Network


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

Hotspot


Dive into the research topics where Alessandra Cirla is active.

Publication


Featured researches published by Alessandra Cirla.


Journal of Medicinal Chemistry | 2008

Cdc7 Kinase Inhibitors : Pyrrolopyridinones as Potential Antitumor Agents. 1. Synthesis and Structure-Activity Relationships

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 | 2013

Alkylsulfanyl-1,2,4-triazoles, a new class of allosteric valosine containing protein inhibitors. Synthesis and structure-activity relationships.

Paolo Polucci; Paola Magnaghi; Mauro Angiolini; Daniela Asa; Nilla Avanzi; Alessandra Badari; Jay Aaron Bertrand; Elena Casale; Silvia Cauteruccio; Alessandra Cirla; Arturo Galvani; Peter K. Jackson; Yichin Liu; Steven Magnuson; Beatrice Malgesini; Stefano Nuvoloni; Christian Orrenius; Federico Riccardi Sirtori; Laura Riceputi; Simona Rizzi; Beatrice Trucchi; Tom O’Brien; Antonella Isacchi; Daniele Donati; Roberto D’Alessio

Valosine containing protein (VCP), also known as p97, is a member of AAA ATPase family that is involved in several biological processes and plays a central role in the ubiquitin-mediated degradation of misfolded proteins. VCP is an ubiquitously expressed, highly abundant protein and has been found overexpressed in many tumor types, sometimes associated with poor prognosis. In this respect, VCP has recently received a great deal of attention as a potential new target for cancer therapy. In this paper, the discovery and structure-activity relationships of alkylsulfanyl-1,2,4-triazoles, a new class of potent, allosteric VCP inhibitors, are described. Medicinal chemistry manipulation of compound 1, identified via HTS, led to the discovery of potent and selective inhibitors with submicromolar activity in cells and clear mechanism of action at consistent doses. This represents a first step toward a new class of potential anticancer agents.


Journal of Medicinal Chemistry | 2009

First Cdc7 kinase inhibitors: pyrrolopyridinones as potent and orally active antitumor agents. 2. Lead discovery.

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.


Expert Opinion on Therapeutic Patents | 2013

PARP inhibitors in cancer therapy: an update

Gianluca Papeo; Elena Casale; Alessia Montagnoli; Alessandra Cirla

Introduction: Inhibitors of the poly(ADP-ribose) polymerases (PARPs) family of proteins are currently being evaluated as potential anticancer medicines at both preclinical and clinical levels. They have the peculiarity to increase the efficacy of DNA-damaging agents and to selectively target tumor cells with specific DNA repair defects. This later development of these drugs should make it possible, in principle, to selectively target neoplastic vs healthy cells, thus realizing the Ehrlichs magic bullet concept of a personalized and tailored cure of diseases. Areas covered: This review is designed to provide the readers with a brief summary and an update on PARP inhibitors in the oncology field, by covering the recent patent literature (2010 – 2012: and Questel Intellectual Property Portal [QPat] database search). Expert opinion: Presently, along with a number of preclinical candidates, there are eight PARP inhibitors in the clinic as either single agents or in combination with various chemotherapy and radiotherapy regimens. The tremendous efforts underneath those results testify the high interest on the target. The investigation and understanding of the cross-reactivity among members of the PARPs family as well as a deeper knowledge of their biological functions may lead to a more profound characterization of the PARP inhibitors profile. This, in turn, will cast additional light on this exciting approach in treating cancer.


Journal of Medicinal Chemistry | 2010

Cdc7 Kinase Inhibitors: 5-Heteroaryl-3-Carboxamido-2-Aryl Pyrroles as Potential Antitumor Agents. 1. Lead Finding

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.


Journal of Medicinal Chemistry | 2015

Discovery of 2-[1-(4,4-Difluorocyclohexyl)Piperidin-4-Yl]-6-Fluoro-3-Oxo-2,3-Dihydro-1H-Isoindole-4-Carboxamide (Nms-P118): A Potent, Orally Available and Highly Selective Parp- 1 Inhibitor for Cancer Therapy.

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.


Journal of Medicinal Chemistry | 2014

Discovery of 2-(cyclohexylmethylamino)pyrimidines as a new class of reversible valosine containing protein inhibitors.

Giovanni Cervi; Paola Magnaghi; Daniela Asa; Nilla Avanzi; Alessandra Badari; Daniela Borghi; Michele Caruso; Alessandra Cirla; Eduard Felder; Arturo Galvani; Fabio Gasparri; Antonio Lomolino; Steven Magnuson; Beatrice Malgesini; Ilaria Motto; Maurizio Pasi; Simona Rizzi; Barbara Salom; Graziella Sorrentino; Sonia Troiani; Barbara Valsasina; Thomas O’Brien; Antonella Isacchi; Daniele Donati; Roberto D’Alessio

Valosine-containing protein (VCP), also known as p97 or cdc48 in yeast, is a highly abundant protein belonging to the AAA ATPase family involved in a number of essential cellular functions, including ubiquitin-proteasome mediated protein degradation, Golgi reassembly, transcription activation, and cell cycle control. Altered expression of VCP has been detected in many cancer types sometimes associated with poor prognosis. Furthermore, VCP mutations are causative of some neurodegenerative disorders. In this paper we report the discovery, synthesis, and structure-activity relationships of substituted 2-aminopyrimidines, representing a new class of reversible VCP inhibitors. This class of compounds, identified in a HTS campaign against recombinant VCP, has been progressively expanded and manipulated to increase biochemical potency and gain cellular activity.


Bioorganic & Medicinal Chemistry | 2014

Pyrrole-3-Carboxamides as Potent and Selective Jak2 Inhibitors.

Maria Gabriella Brasca; Marcella Nesi; Nilla Avanzi; Dario Ballinari; Tiziano Bandiera; Jay Aaron Bertrand; Simona Bindi; Giulia Canevari; Davide Carenzi; Daniele Casero; Lucio Ceriani; Marina Ciomei; Alessandra Cirla; Maristella Colombo; Sabrina Cribioli; Cinzia Cristiani; Franco Della Vedova; Gabriele Fachin; Marina Fasolini; Eduard R. Felder; Arturo Galvani; Antonella Isacchi; Danilo Mirizzi; Ilaria Motto; Achille Panzeri; Enrico Pesenti; Paola Vianello; Paola Gnocchi; Daniele Donati

We report herein the discovery, structure guided design, synthesis and biological evaluation of a novel class of JAK2 inhibitors. Optimization of the series led to the identification of the potent and orally bioavailable JAK2 inhibitor 28 (NMS-P953). Compound 28 displayed significant tumour growth inhibition in SET-2 xenograft tumour model, with a mechanism of action confirmed in vivo by typical modulation of known biomarkers, and with a favourable pharmacokinetic and safety profile.


Cancer Research | 2017

Abstract 2082: NMS-E668, a potent and selective RET kinase inhibitor characterized by specificity towards VEGFR2 and high antitumor efficacy against RET-driven models

Elena Ardini; Patrizia Banfi; Nilla Avanzi; Marina Ciomei; Paolo Polucci; Alessandra Cirla; Antonella Ermoli; Ilaria Motto; Elena Casale; Giulia Canevari; Cinzia Cristiani; Sonia Troiani; Federico Riccardi Sirtori; Nadia Amboldi; Dario Ballinari; Francesco Caprera; Eduard Felder; Arturo Galvani; Daniele Donati; Antonella Isacchi; Maria Menichincheri

RET, a receptor tyrosine kinase (RTK) expressed mainly in neural crest-derived tissues, plays a role in cell growth and differentiation and its physiological activation depends upon binding to the GDNF family. Genetic aberrations leading to constitutive RET activation are well-established as oncogenic events. Activating point mutations of RET, for example, are present in ca. 70% of medullary thyroid carcinoma patients including all hereditary cases, while RET gene rearrangements resulting in production of activated RET fusion proteins occur in approximately 10% of sporadic papillary thyroid carcinomas. More recently, recurring RET gene rearrangements have also been found in 1-2 % of lung adenocarcinomas and subsets of other solid tumors including colorectal and salivary gland carcinomas. Thus RET kinase represents an actionable therapeutic target in multiple clinical settings with high medical need. Consequently several small-molecule inhibitors targeting this kinase have been explored in clinical settings. A common feature of most advanced agents is their lack of selectivity and in particular their potent cross-reactivity against VEGFR2, an RTK whose inhibition is associated with serious, dose-limiting cardiovascular toxicity. Indeed, the high homology between the two kinases renders identification of ATP competitive compounds that selectively inhibit RET over VEGFR2 a highly challenging task. Here we describe the preclinical activity of NMS-E668, a potent and selective ATP-competitive RET inhibitor characterized by favorable activity, selectivity and ADME profiles. Biochemically, NMS-E668 has an excellent selectivity profile against a panel of >50 kinases, notably including >10-fold selectivity over VEGFR2. Selectivity of NMS-E668 for RET vs. VEGFR2 was confirmed in NIH-3T3 cells engineered to express activated forms of these kinases. NMS-E668 potently (IC50 circa 50 nM) and selectively inhibited proliferation of RET-dependent tumor cells, including TT medullary carcinoma cells harboring a RET C634W activating point mutation and LC2/ad lung carcinoma cells bearing the oncogenic fusion protein CCDC6-RET. NMS-E668 also potently inhibited IL3-independent growth of Ba/F3 cells expressing KIF5B-RET, the RET rearrangement that is most commonly found in lung adenocarcinomas. Cellular mechanism studies confirmed that NMS-E668 inhibits RET autophosphorylation and downstream signaling at doses consistent with antiproliferative activity. Tested in vivo against KIF5B-RET-driven Ba/F3 tumors, NMS-E668 displayed >90% tumor growth inhibition accompanied by target modulation following oral administration at 10 and 20 mg/kg, with prolonged tumor regressions observed at the higher dose. Thus NMS-E668, a potent and VEGFR2-sparing RET inhibitor is an innovative and highly promising candidate for further development. Citation Format: Elena Ardini, Patrizia Banfi, Nilla Avanzi, Marina Ciomei, Paolo Polucci, Alessandra Cirla, Antonella Ermoli, Ilaria Motto, Elena Casale, Giulia Canevari, Cinzia Cristiani, Sonia Troiani, Federico Riccardi Sirtori, Nadia Amboldi, Dario Ballinari, Francesco Caprera, Eduard Felder, Arturo Galvani, Daniele Donati, Antonella Isacchi, Maria Menichincheri. NMS-E668, a potent and selective RET kinase inhibitor characterized by specificity towards VEGFR2 and high antitumor efficacy against RET-driven models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2082. doi:10.1158/1538-7445.AM2017-2082


Cancer Research | 2015

Abstract 2851: Preclinical characterization of NMS-P648, a novel and potent PARP-1/-3 inhibitor

Alessia Montagnoli; Gianluca Papeo; Sonia Rainoldi; Alessandra Cirla; Antonella Ciavolella; Clara Albanese; Michele Modugno; Roberta Bosotti; Alessio Somaschini; Giovanni Carapezza; Rosita Lupi; Dario Ballinari; Marina Ciomei; Enrico Pesenti; Daniele Donati; Antonella Isacchi; Arturo Galvani

PARP-1 and PARP-2 are nuclear enzymes activated by DNA strand breaks and are involved in recruitment of DNA repair proteins to sites of damage. PARP-1 and PARP-2 share partially overlapping functions, as shown by the embryonic lethality of double knockout mice compared to animals with single ablation either gene, which are viable. Despite this partial functional redundancy, PARP-1, but not PARP-2, inhibition has been shown to be synthetically lethal with defects in homologous recombination such as BRCA gene mutations. All PARP inhibitors that are currently under clinical investigation inhibit both PARP-1 and PARP-2, due to the high sequence similarity between the catalytic domains of the two enzymes. A third member of the family, PARP-3, has lower homology with PARP-1 and -2 and is much less well characterized, although it is known that its activity is stimulated by double strand breaks and that it promotes DNA repair through an error prone repair pathway, non-homologous end joining (NHEJ). PARP-3 inhibition is thus presumed to be synthetically lethal with other, alternative DNA repair pathway deficiencies. We report preclinical characterization of NMS-P648, a selective PARP-1/-3 inhibitor, with a dissociation constants (K D ), of 0.9 nM on PARP-1 and with greater than four hundred-fold less affinity for PARP-2, as assessed by direct binding assay. In cells, NMS-P648 inhibits hydrogen peroxide induced poly ADP-ribose (PAR) synthesis with an IC 50 of 1 nM, confirming expected mechanism of action and indicating that inhibition of PARP-2 is not required for this process. NMS-P648 has favourable ADME properties, including stability in liver microsome incubation assays, low efflux ratio in the CACO2 assay and an excellent pharmacokinetic profile, with complete oral bioavailability and exposure levels that increase proportionally with dose. Testing NMS-P648 on a panel of more than 100 human tumor cell lines using a 2D colony forming assay format revealed that 7 cell lines were highly sensitive to the compound (IC 50 Citation Format: Alessia Montagnoli, Gianluca Papeo, Sonia Rainoldi, Alessandra Cirla, Antonella Ciavolella, Clara Albanese, Michele Modugno, Roberta Bosotti, Alessio Somaschini, Giovanni Carapezza, Rosita Lupi, Dario Ballinari, Marina Ciomei, Enrico Pesenti, Daniele Donati, Antonella Isacchi, Arturo Galvani. Preclinical characterization of NMS-P648, a novel and potent PARP-1/-3 inhibitor. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2851. doi:10.1158/1538-7445.AM2015-2851

Collaboration


Dive into the Alessandra Cirla's collaboration.

Top Co-Authors

Avatar

Alessandra Scolaro

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Maria Menichincheri

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Barbara Forte

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Alessia Montagnoli

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Paolo Orsini

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonella Ciavolella

National University of Ireland

View shared research outputs
Top Co-Authors

Avatar

Antonio Pillan

National University of Ireland

View shared research outputs
Researchain Logo
Decentralizing Knowledge