Network


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

Hotspot


Dive into the research topics where Federica Gibellini is active.

Publication


Featured researches published by Federica Gibellini.


Blood | 2011

The lymph node microenvironment promotes B-cell receptor signaling, NF-κB activation, and tumor proliferation in chronic lymphocytic leukemia

Yair Herishanu; Patricia Pérez-Galán; Delong Liu; Angélique Biancotto; Stefania Pittaluga; Berengere Vire; Federica Gibellini; Ndegwa Njuguna; Elinor Lee; Lawrence S Stennett; Nalini Raghavachari; Poching Liu; J. Philip McCoy; Mark Raffeld; Maryalice Stetler-Stevenson; Constance Yuan; Richard M. Sherry; Diane C. Arthur; Irina Maric; Therese White; Gerald E. Marti; Peter J. Munson; Wyndham H. Wilson; Adrian Wiestner

Chronic lymphocytic leukemia (CLL), an incurable malignancy of mature B lymphocytes, involves blood, bone marrow, and secondary lymphoid organs such as the lymph nodes (LN). A role of the tissue microenvironment in the pathogenesis of CLL is hypothesized based on in vitro observations, but its contribution in vivo remains ill-defined. To elucidate the effects of tumor-host interactions in vivo, we purified tumor cells from 24 treatment-naive patients. Samples were obtained concurrently from blood, bone marrow, and/or LN and analyzed by gene expression profiling. We identified the LN as a key site in CLL pathogenesis. CLL cells in the LN showed up-regulation of gene signatures, indicating B-cell receptor (BCR) and nuclear factor-κB activation. Consistent with antigen-dependent BCR signaling and canonical nuclear factor-κB activation, we detected phosphorylation of SYK and IκBα, respectively. Expression of BCR target genes was stronger in clinically more aggressive CLL, indicating more effective BCR signaling in this subtype in vivo. Tumor proliferation, quantified by the expression of the E2F and c-MYC target genes and verified with Ki67 staining by flow cytometry, was highest in the LN and was correlated with clinical disease progression. These data identify the disruption of tumor microenvironment interactions and the inhibition of BCR signaling as promising therapeutic strategies in CLL. This study is registered at http://clinicaltrials.gov as NCT00019370.


Haematologica | 2009

Lenalidomide-induced upregulation of CD80 on tumor cells correlates with T-cell activation, the rapid onset of a cytokine release syndrome and leukemic cell clearance in chronic lymphocytic leukemia

Georg Aue; Ndegwa Njuguna; Xin Tian; Susan Soto; Thomas Hughes; Berengere Vire; Keyvan Keyvanfar; Federica Gibellini; Janet Valdez; Carol Boss; Leigh Samsel; J. Philip McCoy; Wyndham H. Wilson; Stefania Pittaluga; Adrian Wiestner

In chronic lymphocytic leukemia (CLL) lenalidomide causes striking immune activation, possibly leading to clearance of tumor cells. This study shows that upregulation of CD80 on tumor cells and T-cell activation, which appears to be dispensable for the drug’s anti-tumor effects, correlate with unique toxicities of lenalidomide in CLL. This provides a rationale for combinations of lenalidomide with immunosuppressive agents. See related perspective article on page 1198. Background In chronic lymphocytic leukemia lenalidomide causes striking immune activation, possibly leading to clearance of tumor cells. We conducted this study to investigate the mechanism of action of lenalidomide and the basis for its unique toxicities in chronic lymphocytic leukemia. Design and Methods Patients with relapsed chronic lymphocytic leukemia were treated with lenalidomide 20 mg (n=10) or 10 mg (n=8) daily for 3 weeks on a 6-week cycle. Correlative studies assessed expression of co-stimulatory molecules on tumor cells, T-cell activation, cytokine levels, and changes in lymphocyte subsets. Results Lenalidomide upregulated the co-stimulatory molecule CD80 on chronic lymphocytic leukemia and mantle cell lymphoma cells but not on normal peripheral blood B cells in vitro. T-cell activation was apparent in chronic lymphocytic leukemia, weak in mantle cell lymphoma, but absent in normal peripheral blood mononuclear cells and correlated with the upregulation of CD80 on B cells. Strong CD80 upregulation and T-cell activation predicted more severe side effects, manifesting in 83% of patients as a cytokine release syndrome within 8–72 h after the first dose of lenalidomide. Serum levels of various cytokines, including tumor necrosis factor-α, increased during treatment. CD80 upregulation on tumor cells correlated with rapid clearance of leukemic cells from the peripheral blood. In contrast, neither the severity of the cytokine release syndrome nor the degree of T-cell activation in vitro correlated with clinical response. Conclusions Upregulation of CD80 on tumor cells and T-cell activation correlate with unique toxicities of lenalidomide in chronic lymphocytic leukemia. However, T-cell activation appears to be dispensable for the drug’s anti-tumor effects. This provides a rationale for combinations of lenalidomide with fludarabine or alemtuzumab.


Leukemia & Lymphoma | 2011

Activation of CD44, a receptor for extracellular matrix components, protects chronic lymphocytic leukemia cells from spontaneous and drug induced apoptosis through MCL-1

Yair Herishanu; Federica Gibellini; Ndegwa Njuguna; Inbal Hazan-Halevy; Mohammed Farooqui; Sarah Bern; Keyvan Keyvanfar; Elinor Lee; Wyndham H. Wilson; Adrian Wiestner

Survival of chronic lymphocytic leukemia (CLL) cells in vivo is supported by the tissue microenvironment, which includes components of the extracellular matrix. Interactions between tumor cells and the extracellular matrix are in part mediated by CD44, whose principal ligand is hyaluronic acid. Here, we show that CD44 is more highly expressed on CLL cells of the clinically more progressive immunglobulin heavy chain variable gene (IGHV)-unmutated subtype than on cells of the IGHV-mutated type. Engagement of CD44 activated the phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen activated protein kinase (MAPK)/ERK pathways and increased myeloid cell leukemia sequence 1 (MCL-1) protein expression. Consistent with the induction of these anti-apoptotic mechanisms, CD44 protected CLL cells from spontaneous and fludarabine-induced apoptosis. Obatoclax, an antagonist of MCL-1, blocked the pro-survival effect of CD44. In addition, obatoclax synergized with fludarabine to induce apoptosis of CLL cells. In conclusion, components of the extracellular matrix may provide survival signals to CLL cells through engagement of CD44. Inhibition of MCL-1 is a promising strategy to reduce the anti-apoptotic effect of the microenvironment on CLL cells.


Leukemia & Lymphoma | 2008

Noxa mediates bortezomib induced apoptosis in both sensitive and intrinsically resistant mantle cell lymphoma cells and this effect is independent of constitutive activity of the AKT and NF-kappaB pathways

Edgar G. Rizzatti; Helena Mora-Jensen; Marc A. Weniger; Federica Gibellini; Elinor Lee; Masanori Daibata; Raymond Lai; Adrian Wiestner

Bortezomib is more active against mantle cell lymphoma (MCL) than against most other lymphoma subtypes. Nevertheless, up to half of patients with MCL have bortezomib resistant disease. Factors contributing to intrinsic resistance to bortezomib have not been determined. Here we used a panel of eight bortezomib sensitive (median IC50 5.9 nM) and three relatively bortezomib resistant cell lines (median IC50 12.9 nM) to investigate differences in tumor biology that could determine sensitivity to bortezomib. Bortezomib effectively inhibited high baseline proteasome activity and induced a comparable degree of proteasome inhibition in both sensitive and resistant cells. At 10 nM, bortezomib induced the proapoptotic BH3-only protein Noxa in sensitive but not resistant cells. At higher concentrations of bortezomib, however, Noxa was also upregulated in resistant cells and this effect was sufficient to induce apoptosis. Silencing of Noxa with siRNA rescued these cells from apoptosis, arguing against a defect in Noxa regulation or function as the basis of bortezomib resistance. Bortezomib was equally effective against cells with high and low constitutive NF-κB signaling. Also, sensitive and resistant MCL cell lines showed comparable activation of the AKT pathway. We conclude that bortezomib can overcome classic mechanisms of resistance to apoptosis and that determinants of bortezomib sensitivity in MCL are due to differences in signaling or stress pathways upstream of Noxa.


Clinical Cancer Research | 2012

ON 01910.Na is selectively cytotoxic for chronic lymphocytic leukemia cells through a dual mechanism of action involving PI3K/AKT inhibition and induction of oxidative stress.

Colby M. Chapman; Xiameng Sun; Mark Roschewski; Georg Aue; Mohamed Farooqui; Lawrence S Stennett; Federica Gibellini; Diane C. Arthur; Patricia Pérez-Galán; Adrian Wiestner

Purpose: Chronic lymphocytic leukemia (CLL), a malignancy of mature B cells, is incurable with chemotherapy. Signals from the microenvironment support leukemic cell survival and proliferation and may confer chemotherapy resistance. ON 01910.Na (Rigosertib), a multikinase phosphoinositide 3-kinase (PI3K) inhibitor, is entering phase III trials for myelodysplastic syndrome. Our aim was to analyze the efficacy of ON 01910.Na against CLL cells in vitro and investigate the molecular effects of this drug on tumor biology. Experimental Design: Cytotoxicity of ON 01910.Na against CLL cells from 34 patients was determined in vitro with flow cytometry of cells stained with Annexin V and CD19. Global gene expression profiling on Affymetrix microarrays, flow cytometry, Western blotting, and cocultures with stroma cells were used to delineate ON 01910.Na mechanism of action. Results: ON 01910.Na induced apoptosis in CLL B cells without significant toxicity against T cells or normal B cells. ON 01910.Na was equally active against leukemic cells associated with a more aggressive disease course [immunoglobulin heavy-chain variable region unmutated, adverse cytogenetics] than against cells without these features. Gene expression profiling revealed two main mechanisms of action: PI3K/AKT inhibition and induction of ROS that resulted in an oxidative stress response through activating protein 1 (AP-1), c-jun-NH2-terminal kinase, and ATF3 culminating in the upregulation of NOXA. ROS scavengers and shRNA mediated knockdown of ATF3- and NOXA-protected cells from drug-induced apoptosis. ON 01910.Na also abrogated the prosurvival effect of follicular dendritic cells on CLL cells and reduced SDF-1–induced migration of leukemic cells. Conclusions: These data support the clinical development of ON 01910.Na in CLL. Clin Cancer Res; 18(7); 1979–91. ©2012 AACR.


British Journal of Haematology | 2007

Decreased TCR ζ-chain expression in T cells from patients with acquired aplastic anaemia

Elena E. Solomou; Susan Wong; Valeria Visconte; Federica Gibellini; Neal S. Young

In aplastic anaemia, T cells have a central role in the pathophysiology of bone marrow destruction. This study showed that T cells from patients with aplastic anaemia expressed decreased T‐cell receptor (TCR) ζ‐chain protein and mRNA levels compared to healthy controls. Patients with decreased TCR ζ‐chain showed an abnormal response in intracellular calcium following stimulation through the TCR. We also observed an altered pattern of the transcription factors CREMα and Elf‐1 that are implicated in ζ‐chain transcription. We concluded that TCR ζ‐chain expression was decreased in the majority of patients with aplastic anaemia, regardless of disease activity or treatment status.


Experimental Hematology | 2009

Role of perforin-mediated cell apoptosis in murine models of infusion-induced bone marrow failure

Annahita Sarcon; Marie J. Desierto; Wenjun Zhou; Valeria Visconte; Federica Gibellini; Jichun Chen; Neal S. Young

OBJECTIVE To investigate the role of perforin-mediated cell apoptosis in murine models of immune-mediated bone marrow (BM) failure. MATERIALS AND METHODS We compared C57BL/6J (B6) mice carrying a perforin gene deletion (Prf(-/-)) with wild-type (WT) controls for cellular composition in lymphohematopoietic tissues. Lymph node (LN) cells from Prf(-/-) mice were coincubated with BM cells from B10-H2(b)/LilMcdJ (C.B10) mice in an apoptosis assay in vitro. We then infused Prf(-/-) and WT B6 LN cells into sublethally irradiated C.B10 and CByB6F1 recipients with mismatches at the minor and major histocompatibility loci, respectively, in order to induce BM failure. Cellular composition was analyzed by flow cytometry. RESULTS Prf(-/-) mice showed normal lymphoid cell composition, but Prf(-/-) LN cells had reduced ability to induce C.B10 BM cell apoptosis in vitro. Infusion of 5 to 10 x 10(6) Prf(-/-) LN cells produced obvious BM failure in C.B10 and CByB6F1 recipients; pancytopenia and BM hypocellularity were only slightly less severe than those caused by infusion of 5 x 10(6) WT B6 LN cells. Infused Prf(-/-) LN cells showed less T-cell expansion, normal T-cell activation, and higher proportions of T cells expressing gamma-interferon, tissue necrosis factor-alpha, and Fas ligand CD178, in comparison to infused WT B6 LN cells. Fas expression was equally high in residual BM cells in recipient of both Prf(-/-) and B6 LN cells. CONCLUSION Perforin deficiency alters T-cell expansion but upregulates T-cell Fas ligand expression. Perforin-mediated cell death appears to play a minor role in mouse models of immune-mediated BM failure.


Cancer Research | 2010

Abstract 3493: ON 01910. Na, a clinical trial stage multi-kinase inhibitor, induces apoptosis in chronic lymphocytic leukemia (CLL) cells through inhibition of PI3K/AKT and activation of the JNK pathway resulting in NOXA and BIM upregulation

Patricia Pérez-Galán; Colby M. Chapman; Xiameng Sun; Federica Gibellini; Poching Liu; Nalini Raghavachari; Adrian Wiestner

Chronic Lymphocytic leukemia (CLL), the most common leukemia in Western countries, is a clinically heterogeneous disease characterized by the accumulation of mature B lymphocytes, where disease progression is driven by survival and proliferation signals provided by the tumor microenvironment. ON 01910. Na (Onconova Therapeutics) belongs to the family of styryl benzyl sulfones, a novel family of non-ATP competitive kinase inhibitors. ON 01910. Na is under clinical development in hematologic and solid tumors. Here we show that ON 01910. Na induced apoptosis in CLL samples (n=28) in vitro, without showing significant toxicity against T-cells or normal B-cells. Twenty four CLL samples were highly sensitive to ON 01910. Na (mean IC50=0.89 μM) a concentration readily achieved in phase I clinical trials, three showed moderate sensitivity (mean IC50=4.26 μM) and one was resistant (IC50 >8 μM). There were no significant differences in ON01910. Na cytotoxicity against CLL cells expressing mutated (mean IC50=1.33 μM) or unmutated (mean IC50 =1.22μM) IgVH sequences. ON 01910. Na was similarly effective against tumor cells carrying 17p or 11q deletions. ON 01910. Na activated Bax and Bak, leading to mitochondrial depolarization. To delineate the upstream pathways underlying ON 01910. Na induced apoptosis, we performed Gene Expression Profiling (GEP) in CLL cells treated in vitro for 4 and 10 hours with ON 01910. Na. Gene Set Enrichment Analysis (GSEA) identified gene sets indicating BCR and PI3K inhibition (FDR Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3493.


Blood | 2007

Point mutations and genomic deletions in CCND1 create stable truncated cyclin D1 mRNAs that are associated with increased proliferation rate and shorter survival

Adrian Wiestner; Mahsa Tehrani; Michael Chiorazzi; George E. Wright; Federica Gibellini; Kazutaka Nakayama; Hui Liu; Andreas Rosenwald; H. Konrad Muller-Hermelink; German Ott; Wing C. Chan; Timothy C. Greiner; Dennis D. Weisenburger; Julie M. Vose; James O. Armitage; Randy D. Gascoyne; Joseph M. Connors; Elias Campo; Emilio Montserrat; Francesc Bosch; Erlend B. Smeland; Stein Kvaløy; Harald Holte; Jan Delabie; Richard I. Fisher; Thomas M. Grogan; Thomas P. Miller; Wyndham H. Wilson; Elaine S. Jaffe; Louis M. Staudt


Blood | 2007

Perforin gene mutations in patients with acquired aplastic anemia

Elena E. Solomou; Federica Gibellini; Brian J. Stewart; Daniela Malide; Maria Berg; Valeria Visconte; Spencer W. Green; Richard Childs; Stephen J. Chanock; Neal S. Young

Collaboration


Dive into the Federica Gibellini's collaboration.

Top Co-Authors

Avatar

Wyndham H. Wilson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elinor Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nalini Raghavachari

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Colby M. Chapman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Delong Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gerald E. Marti

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge