Network


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

Hotspot


Dive into the research topics where Silvia Zappavigna is active.

Publication


Featured researches published by Silvia Zappavigna.


Nanomedicine: Nanotechnology, Biology and Medicine | 2011

Nanotechnologies to use bisphosphonates as potent anticancer agents: the effects of zoledronic acid encapsulated into liposomes.

Monica Marra; Giuseppina Salzano; Carlo Leonetti; Pierfrancesco Tassone; Marco Scarsella; Silvia Zappavigna; Teresa Calimeri; Renato Franco; Giuseppina Liguori; Giovanni Cigliana; Roberta Ascani; Maria Immacolata La Rotonda; Alberto Abbruzzese; Pierosandro Tagliaferri; Michele Caraglia; Giuseppe De Rosa

UNLABELLED Zoledronic acid (ZOL) is a potent amino-bisphosphonate used for the treatment of bone metastases with recently reported antitumor activity. However, the short plasma half-life and rapid accumulation in bone limits the use of ZOL as an antitumor agent in extraskeletal tissues. Therefore, we developed stealth liposomes encapsulating ZOL (LipoZOL) to increase extraskeletal drug availability. Compared to free ZOL, LipoZOL induced a stronger inhibition of growth of a range of different cancer cell lines in vitro. LipoZOL also caused significantly larger inhibition of tumor growth and increased the overall survival in murine models of human prostate cancer and multiple myeloma, in comparison with ZOL. Moreover, a strong inhibition of vasculogenetic events without evidence of necrosis in the tumor xenografts from prostate cancer was recorded after treatment with LipoZOL. We demonstrated both antitumor activity and tolerability of LipoZOL in preclinical animal models of both solid and hematopoietic malignancies, providing a rationale for early exploration of use of LipoZOL as a potential anticancer agent in cancer patients. FROM THE CLINICAL EDITOR The short plasma half-life and rapid accumulation in bone limits the use of zoledronic acid as an antitumor agent in extraskeletal tissues. Therefore, stealth liposomes encapsulating ZOL (LipoZOL) have been developed to increase extraskeletal drug availability.


Biotechnology Advances | 2012

The PPAR-γ agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-β treated pancreatic cancer cells.

Giovanni Vitale; Silvia Zappavigna; Monica Marra; Alessandra Dicitore; Stefania Meschini; Maria Condello; Giuseppe Arancia; Sara Castiglioni; Paola Maroni; Paola Bendinelli; Roberta Piccoletti; Peter M. van Koetsveld; Francesco Cavagnini; Alfredo Budillon; Alberto Abbruzzese; Leo J. Hofland; Michele Caraglia

We have previously shown that cancer cells can protect themselves from apoptosis induced by type I interferons (IFNs) through a ras→MAPK-mediated pathway. In addition, since IFN-mediated signalling components STATs are controlled by PPAR gamma we studied the pharmacological interaction between recombinant IFN-β and the PPAR-γ agonist troglitazone (TGZ). This combination induced a synergistic effect on the growth inhibition of BxPC-3, a pancreatic cancer cell line, through the counteraction of the IFN-β-induced activation of STAT-3, MAPK and AKT and the increase in the binding of both STAT-1 related complexes and PPAR-γ with specific DNA responsive elements. The synergism on cell growth inhibition correlated with a cell cycle arrest in G0/G1 phase, secondary to a long-lasting increase of both p21 and p27 expressions. Blockade of MAPK activation and the effect on p21 and p27 expressions, induced by IFN-β and TGZ combination, were due to the decreased activation of STAT-3 secondary to TGZ. IFN-β alone also increased p21 and p27 expression through STAT-1 phosphorylation and this effect was attenuated by the concomitant activation of IFNbeta-induced STAT-3-activation. The combination induced also an increase in autophagy and a decrease in anti-autophagic bcl-2/beclin-1 complex formation. This effect was mediated by the inactivation of the AKT→mTOR-dependent pathway. To the best of our knowledge this is the first evidence that PPAR-γ activation can counteract STAT-3-dependent escape pathways to IFN-β-induced growth inhibition through cell cycle perturbation and increased autophagic death in pancreatic cancer cells.


Biotechnology Advances | 2012

New self-assembly nanoparticles and stealth liposomes for the delivery of zoledronic acid: a comparative study

Monica Marra; Giuseppina Salzano; Carlo Leonetti; Manuela Porru; Renato Franco; Silvia Zappavigna; Giuseppina Liguori; Gerardo Botti; Paolo Chieffi; Monica Lamberti; Giovanni Vitale; Alberto Abbruzzese; Maria Immacolata La Rotonda; Giuseppe De Rosa; Michele Caraglia

Zoledronic acid (ZOL) is a drug whose potent anti-cancer activity is limited by its short plasma half-life and rapid uptake and accumulation within bone. We have recently proposed new delivery systems to avoid ZOL accumulation into the bone, thus improving extra-skeletal bioavailability. In this work, we have compared the technological and anti-cancer features of either ZOL-containing self-assembly PEGylated nanoparticles (NPs) or ZOL-encapsulating PEGylated liposomes (LIPO-ZOL). ZOL-containing NPs showed superior technological characteristics in terms of mean diameter, size distribution, and ZOL encapsulation efficiency, compared to LIPO-ZOL. Moreover, the anti-cancer activity of NPs in nude mice xenografted with prostate cancer PC3 cells was higher than that one induced by LIPO-ZOL. In addition, NPs induced the complete remission of tumour xenografts and an increase of survival time higher than that one observed with LIPO-ZOL. It has also to be considered that PC3 tumour xenografts were almost completely resistant to the anti-cancer effects induced by free ZOL. Both nanotechnological products did not induce toxic effects not affecting the mice weight nor inducing deaths. Moreover, the histological examination of some vital organs such as liver, kidney and spleen did not find any changes in terms of necrotic effects or modifications in the inflammatory infiltrate. On the other hand, NPs but not LIPO-ZOL caused a statistically significant reduction of the tumour associated macrophages (TAM) in tumour xenografts. This effect was paralleled by a significant increase of both necrotic and apoptotic indexes. The effects of the NPs were also higher in terms of neo-angiogenesis inhibition. These results suggest the future preclinical development of ZOL-encapsulating NPs in the treatment of human cancer.


International Journal of Pharmaceutics | 2011

Self-assembly nanoparticles for the delivery of bisphosphonates into tumors.

Giuseppina Salzano; Monica Marra; Manuela Porru; Silvia Zappavigna; Alberto Abbruzzese; M.I. La Rotonda; Carlo Leonetti; Michele Caraglia; G. De Rosa

Bisphosphonates (BPs) are molecules able to induce apoptosis in several cancer cell lines. However, their short half-life and the rapid uptake and accumulation within bone, limit its use as antitumor agent for extra-skeletal malignancies. Here we proposed a new delivery system to avoid BP accumulation into the bone, thus improving extra-skeletal bioavailability. In this work, we used the zoledronic acid (ZOL), a third generation bisphosphonate, able to induce apoptosis at micromolar concentration. We developed ZOL-containing self-assembly PEGylated nanoparticles (NPs) based on ZOL complexes with calcium phosphate NPs (CaPZ NPs) and cationic liposomes. PEGylation was achieved by two different strategies. CaPZ NPs were covered with PEGylated liposomes (pre-PLCaPZ NPs); alternatively, CaPZ NPs were previously mixed with cationic liposomes and then PEGylated by post-insertion method (post-PLCaPZ NPs). The NPs were fully characterized in terms of mean diameter and size distribution, morphology, ZOL loading, antiproliferative effect on different cell lines. Pre-PLCaPZ NPs showed the best technological characteristics, with a narrow size distribution and a high ZOL loading. Moreover, on different cancer cell lines, these NPs enhanced the antiproliferative effect of ZOL. Finally, in an animal model of prostate cancer, a significant reduction of tumor growth was achieved with pre-PLCaPZ NPs, while the tumor was unaffected by ZOL in solution.


Journal of Experimental & Clinical Cancer Research | 2012

5-Fluorouracil induces apoptosis in rat cardiocytes through intracellular oxidative stress

Monica Lamberti; Stefania Porto; Monica Marra; Silvia Zappavigna; Anna Grimaldi; Daniela Feola; Delia Pesce; Silvio Naviglio; Annamaria Spina; Nicola Sannolo; Michele Caraglia

BackgroundCardiotoxicity is a major complication of anticancer drugs, including anthracyclines and 5-fluorouracil(5-FU) and it can have detrimental effects both in patients and workers involved in the preparation of chemotherapy.MethodsSpecifically, we have assessed the effects of increasing concentrations of 5-FU and doxorubicin (DOXO) on proliferation of H9c2 rat cardiocytes and HT-29 human colon adenocarcinoma cells by MTT assay. Cells were treated for 24, 48 and 72 h with different concentrations of the two drugs alone or with 5-FU in combination with 10-4 M of levofolene (LF).Results5-FU induced a time- and dose-dependent growth inhibition in both cell lines. The 50% growth inhibition (IC:50) was reached at 72 h with concentrations of 4 μM and 400 μM on HT-29 and H9c2, respectively. The addition of LF to 5-FU enhanced this effect. On the other hand, the IC:50 of DOXO was reached at 72 h with concentrations of 0.118 μM on H9c2 and of 0.31 μM for HT-29. We have evaluated the cell death mechanism induced by 50% growth inhibitory concentrations of 5-FU or DOXO in cardiocytes and colon cancer cells. We have found that the treatment with 400 μM 5-FU induced apoptosis in 32% of H9c2 cells. This effect was increased by the addition of LF to 5-FU (38% of apoptotic cells). Apoptosis occurred in only about 10% of HT-29 cells treated with either 5-FU or 5-FU and LF in combination. DOXO induced poor effects on apoptosis of both H9c2 and HT-29 cells (5–7% apoptotic cells, respectively). The apoptosis induced by 5-FU and LF in cardiocytes was paralleled by the activation of caspases 3, 9 and 7 and by the intracellular increase of O2− levels.ConclusionsThese results suggest that cardiotoxic mechanism of chemotherapy agents are different and this disclose a new scenario for prevention of this complication.


Molecular therapy. Nucleic acids | 2015

MicroRNA-423-5p Promotes Autophagy in Cancer Cells and Is Increased in Serum From Hepatocarcinoma Patients Treated With Sorafenib

Paola Stiuso; Nicoletta Potenza; Angela Lombardi; Ida Ferrandino; Antonio Monaco; Silvia Zappavigna; Daniela Vanacore; Nicola Mosca; Filomena Castiello; Stefania Porto; R. Addeo; Salvatore Del Prete; Ferdinando De Vita; Aniello Russo; Michele Caraglia

Hepatocellular carcinoma (HCC) is the third cause of cancer-related deaths worldwide. Sorafenib is the only approved drug for patients with advanced HCC but has shown limited activity. microRNAs (miRs) have been involved in several neoplasms including HCC suggesting their use or targeting as good tools for HCC treatment. The purpose of this study was to identify novel approaches to sensitize HCC cells to sorafenib through miRs. miR-423-5p was validated as positive regulator of autophagy in HCC cell lines by transient transfection of miR and anti-miR molecules. miR-423-5p expression level was evaluated by real-time polymerase chain reaction (PCR) in sera collected from 39 HCC patients before and after treatment with sorafenib. HCC cells were cotreated with sorafenib and miR-423-5p and the effects on cell cycle, apoptosis, and autophagy were evaluated. Secretory miR-423-5p was upregulated both in vitro and in vivo by sorafenib treatment and its increase was correlated with response to therapy since 75% of patients in which an increase of secretory miR423-5p was found were in partial remission or stable disease after 6 moths from the beginning of therapy. HCC cells transfected with miR-423-5p showed an increase of cell percentage in S-phase of cell cycle paralleled by a similar increase of autophagic cells evaluated at both fluorescence activated cell sorter (FACS) and transmission electron microscopy. Our results suggest the miR423-5p can be used as a useful tool to predict response to sorafenib in HCC patients and is involved in autophagy regulation in HCC cells.


Journal of Controlled Release | 2010

In vitro anticancer activity of docetaxel-loaded micelles based on poly(ethylene oxide)-poly(epsilon-caprolactone) block copolymers: Do nanocarrier properties have a role?

Luisanna Ostacolo; Monica Marra; Francesca Ungaro; Silvia Zappavigna; Giovanni Maglio; Fabiana Quaglia; Alberto Abbruzzese; Michele Caraglia

In this paper we have investigated the behavior of core-shell poly(ethylene oxide)-poly(epsilon-caprolactone) (PEO-PCL) micelles derived from copolymers with linear triblock (TR) and 4-arm star-diblock (ST) architectures for the delivery of docetaxel (DTX). DTX was loaded inside micelles (DTX-TR(m) and DTX-ST(m)) with high efficiency and released slowly for more than two weeks. DTX-loaded micelles based on both copolymers had very similar properties in terms of mean size, zeta potential, loading ability and release rate in buffered saline. However, the stability of DTX-ST(m) was very poor in aqueous media with proteins resulting in a strong and progressive aggregation. We studied the effect of increasing concentrations of free DTX or DTX-loaded micelles on growth inhibition of human breast MCF-7 and MDA-MB468 and prostate PC3 and DU145 adenocarcinoma cell lines. DTX-loaded TR micelles induced cell growth inhibition similarly to free DTX whereas DTX-ST(m) showed lower cytotoxicity. On the other hand, by normalizing IC(50) values for the actual amount of DTX released from micelles in the medium, DTX-loaded ST micelles became more active than free DTX in all cell lines tested. Both free DTX and DTX-loaded TR micelles displayed a significantly lower cytotoxic activity in G(2)/M phase synchronized cells, whereas cytotoxicity of DTX-loaded ST micelles did not change. Cytotoxicity was related to micelle stability, uptake and release rate in cell culture media. Our results suggest that for a correct interpretation of cytotoxicity of nanocarriers, the evaluation of their behavior in biologically relevant conditions is of utmost importance to select proper systems for further in vivo testing.


Current Cancer Drug Targets | 2009

Emerging Strategies to Strengthen the Anti-Tumour Activity of Type I Interferons: Overcoming Survival Pathways

Michele Caraglia; Monica Marra; Pierosandro Tagliaferri; Steven W. J. Lamberts; Silvia Zappavigna; G. Misso; F. Cavagnini; Gaetano Facchini; Alberto Abbruzzese; Leo J. Hofland; Giovanni Vitale

Interferon-a (IFN-a) is currently the most used cytokine in the treatment of cancer. However, the potential anti-tumour activity of IFN-a is limited by the activation of tumour resistance mechanisms. In this regard, we have shown that IFN-a, at growth inhibitory concentrations, enhances the EGF-dependent Ras-->Erk signalling and decreases the adenylate cyclase/cAMP pathway activity in cancer cells; both effects represent escape mechanisms to the growth inhibition and apoptosis induced by IFN-a. The selective targeting of these survival pathways might enhance the antitumor activity of IFN-ain cancer cells, as shown by: i) the combination of selective EGF receptor tyrosine kinase inhibitor (gefitinib) and IFN-a having cooperative anti-tumour effects; ii) the farnesyl-transferase inhibitor R115777 strongly potentiating the anti-tumour activity of IFN-a both in vitro and in vivo through the inhibition of different escape mechanisms that are dependent on isoprenylation of intracellular proteins such as ras; iii) the cAMP reconstituting agent (8-Br-cAMP) enhancing the pro-apoptotic activity of IFN-alpha. IFN-beta is a multifunctional cytokine binding the same receptor of IFN-alpha, but with higher affinity (10-fold) and differential structural interactions. We recently showed that IFN-beta is considerably more potent than IFN-alpha in its anti-tumour effect through the induction of apoptosis and/or cell cycle arrest in S-phase. The emergence of long-acting pegylated forms of IFN-beta makes this agent a promising anti-cancer drug. These observations open a new scenario of anticancer intervention able to strengthen the antitumor activity of IFN-alpha or to use more potent type I IFNs.


Expert Opinion on Biological Therapy | 2013

Optimizing treatment of metastatic colorectal cancer patients with anti-EGFR antibodies: overcoming the mechanisms of cancer cell resistance

Teresa Troiani; Silvia Zappavigna; Erika Martinelli; Santolo Rosario Addeo; Paola Stiuso; Fortunato Ciardiello; Michele Caraglia

Introduction: A number of anti-EGFR monoclonal antibodies (mAbs) have been recently developed for the treatment of refractory metastatic colorectal cancer (mCRC). These mAbs, blocking ligand/receptor interactions, exert their biological activity via multiple mechanisms, including inhibition of cell cycle progression, potentiation of cell apoptosis, inhibition of angiogenesis, tumor cell invasion and metastasis and, potentially, induction of immunological effector mechanisms. Areas covered: Cetuximab is an anti-EGFR mAb currently used in mCRC treatment. Despite the evidence of efficacy of cetuximab in the treatment of mCRC patients, the observation of low response rates was the proof of concept of resistance to anti-EGFR mAbs treatment. An increasing number of molecular alterations have been more recently hypothesized to be involved in resistance to anti-EGFR mAbs in CRC: mutations in BRAF, NRAS and PIK3CA, loss of expression of PTEN and, now, activation of HER2 signaling through HER2 gene amplification and/or increased heregulin stimulation. Expert opinion: This review focuses on the development of new strategies such as combination with other agents blocking alternative escape pathways, cancer cell prioritization hyperactivating EGFR pathway, combination with immune system, development of nanotech devices to increase efficacy of antibody-based therapy and overcome the mechanisms of cancer cell resistance.


Biochimica et Biophysica Acta | 2013

Interleukin 18: Friend or foe in cancer

Giuseppe De Palma; Antonio Barbieri; Sabrina Bimonte; Marco Palla; Silvia Zappavigna; Michele Caraglia; Paolo Antonio Ascierto; Gennaro Ciliberto; Claudio Arra

In the last few years, the field of tumor immunology has significantly expanded and its boundaries, never particularly clear, have become less distinct. Although the immune system plays an important role in controlling tumor growth, it has also become clear that tumor growth can be promoted by inflammatory immune responses. A good example that exemplifies the ambiguous role of the immune system in cancer progression is represented by interleukin 18 (IL-18) that was first identified as an interferon-γ-inducing factor (IGIF) involved in T helper type-1 (Th1) immune response. The expression and secretion of IL-18 have been observed in various cell types from immune cells to circulating cancer cells. In this review we highlighted the multiple roles played by IL-18 in immune regulation, cancer progression and angiogenesis and the clinical potential that may result from such understanding.

Collaboration


Dive into the Silvia Zappavigna's collaboration.

Top Co-Authors

Avatar

Michele Caraglia

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Monica Marra

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Alberto Abbruzzese

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Paola Stiuso

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Gabriella Misso

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Carlo Leonetti

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefania Porto

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Manuela Porru

École normale supérieure de Lyon

View shared research outputs
Top Co-Authors

Avatar

Amalia Luce

Seconda Università degli Studi di Napoli

View shared research outputs
Researchain Logo
Decentralizing Knowledge