Network


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

Hotspot


Dive into the research topics where Ezia Bello is active.

Publication


Featured researches published by Ezia Bello.


Cancer Research | 2011

E-3810 Is a Potent Dual Inhibitor of VEGFR and FGFR that Exerts Antitumor Activity in Multiple Preclinical Models

Ezia Bello; Gennaro Colella; Valentina Scarlato; Paolo Oliva; Alexander Berndt; Giovanni Valbusa; Sonia Colombo Serra; Maurizio D'Incalci; Ennio Cavalletti; Raffaella Giavazzi; Giovanna Damia; Gabriella Camboni

Tumor angiogenesis is a degenerate process regulated by a complex network of proangiogenic factors. Existing antiangiogenic drugs used in clinic are characterized by selectivity for specific factors. Antiangiogenic properties might be improved in drugs that target multiple factors and thereby address the inherent mechanistic degeneracy in angiogenesis. Vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) family members and their cognate receptors are key players in promoting tumor angiogenesis. Here we report the pharmacologic profile of E-3810, a novel dual inhibitor of the VEGF and FGF receptors. E-3810 potently and selectively inhibited VEGF receptor (VEGFR)-1, -2, and -3 and FGF receptor (FGFR)-1 and -2 kinases in the nanomolar range. Ligand-dependent phosphorylation of VEGFR-2 and FGFR-1 was suppressed along with human vascular endothelial cell growth at nanomolar concentrations. In contrast, E-3810 lacked cytotoxic effects on cancer cell lines under millimolar concentrations. In a variety of tumor xenograft models, including early- or late-stage subcutaneous and orthotopic models, E-3810 exhibited striking antitumor properties at well-tolerated oral doses administered daily. We found that E-3810 remained active in tumors rendered nonresponsive to the general kinase inhibitor sunitinib resulting from a previous cycle of sunitinib treatment. In Matrigel plug assays performed in nude mice, E-3810 inhibited basic FGF-induced angiogenesis and reduced blood vessel density as assessed by histologic analysis. Dynamic contrast-enhanced magnetic resonance imaging analysis confirmed that E-3810 reduced the distribution of angiogenesis-sensitive contrast agents after only 5 days of treatment. Taken together, our findings identify E-3810 as a potent antiangiogenic small molecule with a favorable pharmacokinetic profile and broad spectrum antitumor activity, providing a strong rationale for its clinical evaluation.


Oncogene | 2014

Mode of action of trabectedin in myxoid liposarcomas

S Di Giandomenico; Roberta Frapolli; Ezia Bello; Sarah Uboldi; S A Licandro; Sergio Marchini; Luca Beltrame; Silvia Brich; V Mauro; Elena Tamborini; Silvana Pilotti; Paolo G. Casali; Federica Grosso; Roberta Sanfilippo; Alessandro Gronchi; Roberto Mantovani; Raffaella Gatta; Carlos M. Galmarini; J M F Sousa-Faro; Maurizio D'Incalci

To elucidate the mechanisms behind the high sensitivity of myxoid/round cell liposarcoma (MRCL) to trabectedin and the suggested selectivity for specific subtypes, we have developed and characterized three MRCL xenografts, namely ML017, ML015 and ML004 differing for the break point of the fusion gene FUS-CHOP, respectively of type I, II and III. FUS-CHOP binding to the promoters of some target genes such as Pentraxin 3 or Fibronectin 1, assessed by chromatin immunoprecipitation, was strongly reduced in the tumor 24 h after the first or the third weekly dose of trabectedin, indicating that the drug at therapeutic doses causes a detachment of the FUS-CHOP chimera from its target promoters as previously shown in vitro. Moreover, the higher sensitivity of MRCL types I and II appears to be related to a more prolonged block of the transactivating activity of the fusion protein. Doxorubicin did not affect the binding of FUS-CHOP to target promoters. Histologically, the response to trabectedin in ML017 and ML015 was associated with a marked depletion of non-lipogenic tumoral cells and vascular component, as well as lipidic maturation as confirmed by PPARγ2 expression in western Blot. By contrast, in ML004 no major changes either in the cellularity or in the amount of mature were found, and consistently PPARγ2 was null. In conclusion, the data support the view that the selective mechanism of action of trabectedin in MRCL is specific and related to its ability to cause a functional inactivation of the oncogenic chimera with consequent derepression of the adypocytic differentiation.


Food and Chemical Toxicology | 2011

Chemical characterization of Iraqi propolis samples and assessing their antioxidant potentials.

Ghassan M. Sulaiman; Khulood W. Al Sammarrae; Ali H. Ad’hiah; Massimo Zucchetti; Roberta Frapolli; Ezia Bello; Eugenio Erba; Maurizio D’Incalci; Renzo Bagnati

Propolis samples, collected from different geographical locations in Iraq (Baghdad, Dahuk, Mosul and Salah ad-Din), were analyzed and assessed for their anti-oxidant activity. Concentrations of phenolic compounds (flavonoids, phenolic acids and their esters) in propolis were estimated using high performance liquid chromatography coupled to electrospray mass spectrometry. Thirty-eight different compounds were identified and 33 of them were polyphenols. Other compounds were tentatively identified as clerodane diterpenoids, and one was considered unknown. Semi-quantitative measurements showed that phenolic acids and their esters were the predominant constituents in propolis extracts, followed by flavones and flavonols, and then flavanones and dihydroflavonols. Propolis samples were further spectrophotometrically characterized using the Folin-Ciocalteu reagent for the determination of total phenolic compounds. The free radical scavenging activities of propolis samples were also evaluated by using the 2,2-diphenyl-1-picrylhydrazyl assay. The results revealed that propolis extracts exhibited strong free radical scavenging activity.


Molecular Cancer Therapeutics | 2013

The Tyrosine Kinase Inhibitor E-3810 Combined with Paclitaxel Inhibits the Growth of Advanced-Stage Triple-Negative Breast Cancer Xenografts

Ezia Bello; Giulia Taraboletti; Gennaro Colella; Massimo Zucchetti; Daniele Forestieri; Simonetta Andrea Licandro; Alexander Berndt; Petra Richter; Maurizio D'Incalci; Ennio Cavalletti; Raffaella Giavazzi; Gabriella Camboni; Giovanna Damia

E-3810 is a novel small molecule that inhibits VEGF receptor-1, -2, and -3 and fibroblast growth factor receptor-1 tyrosine kinases at nmol/L concentrations currently in phase clinical II. In preclinical studies, it had a broad spectrum of antitumor activity when used as monotherapy in a variety of human xenografts. We here investigated the activity of E-3810 combined with different cytotoxic agents in a MDA-MB-231 triple-negative breast cancer xenograft model. The molecule could be safely administered with 5-fluorouracil, cisplatin, and paclitaxel. The E-3810–paclitaxel combination showed a striking activity with complete, lasting tumor regressions; the antitumor activity of the combination was also confirmed in another triple-negative breast xenograft, MX-1. The activity was superior to that of the combinations paclitaxel+brivanib and paclitaxel+sunitinib. Pharmacokinetics studies suggest that the extra antitumor activity of the combination is not due to higher paclitaxel tumor levels, which in fact were lower in mice pretreated with all three kinase inhibitors, and the paclitaxel plasma levels excluded reduced drug availability. Pharmacodynamic studies showed that E-3810, brivanib, and sunitinib given as single agents or in combination with paclitaxel reduced the number of vessels, but did not modify vessel maturation. Reduced tumor collagen IV and increased plasma collagen IV, associated with increased matrix metalloproteinases (MMP), particularly host MMP-9, indicate a proteolytic remodeling of the extracellular matrix caused by E-3810 that in conjunction with the cytotoxic effect of paclitaxel on the tumor cells (caspase-3/7 activity) may contribute to the striking activity of their combination. These data support the therapeutic potential of combining E-3810 with conventional chemotherapy. Mol Cancer Ther; 12(2); 131–40. ©2012 AACR.


International Journal of Cancer | 2013

Comparison of in vitro and in vivo biological effects of trabectedin, lurbinectedin (PM01183) and Zalypsis® (PM00104).

Michela Romano; Roberta Frapolli; Monique Zangarini; Ezia Bello; Luca Porcu; Carlos M. Galmarini; Luis García-Fernández; Carmen Cuevas; Paola Allavena; Eugenio Erba; Maurizio D'Incalci

This study: (i) investigated the in vitro cytotoxicity and mode of action of lurbinectedin (PM01183) and Zalypsis® (PM00104) compared with trabectedin in cell lines deficient in specific mechanisms of repair, (ii) evaluated their in vivo antitumor activity against a series of murine tumors and human xenografts. The antiproliferative activity, the DNA damage and the cell cycle perturbations induced by the three compounds on tumor lines were very similar. Nucleotide Excision Repair (NER) deficient cells were approximately fourfold more resistant to trabectedin, lurbinectedin and Zalypsis®. Cells deficient in non‐homologous end joining (NHEJ), MRN complex and translesion synthesis (TLS) were slightly more sensitive to the three compounds (approximately fivefold) while cells deficient in homologous recombination (HR) were markedly more sensitive (150–200‐fold). All three compounds showed a good antitumor activity in several in vivo models. Lurbinectedin and trabectedin had a similar pattern of antitumor activity in murine tumors and in xenografts, whereas Zalypsis® appeared to have a distinct spectrum of activity. The fact that no relationship whatsoever was found between the in vitro cytotoxic potency and the in vivo antitumor activity, suggests that in addition to direct cytotoxic mechanisms other host‐mediated effects are involved in the in vivo pharmacological effects.


Clinical Cancer Research | 2010

Novel Models of Myxoid Liposarcoma Xenografts Mimicking the Biological and Pharmacologic Features of Human Tumors

Roberta Frapolli; Elena Tamborini; Emanuela Virdis; Ezia Bello; Eva Tarantino; Sergio Marchini; Federica Grosso; Roberta Sanfilippo; Alessandro Gronchi; Juan Carlos Tercero; G. Peloso; Paolo G. Casali; Silvana Pilotti; Maurizio D'Incalci

Purpose: Myxoid liposarcoma is a common subtype of liposarcoma. It is associated in more than 90% of cases with the chromosomal translocation t(12;16)(q13;p11) leading to the fusion FUS-CHOP gene that is responsible for the oncogenic transformation of preadipocytes. Recently the marine natural product trabectedin has shown highly selective activity for myxoid liposarcoma, even in the most aggressive round-cell subtype. Experimental Design: Fragments of 17 sarcomas were transplanted s.c. in female athymic NCr-nu/nu mice. Xenografts were established and characterized by morphology, fluorescence in situ hybridization analysis for the translocation and reverse transcriptase-PCR analysis for fusion transcripts. Trabectedin was injected i.v. Results: Seven of 17 tumors grew as continuous xenografts, five of them being myxoid liposarcoma of the round-cell subtype. The chromosomal rearrangement and fusion transcripts in different passages were the same as in the human tumors from which they were derived. The responsiveness to trabectedin in type II myxoid liposarcoma xenografts was as high as in patients. The pathologic response was associated with the presence of the FUS-CHOP fusion gene, indicating that the drug does not totally eradicate the disease. Type III myxoid liposarcoma xenografts seemed much less sensitive to trabectedin, confirming previous clinical observations. Conclusions: This study reports for the first time the characterization of human myxoid liposarcoma xenografts that adequately mimic the biological and pharmacologic features of the human tumor. These models offer a useful tool for investigating the mechanism of selectivity of trabectedin, testing new combinations with this drug and evaluating novel therapies for myxoid liposarcoma. Clin Cancer Res; 16(20); 4958–67. ©2010 AACR.


Molecular Pharmaceutics | 2016

PEGylated Nanoparticles Obtained through Emulsion Polymerization as Paclitaxel Carriers

Claudio Colombo; Lavinia Morosi; Ezia Bello; Raffaele Ferrari; Simonetta Andrea Licandro; Monica Lupi; Paolo Ubezio; Massimo Morbidelli; Massimo Zucchetti; Maurizio D’Incalci; Davide Moscatelli; Roberta Frapolli

Polymer nanoparticles (NPs) represent a promising way to deliver poorly water-soluble anticancer drugs without the use of unwanted excipients, whose presence can be the cause of severe side effects. In this work, a Cremophor-free formulation for paclitaxel (PTX) has been developed by employing PEGylated polymer nanoparticles (NPs) as drug delivery carriers based on modified poly(ε-caprolactone) macromonomers and synthesized through free radical emulsion polymerization. Paclitaxel was loaded in the NPs in a postsynthesis process which allowed to obtain a drug concentration suitable for in vivo use. In vivo experiments on drug biodistribution and therapeutic efficacy show comparable behavior between the NPs and the Cremophor formulation, also showing good tolerability of the new formulation proposed.


Cancer Research | 2006

Biological Properties of IDN5174, a New Synthetic Camptothecin with the Open Lactone Ring

Giovanni Luca Beretta; Giovanna Petrangolini; Michelandrea De Cesare; Graziella Pratesi; Paola Perego; Stella Tinelli; Monica Tortoreto; Massimo Zucchetti; Roberta Frapolli; Ezia Bello; Carla Manzotti; Gabriele Fontana; Ezio Bombardelli; Arturo Battaglia; Cristian Samorì; Franco Zunino

A series of water-soluble camptothecins obtained by linking a spermidine moiety to the 21-position of the open form through an amidic bond have been tested for their biochemical and biological activities. Growth inhibition assay on the human non-small cell lung cancer carcinoma NCI-H460 cell line revealed that the camptothecin analogues were less potent than topotecan and SN38 after 1 hour of treatment. The potency increased after 72 hours of exposure, being similar to that of reference camptothecins. The analysis of topoisomerase I-mediated DNA cleavage using the purified enzyme indicated that the novel camptothecin analogues retained ability to poison topoisomerase I and displayed the same cleavage pattern of SN38. Persistence of the DNA cleavage was comparable with that of SN38. Stabilization of the cleavable complex was not the result of hydrolysis of the N-C bond between polyamine and the drug because no free camptothecin was recovered at the end of DNA cleavage in presence of IDN5174, the analogue selected for detailed studies. IDN5174 exhibited an antitumor activity comparable with that of topotecan and irinotecan against NCI-H460 tumor xenograft. The pharmacokinetics in mice showed a favorable disposition in tumor tissue with low amount of camptothecin detectable in plasma and tumor (around 5-10%), thus supporting the efficacy of intact IDN5174. In conclusion, we found that IDN5174 maintained the biological and antitumor properties, in spite of lack of the closed E ring. The available results support the interpretation that the polyamine linked at the 21-position may allow a favorable drug interaction in the ternary complex.


Nanotechnology | 2014

A biodistribution study of PEGylated PCL-based nanoparticles in C57BL/6 mice bearing B16/F10 melanoma

Monica Lupi; Claudio Colombo; Roberta Frapolli; Raffaele Ferrari; Leopoldo Sitia; Luca Dragoni; Ezia Bello; Simonetta Andrea Licandro; Francesca Falcetta; Paolo Ubezio; Paolo Bigini; Mario Salmona; Maurizio D'Incalci; Massimo Morbidelli; Davide Moscatelli

One of the major drawbacks that limits the clinical application of nanoparticles is the lack of preliminary investigations related to their biocompatibility, biodegradability and biodistribution. In this work, biodegradable PEGylated polymer nanoparticles (NPs) have been synthesized by using macromonomers based on poly(ε-caprolaconte) oligomers. More in detail, NPs have been produced by adopting a surfactant-free semibatch emulsion polymerization process using PEG chains as a stabilizing agent. The NPs were also labeled with rhodamine B covalently bound to the NPs to quantitatively study their biodistribution in vivo. NPs were investigated in both in vitro and in vivo preclinical systems to study their biodistribution in mice bearing B16/F10 melanoma, as well as their biocompatibility and biodegradability. The NP concentration was evaluated in different tissues at several times after intravenous injection. The disappearance of the NPs from the plasma was biphasic, with distribution and elimination half-lives of 30 min and 15 h, respectively. NPs were retained in tumors and in filter organs for a long time, were still detectable after 7 d and maintained a steady concentration in the tumor for 120 h. 48 h after injection, 70 ± 15% of the inoculated NPs were excreted in the feces. The favorable tumor uptake, fast excretion and absence of cytotoxicity foster the further development of produced NPs as drug delivery carriers.


Food and Chemical Toxicology | 2012

Assessing the anti-tumour properties of Iraqi propolis in vitro and in vivo

Ghassan M. Sulaiman; Ali H. Ad'hiah; Khulood W. Al-Sammarrae; Renzo Bagnati; Roberta Frapolli; Ezia Bello; Sarah Uboldi; Michela Romano; Nicolò Panini; Eugenio Scanziani; Marzia Pezzolato; Eugenio Erba; Maurizio D'Incalci

The study was designed to evaluate anti-tumour properties of Iraqi propolis collected from Mosul region (M) on HL-60 and HCT-116 cell lines and on HCT-116 in vivo. M induced an inhibitory effect against the proliferation of HL-60 and colony potential of HCT-116 cells. The apoptosis in HL-60 cells was associated with down-regulation of Bcl-2 and activation of Bax, while in HCT-116 cells, necrotic features were observed; size of cells was dramatically increased by swelling of cytoplasm and loss of membrane integrity, cell rupture and release of cellular contents. Analysis of BrdU/DNA cell cycle in both cell lines showed that M induced cell cycle perturbations in both BrdU positive and BrdU negative cells. The exposure of HL-60 to M caused γ-H2AX in a dose dependent manner and was associated with induction of apoptosis. The experiments in HCT-116 tumor-bearing mice showed that oral administration of propolis at doses that caused no detectable toxicity was associated with a decrease in mitotic cells and an increase in endoreduplications, increased p53 and decreased Ki-67 expression of cells in tumor sections. This study provides the rationale to investigate the potential beneficial effect of propolis in the diet of patients receiving anti-cancer therapies.

Collaboration


Dive into the Ezia Bello's collaboration.

Top Co-Authors

Avatar

Roberta Frapolli

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Maurizio D'Incalci

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Massimo Zucchetti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Giovanna Damia

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Eugenio Erba

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Maurizio D’Incalci

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Michela Romano

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Federica Grosso

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sergio Marchini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge