Network


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

Hotspot


Dive into the research topics where Arianna Giacomini is active.

Publication


Featured researches published by Arianna Giacomini.


Cancer Cell | 2015

Long-Pentraxin 3 Derivative as a Small-Molecule FGF Trap for Cancer Therapy

Roberto Ronca; Arianna Giacomini; Emanuela Di Salle; Daniela Coltrini; Katiuscia Pagano; Laura Ragona; Sara Matarazzo; Sara Rezzola; Daniele Maiolo; Rubben Torella; Elisabetta Moroni; Roberta Mazzieri; Giulia Escobar; Marco Mor; Giorgio Colombo; Marco Presta

The fibroblast growth factor (FGF)/FGF receptor (FGFR) system plays a crucial role in cancer by affecting tumor growth, angiogenesis, drug resistance, and escape from anti-angiogenic anti-vascular endothelial growth factor therapy. The soluble pattern recognition receptor long-pentraxin 3 (PTX3) acts as a multi-FGF antagonist. Here we demonstrate that human PTX3 overexpression in transgenic mice driven by the Tie2 promoter inhibits tumor growth, angiogenesis, and metastasis in heterotopic, orthotopic, and autochthonous FGF-dependent tumor models. Using pharmacophore modeling of the interaction of a minimal PTX3-derived FGF-binding pentapeptide with FGF2, we identified a small-molecule chemical (NSC12) that acts as an extracellular FGF trap with significant implications in cancer therapy.


Expert Opinion on Therapeutic Targets | 2015

The potential of fibroblast growth factor/fibroblast growth factor receptor signaling as a therapeutic target in tumor angiogenesis

Roberto Ronca; Arianna Giacomini; Marco Rusnati; Marco Presta

Introduction: Fibroblast growth factors (FGFs) are endowed with a potent pro-angiogenic activity. Activation of the FGF/FGF receptor (FGFR) system occurs in a variety of human tumors. This may lead to neovascularization, supporting tumor progression and metastatic dissemination. Thus, a compelling biologic rationale exists for the development of anti-FGF/FGFR agents for the inhibition of tumor angiogenesis in cancer therapy. Areas covered: A comprehensive search on PubMed was performed to identify studies on the role of the FGF/FGFR system in angiogenesis. Endothelial FGFR signaling, the pro-angiogenic function of canonical FGFs, and their role in human tumors are described. In addition, experimental approaches aimed at the identification and characterization of nonselective and selective FGF/FGFR inhibitors and their evaluation in clinical trials are summarized. Expert opinion: Different approaches can be envisaged to inhibit the FGF/FGFR system, a target for the development of ‘two-compartment’ anti-angiogenic/anti-tumor agents, including FGFR selective and nonselective small-molecule tyrosine kinase inhibitors, anti-FGFR antibodies, and FGF ligand traps. Further studies are required to define the correlation between tumor vascularization and activation of the FGF/FGFR system and for the identification of cancer patients more likely to benefit from anti-FGF/FGFR treatments. In addition, advantages and disadvantages about the use of selective versus non-selective FGF inhibitors remain to be elucidated.


The Journal of Pathology | 2013

Long pentraxin-3 as an epithelial–stromal fibroblast growth factor-targeting inhibitor in prostate cancer

Roberto Ronca; Patrizia Alessi; Daniela Coltrini; Emanuela Di Salle; Arianna Giacomini; Daria Leali; Michela Corsini; Mirella Belleri; Chiara Tobia; Cecilia Garlanda; Elisa Bonomi; Regina Tardanico; William Vermi; Marco Presta

Fibroblast growth factors (FGFs) exert autocrine/paracrine functions in prostate cancer by stimulating angiogenesis and tumour growth. Here dihydrotestosterone (DHT) up‐regulates FGF2 and FGF8b production in murine TRAMP‐C2 prostate cancer cells, activating a FGF‐dependent autocrine loop of stimulation. The soluble pattern recognition receptor long pentraxin‐3 (PTX3) acts as a natural FGF antagonist that binds FGF2 and FGF8b via its N‐terminal domain. We demonstrate that recombinant PTX3 protein and the PTX3‐derived pentapeptide Ac‐ARPCA‐NH2 abolish the mitogenic response of murine TRAMP‐C2 cells and human LNCaP prostate cancer cells to DHT and FGFs. Also, PTX3 hampers the angiogenic activity of DHT‐activated TRAMP‐C2 cells on the chick embryo chorioallantoic membrane (CAM). Accordingly, human PTX3 overexpression inhibits the mitogenic activity exerted by DHT or FGFs on hPTX3_TRAMP‐C2 cell transfectants and their angiogenic activity. Also, hPTX3_TRAMP‐C2 cells show a dramatic decrease of their angiogenic and tumourigenic potential when grafted in syngeneic or immunodeficient athymic male mice. A similar inhibitory effect is observed when TRAMP‐C2 cells overexpress only the FGF‐binding N‐terminal PTX3 domain. In keeping with the anti‐tumour activity of PTX3 in experimental prostate cancer, immunohistochemical analysis of prostate needle biopsies from primary prostate adenocarcinoma patients shows that parenchymal PTX3 expression, abundant in basal cells of normal glands, is lost in high‐grade prostatic intraepithelial neoplasia and in invasive tumour areas. These results identify PTX3 as a potent FGF antagonist endowed with anti‐angiogenic and anti‐neoplastic activity in prostate cancer. Copyright


Molecular Cancer Therapeutics | 2013

Long Pentraxin-3 Inhibits Epithelial–Mesenchymal Transition in Melanoma Cells

Roberto Ronca; Emanuela Di Salle; Arianna Giacomini; Daria Leali; Patrizia Alessi; Daniela Coltrini; Cosetta Ravelli; Sara Matarazzo; Domenico Ribatti; William Vermi; Marco Presta

During melanoma progression, malignant melanocytes are reprogrammed into mesenchymal-like cells through to an epithelial–mesenchymal transition (EMT) process associated with the acquisition of an invasive, prometastatic phenotype. The fibroblast growth factor-2 (FGF2)/FGF receptor (FGFR) system plays a pivotal role in melanoma, leading to autocrine/paracrine induction of tumor cell proliferation and angiogenesis. Long pentraxin-3 (PTX3) interacts with FGF2, and other FGF family members, inhibiting FGF-dependent neovascularization and tumor growth. Here, PTX3 protein and the PTX3-derived acetylated pentapeptide Ac-ARPCA-NH2 inhibit FGF2-driven proliferation and downstream FGFR signaling in murine melanoma B16-F10 cells. Moreover, human PTX3-overexpressing hPTX_B16-F10 cells are characterized by the reversed transition from a mesenchymal to an epithelial-like appearance, inhibition of cell proliferation, loss of clonogenic potential, reduced motility and invasive capacity, downregulation of various mesenchymal markers, and upregulation of the epithelial marker E-cadherin. Accordingly, PTX3 affects cell proliferation and EMT transition in human A375 and A2058 melanoma cells. Also, hPTX_B16-F10 cells showed a reduced tumorigenic and metastatic activity in syngeneic C57BL/6 mice. In conclusion, PTX3 inhibits FGF/FGFR-driven EMT in melanoma cells, hampering their tumorigenic and metastatic potential. These data represent the first experimental evidence about a nonredundant role of the FGF/FGFR system in the modulation of the EMT process in melanoma and indicate that PTX3 or its derivatives may represent the basis for the design of novel therapeutic approaches in FGF/FGFR-dependent tumors, including melanoma. Mol Cancer Ther; 12(12); 2760–71. ©2013 AACR.


Brain | 2013

Inhibition of angiogenesis by β-galactosylceramidase deficiency in globoid cell leukodystrophy

Mirella Belleri; Roberto Ronca; Daniela Coltrini; Beatrice Nico; Domenico Ribatti; Pietro Luigi Poliani; Arianna Giacomini; Patrizia Alessi; Sergio Marchesini; Marta Santos; Ernesto R. Bongarzone; Marco Presta

Globoid cell leukodystrophy (Krabbe disease) is a neurological disorder of infants caused by genetic deficiency of the lysosomal enzyme β-galactosylceramidase leading to accumulation of the neurotoxic metabolite 1-β-d-galactosylsphingosine (psychosine) in the central nervous system. Angiogenesis plays a pivotal role in the physiology and pathology of the brain. Here, we demonstrate that psychosine has anti-angiogenic properties by causing the disassembling of endothelial cell actin structures at micromolar concentrations as found in the brain of patients with globoid cell leukodystrophy. Accordingly, significant alterations of microvascular endothelium were observed in the post-natal brain of twitcher mice, an authentic model of globoid cell leukodystrophy. Also, twitcher endothelium showed a progressively reduced capacity to respond to pro-angiogenic factors, defect that was corrected after transduction with a lentiviral vector harbouring the murine β-galactosylceramidase complementary DNA. Finally, RNA interference-mediated β-galactosylceramidase gene silencing causes psychosine accumulation in human endothelial cells and hampers their mitogenic and motogenic response to vascular endothelial growth factor. Accordingly, significant alterations were observed in human microvasculature from brain biopsy of a globoid cell leukodystrophy case. Together these data demonstrate that β-galactosylceramidase deficiency induces significant alterations in endothelial neovascular responses that may contribute to central nervous system and systemic damages that occur in globoid cell leukodystrophy.


Cancer Research | 2013

Abstract 5609: Bortezomib enhanced cytotoxic effects of novel sTRAIL-targeted nanocarriers against neuroectodermal tumors .

Monica Loi; Arianna Giacomini; Pamela Becherini; Fabio Pastorino; Mirco Ponzoni

The soluble form of the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (sTRAIL) is a cancer cell-specific molecule exerting anti-tumor activity in a variety of transformed cells. sTRAIL elicits apoptosis by activating the pro-apoptotic receptors 1 (TRAIL-R1) and 2 (TRAIL-R2). However, clinical studies in cancer patients demonstrated that recombinant human sTRAIL showed short serum half-life that does not lead to a subsequent tumor accumulation over time. The aim of this work was to develop a novel liposomal formulation carrying the sTRAIL at the outer surface (sTRAIL-SL) in order to improve its pharmacokinetic features, while preserving its anti-tumor activity and increasing its in vivo efficacy. Neuroectodermal tumor cells (i.e neuroblastoma (NB) and melanoma) can be either sensitive or resistant to sTRAIL-driven death. In this regard, we investigated the possibility to combine our novel sTRAIL liposomal formulation with the proteasome inhibitor Bortezomib (BTZ), known to be able to sensitize tumor cells to sTRAIL-induced apoptosis, by up-regulating TRAIL-R2. The expression of TRAIL receptors was validated by FACS analysis in a panel of human NB and melanoma cell lines. Receptors expression after BTZ treatment was also evaluated, confirming the BTZ-driven up-regulation of TRAIL-R2 in all the tumor cells tested. No effect on TRAIL-R1 expression was pointed out. Cellular association of sTRAIL-SL was evaluated by FACS analysis on both NB and melanoma cell lines. In vitro cytotoxicity and selective apoptosis induction by sTRAIL-SL were examined in both sTRAIL-sensitive and -resistant NB and melanoma cell lines, by using annexin V/PI assay. Treatments were carried out with sTRAIL, either free or coupled to SL, alone and in combination with BTZ. In sTRAIL-sensitive cells, we observed that sTRAIL-SL treatment resulted in a dose-dependent induction of apoptosis with an efficacy slightly more pronounced than free sTRAIL. Moreover, experiments of combination treatments showed that BTZ effectively sensitized cells to sTRAIL-induced apoptosis. Upon BTZ priming, sTRAIL-resistant cells undergo enhanced apoptotic death by triggering with sTRAIL-SL compared to treatment with free sTRAIL. In preliminary mechanistic experiments sTRAIL-induced cytotoxicity is dependent on β-catenin degradation and Caspase-3 and 8 activation. No effect was exerted on normal fibroblasts. In vivo experiments investigating the pharmacokinetic profile of sTRAIL when coupled to nanocarriers and evaluating its anti-tumor activity against both NB and melanoma animal models are on running. Our data show that sTRAIL, when coupled to nanocarriers preserved and improved its tumor-selective activity on both NB and melanoma, suggesting that it might be an efficient vehicle for sTRAIL delivery and, in combination with BTZ, a valid strategy for the treatment of sTRAIL-resistant tumors. Citation Format: Monica Loi, Arianna Giacomini, Pamela Becherini, Fabio Pastorino, Mirco Ponzoni. Bortezomib enhanced cytotoxic effects of novel sTRAIL-targeted nanocarriers against neuroectodermal tumors . [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 5609. doi:10.1158/1538-7445.AM2013-5609


The FASEB Journal | 2014

Antiangiogenic effects of N6-isopentenyladenosine, an endogenous isoprenoid end product, mediated by AMPK activation

Simona Pisanti; Paola Picardi; Elena Ciaglia; Luigi Margarucci; Roberto Ronca; Arianna Giacomini; Anna Maria Malfitano; Agostino Casapullo; Chiara Laezza; Patrizia Gazzerro; Maurizio Bifulco

N6‐isopentenyladenosine (iPA), an end product of the mevalonate pathway with an isopentenyl chain, is already known to exert a suppressor effect against various tumors. In this work, we investigated whether iPA also directly interferes with the angiogenic process, which is fundamental to tumor growth and progression. To this end, using human umbilical vein endothelial cells (HUVECs) as a suitable in vitro model of angiogenesis, we evaluated their viability, proliferation, migration, invasion, tube formation in response to iPA, and molecular mechanisms involved. Data were corroborated in mice by using a gel plug assay. iPA dose‐ and time‐dependently inhibited all the neoangiogenesis stages, with an IC50 of 0.98 μM. We demonstrated for the first time, by liquid chromatography–coupled tandem mass spectrometry (LC‐MS/MS), that iPA was monophosphorylated into 5′‐iPA‐monophosphate (iPAMP) by the adenosine kinase (ADK) inside the cells. iPAMP is the active form that inhibits angiogenesis through the direct activation of AMP‐kinase (AMPK). Indeed, all effects were completely reversed by pretreatment with 5‐iodotubercidin (5‐Itu), an ADK inhibitor. The isoprenoid intermediate isopentenyl pyrophosphate (IPP), which shares the isopentenyl moiety with iPA, was ineffective in the inhibition of angiogenesis, thus showing that the iPA structure is specific for the observed effects. In conclusion, iPA is a novel AMPK activator and could represent a useful tool for the treatment of diseases where excessive neoangiogenesis is the underlying pathology.—Pisanti, S., Picardi, P., Ciaglia, E., Margarucci, L., Ronca, R., Giacomini, A., Malfitano, A. M., Casapullo, A., Laezza, C., Gazzerro, P., Bifulco, M. Antiangiogenic effects of N6‐isopentenyladenosine, an endogenous isoprenoid end‐product, mediated by AMPK activation. FASEB J. 28, 1132–1144 (2014). www.fasebj.org


Angiogenesis | 2015

Brain angioarchitecture and intussusceptive microvascular growth in a murine model of Krabbe disease.

Arianna Giacomini; Maximilian Ackermann; Mirella Belleri; Daniela Coltrini; Beatrice Nico; Domenico Ribatti; Moritz A. Konerding; Marco Presta; Marco Righi

Defects of the angiogenic process occur in the brain of twitcher mouse, an authentic model of human Krabbe disease caused by genetic deficiency of lysosomal β-galactosylceramidase (GALC), leading to lethal neurological dysfunctions and accumulation of neurotoxic psychosine in the central nervous system. Here, quantitative computational analysis was used to explore the alterations of brain angioarchitecture in twitcher mice. To this aim, customized ImageJ routines were used to assess calibers, amounts, lengths and spatial dispersion of CD31+ vessels in 3D volumes from the postnatal frontal cortex of twitcher animals. The results showed a decrease in CD31 immunoreactivity in twitcher brain with a marked reduction in total vessel lengths coupled with increased vessel fragmentation. No significant changes were instead observed for the spatial dispersion of brain vessels throughout volumes or in vascular calibers. Notably, no CD31+ vessel changes were detected in twitcher kidneys in which psychosine accumulates at very low levels, thus confirming the specificity of the effect. Microvascular corrosion casting followed by scanning electron microscopy morphometry confirmed the presence of significant alterations of the functional angioarchitecture of the brain cortex of twitcher mice with reduction in microvascular density, vascular branch remodeling and intussusceptive angiogenesis. Intussusceptive microvascular growth, confirmed by histological analysis, was paralleled by alterations of the expression of intussusception-related genes in twitcher brain. Our data support the hypothesis that a marked decrease in vascular development concurs to the onset of neuropathological lesions in twitcher brain and suggest that neuroinflammation-driven intussusceptive responses may represent an attempt to compensate impaired sprouting angiogenesis.


Journal of Medicinal Chemistry | 2016

Synthesis, Structural Elucidation, and Biological Evaluation of NSC12, an Orally Available Fibroblast Growth Factor (FGF) Ligand Trap for the Treatment of FGF-Dependent Lung Tumors

Riccardo Castelli; Arianna Giacomini; Mattia Anselmi; Nicole Bozza; Federica Vacondio; Silvia Rivara; Sara Matarazzo; Marco Presta; Marco Mor; Roberto Ronca

NSC12 is an orally available pan-FGF trap able to inhibit FGF2/FGFR interaction and endowed with promising antitumor activity. It was identified by virtual screening from a NCI small molecule library, but no data were available about its synthesis, stereochemistry, and physicochemical properties. We report here a synthetic route that allowed us to characterize and unambiguously identify the structure of the active compound by a combination of NMR spectroscopy and in silico conformational analysis. The synthetic protocol allowed us to sustain experiments aimed at assessing its therapeutic potential for the treatment of FGF-dependent lung cancers. A crucial step in the synthesis generated a couple of diastereoisomers, with only one able to act as a FGF trap molecule and to inhibit FGF-dependent receptor activation, cell proliferation, and tumor growth when tested in vitro and in vivo on murine and human lung cancer cells.


Biochimica et Biophysica Acta | 2018

Long pentraxin 3: A novel multifaceted player in cancer

Arianna Giacomini; Gaia C. Ghedini; Marco Presta; Roberto Ronca

Since its discovery in 1992, long pentraxin 3 (PTX3) has been characterized as soluble patter recognition receptor, a key player of the innate immunity arm with non-redundant functions in pathogen recognition and inflammatory responses. As a component of the extra-cellular matrix milieu, PTX3 has been implicated also in wound healing/tissue remodeling, cardiovascular diseases, fertility, and infectious diseases. Consequently, PTX3 levels in biological fluids have been proposed as a fluid-phase biomarker in different pathological conditions. In the last decade, experimental evidences have shown that PTX3 may exert a significant impact also on different aspects of cancer biology, including tumor onset, angiogenesis, metastatic dissemination and immune-modulation. However, it remains unclear whether PTX3 acts as a good cop or bad cop in cancer. In this review, we will summarize and discuss the scientific literature data focusing on the role of PTX3 in experimental and human tumors, including its putative translational implications.

Collaboration


Dive into the Arianna Giacomini'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