endothelial cells and then used siRNA (small interfering RNA) screens to investigate the function of a subset of these phosphorylated proteins in VEGF responses. The PI3K (phosphatidylinositol 3-kinase)–mTORC2 (mammalian target of rapamycin complex 2) axis emerged as central in activating VEGF-regulated phosphorylation and increasing endothelial cell viability by suppressing the activity of the transcription factor FoxO1 (forkhead box protein O1), an effect that limited cellular apoptosis and feedback activation of receptor tyrosine kinases. This FoxO1-mediated feedback loop not only reduced the effectiveness of mTOR inhibitors at decreasing protein phosphorylation and cell survival but also rendered cells more susceptible to PI3K inhibition. Collectively, our study provides a global and dynamic view of VEGF-regulated phosphorylation events and implicates the mTORC2-FoxO1 axis in VEGF receptor signaling and reprogramming of receptor tyrosine kinases in human endothelial cells.


Clinical Cancer Research | 2015

Upregulation of Periostin and Reactive Stroma Is Associated with Primary Chemoresistance and Predicts Clinical Outcomes in Epithelial Ovarian Cancer

Lisa Ryner; Yinghui Guan; Ron Firestein; Yuanyuan Xiao; YounJeong Choi; Christina Rabe; Shan Lu; Eloisa Fuentes; Ling Huw; Mark R. Lackner; Ling Fu; Lukas Amler; Carlos Bais; Yulei Wang

Purpose: Up to one third of ovarian cancer patients are intrinsically resistant to platinum-based treatment. However, predictive and therapeutic strategies are lacking due to a poor understanding of the underlying molecular mechanisms. This study aimed to identify key molecular characteristics that are associated with primary chemoresistance in epithelial ovarian cancers. Experimental Design: Gene expression profiling was performed on a discovery set of 85 ovarian tumors with clinically well-defined response to chemotherapies as well as on an independent validation dataset containing 138 ovarian patients from the chemotreatment arm of the ICON7 trial. Results: We identified a distinct “reactive stroma” gene signature that is specifically associated with primary chemoresistant tumors and was further upregulated in posttreatment recurrent tumors. Immunohistochemistry (IHC) and RNA in situ hybridization (RNA ISH) analyses on three of the highest-ranked signature genes (POSTN, LOX, and FAP) confirmed that modulation of the reactive stroma signature genes within the peritumoral stromal compartments was specifically associated with the clinical chemoresistance. Consistent with these findings, chemosensitive ovarian cells grown in the presence of recombinant POSTN promoted resistance to carboplatin and paclitaxel treatment in vitro. Finally, we validated the reactive stroma signature in an independent dataset and demonstrated that a high POSTN expression level predicts shorter progression-free survival following first-line chemotherapy. Conclusions: Our findings highlight the important interplay between cancer and the tumor microenvironment in ovarian cancer biology and treatment. The identified reactive stromal components in this study provide a molecular basis to the further development of novel diagnostic and therapeutic strategies for overcoming chemoresistance in ovarian cancer. Clin Cancer Res; 21(13); 2941–51. ©2015 AACR.

Collaboration


Dive into the Carlos Bais's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olivier Chinot

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge