Network


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

Hotspot


Dive into the research topics where Romina Alfonsi is active.

Publication


Featured researches published by Romina Alfonsi.


The EMBO Journal | 2015

Gli1/DNA interaction is a druggable target for Hedgehog‐dependent tumors

Paola Infante; Mattia Mori; Romina Alfonsi; Francesca Ghirga; Federica Aiello; Sara Toscano; Cinzia Ingallina; Mariangela Siler; Danilo Cucchi; Agnese Po; Evelina Miele; Davide D'Amico; Gianluca Canettieri; Enrico De Smaele; Elisabetta Ferretti; Isabella Screpanti; Gloria Uccello Barretta; Maurizio Botta; Bruno Botta; Alberto Gulino; Lucia Di Marcotullio

Hedgehog signaling is essential for tissue development and stemness, and its deregulation has been observed in many tumors. Aberrant activation of Hedgehog signaling is the result of genetic mutations of pathway components or other Smo‐dependent or independent mechanisms, all triggering the downstream effector Gli1. For this reason, understanding the poorly elucidated mechanism of Gli1‐mediated transcription allows to identify novel molecules blocking the pathway at a downstream level, representing a critical goal in tumor biology. Here, we clarify the structural requirements of the pathway effector Gli1 for binding to DNA and identify Glabrescione B as the first small molecule binding to Gli1 zinc finger and impairing Gli1 activity by interfering with its interaction with DNA. Remarkably, as a consequence of its robust inhibitory effect on Gli1 activity, Glabrescione B inhibited the growth of Hedgehog‐dependent tumor cells in vitro and in vivo as well as the self‐renewal ability and clonogenicity of tumor‐derived stem cells. The identification of the structural requirements of Gli1/DNA interaction highlights their relevance for pharmacologic interference of Gli signaling.


Cell Death & Differentiation | 2013

PCAF ubiquitin ligase activity inhibits Hedgehog/Gli1 signaling in p53-dependent response to genotoxic stress

Daniela Mazzà; Paola Infante; Valeria Colicchia; A Greco; Romina Alfonsi; Mariangela Siler; Laura Antonucci; Agnese Po; E De Smaele; Elisabetta Ferretti; Carlo Capalbo; Diana Bellavia; Gianluca Canettieri; Giuseppe Giannini; Isabella Screpanti; Alberto Gulino; L Di Marcotullio

The Hedgehog (Hh) signaling regulates tissue development, and its aberrant activation is a leading cause of malignancies, including medulloblastoma (Mb). Hh-dependent tumorigenesis often occurs in synergy with other mechanisms, such as loss of p53, the master regulator of the DNA damage response. To date, little is known about mechanisms connecting DNA-damaging events to morphogen-dependent processes. Here, we show that genotoxic stress triggers a cascade of signals, culminating with inhibition of the activity of Gli1, the final transcriptional effector of Hh signaling. This inhibition is dependent on the p53-mediated elevation of the acetyltransferase p300/CBP-associated factor (PCAF). Notably, we identify PCAF as a novel E3 ubiquitin ligase of Gli1. Indeed PCAF, but not a mutant with a deletion of its ubiquitination domain, represses Hh signaling in response to DNA damage by promoting Gli1 ubiquitination and its proteasome-dependent degradation. Restoring Gli1 levels rescues the growth arrest and apoptosis effect triggered by genotoxic drugs. Consistently, DNA-damaging agents fail to inhibit Gli1 activity in the absence of either p53 or PCAF. Finally, Mb samples from p53-null mice display low levels of PCAF and upregulation of Gli1 in vivo, suggesting PCAF as potential therapeutic target in Hh-dependent tumors. Together, our data define a mechanism of inactivation of a morphogenic signaling in response to genotoxic stress and unveil a p53/PCAF/Gli1 circuitry centered on PCAF that limits Gli1-enhanced mitogenic and prosurvival response.


Trends in Pharmacological Sciences | 2015

Targeting GLI factors to inhibit the Hedgehog pathway

Paola Infante; Romina Alfonsi; Bruno Botta; Mattia Mori; Lucia Di Marcotullio

Hedgehog (Hh) signaling has emerged in recent years as an attractive target for anticancer therapy because its aberrant activation is implicated in several cancers. Major progress has been made in the development of SMOOTHENED (SMO) antagonists, although they have shown several limitations due to downstream SMO pathway activation or the occurrence of drug-resistant SMO mutations. Recently, particular interest has been elicited by the identification of molecules able to hit glioma-associated oncogene (GLI) factors, the final effectors of the Hh pathway, which provide a valid tool to overcome anti-SMO resistance. Here, we review results achieved in developing GLI antagonists, explaining their mechanisms of action and highlighting their therapeutic potential. We also underline the relevance of structural details in their discovery and optimization.


Journal of Medicinal Chemistry | 2014

New pyrrole derivatives with potent tubulin polymerization inhibiting activity as anticancer agents including hedgehog-dependent cancer

Giuseppe La Regina; Ruoli Bai; Antonio Coluccia; Valeria Famiglini; Sveva Pelliccia; Sara Passacantilli; Carmela Mazzoccoli; Vitalba Ruggieri; Lorenza Sisinni; Alessio Bolognesi; Whilelmina Maria Rensen; Andrea Miele; Marianna Nalli; Romina Alfonsi; Lucia Di Marcotullio; Alberto Gulino; Andrea Brancale; Ettore Novellino; Giulio Dondio; Stefania Vultaggio; Mario Varasi; Ciro Mercurio; Ernest Hamel; Patrizia Lavia; Romano Silvestri

We synthesized 3-aroyl-1-arylpyrrole (ARAP) derivatives as potential anticancer agents having different substituents at the pendant 1-phenyl ring. Both the 1-phenyl ring and 3-(3,4,5-trimethoxyphenyl)carbonyl moieties were mandatory to achieve potent inhibition of tubulin polymerization, binding of colchicine to tubulin, and cancer cell growth. ARAP 22 showed strong inhibition of the P-glycoprotein-overexpressing NCI-ADR-RES and Messa/Dx5MDR cell lines. Compounds 22 and 27 suppressed in vitro the Hedgehog signaling pathway, strongly reducing luciferase activity in SAG treated NIH3T3 Shh-Light II cells, and inhibited the growth of medulloblastoma D283 cells at nanomolar concentrations. ARAPs 22 and 27 represent a new potent class of tubulin polymerization and cancer cell growth inhibitors with the potential to inhibit the Hedgehog signaling pathway.


Journal of Medicinal Chemistry | 2015

New Indole Tubulin Assembly Inhibitors Cause Stable Arrest of Mitotic Progression, Enhanced Stimulation of Natural Killer Cell Cytotoxic Activity, and Repression of Hedgehog-Dependent Cancer

Giuseppe La Regina; Ruoli Bai; Antonio Coluccia; Valeria Famiglini; Sveva Pelliccia; Sara Passacantilli; Carmela Mazzoccoli; Vitalba Ruggieri; Annalisa Verrico; Andrea Miele; Ludovica Monti; Marianna Nalli; Romina Alfonsi; Lucia Di Marcotullio; Alberto Gulino; Biancamaria Ricci; Alessandra Soriani; Angela Santoni; Michele Caraglia; Stefania Porto; Eleonora Da Pozzo; Claudia Martini; Andrea Brancale; Luciana Marinelli; Ettore Novellino; Stefania Vultaggio; Mario Varasi; Ciro Mercurio; Chiara Bigogno; Giulio Dondio

We designed 39 new 2-phenylindole derivatives as potential anticancer agents bearing the 3,4,5-trimethoxyphenyl moiety with a sulfur, ketone, or methylene bridging group at position 3 of the indole and with halogen or methoxy substituent(s) at positions 4-7. Compounds 33 and 44 strongly inhibited the growth of the P-glycoprotein-overexpressing multi-drug-resistant cell lines NCI/ADR-RES and Messa/Dx5. At 10 nM, 33 and 44 stimulated the cytotoxic activity of NK cells. At 20-50 nM, 33 and 44 arrested >80% of HeLa cells in the G2/M phase of the cell cycle, with stable arrest of mitotic progression. Cell cycle arrest was followed by cell death. Indoles 33, 44, and 81 showed strong inhibition of the SAG-induced Hedgehog signaling activation in NIH3T3 Shh-Light II cells with IC50 values of 19, 72, and 38 nM, respectively. Compounds of this class potently inhibited tubulin polymerization and cancer cell growth, including stimulation of natural killer cell cytotoxic activity and repression of Hedgehog-dependent cancer.


Cell Death and Disease | 2016

Inhibition of Hedgehog-dependent tumors and cancer stem cells by a newly identified naturally occurring chemotype

Paola Infante; Romina Alfonsi; Cinzia Ingallina; Deborah Quaglio; Francesca Ghirga; Ilaria D'Acquarica; Flavia Bernardi; Laura Di Magno; Gianluca Canettieri; Isabella Screpanti; Alberto Gulino; Bruno Botta; Mattia Mori; Lucia Di Marcotullio

Hedgehog (Hh) inhibitors have emerged as valid tools in the treatment of a wide range of cancers. Indeed, aberrant activation of the Hh pathway occurring either by ligand-dependent or -independent mechanisms is a key driver in tumorigenesis. The smoothened (Smo) receptor is one of the main upstream transducers of the Hh signaling and is a validated target for the development of anticancer compounds, as underlined by the FDA-approved Smo antagonist Vismodegib (GDC-0449/Erivedge) for the treatment of basal cell carcinoma. However, Smo mutations that confer constitutive activity and drug resistance have emerged during treatment with Vismodegib. For this reason, the development of new effective Hh inhibitors represents a major challenge for cancer therapy. Natural products have always represented a unique source of lead structures in drug discovery, and in recent years have been used to modulate the Hh pathway at multiple levels. Here, starting from an in house library of natural compounds and their derivatives, we discovered novel chemotypes of Hh inhibitors by mean of virtual screening against the crystallographic structure of Smo. Hh functional based assay identified the chalcone derivative 12 as the most effective Hh inhibitor within the test set. The chalcone 12 binds the Smo receptor and promotes the displacement of Bodipy-Cyclopamine in both Smo WT and drug-resistant Smo mutant. Our molecule stands as a promising Smo antagonist able to specifically impair the growth of Hh-dependent tumor cells in vitro and in vivo and medulloblastoma stem-like cells and potentially overcome the associated drug resistance.


Journal of Medicinal Chemistry | 2017

Design, Palladium-Catalyzed Synthesis, and Biological Investigation of 2-Substituted 3-Aroylquinolin-4(1H)-ones as Inhibitors of the Hedgehog Signaling Pathway

Romina Alfonsi; Bruno Botta; Sandro Cacchi; Lucia Di Marcotullio; Giancarlo Fabrizi; Roberta Faedda; Antonella Goggiamani; Antonia Iazzetti; Mattia Mori

2-Substituted 3-aroylquinolin-4(1H)-ones, prepared through a palladium-catalyzed carbonylative cyclization of N-(2-iodoaryl)enaminones, proved to inhibit efficiently the Hedgehog pathway through direct antagonism of the wild-type and drug-resistant form of the Smoothened receptor. Notably, these compounds repressed the Hh-dependent growth events and the proliferation of tumor cells with aberrant activation of the Hh pathway, which plays a crucial role in development and tumorigenesis.


International Journal of Molecular Sciences | 2018

The Double Face of Exosome-Carried MicroRNAs in Cancer Immunomodulation

Romina Alfonsi; Ludovica Grassi; Michele Signore; Désirée Bonci

In recent years many articles have underlined the key role of nanovesicles, i.e., exosomes, as information carriers among biological systems including cancer. Tumor-derived exosomes (TEXs) are key players in the dynamic crosstalk between cancer cells and the microenvironment while promote immune system control evasion. In fact, tumors are undoubtedly capable of silencing the immune response through multiple mechanisms, including the release of exosomes. TEXs have been shown to boost tumor growth and promote progression and metastatic spreading via suppression or stimulation of the immune response towards cancer cells. The advantage of immunotherapeutic treatment alone over combining immuno- and conventional therapy is currently debated. Understanding the role of tumor exosome-cargo is of crucial importance for our full comprehension of neoplastic immonosuppression and for the construction of novel therapies and vaccines based on (nano-) vesicles. Furthermore, to devise new anti-cancer approaches, diverse groups investigated the possibility of engineering TEXs by conditioning cancer cells’ own cargo. In this review, we summarize the state of art of TEX-based immunomodulation with a particular focus on the molecular function of non-coding family genes, microRNAs. Finally, we will report on recent efforts in the study of potential applications of engineered exosomes in cancer immunotherapy.


Journal of Enzyme Inhibition and Medicinal Chemistry | 2018

Chemical, computational and functional insights into the chemical stability of the Hedgehog pathway inhibitor GANT61

Andrea Calcaterra; Valentina Iovine; Bruno Botta; Deborah Quaglio; Ilaria D’Acquarica; Alessia Ciogli; Antonia Iazzetti; Romina Alfonsi; Ludovica Lospinoso Severini; Paola Infante; Lucia Di Marcotullio; Mattia Mori; Francesca Ghirga

Abstract This work aims at elucidating the mechanism and kinetics of hydrolysis of GANT61, the first and most-widely used inhibitor of the Hedgehog (Hh) signalling pathway that targets Glioma-associated oncogene homologue (Gli) proteins, and at confirming the chemical nature of its bioactive form. GANT61 is poorly stable under physiological conditions and rapidly hydrolyses into an aldehyde species (GANT61-A), which is devoid of the biological activity against Hh signalling, and a diamine derivative (GANT61-D), which has shown inhibition of Gli-mediated transcription. Here, we combined chemical synthesis, NMR spectroscopy, analytical studies, molecular modelling and functional cell assays to characterise the GANT61 hydrolysis pathway. Our results show that GANT61-D is the bioactive form of GANT61 in NIH3T3 Shh-Light II cells and SuFu−/− mouse embryonic fibroblasts, and clarify the structural requirements for GANT61-D binding to Gli1. This study paves the way to the design of GANT61 derivatives with improved potency and chemical stability. Graphical Abstract


Molecular Cancer Therapeutics | 2016

MK-4101 - a potent inhibitor of the hedgehog pathway - is highly active against medulloblastoma and basal cell carcinoma

Gessica Filocamo; Mirko Brunetti; Fabrizio Colaceci; Romina Sasso; Mirella Tanori; Emanuela Pasquali; Romina Alfonsi; Mariateresa Mancuso; Anna Saran; Armin Lahm; Lucia Di Marcotullio; Christian Steinkühler; Simonetta Pazzaglia

Aberrant activation of the Hedgehog (Hh) signaling pathway is implicated in the pathogenesis of many cancers, including medulloblastoma and basal cell carcinoma (BCC). In this study, using neonatally irradiated Ptch1+/− mice as a model of Hh-dependent tumors, we investigated the in vivo effects of MK-4101, a novel SMO antagonist, for the treatment of medulloblastoma and BCC. Results clearly demonstrated a robust antitumor activity of MK-4101, achieved through the inhibition of proliferation and induction of extensive apoptosis in tumor cells. Of note, beside antitumor activity on transplanted tumors, MK-4101 was highly efficacious against primary medulloblastoma and BCC developing in the cerebellum and skin of Ptch1+/− mice. By identifying the changes induced by MK-4101 in gene expression profiles in tumors, we also elucidated the mechanism of action of this novel, orally administrable compound. MK-4101 targets the Hh pathway in tumor cells, showing the maximum inhibitory effect on Gli1. MK-4101 also induced deregulation of cell cycle and block of DNA replication in tumors. Members of the IGF and Wnt signaling pathways were among the most highly deregulated genes by MK-4101, suggesting that the interplay among Hh, IGF, and Wnt is crucial in Hh-dependent tumorigenesis. Altogether, the results of this preclinical study support a therapeutic opportunity for MK-4101 in the treatment of Hh-driven cancers, also providing useful information for combination therapy with drugs targeting pathways cooperating with Hh oncogenic activity. Mol Cancer Ther; 15(6); 1177–89. ©2016 AACR.

Collaboration


Dive into the Romina Alfonsi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paola Infante

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Mattia Mori

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Bruno Botta

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Alberto Gulino

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Francesca Ghirga

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Cinzia Ingallina

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnese Po

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge