Network


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

Hotspot


Dive into the research topics where Maurizio D’Incalci is active.

Publication


Featured researches published by Maurizio D’Incalci.


Cancer Cell | 2013

Role of Macrophage Targeting in the Antitumor Activity of Trabectedin

Giovanni Germano; Roberta Frapolli; Cristina Belgiovine; Achille Anselmo; Samantha Pesce; Manuela Liguori; Eugenio Erba; Sarah Uboldi; Massimo Zucchetti; Fabio Pasqualini; Manuela Nebuloni; Nico van Rooijen; Roberta Mortarini; Luca Beltrame; Sergio Marchini; Ilaria Fuso Nerini; Roberta Sanfilippo; Paolo G. Casali; Silvana Pilotti; Carlos M. Galmarini; Andrea Anichini; Alberto Mantovani; Maurizio D’Incalci; Paola Allavena

There is widespread interest in macrophages as a therapeutic target in cancer. Here, we demonstrate that trabectedin, a recently approved chemotherapeutic agent, induces rapid apoptosis exclusively in mononuclear phagocytes. In four mouse tumor models, trabectedin caused selective depletion of monocytes/macrophages in blood, spleens, and tumors, with an associated reduction of angiogenesis. By using trabectedin-resistant tumor cells and myeloid cell transfer or depletion experiments, we demonstrate that cytotoxicity on mononuclear phagocytes is a key component of its antitumor activity. Monocyte depletion, including tumor-associated macrophages, was observed in treated tumor patients. Trabectedin activates caspase-8-dependent apoptosis; selectivity for monocytes versus neutrophils and lymphocytes is due to differential expression of signaling and decoy TRAIL receptors. This unexpected property may be exploited in different therapeutic strategies.


Nature Communications | 2014

microRNA-181a has a critical role in ovarian cancer progression through the regulation of the epithelial-mesenchymal transition.

Aditya Parikh; Christine Elaine Lee; Peronne Joseph; Sergio Marchini; Alessia Baccarini; V. Kolev; Chiara Romualdi; Robert Fruscio; Hardik Shah; Feng Wang; Gavriel Mullokandov; David A. Fishman; Maurizio D’Incalci; Jamal Rahaman; Tamara Kalir; Raymond W. Redline; Brian D. Brown; Goutham Narla; Analisa DiFeo

Ovarian cancer is a leading cause of cancer deaths among women. Effective targets to treat advanced epithelial ovarian cancer (EOC) and biomarkers to predict treatment response are still lacking because of the complexity of pathways involved in ovarian cancer progression. Here we show that miR-181a promotes TGF-β-mediated epithelial-to-mesenchymal transition via repression of its functional target, Smad7. miR-181a and phosphorylated Smad2 are enriched in recurrent compared with matched-primary ovarian tumours and their expression is associated with shorter time to recurrence and poor outcome in patients with EOC. Furthermore, ectopic expression of miR-181a results in increased cellular survival, migration, invasion, drug resistance and in vivo tumour burden and dissemination. In contrast, miR-181a inhibition via decoy vector suppression and Smad7 re-expression results in significant reversion of these phenotypes. Combined, our findings highlight an unappreciated role for miR-181a, Smad7, and the TGF-β signalling pathway in high-grade serous ovarian cancer.


Nucleic Acids Research | 2009

Stabilization of quadruplex DNA perturbs telomere replication leading to the activation of an ATR-dependent ATM signaling pathway.

Angela Rizzo; Erica Salvati; Manuela Porru; Carmen D’Angelo; Malcolm F. G. Stevens; Maurizio D’Incalci; Carlo Leonetti; Eric Gilson; Gabriella Zupi; Annamaria Biroccio

Functional telomeres are required to maintain the replicative ability of cancer cells and represent putative targets for G-quadruplex (G4) ligands. Here, we show that the pentacyclic acridinium salt RHPS4, one of the most effective and selective G4 ligands, triggers damages in cells traversing S phase by interfering with telomere replication. Indeed, we found that RHPS4 markedly reduced BrdU incorporation at telomeres and altered the dynamic association of the telomeric proteins TRF1, TRF2 and POT1, leading to chromosome aberrations such as telomere fusions and telomere doublets. Analysis of the molecular damage pathway revealed that RHPS4 induced an ATR-dependent ATM signaling that plays a functional role in the cellular response to RHPS4 treatment. We propose that RHPS4, by stabilizing G4 DNA at telomeres, impairs fork progression and/or telomere processing resulting in telomere dysfunction and activation of a replication stress response pathway. The detailed understanding of the molecular mode of action of this class of compounds makes them attractive tools to understand telomere biology and provides the basis for a rational use of G4 ligands for the therapy of cancer.


European Journal of Cancer | 2009

Contemporary pre-clinical development of anticancer agents – What are the optimal preclinical models?

Giovanna Damia; Maurizio D’Incalci

The successful identification of novel effective anticancer drugs is largely dependent on the use of appropriate preclinical experimental models that should possibly mimic the complexity of different cancer diseases. The huge number of targets suitable for the design of new anticancer drugs is producing hundreds of novel molecules that require appropriate experimental models to investigate their mode of action and antitumour activity in order to select for clinical investigation the ones with higher chances of being clinically effective. However, our ability to predict the clinical efficacy of a new compound in the clinic based on preclinical data is still limited. This paper overviews the in vitro/in vivo preclinical systems that are currently used to test either compounds with an unknown mechanism of action or compounds designed to hit cancer-specific or cancer-related molecular targets. Examples of experimental models successfully used to identify novel compounds are provided. Xenografts are still the most commonly used in vivo models in drug development due to their high degree of reproducibility and because, in some cases, particularly when orthotopically transplanted, they maintain several biological properties of the human tumours they derive from. Genetic models are very useful for target validation, but are often not sufficiently reproducible to be used for drug evaluation. The variety of animal models can be effectively used to optimally test drugs that presumably act by a defined mode of action, but final success is highly dependent on the ability of drug development teams to integrate different expertises such as biology, chemistry, pharmacology, toxicology and clinical oncology into a clever and well orchestrated plan that keeps in consideration both the complexity of cancer diseases, involving alterations of different pathways, and the complexity of drugs whose pharmacological properties are crucial to obtain the desired effects.


European Journal of Cancer | 2013

Resistance to platinum-based chemotherapy is associated with epithelial to mesenchymal transition in epithelial ovarian cancer

Sergio Marchini; Robert Fruscio; Luca Clivio; Luca Beltrame; Luca Porcu; Ilaria Fuso Nerini; Duccio Cavalieri; Giovanna Chiorino; Giorgio Cattoretti; Costantino Mangioni; Rodolfo Milani; Valter Torri; Chiara Romualdi; Alberto Zambelli; Michela Romano; M. Signorelli; Silvana Di Giandomenico; Maurizio D’Incalci

BACKGROUND The present study is aimed to identify genetic pathways correlated with chemoresistance in epithelial ovarian cancer (EOC). METHODS We compared the molecular profiles of 23 tumour biopsies of stage III-IV (training set) at primary surgery, before chemotherapy, to the profile from the same patients at second surgery, after several lines of platinum (Pt)-based chemotherapy when the tumours were resistant. In the hypothesis that identified markers were related to Pt-resistance and to prognosis, we validated this signature in 52 EOC taken at primary surgery (validation set) selected to be either very sensitive to the first line therapy, i.e. not relapsing before one year from the end of therapy, or resistant, i.e. relapsing within 6 months from the end of therapy. RESULTS In the training set, we identified a resistance signature indicative of the activation of epithelial to mesenchymal transition (EMT) by transforming growth factor (TGF)-beta pathway. We then validated this signature in 52 EOC taken at primary surgery (validation set). Some genes involved in EMT, such as BMP and activin membrane-bound inhibitor (BAMBI), and mir-141 resulted in association with overall or progression free survival. CONCLUSION Some genes involved in EMT were associated to overall or progression free survival, suggesting EMT as vital to the resistance mechanisms.


Cancer Treatment Reviews | 2013

Targeting triple negative breast cancer: Is p53 the answer?

Natalie Turner; Erica Moretti; Olimpia Siclari; Ilenia Migliaccio; Libero Santarpia; Maurizio D’Incalci; Stefano Piccolo; Andrea Veronesi; Alberto Zambelli; Gianni Del Sal; Angelo Di Leo

Triple negative breast cancers, which are defined by lack of expression of estrogen, progesterone, or HER2 receptors, represent approximately 15% of all breast cancers, although they account for a much higher proportional of breast cancer mortality. This is due both to their innate aggressive biological characteristics, but also to lack of effective therapies. Conventional chemotherapy is currently the only treatment option, thus there is a critical need to find new and effective targeted therapies in this disease. While investigation of agents such as poly (ADP-ribose) polymerase (PARP) inhibitors and EGFR inhibitors continues, results from recent clinical trials indicate that these therapies are not as active in sporadic triple negative breast cancers as initially hoped. It is important therefore to consider other emerging therapeutic agents. Mutation in p53 is found in the vast majority of triple negative breast cancers, and as such is a target of particular interest. Within this review, several agents with potential activity against aberrant p53 signaling have been considered, as a novel approach to finding an effective targeted therapy for this aggressive breast cancer subtype.


Expert Opinion on Investigational Drugs | 2003

Preclinical and clinical results with the natural marine product ET-743

Maurizio D’Incalci; José Jimeno

ET-743 (Yondelis™, trabectedin) is a natural marine product with antitumour properties derived from the tunicate Ecteinascidia turbinata. ET-743 binds to the N2 position of guanine in the minor groove of DNA with some degree of sequence specificity, altering the transcription regulation of induced genes. Cells that are deficient in nucleotide excision repair, hypersensitive to UV rays, cisplatin and conventional alkylating agents, are resistant to ET-743. This is a unique property of ET-743 and is of potential importance for the drug activity when administered alone or in combination with other drugs. ET-743 showed striking antitumour activity against sensitive and resistant human xenografts. The dose-limiting toxicities in animal models, hepatobiliary events, were of concern, but the pattern of the reversibility noted in monkeys and the evidence of a positive therapeutic index in tumour-bearing nude mice prompted its clinical development. The Phase I programme investigated different schedules of administration, with the dose-limiting toxicities being neutropenia and fatigue. As anticipated in the preclinical models, reversible non-cumulative transaminitis was a prevalent finding from one-third of the maximum tolerated dose level; long-lasting objective responses in pretreated resistant patients were noted, including consistent efficacy data in mesenchymal tumours. The Phase II data for ET-743 administered as a single agent has established a clinical role for the compound in advanced pretreated soft tissue sarcoma and a promising potential in pretreated ovarian and breast cancer. ET-743 combined with other drugs (i.e., cisplatin, paclitaxel or doxorubicin) showed more than additive effects in several preclinical systems and initial clinical results (e.g., a combination of ET-743 with cisplatin) appear to confirm the preclinical findings. In summary, ET-743 is a new drug with a novel mode of action, which has demonstrated activity in human tumours resistant to the available anticancer drugs. Further comparative studies are needed to define the role of ET-743 alone or in combination in cancer chemotherapy.


Food and Chemical Toxicology | 2011

Chemical characterization of Iraqi propolis samples and assessing their antioxidant potentials.

Ghassan M. Sulaiman; Khulood W. Al Sammarrae; Ali H. Ad’hiah; Massimo Zucchetti; Roberta Frapolli; Ezia Bello; Eugenio Erba; Maurizio D’Incalci; Renzo Bagnati

Propolis samples, collected from different geographical locations in Iraq (Baghdad, Dahuk, Mosul and Salah ad-Din), were analyzed and assessed for their anti-oxidant activity. Concentrations of phenolic compounds (flavonoids, phenolic acids and their esters) in propolis were estimated using high performance liquid chromatography coupled to electrospray mass spectrometry. Thirty-eight different compounds were identified and 33 of them were polyphenols. Other compounds were tentatively identified as clerodane diterpenoids, and one was considered unknown. Semi-quantitative measurements showed that phenolic acids and their esters were the predominant constituents in propolis extracts, followed by flavones and flavonols, and then flavanones and dihydroflavonols. Propolis samples were further spectrophotometrically characterized using the Folin-Ciocalteu reagent for the determination of total phenolic compounds. The free radical scavenging activities of propolis samples were also evaluated by using the 2,2-diphenyl-1-picrylhydrazyl assay. The results revealed that propolis extracts exhibited strong free radical scavenging activity.


European Journal of Cancer | 2009

Phase I clinical and pharmacokinetic study of trabectedin and doxorubicin in advanced soft tissue sarcoma and breast cancer

Cristiana Sessa; Antonella Perotti; Cristina Noberasco; F. De Braud; E. Gallerani; Sara Cresta; Massimo Zucchetti; L. Viganò; A. Locatelli; Jose Jimeno; J.W. Feilchenfeldt; Maurizio D’Incalci; G Capri; N. Ielmini; Luca Gianni

The combination of trabectedin (T) and doxorubicin (D) was brought into clinical development in soft tissue sarcoma (STS) and advanced breast cancer (ABC) because of its in vitro and in vivo additive anti-tumour effect, the fact that there are no overlapping toxicities and the anti-tumour activity of T in those tumours. Feasibility and anti-tumour activity of T+D administered every 3 weeks were evaluated in 38 patients (STS=29, ABC=9) untreated for advanced disease. D was given at 60 mg/m(2) and T at escalating doses from 600 to 800 microg/m(2), which was the maximum tolerated dose due to dose-limiting febrile neutropenia and asthenia. The recommended dose--given to 18 patients in total--was 700 microg/m(2) T with 60 mg/m(2) D. The pharmacokinetic profile of T and D at cycle 1 was analysed in 20 patients. The most common toxicities included a severe but reversible ASAT/ALAT increase (94%), nausea/vomiting, neutropenia, asthenia/fatigue, stomatitis. Partial response and stable disease were assessed in 18% and 56% of STS patients and in 55% and 33% of ABC patients. No pharmacokinetic interaction between T and D was observed. The lack of cumulative toxicity and related complications and the promising activity in STS support further development of T+D.


European Journal of Cancer | 2013

A first in human phase I study of the proteasome inhibitor CEP-18770 in patients with advanced solid tumours and multiple myeloma

E. Gallerani; Massimo Zucchetti; Dario Brunelli; Elena Marangon; Cristina Noberasco; Dagmar Hess; Angelo Delmonte; Giovanni Martinelli; Steffen Böhm; Christopher Driessen; Filippo de Braud; Silvia Marsoni; Roberta Cereda; Federica Sala; Maurizio D’Incalci; Cristiana Sessa

BACKGROUND The safety, pharmacokinetics (PK) and pharmacodynamics of CEP-18770, a new peptide boronic acid proteasome inhibitor, have been investigated after intravenous administration on days 1, 4, 8 and 11 of every 21d cycle in patients with solid tumours and multiple myeloma (MM). PATIENTS AND METHODS Thirty-eight patients were treated with CEP-18770 at escalating doses from 0.1 to 1.8mg/m(2) where 2 out of 5 patients showed dose limiting toxicities. The maximum tolerated/recommended dose (MTD/RD) of 1.5mg/m(2) was tested in 12 additional patients. Skin rash was dose-limiting and occurred in 53% of patients; other frequent toxicities were asthenia (29%), stomatitis (21%) and pyrexia (16%). No significant peripheral neuropathy was observed. PK in plasma was linear with a half-life of the elimination phase of 62.0±43.5h. Proteasome inhibition in peripheral blood mononuclear cells was dose related in MM patients; it was of 45.4±11.5% at the RD. CONCLUSIONS CEP-18770 showed a favourable safety profile with lack of neurotoxicity and linear plasma PK. The definition of the optimal biological dose and schedule of treatment is actively pursued because of the high incidence of skin toxicity of the twice a week schedule.

Collaboration


Dive into the Maurizio D’Incalci's collaboration.

Top Co-Authors

Avatar

Massimo Zucchetti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Roberta Frapolli

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Sergio Marchini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Eugenio Erba

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Luca Beltrame

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Paolo Ubezio

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Ezia Bello

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Lavinia Morosi

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Massimo Broggini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge