Network


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

Hotspot


Dive into the research topics where Chiara Fazio is active.

Publication


Featured researches published by Chiara Fazio.


Gut | 2014

Anticolorectal cancer activity of the omega-3 polyunsaturated fatty acid eicosapentaenoic acid

Andrew J. Cockbain; Milene Volpato; Amanda Race; Alessandra Munarini; Chiara Fazio; Andrea Belluzzi; Paul M. Loadman; Giles J. Toogood; Mark A. Hull

Background Oral administration of the omega-3 fatty acid eicosapentaenoic acid (EPA), as the free fatty acid (FFA), leads to EPA incorporation into, and reduced growth of, experimental colorectal cancer liver metastases (CRCLM). Design: We performed a Phase II double-blind, randomised, placebo-controlled trial of EPA-FFA 2 g daily in patients undergoing liver resection surgery for CRCLM. The patients took EPA-FFA (n=43) or placebo (n=45) prior to surgery. The primary end-point was the CRCLM Ki67 proliferation index (PI). Secondary end-points included safety and tolerability of EPA-FFA, tumour fatty acid content and CD31-positive vascularity. We also analysed overall survival (OS) and disease-free survival (DFS). Results The median (range) duration of EPA-FFA treatment was 30 (12–65) days. Treatment groups were well matched with no significant difference in disease burden at surgery or preoperative chemotherapy. EPA-FFA treatment was well tolerated with no excess of postoperative complications. Tumour tissue from EPA-FFA-treated patients demonstrated a 40% increase in EPA content (p=0.0008), no difference in Ki67 PI, but reduced vascularity in ‘EPA-naïve’ individuals (p=0.075). EPA-FFA also demonstrated antiangiogenic activity in vitro. In the first 18 months after CRCLM resection, EPA-FFA-treated individuals obtained OS benefit compared with placebo, although early CRC recurrence rates were similar. Conclusions EPA-FFA therapy is safe and well tolerated in patients with advanced CRC undergoing liver surgery. EPA-FFA may have antiangiogenic properties. Remarkably, limited preoperative treatment may provide postoperative OS benefit. Phase III clinical evaluation of prolonged EPA-FFA treatment in CRCLM patients is warranted. Trial Identifier: ClinicalTrials.gov NCT01070355.


International Journal of Cancer | 2014

Eicosapentaenoic acid free fatty acid prevents and suppresses colonic neoplasia in colitis-associated colorectal cancer acting on Notch signaling and gut microbiota

Giulia Piazzi; Giuseppe D'Argenio; Anna Prossomariti; Vincenzo Lembo; Giovanna Mazzone; Marco Candela; Elena Biagi; Patrizia Brigidi; Paola Vitaglione; Vincenzo Fogliano; Leonarda D'Angelo; Chiara Fazio; Alessandra Munarini; Andrea Belluzzi; Claudio Ceccarelli; Pasquale Chieco; Tiziana Balbi; Paul M. Loadman; Mark A. Hull; Marco Romano; Franco Bazzoli; Luigi Ricciardiello

Inflammatory bowel diseases are associated with increased risk of developing colitis‐associated colorectal cancer (CAC). Epidemiological data show that the consumption of ω‐3 polyunsaturated fatty acids (ω‐3 PUFAs) decreases the risk of sporadic colorectal cancer (CRC). Importantly, recent data have shown that eicosapentaenoic acid‐free fatty acid (EPA‐FFA) reduces polyp formation and growth in models of familial adenomatous polyposis. However, the effects of dietary EPA‐FFA are unknown in CAC. We tested the effectiveness of substituting EPA‐FFA, for other dietary fats, in preventing inflammation and cancer in the AOM‐DSS model of CAC. The AOM‐DSS protocols were designed to evaluate the effect of EPA‐FFA on both initiation and promotion of carcinogenesis. We found that EPA‐FFA diet strongly decreased tumor multiplicity, incidence and maximum tumor size in the promotion and initiation arms. Moreover EPA–FFA, in particular in the initiation arm, led to reduced cell proliferation and nuclear β‐catenin expression, whilst it increased apoptosis. In both arms, EPA‐FFA treatment led to increased membrane switch from ω‐6 to ω‐3 PUFAs and a concomitant reduction in PGE2 production. We observed no significant changes in intestinal inflammation between EPA‐FFA treated arms and AOM‐DSS controls. Importantly, we found that EPA‐FFA treatment restored the loss of Notch signaling found in the AOM‐DSS control and resulted in the enrichment of Lactobacillus species in the gut microbiota. Taken together, our data suggest that EPA‐FFA is an excellent candidate for CRC chemoprevention in CAC.


Carcinogenesis | 2014

A combination of eicosapentaenoic acid-free fatty acid, epigallocatechin-3-gallate and proanthocyanidins has a strong effect on mTOR signaling in colorectal cancer cells

Leonarda D’Angelo; Giulia Piazzi; Annalisa Pacilli; Anna Prossomariti; Chiara Fazio; Lorenzo Montanaro; Giulia Graziani; Vincenzo Fogliano; Alessandra Munarini; Francesca Bianchi; Andrea Belluzzi; Franco Bazzoli; Luigi Ricciardiello

Colorectal cancer (CRC) is one of the major causes of cancer death worldwide. The development of novel anti-CRC agents able to overcome drug resistance and/or off-target toxicity is of pivotal importance. The mammalian target of rapamycin (mTOR) plays a critical role in CRC, regulating protein translation and controlling cell growth, proliferation, metabolism and survival. The aim of this study was to explore the effect of a combination of three natural compounds, eicosapentaenoic acid-free fatty acid (EPA-FFA), epigallocatechin-3-gallate (EGCG) and proanthocyanidins (grape seed [GS] extract) at low cytotoxic concentrations on CRC cells and test their activity on mTOR and translational regulation. The CRC cell lines HCT116 and SW480 were treated for 24h with combinations of EPA-FFA (0-150 µM), EGCG (0-175 µM) and GS extract (0-15 µM) to evaluate the effect on cell viability. The low cytotoxic combination of EPA-FFA 150 µM, EGCG 175 µM and GS extract 15 µM completely inhibited the mTOR signaling in HCT116 and SW480 cells, reaching an effect stronger than or comparable to that of the mTOR inhibitor Rapamycin in HCT116 or SW480 cells, respectively. Moreover, the treatment led to changes of protein translation of ribosomal proteins, c-Myc and cyclin D1. In addition, we found a reduction of clonal capability in both cell lines, with block of cell cycle in G0G1 and induction of apoptosis. Our data suggest that the low cytotoxic combination of EPA-FFA, EGCG and GS extract, tested for the first time here, inhibits mTOR signaling and thus could be considered for CRC treatment.


Scientific Reports | 2016

Inflammation increases NOTCH1 activity via MMP9 and is counteracted by Eicosapentaenoic Acid-free fatty acid in colon cancer cells

Chiara Fazio; Giulia Piazzi; Paola Vitaglione; Vincenzo Fogliano; Alessandra Munarini; Anna Prossomariti; Maddalena Milazzo; Leonarda D’Angelo; Manuela Napolitano; Pasquale Chieco; Andrea Belluzzi; Franco Bazzoli; Luigi Ricciardiello

Aberrant NOTCH1 signalling is critically involved in multiple models of colorectal cancer (CRC) and a prominent role of NOTCH1 activity during inflammation has emerged. Epithelial to Mesenchymal Transition (EMT), a crucial event promoting malignant transformation, is regulated by inflammation and Metalloproteinase-9 (MMP9) plays an important role in this process. Eicosapentaenoic Acid (EPA), an omega-3 polyunsaturated fatty acid, was shown to prevent colonic tumors in different settings. We recently found that an extra-pure formulation of EPA as Free Fatty Acid (EPA-FFA) protects from colon cancer development in a mouse model of Colitis-Associated Cancer (CAC) through modulation of NOTCH1 signalling. In this study, we exposed colon cancer cells to an inflammatory stimulus represented by a cytokine-enriched Conditioned Medium (CM), obtained from THP1-differentiated macrophages. We found, for the first time, that CM strongly up-regulated NOTCH1 signalling and EMT markers, leading to increased invasiveness. Importantly, NOTCH1 signalling was dependent on MMP9 activity, upon CM exposure. We show that a non-cytotoxic pre-treatment with EPA-FFA antagonizes the effect of inflammation on NOTCH1 signalling, with reduction of MMP9 activity and invasiveness. In conclusion, our data suggest that, in CRC cells, inflammation induces NOTCH1 activity through MMP9 up-regulation and that this mechanism can be counteracted by EPA-FFA.


Cell Death and Disease | 2016

Inflammation and Notch signaling: a crosstalk with opposite effects on tumorigenesis

Chiara Fazio; Luigi Ricciardiello

The Notch cascade is a fundamental and highly conserved pathway able to control cell-fate. The Notch pathway arises from the interaction of one of the Notch receptors (Notch1–4) with different types of ligands; in particular, the Notch pathway can be activated canonically (through the ligands Jagged1, Jagged2, DLL1, DLL3 or DLL4) or non-canonically (through various molecules shared by other pathways). In the context of tumor biology, the deregulation of Notch signaling is found to be crucial, but it is still not clear if the activation of this pathway exerts a tumor-promoting or a tumor suppressing function in different cancer settings. Untill now, it is well known that the inflammatory compartment is critically involved in tumor progression; however, inflammation, which occurs as a physiological response to damage, can also drive protective processes toward carcinogenesis. Therefore, the role of inflammation in cancer is still controversial and needs to be further clarified. Interestingly, recent literature reports that some of the signaling molecules modulated by the cells of the immune system also belong to or interact with the canonical and non-canonical Notch pathways, delineating a possible link between Notch activation and inflammatory environment. In this review we analyze the hypothesis that specific inflammatory conditions can control the activation of the Notch pathway in terms of biological effect, partially explaining the dichotomy of both phenomena. For this purpose, we detail the molecular links reported in the literature connecting inflammation and Notch signaling in different types of tumor, with a particular focus on colorectal carcinogenesis, which represents a perfect example of context-dependent interaction between malignant transformation and immune response.


Scientific Reports | 2017

Short-term treatment with eicosapentaenoic acid improves inflammation and affects colonic differentiation markers and microbiota in patients with ulcerative colitis

Anna Prossomariti; Eleonora Scaioli; Giulia Piazzi; Chiara Fazio; Matteo Bellanova; Elena Biagi; Marco Candela; Patrizia Brigidi; Clarissa Consolandi; Tiziana Balbi; Pasquale Chieco; Alessandra Munarini; Milena Pariali; Manuela Minguzzi; Franco Bazzoli; Andrea Belluzzi; Luigi Ricciardiello

Patients with long-standing ulcerative colitis (UC) have an increased colorectal cancer (CRC) risk. In this pilot study we evaluated the effect of Eicosapentaenoic acid as free fatty acid (EPA-FFA) supplementation on mucosal disease activity, colonic differentiation markers and microbiota composition in UC patients. Twenty long-standing UC patients in stable clinical remission and with fecal calprotectin (FC) > 150 µg/g were enrolled (T0) and supplemented with EPA-FFA 2 g/daily for 90 days (T3). Endoscopic and histologic disease activities were measured by Mayo and Geboes scores, respectively. HES1, KLF4, STAT3, IL-10 and SOCS3 levels were determined using western blotting and qRT-PCR, while phospho-STAT3 levels were assessed by western blotting. Goblet cells were stained by Alcian blue. Microbiota analyses were performed on both fecal and colonic samples. Nineteen patients completed the study; seventeen (89.5%) were compliant. EPA-FFA treatment reduced FC levels at T3. Patients with FC > 150 µg/g at T3 (n = 2) were assumed as non-responders. EPA-FFA improved endoscopic and histological inflammation and induced IL-10, SOCS3, HES1 and KLF4 in compliant and responder patients. Importantly, long-term UC-driven microbiota composition was partially redressed by EPA-FFA. In conclusion, EPA-FFA supplementation reduced mucosal inflammation, promoted goblet cells differentiation and modulated intestinal microbiota composition in long-standing UC patients.


Phytochemistry Reviews | 2014

Components of the Mediterranean Diet with chemopreventive activity toward colorectal cancer

Chiara Fazio; Luigi Ricciardiello


Gastroenterology | 2013

935 A Combination of Eicosapentaenoic Acid-Free Fatty Acid, Epigallocatechin-3-Gallate and Proanthocyanidins Has a Strong Effect on mTOR Signaling in Colorectal Cancer Cells

Leonarda D'Angelo; Giulia Piazzi; Anna Prossomariti; Chiara Fazio; Annalisa Pacilli; Lorenzo Montanaro; Giulia Graziani; Vincenzo Fogliano; Alessandra Munarini; Andrea Belluzzi; Franco Bazzoli; Luigi Ricciardiello


Gastroenterology | 2017

Eicosapentaenoic Acid-Free Fatty Reduces Fecal Calprotectin Levels and Affects Colitis-Associated Colorectal Cancer Pathways in Patients with Long-Standing Ulcerative Colitis

Anna Prossomariti; Eleonora Scaioli; Giulia Piazzi; Chiara Fazio; Matteo Bellanova; Elena Biagi; Marco Candela; Patrizia Brigidi; Clarissa Consolandi; Tiziana Balbi; Pasquale Chieco; Alessandra Munarini; Milena Pariali; Manuela Minguzzi; Franco Bazzoli; Andrea Belluzzi; Luigi Ricciardiello


Gastroenterology | 2013

Mo1165 Eicosapentaenoic Acid-Free Fatty Acid Strongly Prevents and Suppresses Colonic Tumors in Colitis-Associated Colorectal Cancer

Giulia Piazzi; Giuseppe D'Argenio; Vincenzo Lembo; Giovanna Mazzone; Anna Prossomariti; Paola Vitaglione; Vincenzo Fogliano; Alessandra Munarini; Claudio Ceccarelli; Chiara Fazio; Leonarda D'Angelo; Andrea Belluzzi; Pasquale Chieco; Mark A. Hull; Marco Romano; Franco Bazzoli; Luigi Ricciardiello

Collaboration


Dive into the Chiara Fazio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vincenzo Fogliano

Wageningen University and Research Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paola Vitaglione

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge