Barbara Forte
National University of Ireland, Galway
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Barbara Forte.
Marine Drugs | 2009
Barbara Forte; Beatrice Malgesini; Claudia Piutti; Francesca Quartieri; Alessandra Scolaro; Gianluca Papeo
In his most celebrated tale “The Picture of Dorian Gray”, Oscar Wilde stated that “those who go beneath the surface do so at their peril”. This sentence could be a prophetical warning for the practitioner who voluntarily challenges himself with trying to synthesize marine sponge-deriving pyrrole-imidazole alkaloids. This now nearly triple-digit membered community has been growing exponentially in the last 20 years, both in terms of new representatives and topological complexity – from simple, achiral oroidin to the breathtaking 12-ring stylissadines A and B, each possessing 16 stereocenters. While the biosynthesis and the role in the sponge economy of most of these alkaloids still lies in the realm of speculations, significant biological activities for some of them have clearly emerged. This review will account for the progress in achieving the total synthesis of the more biologically enticing members of this class of natural products.
Journal of Medicinal Chemistry | 2008
Ermes Vanotti; Raffaella Amici; Alberto Bargiotti; Jens Berthelsen; Roberta Bosotti; Antonella Ciavolella; Alessandra Cirla; Cinzia Cristiani; Roberto D'alessio; Barbara Forte; Antonella Isacchi; Katia Martina; Maria Menichincheri; Antonio Molinari; Alessia Montagnoli; Paolo Orsini; Antonio Pillan; Fulvia Roletto; Alessandra Scolaro; Marcellino Tibolla; Barbara Valsasina; Mario Varasi; Daniele Volpi; Corrado Santocanale
Cdc7 kinase is an essential protein that promotes DNA replication in eukaryotic organisms. Genetic evidence indicates that Cdc7 inhibition can cause selective tumor-cell death in a p53-independent manner, supporting the rationale for developing Cdc7 small-molecule inhibitors for the treatment of cancers. In this paper, the synthesis and structure-activity relationships of 2-heteroaryl-pyrrolopyridinones, the first potent Cdc7 kinase inhibitors, are described. Starting from 2-pyridin-4-yl-1,5,6,7-tetrahydro-pyrrolo[3,2-c]pyridin-4-one, progress toward a simple scaffold, tailored for Cdc7 inhibition, is reported.
Journal of Medicinal Chemistry | 2009
Maria Menichincheri; Alberto Bargiotti; Jens Berthelsen; Jay Aaron Bertrand; Roberto Bossi; Antonella Ciavolella; Alessandra Cirla; Cinzia Cristiani; Croci; Roberto D'alessio; Marina Fasolini; Francesco Fiorentini; Barbara Forte; Antonella Isacchi; Katia Martina; A Molinari; Alessia Montagnoli; Paolo Orsini; Fabrizio Orzi; Enrico Pesenti; Daniele Pezzetta; Antonio Pillan; Italo Poggesi; Fulvia Roletto; Alessandra Scolaro; Marco Tato; Marcellino Tibolla; Barbara Valsasina; Mario Varasi; Daniele Volpi
Cdc7 kinase is a key regulator of the S-phase of the cell cycle, known to promote the activation of DNA replication origins in eukaryotic organisms. Cdc7 inhibition can cause tumor-cell death in a p53-independent manner, supporting the rationale for developing Cdc7 inhibitors for the treatment of cancer. In this paper, we conclude the structure-activity relationships study of the 2-heteroaryl-pyrrolopyridinone class of compounds that display potent inhibitory activity against Cdc7 kinase. Furthermore, we also describe the discovery of 89S, [(S)-2-(2-aminopyrimidin-4-yl)-7-(2-fluoro-ethyl)-1,5,6,7-tetrahydropyrrolo[3,2-c]pyridin-4-one], as a potent ATP mimetic inhibitor of Cdc7. Compound 89S has a Ki value of 0.5 nM, inhibits cell proliferation of different tumor cell lines with an IC50 in the submicromolar range, and exhibits in vivo tumor growth inhibition of 68% in the A2780 xenograft model.
Bioorganic & Medicinal Chemistry Letters | 2011
Italo Beria; Roberto Bossi; Maria Gabriella Brasca; Michele Caruso; Walter Ceccarelli; Gabriele Fachin; Marina Fasolini; Barbara Forte; Francesco Fiorentini; Enrico Pesenti; Daniele Pezzetta; Helena Posteri; Alessandra Scolaro; Stefania Re Depaolini; Barbara Valsasina
As part of our drug discovery effort, we identified and developed 4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline derivatives as PLK1 inhibitors. We now report the optimization of this class that led to the identification of NMS-P937, a potent, selective and orally available PLK1 inhibitor. Also, in order to understand the source of PLK1 selectivity, we determined the crystal structure of PLK1 with NMS-P937. The compound was active in vivo in HCT116 xenograft model after oral administration and is presently in Phase I clinical trials evaluation.
Journal of Medicinal Chemistry | 2010
Maria Menichincheri; Clara Albanese; Cristina Alli; Dario Ballinari; Alberto Bargiotti; Marina Caldarelli; Antonella Ciavolella; Alessandra Cirla; Maristella Colombo; Francesco Colotta; Valter Croci; Roberto D’Alessio; Matteo D’Anello; Antonella Ermoli; Francesco Fiorentini; Barbara Forte; Arturo Galvani; Patrizia Giordano; Antonella Isacchi; Katia Martina; Antonio Molinari; Jürgen Moll; Alessia Montagnoli; Paolo Orsini; Fabrizio Orzi; Enrico Pesenti; Antonio Pillan; Fulvia Roletto; Alessandra Scolaro; Marco Tato
Cdc7 serine/threonine kinase is a key regulator of DNA synthesis in eukaryotic organisms. Cdc7 inhibition through siRNA or prototype small molecules causes p53 independent apoptosis in tumor cells while reversibly arresting cell cycle progression in primary fibroblasts. This implies that Cdc7 kinase could be considered a potential target for anticancer therapy. We previously reported that pyrrolopyridinones (e.g., 1) are potent and selective inhibitors of Cdc7 kinase, with good cellular potency and in vitro ADME properties but with suboptimal pharmacokinetic profiles. Here we report on a new chemical class of 5-heteroaryl-3-carboxamido-2-substituted pyrroles (1A) that offers advantages of chemistry diversification and synthetic simplification. This work led to the identification of compound 18, with biochemical data and ADME profile similar to those of compound 1 but characterized by superior efficacy in an in vivo model. Derivative 18 represents a new lead compound worthy of further investigation toward the ultimate goal of identifying a clinical candidate.
Expert Opinion on Therapeutic Patents | 2009
Gianluca Papeo; Barbara Forte; Paolo Orsini; Claudia Perrera; Helena Posteri; Alessandra Scolaro; Alessia Montagnoli
Background: During the last few years an increasing number of poly(ADP-ribose) polymerase (PARP) inhibitors have been appearing in the context of cancer therapy. This is mainly due to a better knowledge of the best-characterized member of the PARP family of enzymes, PARP-1, further reinforced by the recognition of the clinical benefits arising from its inhibition. Objective/method: The aim of this review is to give the reader an update on PARP inhibition in cancer therapy, by covering both the scientific (SciFinder® search) and the patent literature (Chemical Abstract®/Derwent® search) published recently (2005 – 2008). Conclusions: More patient-compliant orally available PARP-1 inhibitor clinical candidates, along with their possible use as single agents in specific, molecularly defined cancer indications, increase the expectations for this therapeutic approach. The growing understanding of the biological role of other PARPs, such as Tankyrase 1, may be of interest as new potential targets. Besides the classical NAD-mimicking pharmacophore, additional compounds, which either do not resemble nicotinamide or exploit different binding sites, are emerging.
Journal of Medicinal Chemistry | 2015
G.M.E Papeo; Helena Posteri; Daniela Borghi; A.A Busel; F Caprera; Elena Casale; M Ciomei; Alessandra Cirla; E Corti; M D'Anello; Marina Fasolini; Barbara Forte; Arturo Galvani; Antonella Isacchi; A Khvat; M.Y Krasavin; R Lupi; Paolo Orsini; Rita Perego; Enrico Pesenti; Daniele Pezzetta; Sonia Rainoldi; F Riccardi-Sirtori; Alessandra Scolaro; Francesco Sola; Fabio Zuccotto; Eduard Felder; Daniele Donati; Alessia Montagnoli
The nuclear protein poly(ADP-ribose) polymerase-1 (PARP-1) has a well-established role in the signaling and repair of DNA and is a prominent target in oncology, as testified by the number of candidates in clinical testing that unselectively target both PARP-1 and its closest isoform PARP-2. The goal of our program was to find a PARP-1 selective inhibitor that would potentially mitigate toxicities arising from cross-inhibition of PARP-2. Thus, an HTS campaign on the proprietary Nerviano Medical Sciences (NMS) chemical collection, followed by SAR optimization, allowed us to discover 2-[1-(4,4-difluorocyclohexyl)piperidin-4-yl]-6-fluoro-3-oxo-2,3-dihydro-1H-isoindole-4-carboxamide (NMS-P118, 20by). NMS-P118 proved to be a potent, orally available, and highly selective PARP-1 inhibitor endowed with excellent ADME and pharmacokinetic profiles and high efficacy in vivo both as a single agent and in combination with Temozolomide in MDA-MB-436 and Capan-1 xenograft models, respectively. Cocrystal structures of 20by with both PARP-1 and PARP-2 catalytic domain proteins allowed rationalization of the observed selectivity.
Bioorganic & Medicinal Chemistry | 2010
Simona Bindi; Daniele Fancelli; Cristina Alli; Daniela Berta; Jay Aaron Bertrand; Alexander D. Cameron; Paolo Cappella; Patrizia Carpinelli; Giovanni Cervi; Valter Croci; Matteo D’Anello; Barbara Forte; M.Laura Giorgini; Aurelio Marsiglio; Juergen Moll; Enrico Pesenti; Valeria Pittalà; Maurizio Pulici; Federico Riccardi-Sirtori; Fulvia Roletto; Chiara Soncini; Paola Storici; Mario Varasi; Daniele Volpi; Paola Zugnoni; Paola Vianello
A novel series of 3-amino-1H-thieno[3,2-c]pyrazole derivatives demonstrating high potency in inhibiting Aurora kinases was developed. Here we describe the synthesis and a preliminary structure-activity relationship, which led to the discovery of a representative compound (38), which showed low nanomolar inhibitory activity in the anti-proliferation assay and was able to block the cell cycle in HCT-116 cell line. This compound demonstrated favorable pharmacokinetic properties and good efficacy in the HL-60 xenograft tumor model.
Biochemistry | 2013
Giulia Canevari; S Re Depaolini; Ulisse Cucchi; Jay Aaron Bertrand; Elena Casale; Claudia Perrera; Barbara Forte; Patrizia Carpinelli; Eduard R. Felder
Maternal embryonic leucine zipper kinase (MELK) is upregulated in several types of tumor, including breast, prostate, and brain tumors. Its expression is generally associated with cell survival, cell proliferation, and resistance to apoptosis. Therefore, the potential of MELK inhibitors as therapeutic agents is recently attracting considerable interest. Here we report the first structures of MELK in complex with AMP-PNP and with nanomolar inhibitors. Our studies shed light on the role of the MELK UBA domain, provide a characterization of the kinase active site, and identify key residues for achieving high potency, laying the groundwork for structure-based drug design efforts.
Cancer Research | 2010
Gianluca Papeo; Sonia Rainoldi; Laura Gianellini; Antonella Ciavolella; Antonella Leone; Rosita Lupi; Francesco Sola; Francesco Caprera; Mauro Paolucci; Sandrine Thieffine; Rita Perego; Clara Albanese; Elena Casale; Barbara Forte; Marco Guanci; Paolo Orsini; Helena Posteri; Alessandra Scolaro; Federico Riccardi-Sirtori; Fabio Zuccotto; Daniele Donati; Jurgen Moll; Alessia Montagnoli
Poly (ADP-ribose) polymerase 1 and 2 (Parp-1 and Parp-2) are nuclear enzymes responsible for signaling the presence of DNA damages by catalyzing the addition of ADP-ribose units to DNA, histones and various DNA repair enzymes, thus facilitating DNA repair. Parp-1 has been gaining increasing interest as a therapeutic target for cancer in combination with DNA damaging agents but also as single agent in particular tumor settings, such as BRCA mutated tumors. While Parp-1 knock out mice are viable and display only defects in DNA repair, double Parp-1 and Parp-2 knock-out are embryonic lethal indicating that the absence of Parp-1 and Parp-2 is less tolerated in normal cells. We confirmed this concept in vitro by RNAi of Parp-1 and Parp-2, showing that the inhibition of Parp-1 alone is enough to kill tumor cells but spares normal cells. Hence a selective Parp-1 inhibitor might have a better safety profile, particularly in view of a long treatment. Here we report the discovery and the in vitro and in vivo characterization of a novel class of potent, orally available Parp-1 selective small molecule inhibitors with no activity on other Parp isoforms such as Parp-2, −3 and −5a (tankyrase), which differentiates it from current clinical Parp inhibitors. The most potent compounds were further evaluated in vitro and in vivo. These compounds are highly potent in inhibiting DNA damage-induced Parp-1 activity in cells and exhibit selective anti-proliferative and pro-apoptotic activity in tumor cell lines harbouring defects in DNA repair. In vivo, the compounds show an excellent pharmacokinetic profile with almost complete oral bioavailability and demonstrate efficacy as a single agent in xenograft tumor models with DNA repair deficiencies superior to reference Parp inhibitors. These compounds are very well tolerated with no overt toxicity even after prolonged exposure and their mechanism of action is confirmed both in tumors and peripheral blood cells of treated mice. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 691.