Network


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

Hotspot


Dive into the research topics where Antonio Basile is active.

Publication


Featured researches published by Antonio Basile.


Clinical Cancer Research | 2011

Lenalidomide restrains motility and overangiogenic potential of bone marrow endothelial cells in patients with active multiple myeloma

Annunziata De Luisi; Arianna Ferrucci; Addolorata Coluccia; Roberto Ria; Michele Moschetta; Emanuela de Luca; Luisa Pieroni; Michele Maffia; Andrea Urbani; Giulia Di Pietro; Attilio Guarini; Girolamo Ranieri; Paolo Ditonno; Simona Berardi; Antonella Caivano; Antonio Basile; Nicola Cascavilla; Silvana Capalbo; Giovanni Quarta; Franco Dammacco; Domenico Ribatti; Angelo Vacca

Purpose: To determine the in vivo and in vitro antiangiogenic power of lenalidomide, a “lead compound” of IMiD immunomodulatory drugs in bone marrow (BM) endothelial cells (EC) of patients with multiple myeloma (MM) in active phase (MMEC). Experimental Design: The antiangiogenic effect in vivo was studied using the chorioallantoic membrane (CAM) assay. Functional studies in vitro (angiogenesis, “wound” healing and chemotaxis, cell viability, adhesion, and apoptosis) were conducted in both primary MMECs and ECs of patients with monoclonal gammopathies (MGUS) of undetermined significance (MGEC) or healthy human umbilical vein endothelial cells (HUVEC). Real-time reverse transcriptase PCR, Western blotting, and differential proteomic analysis were used to correlate morphologic and biological EC features with the lenalidomide effects at the gene and protein levels. Results: Lenalidomide exerted a relevant antiangiogenic effect in vivo at 1.75 μmol/L, a dose reached in interstitial fluids of patients treated with 25 mg/d. In vitro, lenalidomide inhibited angiogenesis and migration of MMECs, but not of MGECs or control HUVECs, and had no effect on MMEC viability, apoptosis, or fibronectin- and vitronectin-mediated adhesion. Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways. Conclusions: This study provides information on the molecular mechanisms associated with the antimigratory and antiangiogenic effects of lenalidomide in primary MMECs, thus giving new avenues for effective endothelium-targeted therapies in MM. Clin Cancer Res; 17(7); 1935–46. ©2011 AACR.


Leukemia | 2014

Bone marrow fibroblasts parallel multiple myeloma progression in patients and mice: in vitro and in vivo studies

Maria Antonia Frassanito; Luigia Rao; Michele Moschetta; Roberto Ria; L. Di Marzo; A De Luisi; Vito Racanelli; Ivana Catacchio; Simona Berardi; Antonio Basile; Eline Menu; Simona Ruggieri; Beatrice Nico; Domenico Ribatti; Ruggiero Fumarulo; Franco Dammacco; Karin Vanderkerken; Angelo Vacca

The role of cancer-associated fibroblasts (CAFs) has not been previously studied in multiple myeloma (MM). Here, cytofluorimetric analysis revealed higher proportions of bone marrow (BM) CAFs in patients with active MM (both at diagnosis and relapse) compared with patients in remission or those with monoclonal gammopathy of undetermined significance or deficiency anemia (controls). CAFs from MM patients produced increased levels of transforming growth factor-β, interleukin-6, stromal cell-derived factor-1α, insulin-like growth factor-1, vascular endothelial growth factor and fibroblast growth factor-2 and displayed an activated and heterogeneous phenotype, which supported their origin from resident fibroblasts, endothelial cells and hematopoietic stem and progenitor cells via the endothelial–mesenchymal transition as well as mesenchymal stem cells via the mesenchymal transition, as both of these processes are induced by MM cells and CAFs. Active MM CAFs fostered chemotaxis, adhesion, proliferation and apoptosis resistance in MM cells through cytokine signals and cell-to-cell contact, which were inhibited by blocking CXCR4, several integrins and fibronectin. MM cells also induced the CAFs proliferation. In syngeneic 5T33MM and xenograft mouse models, MM cells induced the expansion of CAFs, which, in turn, promoted MM initiation and progression as well as angiogenesis. In BM biopsies from patients and mice, nests of CAFs were found in close contact with MM cells, suggesting a supportive niche. Therefore, the targeting of CAFs in MM patients may be envisaged as a novel therapeutic strategy.


Clinical Cancer Research | 2013

Novel targeting of phospho-cMET overcomes drug resistance and induces antitumor activity in multiple myeloma

Michele Moschetta; Antonio Basile; Arianna Ferrucci; Maria Antonia Frassanito; Luigia Rao; Roberto Ria; Antonio Giovanni Solimando; Nicola Giuliani; Angelina Boccarelli; Fabio Fumarola; Mauro Coluccia; Bernardo Rossini; Simona Ruggieri; Beatrice Nico; Eugenio Maiorano; Domenico Ribatti; Aldo M. Roccaro; Angelo Vacca

Purpose: The aim of the study was to verify the hypothesis that the cMet oncogene is implicated in chemio- and novel drug resistance in multiple myeloma. Experimental Design: We have evaluated the expression levels of cMET/phospho-cMET (p-cMET) and the activity of the novel selective p-cMET inhibitor (SU11274) in multiple myeloma cells, either sensitive (RPMI-8226 and MM.1S) or resistant (R5 and MM.1R) to anti–multiple myeloma drugs, in primary plasma cells and in multiple myeloma xenograft models. Results: We found that resistant R5 and MM.1R cells presented with higher cMET phosphorylation, thus leading to constitutive activation of cMET-dependent signaling pathways. R5 cells exhibited a higher susceptibility to the SU11274 inhibitory effects on viability, proliferation, chemotaxis, adhesion, and to its apoptogenic effects. SU11274 was able to revert drug resistance in R5 cells. R5 but not RPMI-8226 cells displayed cMET-dependent activation of mitogen-activated protein kinase pathway. The cMET and p-cMET expression was higher on plasma cells from patients with multiple myeloma at relapse or on drug resistance than on those from patients at diagnosis, complete/partial remission, or from patients with monoclonal gammopathy of unknown significance. Viability, chemotaxis, adhesion to fibronectin or paired bone marrow stromal cells of plasma cells from relapsed or resistant patients was markedly inhibited by SU11274. Importantly, SU11274 showed higher therapeutic activity in R5- than in RPMI-8226–induced plasmocytomas. In R5 tumors, it caused apoptosis and necrosis and reverted bortezomib resistance. Conclusion: Our findings suggest that the cMET pathway is constitutively activated in relapsed and resistant multiple myeloma where it may also be responsible for induction of drug resistance, thus providing the preclinical rationale for targeting cMET in patients with relapsed/refractory multiple myeloma. Clin Cancer Res; 19(16); 4371–82. ©2013 AACR.


Oncogene | 2012

Four proteins governing overangiogenic endothelial cell phenotype in patients with multiple myeloma are plausible therapeutic targets

Simona Berardi; Antonella Caivano; Roberto Ria; Beatrice Nico; R Savino; R Terracciano; G De Tullio; Arianna Ferrucci; A De Luisi; Michele Moschetta; G. Mangialardi; Ivana Catacchio; Antonio Basile; Attilio Guarini; Alfredo Zito; Pasquale Ditonno; P Musto; Franco Dammacco; Domenico Ribatti; Angelo Vacca

Bone marrow (BM) angiogenesis has an important role in the initiation and progression of multiple myeloma (MM). We looked at novel mechanisms of vessel formation in patients with MM through a comparative proteomic analysis between BM endothelial cells (ECs) of patients with active MM (MMECs) and ECs of patients with monoclonal gammopathy of undetermined significance (MGECs) and of subjects with benign anemia (normal ECs). Four proteins were found overexpressed in MMECs: filamin A, vimentin, α-crystallin B and 14-3-3ζ/δ protein, not yet linked to overangiogenic phenotype. These proteins gave a typical distribution in the BM of MM patients and in MMECs versus MGECs, plausibly according to a different functional state. Their expression was enhanced by vascular endothelial growth factor, fibroblast growth factor 2, hepatocyte growth factor and MM plasma cell conditioned medium in step with enhancement of MMEC angiogenesis. Their silencing RNA knockdown affected critical MMEC angiogenesis-related functions, such as spreading, migration and tubular morphogenesis. A gradual stabilization of 14-3-3ζ/δ protein was observed, with transition from normal ECs to MGECs and MMECs that may be a critical step for the angiogenic switch in MMECs and maintenance of the cell overangiogenic phenotype. These proteins were substantially impacted by anti-MM drugs, such as bortezomib, lenalidomide and panobinostat. Results suggest that these four proteins could be new targets for the antiangiogenic management of MM patients.


Expert Opinion on Therapeutic Targets | 2012

Tumor endothelial markers as a target in cancer

Domenico Ribatti; Girolamo Ranieri; Antonio Basile; Amalia Azzariti; Angelo Paradiso; Angelo Vacca

Introduction: Several anti-angiogenic agents have been developed and some of them have been clinically applied in the tumor therapy. Anti-angiogenic therapy faces some hurdles: inherent or acquired resistance, increased invasiveness, and lack of biomarkers. Characterization of tumor endothelial markers may help to target endothelium and to identify potential predictive factors of response to anti-angiogenic therapies. Numerous surrogates, angiogenic and endothelium markers have emerged from recent pre-clinical studies, including physiological and soluble molecules in plasma and from platelets, circulating cells, tumor tissue factors and imaging markers. However, no wholly validated biomarkers currently exist to predict the success or the failure of the anti-angiogenic therapy of cancer. Therefore, the research of suitable and validate biomarkers is currently ongoing. Areas covered: This review provides an overview of the status of our knowledge concerning tumor endothelial markers, therapeutics targeting, possible resistance mechanisms and predictive value of these biomarkers and discuss future strategies to use and identify them in the anti-angiogenic therapy. Expert opinion: Anti-angiogenesis is a milestone to improve the treatment of several types of cancer and predictive biomarkers for a response to anti-endothelium therapy are one of the most important challenges for anti-angiogenesis research.


The Journal of Pathology | 2013

Pentraxin 3 (PTX3) inhibits plasma cell/stromal cell cross‐talk in the bone marrow of multiple myeloma patients

Antonio Basile; Michele Moschetta; Paolo Ditonno; Roberto Ria; Ilaria Marech; Annunziata De Luisi; Simona Berardi; Maria Antonia Frassanito; Emanuele Angelucci; Daniele Derudas; Giorgina Specchia; Paola Curci; Vincenzo Pavone; Bernardo Rossini; Domenico Ribatti; Barbara Bottazzi; Alberto Mantovani; Marco Presta; Franco Dammacco; Angelo Vacca

Pentraxin 3 (PTX3) is a soluble pattern recognition receptor that binds with high affinity and selectivity to fibroblast growth factor‐2 (FGF2), thus inhibiting its pro‐angiogenic activity. Here we investigated the effects of PTX3 on monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma (MM) patient‐derived bone marrow (BM) plasma cells (PCs), endothelial cells (ECs), and fibroblasts (FBs), and assessed whether PTX3 can modulate the cross‐talk between PCs and those microenvironment cells. PTX3 and FGF2 expression was evaluated by ELISA. Functional studies, including cell viability, wound healing, chemotaxis, and Matrigel® assays, were performed on MGUS and MM ECs and FBs upon the PTX3 treatment. Through western blot PTX3‐induced modulation in FGF2/FGF receptor signalling pathways was evaluated in MGUS and MM ECs and FBs through western blot. Co‐cultures between MM ECs/FBs and human PC lines were used to evaluate possible PTX3 indirect effects on MM PCs. Adhesion molecules were studied by flow cytometry. PTX3 provides a direct time‐ and dose‐dependent apoptotic effect on MM ECs and FBs, but not on either MM primary PCs or human PC lines. PTX3 inhibits migration of MM ECs and FBs in a dose‐dependent manner, and impacts in vitro and in vivo FGF2‐mediated MM angiogenesis. Co‐cultures of PCs and ECs/FBs show that PTX3 treatment indirectly impairs PC viability and adhesion. We conclude that PTX3 is an anti‐angiogenic factor in MM and behaves as a cytotoxic molecule on MM cells by inhibiting the cross‐talk between PCs and ECs/FBs. Copyright


Oncotarget | 2017

Epha3 acts as proangiogenic factor in multiple myeloma

Antonella Caivano; Francesco La Rocca; Ilaria Laurenzana; Tiziana Annese; Roberto Tamma; Ubaldo Famigliari; Vittorio Simeon; Stefania Trino; Luciana De Luca; Oreste Villani; Simona Berardi; Antonio Basile; Angelo Vacca; Giuseppe Saglio; Luigi Del Vecchio; Pellegrino Musto; Daniela Cilloni

This study investigates the role of ephrin receptor A3 (EphA3) in the angiogenesis of Multiple Myeloma (MM) and the effects of a selective target of EphA3 by a specific monoclonal antibody on primary bone marrow endothelial cells (ECs) of MM patients. EphA3 mRNA and protein were evaluated in ECs of MM patients (MMECs), in ECs of patients with monoclonal gammopathies of undetermined significance (MGECs) and in ECs of healthy subjects (control ECs). The effects of EphA3 targeting by mRNA silencing (siRNA) or by the anti EphA3 antibody on the angiogenesis were evaluated. We found that EphA3 is highly expressed in MMECs compared to the other EC types. Loss of function of EphA3 by siRNA significantly inhibited the ability of MMECs to adhere to fibronectin, to migrate and to form tube like structures in vitro, without affecting cell proliferation or viability. In addition, gene expression profiling showed that knockdown of EphA3 down modulated some molecules that regulate adhesion, migration and invasion processes. Interestingly, EphA3 targeting by an anti EphA3 antibody reduced all the MMEC angiogenesis-related functions in vitro. In conclusion, our findings suggest that EphA3 plays an important role in MM angiogenesis.


Frontiers in Bioscience | 2014

Bone marrow vascular niche and the control of angiogenesis in multiple myeloma.

Domenico Ribatti; Antonio Basile; Simona Ruggieri; Angelo Vacca


Blood | 2013

EphA3 As a Molecular Target In Multiple Myeloma: Opportunity For a Novel Therapeutic Approach With a Specific Monoclonal Antibody

Francesco La Rocca; Ilaria Laurenzana; Alessandra Favole; Irma Airoldi; Emma Di Carlo; Valentina Gaidano; Enrico Bracco; Oreste Villani; Giuseppe Pietrantuono; Luciana De Luca; Annalisa Morano; Vittorio Simeon; Stefania Trino; Simona Berardi; Antonio Basile; Martin Lackmann; Angelo Vacca; Pellegrino Musto; Francesco Frassoni; Daniela Cilloni


European Journal of Internal Medicine | 2013

Proangiogenic effect of bone marrow fibroblasts in patients with multiple myeloma

Antonio Giovanni Solimando; L. Di Marzo; Luigia Rao; Antonio Basile; Roberto Ria; Maria Antonia Frassanito; Angelo Vacca

Collaboration


Dive into the Antonio Basile's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge