Network


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

Hotspot


Dive into the research topics where Giulia Stazi is active.

Publication


Featured researches published by Giulia Stazi.


Journal of Medicinal Chemistry | 2016

Discovery of a Novel Inhibitor of Histone Lysine-Specific Demethylase 1A (KDM1A/LSD1) as Orally Active Antitumor Agent

Paola Vianello; Oronza A. Botrugno; Anna Cappa; Roberto Dal Zuffo; Paola Dessanti; Antonello Mai; Biagina Marrocco; Andrea Mattevi; Giuseppe Meroni; Saverio Minucci; Giulia Stazi; Florian Thaler; Paolo Trifiró; Sergio Valente; Manuela Villa; Mario Varasi; Ciro Mercurio

We report the stereoselective synthesis and biological activity of a novel series of tranylcypromine (TCPA) derivatives (14a-k, 15, 16), potent inhibitors of KDM1A. The new compounds strongly inhibit the clonogenic potential of acute leukemia cell lines. In particular three molecules (14d, 14e, and 14g) showing selectivity versus MAO A and remarkably inhibiting colony formation in THP-1 human leukemia cells, were assessed in mouse for their preliminary pharmacokinetic. 14d and 14e were further tested in vivo in a murine acute promyelocytic leukemia model, resulting 14d the most effective. Its two enantiomers were synthesized: the (1S,2R) enantiomer 15 showed higher activity than its (1R,2S) analogue 16, in both biochemical and cellular assays. Compound 15 exhibited in vivo efficacy after oral administration, determining a 62% increased survival in mouse leukemia model with evidence of KDM1A inhibition. The biological profile of compound 15 supports its further investigation as a cancer therapeutic.


Expert Opinion on Therapeutic Patents | 2016

LSD1 inhibitors: a patent review (2010-2015)

Giulia Stazi; Clemens Zwergel; Sergio Valente; Antonello Mai

ABSTRACT Introduction: Lysine demethylase 1 (LSD1) plays an important role in mediating the expression of genes involved in cancer and non-cancer diseases such as viral infections, cardiovascular and neurodegenerative disorders. It is involved in a number of processes from adipogenesis to cell adhesion to viral latency, regulating several cell pathways related with proliferation, development, and cell cycle control. Numerous chemical entities have been studied in recent years and some of them entered the clinical arena. Areas covered: This review summarizes recent efforts in the drug development of LSD1 inhibitors reported in the patent literature covering the 2010-2015 period, including their potential use as therapeutics in cancerous, neurological, inflammatory, cardiovascular, and viral diseases. Expert opinion: The development of novel potent and selective LSD1 inhibitors is ongoing, in order to improve their potency and selectivity against specific types of cancer or non-cancer diseases. More in-depth studies are required to assess the role of LSD1 inhibitors in the expression of LSD1 target genes, for a better assessment of the biochemistry underlying their efficacy, and to provide evidence for any possible side effects. Furthermore, an interesting therapeutic approach is the use of LSD1 inhibitors in conjunction with other epidrugs to combine their therapeutic potential, leading to innovative, personalized treatments.


MedChemComm | 2015

Pyrrole- and indole-containing tranylcypromine derivatives as novel lysine-specific demethylase 1 inhibitors active on cancer cells

Veronica Rodriguez; Sergio Valente; Stefano Rovida; Dante Rotili; Giulia Stazi; Alessia Lucidi; Giuseppe Ciossani; Andrea Mattevi; Oronza A. Botrugno; Paola Dessanti; Ciro Mercurio; Paola Vianello; Saverio Minucci; Mario Varasi; Antonello Mai

On the basis of previous research showing the capability of N-carbobenzyloxy-(Z-)amino acid-tranylcypromine (-TCPA) derivatives to inhibit LSD1, we inserted at the 4-amino-TCPA moiety first a Z-Pro (9) and a Z-Gly (10) residue and then, after the encouraging data obtained for 9, a pyrrole and an indole ring in which the relative N1 position carried a acetophenone, a N-phenyl/benzylacetamide, or a Z chain (11a–f and 12a–f, respectively). In both series, the Z-pyrrole and indole derivatives 11e, f and 12e, f displayed high LSD1 inhibitory activity. The compounds are able to inhibit LSD1 in NB4 cells, increasing the expression of two related genes, GFI-1b and ITGAM, and to induce cell growth arrest in the AML MB4-11 and APL NB4 cell lines.


Oncotarget | 2017

The histone methyltransferase EZH2 as a druggable target in SHH medulloblastoma cancer stem cells

Evelina Miele; Sergio Valente; Vincenzo Alfano; Marianna Silvano; Paolo Mellini; Diana Borovika; Biagina Marrocco; Agnese Po; Zein Mersini Besharat; Giuseppina Catanzaro; Giuseppe Battaglia; Luana Abballe; Clemens Zwergel; Giulia Stazi; Ciro Milite; Sabrina Castellano; Marco Tafani; Peteris Trapencieris; Antonello Mai; Elisabetta Ferretti

The histone methyltransferase EZH2 plays a role in maintenance of the stem component of cancer, and its overexpression and/or mutation typically drives tumor aggressiveness, drug resistance and patients’ poor prognosis. In this study, we use mouse and human medulloblastoma stem-like cells belonging to the Sonic Hedgehog subgroup (SHH MB-SLCs) and demonstrate that genetic suppression of EZH2 reduces the level of its histone mark H3K27me3 and lowers proliferation and self-renewal. We designed an EZH2 inhibitor (EZH2i) as a simplified analog of EPZ005687 and GSK2816126, MC3629, and we tested its biological activity in SHH MB-SLCs. Pharmacological inhibition of EZH2 impairs SHH MB cells proliferation and self-renewal, and induces apoptosis in vitro. Finally, we generated xenograft MB-SLCs orthotopic tumors in nude mice to test MC3629 in vivo. In treated mice, we observed impairment of tumor growth, together with induction of apoptosis and reduction of proliferation and stemness. Overall, these findings describe EZH2 as a druggable target in MB and provide insight into the biological activity of MC3629 as an EZH2i.


European Journal of Medicinal Chemistry | 2017

Novel coumarin- and quinolinone-based polycycles as cell division cycle 25-A and -C phosphatases inhibitors induce proliferation arrest and apoptosis in cancer cells

Clemens Zwergel; Brigitte Czepukojc; Emilie Evain-Bana; Zhanjie Xu; Giulia Stazi; Mattia Mori; Alexandros Patsilinakos; Antonello Mai; Bruno Botta; Rino Ragno; Denise Bagrel; Gilbert Kirsch; Peter Meiser; Claus Jacob; Mathias Montenarh; Sergio Valente

Cell division cycle phosphatases CDC25 A, B and C are involved in modulating cell cycle processes and are found overexpressed in a large panel of cancer typology. Here, we describe the development of two novel quinone-polycycle series of CDC25A and C inhibitors on the one hand 1a-k, coumarin-based, and on the other 2a-g, quinolinone-based, which inhibit either enzymes up to a sub-micro molar level and at single-digit micro molar concentrations, respectively. When tested in six different cancer cell lines, compound 2c displayed the highest efficacy to arrest cell viability, showing in almost all cell lines sub-micro molar IC50 values, a profile even better than the reference compound NCS95397. To investigate the putative binding mode of the inhibitors and to develop quantitative structure-activity relationships, molecular docking and 3-D QSAR studies were also carried out. Four selected inhibitors, 1a, 1d, 2a and 2c have been also tested in A431 cancer cells; among them, compound 2c was the most potent one leading to cell proliferation arrest and decreased CDC25C protein levels together with its splicing variant. Compound 2c displayed increased phosphorylation levels of histone H3, induction of PARP and caspase 3 cleavage, highlighting its contribution to cell death through pro-apoptotic effects.


Expert Opinion on Therapeutic Patents | 2017

EZH2 inhibitors: a patent review (2014-2016)

Giulia Stazi; Clemens Zwergel; Antonello Mai; Sergio Valente

ABSTRACT Introduction: The histone methyltransferase EZH2 is the catalytic subunit of the PRC2 complex involved in H3K27 trimethylation. Aberrant PRC2 activity has been reported in several cancers and EZH2 overexpression has been associated with poor outcome in different tumors. EZH2 somatic mutations and deletions was found in lymphomas, myelodysplastic and myeloproliferative disorders and associated with higher H3K27me3 levels. Numerous chemical entities have been studied as EZH2 inhibitors in the recent years and some of them entered the cancer clinical arena. Areas covered: This review summarizes recent efforts in the drug development of EZH2 inhibitors reported in the patent literature covering the 2014–2016 period, and their potential use as therapeutics mainly in cancerous diseases. Expert opinion: Despite the number of compounds described, only a few of them entered the clinical arena. Moreover, most of the compounds developed share a common 2-pyridone ring pharmacophore. Recently, secondary mutants have been described to be resistant to the standard EZH2 inhibitors treatment. Based on these data a lot of effort is still required to find new chemical entities that inhibit EZH2 directly, or indirectly (via PRC2 disruption). Several issues are still to be settled, such as drug resistance and the importance of selectivity over EZH1 or somatic EZH2 mutants.


Philosophical Transactions of the Royal Society B | 2018

Pyrazole-based inhibitors of enhancer of zeste homologue 2 induce apoptosis and autophagy in cancer cells

Paolo Mellini; Biagina Marrocco; Diana Borovika; Lucia Polletta; Ilaria Carnevale; Serena Saladini; Giulia Stazi; Clemens Zwergel; Peteris Trapencieris; Elisabetta Ferretti; Marco Tafani; Sergio Valente; Antonello Mai

Novel pyrazole-based EZH2 inhibitors have been prepared through a molecular pruning approach from known inhibitors bearing a bicyclic moiety as a central scaffold. The hit compound 1o (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-methyl-1-phenyl-1H-pyrazole-4-carboxamide) showed low micromolar EZH2/PRC2 inhibition and high selectivity towards a panel of other methyltransferases. Moreover, 1o displayed cell growth arrest in breast MDA-MB231, leukaemia K562, and neuroblastoma SK-N-BE cancer cells joined to reduction of H3K27me3 levels and induction of apoptosis and autophagy. This article is part of a discussion meeting issue ‘Frontiers in epigenetic chemical biology’.


Philosophical Transactions of the Royal Society B | 2018

Correction to ‘Pyrazole-based inhibitors of enhancer of zeste homologue 2 induce apoptosis and autophagy in cancer cells’

Paolo Mellini; Biagina Marrocco; Diana Borovika; Lucia Polletta; Ilaria Carnevale; Serena Saladini; Giulia Stazi; Clemens Zwergel; Peteris Trapencieris; Elisabetta Ferretti; Marco Tafani; Sergio Valente; Antonello Mai

Phil. Trans. R. Soc. B 373, 20170150. (Published 23 April 2018) (doi:10.1098/rstb.2017.0150) An affiliation was missing for Elisabetta Ferretti. The correct author list and affiliations is as follows: Paolo Mellini1, Biagina Marrocco1, Diana Borovika2, Lucia Polletta …


Molecular Cancer Therapeutics | 2018

A Quinoline-Based DNA Methyltransferase Inhibitor as a Possible Adjuvant in Osteosarcoma Therapy

Maria Cristina Manara; Sergio Valente; Camilla Cristalli; Giordano Nicoletti; Lorena Landuzzi; Clemens Zwergel; Roberta Mazzone; Giulia Stazi; Paola B. Arimondo; Michela Pasello; Clara Guerzoni; Piero Picci; Patrizia Nanni; Pier Luigi Lollini; Antonello Mai; Katia Scotlandi

The identification of new therapeutic strategies against osteosarcoma, the most common primary bone tumor, continues to be a primary goal to improve the outcomes of patients refractory to conventional chemotherapy. Osteosarcoma originates from the transformation of mesenchymal stem cells (MSC) and/or osteoblast progenitors, and the loss of differentiation is a common biological osteosarcoma feature, which has strong significance in predicting tumor aggressiveness. Thus, restoring differentiation through epigenetic reprogramming is potentially exploitable for therapeutic benefits. Here, we demonstrated that the novel nonnucleoside DNMT inhibitor (DNMTi) MC3343 affected tumor proliferation by blocking osteosarcoma cells in G1 or G2–M phases and induced osteoblastic differentiation through the specific reexpression of genes regulating this physiologic process. Although MC3343 has a similar antiproliferative effect as 5azadC, the conventional FDA-approved nucleoside inhibitor of DNA methylation, its effects on cell differentiation are distinct. Induction of the mature osteoblast phenotype coupled with a sustained cytostatic response was also confirmed in vivo when MC3343 was used against a patient-derived xenograft (PDX). In addition, MC3343 displayed synergistic effects with doxorubicin and cisplatin (CDDP), two major chemotherapeutic agents used to treat osteosarcoma. Specifically, MC3343 increased stable doxorubicin bonds to DNA, and combined treatment resulted in sustained DNA damage and increased cell death. Overall, this nonnucleoside DNMTi is an effective novel agent and is thus a potential therapeutic option for patients with osteosarcoma who respond poorly to preadjuvant chemotherapy. Mol Cancer Ther; 17(9); 1881–92. ©2018 AACR.


Future Medicinal Chemistry | 2018

Trends of LSD1 inhibitors in viral infections

Clemens Zwergel; Giulia Stazi; Antonello Mai; Sergio Valente

LSD1 was described as an FAD-dependent MAO homolog by Shi et al. in 2004 as the first histone demethylase [1], confirming that histone methylation is a reversible epigenetic mark. LSD1 possesses three main domains containing the N-terminal SWIRM domain, the C-terminal AOL catalytic domain and a central tower domain that contains the binding sites for the following interacting proteins: CoREST, CtBP1, HDAC1/2 and Snail1 [2]. LSD1 is capable of demethylating monoand dimethyl H3K4 and H3K9 [1,2], although more recently it has been shown that the neuron-specific isoform LSD1n has a different substrate, histone H4K20 [3]. Moreover, several nonhistone proteins, such as p53, E2F1, DNMT and MYPT1, have been reported as LSD1 substrates [4]. LSD1 can also interact with several transcription factors that modulate LSD1 involvement in different biological contexts [4]. LSD1 is currently studied mainly in cancer, but due to its wide biological role, its dysfunction is more and more investigated also in neurodegenerative diseases, protein conformation disorders, inflammation, adipogenesis, muscle differentiation, cardiovascular diseases and in viral infections and latency [4]. DNA viruses are encapsulated without histones, but rapidly acquire chromatin structure upon infection [5], making it feasible that a histone methyltransferase or demethylase could be involved in the regulation of DNA virus replication. In the last years, a number of studies confirmed that inhibition of LSD1 can block viral genome transcription and replication of DNA viruses. In contrast, RNA viruses do not rely on chromatin structure and histone capsid for their replication. As a result, LSD1 is believed to possess little influence on RNA virus replication via histone demethylation. Nevertheless, it cannot be ruled out that LSD1 downregulates RNA virus replication via the demethylation of other host or viral proteins [5].

Collaboration


Dive into the Giulia Stazi's collaboration.

Top Co-Authors

Avatar

Sergio Valente

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Antonello Mai

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Clemens Zwergel

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Biagina Marrocco

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Tafani

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Paolo Mellini

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ciro Mercurio

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Ilaria Carnevale

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge