Network


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

Hotspot


Dive into the research topics where Enzo Bonmassar is active.

Publication


Featured researches published by Enzo Bonmassar.


Biochimica et Biophysica Acta | 2009

miR-155 gene: a typical multifunctional microRNA.

Isabella Faraoni; Francesca Romana Antonetti; John Cardone; Enzo Bonmassar

In the last years small RNA molecules, i.e. microRNA (miRNA) encoded by miR genes, have been found to play a crucial role in regulating gene expression of a considerable part of plants and animals genome. Here, we report the essential information on biogenesis of miRNAs and recent evidence on their important role in human diseases. Emphasis has been given to miR-155, since this molecule represents a typical multifunctional miRNA. Recent data indicate that miR-155 has distinct expression profiles and plays a crucial role in various physiological and pathological processes such as haematopoietic lineage differentiation, immunity, inflammation, cancer, and cardiovascular diseases. Moreover, miR-155 has been found to be implicated in viral infections, particularly in those caused by DNA viruses. The available experimental evidence indicating that miR-155 is over expressed in a variety of malignant tumors allows us to include this miRNA in the list of genes of paramount importance in cancer diagnosis and prognosis. Exogenous molecular control in vivo of miR-155 expression could open up new ways to restrain malignant growth and viral infections, or to attenuate the progression of cardiovascular diseases.


Journal of Clinical Oncology | 2005

Chemo-Immunotherapy of Metastatic Colorectal Carcinoma With Gemcitabine Plus FOLFOX 4 Followed by Subcutaneous Granulocyte Macrophage Colony-Stimulating Factor and Interleukin-2 Induces Strong Immunologic and Antitumor Activity in Metastatic Colon Cancer Patients

Pierpaolo Correale; Maria Grazia Cusi; Kwong Y. Tsang; Maria Teresa Del Vecchio; Stefania Marsili; Marco La Placa; Chiara Intrivici; Angelo Aquino; Lucia Micheli; Cristina Nencini; Francesco Ferrari; Giorgio Giorgi; Enzo Bonmassar; Guido Francini

PURPOSE Tumor cell killing by anticancer drugs may be supported by their immuno- and pharmacologic effects. Chemotherapy is in fact able to (A) upregulate tumor-associated antigen expression, including carcinoembryonic antigen (CEA) or other target molecules such as thymidylate synthase (TS); and (B) downregulate tumor cell resistance to the death signals induced by tumor antigen-specific cytotoxic T lymphocytes. This provides the rationale for combining chemo- and immunotherapy. MATERIALS AND METHODS We describe the results of a translational phase II trial designed to evaluate the toxicity, antitumor activity and immunologic effects of gemcitabine + FOLFOX-4 (oxaliplatin, fluorouracil, and folinic acid) polychemotherapy followed by the subcutaneous administration of granulocyte macrophage colony-stimulating factor and low-dose interleukin-2 in colorectal carcinoma patients. The study involved 29 patients (16 males and 13 females with a mean age of 69 years), 21 of whom had received a previous line of treatment, and 19 had liver involvement. RESULTS The treatment was well tolerated and induced very high objective response (68.9%) and disease control rates (96.5%), with an average time to progression of 12.5 months. An immunologic study of peripheral blood mononuclear cells (PBMCs) taken from 20 patients showed an enhanced proliferative response to colon carcinoma antigen and a significant reduction in suppressive regulatory T lymphocytes (CD4+CD25T-reg+). A cytofluorimetric study of the PBMCs of five HLA-A(*)02.01+ patients who achieved an objective response showed an increased frequency of cytolytic T lymphocyte precursors specific for known CEA- and TS-derived epitopes. CONCLUSION The results show that our regimen has strong immunologic and antitumor activity in colorectal cancer patients and deserves to be investigated in phase III trials.


Cancer Research | 2006

Immunization of Stage IV Melanoma Patients with Melan-A/MART-1 and gp100 Peptides plus IFN-α Results in the Activation of Specific CD8+ T Cells and Monocyte/Dendritic Cell Precursors

Tiziana Di Pucchio; Lorenzo Pilla; Imerio Capone; Maria Ferrantini; Enrica Montefiore; Francesca Urbani; Roberto Patuzzo; Elisabetta Pennacchioli; Mario Santinami; Agata Cova; Gloria Sovena; Claudia Lombardo; Arianna Lombardi; Patrizia Caporaso; Stefania D'Atri; Paolo Marchetti; Enzo Bonmassar; Giorgio Parmiani; Filippo Belardelli; Licia Rivoltini

The use of IFN-alpha in clinical oncology has generally been based on the rationale of exploiting its antiproliferative and antiangiogenic activities. However, IFN-alpha also exhibits enhancing effects on T-cell and dendritic cell functions, which may suggest a novel use as a vaccine adjuvant. We have carried out a pilot phase I-II trial to determine the effects of IFN-alpha, administered as an adjuvant of Melan-A/MART-1:26-35(27L) and gp100:209-217(210M) peptides, on immune responses in stage IV melanoma patients. In five of the seven evaluable patients, a consistent enhancement of CD8(+) T cells recognizing modified and native MART-1 and gp100 peptides and MART-1(+)gp100(+) melanoma cells was observed. Moreover, vaccination induced an increase in CD8(+) T-cell binding to HLA tetramers containing the relevant peptides and an increased frequency of CD45RA(+)CCR7(-) (terminally differentiated effectors) and CD45RA(-)CCR7(-) (effector memory) cells. In all patients, treatment augmented significantly the percentage of CD14(+) monocytes and particularly of the CD14(+)CD16(+) cell fraction. An increased expression of CD40 and CD86 costimulatory molecules in monocytes was also observed. Notably, postvaccination monocytes from two of the three patients showing stable disease or long disease-free survival showed an enhanced antigen-presenting cell function and capability to secrete IP10/CXCL10 when tested in mixed leukocyte reaction assays, associated to a boost of antigen and melanoma-specific CD8(+) T cells. Although further clinical studies are needed to show the adjuvant activity of IFN-alpha, the present data represent an important starting point for considering a new clinical use of IFN-alpha and new immunologic end points, potentially predictive of clinical response.


International Journal of Cancer | 2003

Treatment of colon and breast carcinoma cells with 5-fluorouracil enhances expression of carcinoembryonic antigen and susceptibility to HLA-A(*)02.01 restricted, CEA-peptide-specific cytotoxic T cells in vitro

Pierpaolo Correale; Angelo Aquino; Antonio Giuliani; Monia Pellegrini; Lucia Micheli; Maria Grazia Cusi; Cristina Nencini; Roberto Petrioli; Salvatore Prete; Liana De Vecchis; Mario Turriziani; Giorgio Giorgi; Enzo Bonmassar; Guido Francini

Cancer vaccines directed against tumor associate antigen (TAA) have produced encouraging results in preclinical models but not in cancer patients. A major limitation of this strategy is the relative degree of tolerance to these antigens and the low and heterogeneous tumor cell expression of TAA and major histocompatibility complex (MHC). Previous studies have shown that 5‐fluorouracil (5‐FU) can upregulate the expression of membrane‐associated carcino‐embryonic antigen (CEA), and MHC molecules in colon and breast carcinoma cell lines. We have investigated whether this drug can also enhance their sensitivity to the lytic effects of CEA‐peptide specific Cytotoxic T cell lymphocytes (CTL). The CEA peptide‐specific CTLs generated in our laboratory from normal HLA‐A(*)02.01+ donor PBMCs, were able to kill HLA‐A(*)02.01+/CEA+ breast (MCF‐7‐T103) and colon (HLA‐A(*)02.01 gene‐transfected HT‐29 and C22.20) carcinoma cells in HLA‐A(*)02.01 restricted manner. The treatment of target cells with 5‐FU, enhanced their CEA expression and susceptibility to CTL‐mediated lysis. Cold competition assays confirmed these results, thus supporting the hypothesis that immune target cell lysis and 5‐FU mediated enhancement were dependent on CEA peptide presentation by cancer cells. 5‐FU treatment of functionally “mature” CTL after in vitro expansion, did not reduce their cytolytic activity against MT‐2 target cells but, when the anti‐metabolite was added during the immune‐sensitization phase, CTL generation was significantly inhibited. These results provide a rationale for investigating a possible new role of 5‐FU as an immuno targeting amplifier agent in breast and colorectal cancer patients immunized with CEA‐directed cancer vaccines.


Journal of Immunology | 2005

Dendritic Cell-Mediated Cross-Presentation of Antigens Derived from Colon Carcinoma Cells Exposed to a Highly Cytotoxic Multidrug Regimen with Gemcitabine, Oxaliplatin, 5-Fluorouracil, and Leucovorin, Elicits a Powerful Human Antigen-Specific CTL Response with Antitumor Activity in Vitro

Pierpaolo Correale; Maria Grazia Cusi; Maria Teresa Del Vecchio; Angelo Aquino; Salvatore Prete; Kwong Y. Tsang; Lucia Micheli; Cristina Nencini; Marco La Placa; Francesco Montagnani; Chiara Terrosi; Michele Caraglia; Vincenzo Formica; Giorgio Giorgi; Enzo Bonmassar; Guido Francini

Gemcitabine, oxaliplatin, leucovorin, and 5-fluorouracil (GOLF) is a novel multidrug regimen inducing high levels of necrosis and apoptosis in colon carcinoma cells. This regimen is also able to promote a process of Ag remodeling including up-regulation of immunotherapy targets like carcinoembryonic Ag (CEA), thymidylate synthase (TS). We have conducted a preclinical study aimed to investigate whether these drug-induced modifications would also enhance colon cancer cell immunogenicity. Several CTL lines were thus generated by in vitro stimulating human HLA-A(*)02.01+ PBMCs, from normal donors and colon cancer patients, with autologous dendritic cells cross-primed with cell lysates of colon cancer cells untreated, irradiated, or previously exposed to different drug treatments including the GOLF regimen. Class I HLA-restricted cytolytic activity of these CTL lines was tested against colon cancer cells and CEA and TS gene transfected target cells. These experiments revealed that CTLs sensitized with GOLF-treated cancer cells were much more effective than those sensitized with the untreated colon carcinoma cells or those exposed to the other treatments. CTL lines sensitized against the GOLF-treated colon cancer cells, also expressed a greater percentage of T-lymphocyte precursors able to recognize TS- and CEA-derived peptides. These results suggest that GOLF regimen is a powerful antitumor and immunomodulating regimen that can make the tumor cells a suitable means to induce an Ag-specific CTL response. These results suggest that a rationale combination of GOLF chemotherapy with cytokine-based immunotherapy could generate a chemotherapy-modulated Ag-specific T-lymphocyte response in cancer patients able to destroy the residual disease survived to the cytotoxic drugs.


Molecular Pharmacology | 2008

AKT Is Activated in an Ataxia-Telangiectasia and Rad3-Related-Dependent Manner in Response to Temozolomide and Confers Protection against Drug-Induced Cell Growth Inhibition

Simona Caporali; Lauretta Levati; Giuseppe Starace; Gianluca Ragone; Enzo Bonmassar; Ester Alvino; Stefania D'Atri

The phosphatidylinositol 3-kinase/AKT pathway is activated frequently in human cancer, and it has been implicated in tumor cell proliferation, survival, and chemoresistance. In this study, we addressed the role of AKT in cellular responses to the therapeutic methylating agent temozolomide (TMZ), and we investigated the possible link between TMZ-induced modulation of AKT function and activation of ataxia-telangiectasia and Rad3-related (ATR)- and ataxia telangiectasia mutated (ATM)-dependent signaling pathways. We found that clinically relevant concentrations of TMZ caused activation of endogenous AKT in lymphoblastoid cells, and in colon and breast cancer cells, and that this molecular event required a functional mismatch repair system. Transfection of a dominant-negative kinase-dead form of AKT1 into breast cancer cells abrogated TMZ-induced activation of endogenous AKT, and it markedly enhanced cell sensitivity to the drug. Likewise, exposure of the MMR-proficient cell lines to the AKT inhibitor d-3-deoxy-2-O-methyl-myo inositol 1-[(R)-2-methoxy-3-(octadecyloxy)-propyl hydrogen phosphate] (SH-5) impaired AKT phosphorylation in response to TMZ, and it significantly increased cell chemosensitivity. Furthermore, small interfering RNA (siRNA)-mediated reduction of AKT1 expression in colon cancer cells potentiated the growth inhibitory effects of TMZ. Inhibition of ATM expression in colon cancer cells by siRNA did not impair TMZ-induced activation of AKT, whereas siRNA-mediated inhibition of ATR prevented AKT activation in response to the drug and increased cell chemosensitivity. These results strongly support the hypothesis that clinical benefit could be obtained by combining TMZ with inhibitors of the AKT pathway. Moreover, they provide the first evidence of a novel function of ATR as an upstream activator of AKT in response to DNA damage induced by O6-guanine-methylating agents.


Leukemia | 1999

Treatment with temozolomide and poly(ADP-ribose) polymerase inhibitors induces early apoptosis and increases base excision repair gene transcripts in leukemic cells resistant to triazene compounds.

Lucio Tentori; Mario Turriziani; D Franco; A Serafino; L Levati; R Roy; Enzo Bonmassar; Grazia Graziani

Methylating triazenes have shown marked antileukemic effects, possibly through generation of a variety of DNA adducts. Cells tolerant to O6-methylguanine due to a defect in the mismatch repair system (MRS), might become sensitive to other methyl adducts, by inhibiting the N-methylpurine repair, which requires base excision repair (BER) and poly(ADP-ribose) polymerase (PADPRP). Therefore, MRS-deficient Jurkat leukemic cells resistant to methylating triazenes, have been treated with temozolomide (TZM) and PADPRP inhibitors. Expression of PADPRP or molecules involved in the BER system [3-methylpurine-DNA glycosylase (MPG) and X-ray repair cross-complementing 1 (XRCC1)], have been explored. Cytotoxic effects of TZM associated with PADPRP inhibitors are evident shortly after treatment, suggesting that completion of cell division is not required for the lethal effect of the drug combination. Increase of PADPRP or MPG transcripts was found after treatment with TZM alone or combined with PADPRP inhibitor. XRCC1 transcript was positively modulated only in the case of drug combination. This could suggest that in the presence of PADPRP inhibitor, persistence of DNA damage triggers XRCC1 transcription. Our results suggest that association of TZM and PADPRP inhibitors might be of benefit for MRS-deficient malignancies unresponsive to the methylating agent.


Pigment Cell & Melanoma Research | 2011

MicroRNA‐155 targets the SKI gene in human melanoma cell lines

Lauretta Levati; Elena Pagani; Sveva Romani; D. Castiglia; Eugenia Piccinni; Claudia Covaciu; Patrizia Caporaso; Sergio Bondanza; Francesca Romana Antonetti; Enzo Bonmassar; Fabio Martelli; Ester Alvino; Stefania D'Atri

The SKI protein is a transcriptional coregulator over‐expressed in melanoma. Experimentally induced down‐regulation of SKI inhibits melanoma cell growth in vitro and in vivo. MicroRNAs (miRNAs) negatively modulate gene expression and have been implicated in oncogenesis. We previously showed that microRNA‐155 (miR‐155) is down‐regulated in melanoma cells as compared with normal melanocytes and that its ectopic expression impairs proliferation and induces apoptosis. Here, we investigated whether miR‐155 could mediate melanoma growth inhibition via SKI gene silencing. Luciferase reporter assays demonstrated that miR‐155 interacted with SKI 3′UTR and impaired gene expression. Transfection of melanoma cells with miR‐155 reduced SKI levels, while inhibition of endogenous miR‐155 up‐regulated SKI expression. Specifically designed small interfering RNAs reduced SKI expression and inhibited proliferation. However, melanoma cells over‐expressing a 3′UTR‐deleted SKI were still susceptible to the antiproliferative effect of miR‐155. Our data demonstrate for the first time that SKI is a target of miR‐155 in melanoma. However, impairment of SKI expression is not the leading mechanism involved in the growth‐suppressive effect of miR‐155 found in this malignancy.


Genes, Chromosomes and Cancer | 1998

Mutation of the mismatch repair gene hMSH2 and hMSH6 in a human T-cell leukemia line tolerant to methylating agents.

Lauretta Levati; Giancarlo Marra; Teresa Lettieri; Stefania D'Atri; Patrizia Vernole; Lucio Tentori; Pedro Miguel Lacal; Elena Pagani; Enzo Bonmassar; Josef Jiricny; Grazia Graziani

Cell killing by monofunctional methylating agents is due mainly to the formation of adducts at the O6 position of guanine. These methyl adducts are removed from DNA by the O6‐alkylguanine DNA alkyltransferase (OGAT). The mechanism by which O6‐methylguanine (O6meG) induces cell death in OGAT‐deficient cells requires a functional mismatch repair system (MRS). We have previously reported that depletion of OGAT activity in the human T‐cell leukemic Jurkat line does not sensitize these cells to the cytotoxic and apoptotic effects of the methylating triazene temozolomide (Tentori et al., 1995). We therefore decided to establish whether the tolerance of Jurkat cells to O6meG could be associated with a defect in MRS. The results of mismatch repair complementation studies indicated that Jurkat cells are defective in hMutSα, a heterodimer of the hMSH2 and hMSH6 proteins. Cytogenetic analysis of two Jurkat clones revealed a deletion in the short arm of chromosome region 2p15–21, indicating an allelic loss of both hMSH2 and hMSH6 genes. DNA sequencing revealed that exon 13 of the second hMSH2 allele contains a base substitution at codon 711, which changes an arginine to a termination codon (CGA→TGA). In addition, a (C)8→(C)7 frameshift mutation in codon 1085–1087 of the hMSH6 gene was also found. Although both hMSH2 and hMSH6 transcripts could be detected in Jurkat clones, the respective polypeptides were absent. Taken together, these data indicate that tolerance of Jurkat cells to methylation damage is linked to a loss of functional hMutSα. Genes Chromosomes Cancer 23:159–166, 1998.


Cancer Immunology, Immunotherapy | 1981

Immunoadjuvant effects of Candida albicans and its cell wall fractions in a mouse lymphoma model

Antonio Cassone; Pierfrancesco Marconi; Francesco Bistoni; E. Mattia; G. Sbaraglia; Enrico Garaci; Enzo Bonmassar

SummaryChemical, ultrastructural, and immunoadjuvant properties of Candida albicans (CA) and of a number of its fractions have been characterized through the analysis of the antitumor activity of soluble and insoluble cell wall components.CD2F1 mice were inoculated IP with Moloney virus-induced lymphoma LSTRA and treated with bis-1-chloroethyl-nitrosurea (BCNU) on day +5 after tumor challenge. A significant increase of the antitumor efficacy of BCNU treatment was found in mice inoculated with CA as immunoadjuvant on days −14 and +1 (−14/+1 schedule) with respect to tumor challenge.However, no significant difference in survival time was found between mice treated with BCNU alone and those treated with BCNU plus either soluble mannan or glucan-protein fractions extracted from CA and administered according to −14/+1 or −7/+1 schedules. On the other hand, mice treated with BCNU plus the insoluble glucan fraction (wall ‘ghosts’) given on days −14/+1 or even on day −7 only (i.e., without boosting after tumor challenge) survived longer than animals treated with BCNU alone.The immunoadjuvant effect of CA and of other ‘classic’ immunoadjuvants, such as BCG and Corynebacterium parvum, was completely abolished by total-body irradiation (400 R) given 5 h before the first administration of the agent on day −7 prior to tumor challenge.These results indicate that: (a) the minimal structure required for the expression of the immunoadjuvant effect of CA is the insoluble, β-glucan component of the cell wall; (b) the soluble components of CA cell wall (i.e., mannan and glucan-protein) per se do not show any detectable immunoadjuvant effect in the present animal-tumor system; they may, however, ‘modulate’ this effect, as shown by the fact that whole CA, but not the insoluble β-glucan, needs a boosting injection for the expression of its immunoadjuvant properties; (c) the immunoadjuvanticity of CA is radiosensitive.

Collaboration


Dive into the Enzo Bonmassar's collaboration.

Top Co-Authors

Avatar

Abraham Goldin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Grazia Graziani

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabella Faraoni

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Lucio Tentori

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Ornella Franzese

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Angelo Aquino

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Mario Turriziani

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Ester Alvino

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Pedro Miguel Lacal

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge