Network


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

Hotspot


Dive into the research topics where Tiziana Ottone is active.

Publication


Featured researches published by Tiziana Ottone.


Blood | 2010

Cytogenetic and molecular diagnostic characterization combined to postconsolidation minimal residual disease assessment by flow cytometry improves risk stratification in adult acute myeloid leukemia

Francesco Buccisano; Luca Maurillo; Alessandra Spagnoli; Maria Ilaria Del Principe; Daniela Fraboni; Paola Panetta; Tiziana Ottone; Maria Irno Consalvo; Serena Lavorgna; Pietro Bulian; Emanuele Ammatuna; Daniela F. Angelini; Adamo Diamantini; Selenia Campagna; Licia Ottaviani; Chiara Sarlo; Valter Gattei; Giovanni Del Poeta; William Arcese; Sergio Amadori; Francesco Lo Coco; Adriano Venditti

A total of 143 adult acute myeloid leukemia (AML) patients with available karyotype (K) and FLT3 gene mutational status were assessed for minimal residual disease (MRD) by flow cytometry. Twenty-two (16%) patients had favorable, 115 (80%) intermediate, and 6 (4%) poor risk K; 19 of 129 (15%) carried FLT3-ITD mutation. Considering postconsolidation MRD status, patients with good/intermediate-risk K who were MRD(-) had 4-year relapse-free survival (RFS) of 70% and 63%, and overall survival (OS) of 84% and 67%, respectively. Patients with good- and intermediate-risk K who were MRD(+) had 4-year RFS of 15% and 17%, and OS of 38% and 23%, respectively (P < .001 for all comparisons). FLT3 wild-type patients achieving an MRD(-) status, had a better outcome than those who remained MRD(+) (4-year RFS, 54% vs 17% P < .001; OS, 60% vs 23%, P = .002). Such an approach redefined cytogenetic/genetic categories in 2 groups: (1) low-risk, including good/intermediate K-MRD(-) with 4-year RFS and OS of 58% and 73%, respectively; and (2) high risk, including poor-risk K, FLT3-ITD mutated cases, good/intermediate K-MRD(+) categories, with RFS and OS of 22% and 17%, respectively (P < .001 for all comparisons). In AML, the integrated evaluation of baseline prognosticators and MRD improves risk-assessment and optimizes postremission therapy.


The Journal of Molecular Diagnostics | 2008

An Allele-Specific RT-PCR Assay to Detect Type A Mutation of the Nucleophosmin-1 Gene in Acute Myeloid Leukemia

Tiziana Ottone; Emanuele Ammatuna; Serena Lavorgna; Nélida I. Noguera; Francesco Buccisano; Adriano Venditti; Laura Giannì; Massimiliano Postorino; Giorgio Federici; Sergio Amadori; Francesco Lo-Coco

Nucleophosmin-1 (NPM1) mutations represent the most frequent gene alteration in acute myeloid leukemia (AML). The most common NPM1 mutation type, accounting for 75 to 80% of cases, is referred to as mutation A (NPM1-mutA). These NPM1 alterations have been shown to possess prognostic significance because they appear to identify patients who will benefit from chemotherapy. Given the high prevalence and stability of these mutations over the course of disease, NPM1 mutations may serve as ideal targets for minimal residual disease (MRD) assessment in AML. Current detection methods are costly, require sophisticated equipment, and are often not sufficiently sensitive. We report here an allele-specific (ASO)-RT-PCR assay that enables rapid and sensitive detection of NPM1-mutA. A semi-nested ASO-PCR method was also designed to increase the sensitivity of our assay for the monitoring of MRD. We analyzed bone marrow cells collected from 52 patients with AML at presentation. NPM1-mutA was detected in leukemic cells from 21 patients. Assay specificity was confirmed by capillary electrophoresis and DNA sequencing. ASO-RT-PCR and semi-nested ASO-PCR assays could detect NPM1-mutA with sensitivities of 10(-2) and 10(-5), respectively. Results obtained here verify that our ASO-RT-PCR assay is a specific and sensitive method for the routine screening of NPM1-mutA, as well as for MRD monitoring of AML patients with this alteration. This method is convenient and easily applicable in countries with limited resources and no access to real-time quantitative PCR-based technologies.


Neurology | 2011

Risk of acute promyelocytic leukemia in multiple sclerosis Coding variants of DNA repair genes

Syed Khizer Hasan; F. Buttari; Tiziana Ottone; M.T. Voso; Stefan Hohaus; Elena Marasco; Vilma Mantovani; Paolo Garagnani; Miguel A. Sanz; Laura Cicconi; Giorgio Bernardi; D. Centonze; Francesco Lo-Coco

Background: Single nucleotide polymorphisms (SNPs) in double-strand break repair genes may alter DNA repair capacity and, in turn, confer predisposition to leukemia. We analyzed polymorphic variants of DNA repair and detoxification genes in patients with multiple sclerosis (MS) who developed secondary acute promyelocytic leukemia (sAPL), in most cases after treatment with mitoxantrone (MTZ). Methods: Using MassARRAY high-throughput DNA analysis with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, we genotyped patients with sAPL (n = 20) developed after treatment of MS (18 out 20 treated with MTZ) for the presence of 210 SNPs of 22 genes mostly involved in DNA repair and drug detoxification. Patients with MS who did not develop sAPL including 41 treated with MTZ (n = 253 and 41, respectively) and healthy blood donors (n = 310) were also genotyped as controls. Results: We observed risk allele frequency between MS and sAPL for BRCA2 (rs1801406): 6% and 26%, p = 0.007; XRCC5 (rs207906): 2.5% and 15%, p = 0.016; CYP3A4 (rs2740574): 4.5% and 25%, p = 0.0035. The association of homozygous variants of BRCA2 and XRCC5 yielded higher risk of sAPL (MS vs sAPL: 0.4% and 18%, p = 0.001). We also observed a significant association between a SNP in the promoter region (rs2740574) of CYP3A4, an enzyme involved in the metabolism of chemotherapeutic agents and development of sAPL. Conclusions: Increased susceptibility to develop sAPL in patients with MS receiving MTZ may be linked to genetic variants in DNA repair and drug-metabolizing enzymes that result in impaired detoxification of chemotherapy or inefficient repair of drug-induced genetic damage.


Genes, Chromosomes and Cancer | 2010

Analysis of t(15;17) chromosomal breakpoint sequences in therapy-related versus de novo acute promyelocytic leukemia: association of DNA breaks with specific DNA motifs at PML and RARA loci.

Syed Khizer Hasan; Tiziana Ottone; Richard F. Schlenk; Yuanyuan Xiao; Joseph L. Wiemels; Maria Enza Mitra; Paolo Bernasconi; Francesco Di Raimondo; Maria Teresa Lupo Stanghellini; Pepa Marco; Ashley N. Mays; Hartmut Döhner; Miguel A. Sanz; S. Amadori; David Grimwade; Francesco Lo-Coco

We compared genomic breakpoints at the PML and RARA loci in 23 patients with therapy‐related acute promyelocytic leukemia (t‐APL) and 25 de novo APL cases.Eighteen of 23 t‐APL cases received the topoisomerase II poison mitoxantrone for their primary disorder. DNA breaks were clustered in a previously reported 8 bp “hot spot” region of PML corresponding to a preferred site of mitoxantrone‐induced DNA topoisomerase II‐mediated cleavage in 39% of t‐APL occurring in patients exposed to this agent and in none of the cases arising de novo (P = 0.007). As to RARA breakpoints, clustering in a 3′ region of intron 2 (region B) was found in 65% of t‐APL and 28% of de novo APL patients, respectively. Scan statistics revealed significant clustering of RARA breakpoints in region B in t‐APL cases (P = 0.001) as compared to de novo APL (P = 1). Furthermore, ∼300 bp downstream of RARA region B contained a sequence highly homologous to a topoisomerase II consensus sequence. Biased distribution of DNA breakpoints at both PML and RARA loci suggest the existence of different pathogenetic mechanisms in t‐APL as compared with de novo APL.


Leukemia | 2013

Nucleophosmin/B26 regulates PTEN through interaction with HAUSP in acute myeloid leukemia.

N. I. Noguera; Min Sup Song; Mariadomenica Divona; G. Catalano; K. L. Calvo; F. García; Tiziana Ottone; F. Florenzano; I. Faraoni; L. Battistini; E. Colombo; S. Amadori; Pier Paolo Pandolfi; Francesco Lo-Coco

PTEN (phosphatase and tensin homolog deleted in chromosome 10) is a bona fide dual lipid and protein phosphatase with cytoplasmic (Cy) and nuclear localization. PTEN nuclear exclusion has been associated with tumorigenesis. Nucleophosmin (NPM1) is frequently mutated in acute myeloid leukemia (AML) and displays Cy localization in mutated nucleophosmin (NPMc+) AML. Here we show that NPM1 directly interacts with herpes virus-associated ubiquitin specific protease (HAUSP), which is known as a PTEN deubiquitinating enzyme. Strikingly, PTEN is aberrantly localized in AML carrying NPMc+. Mechanistically, NPM1 in the nucleus opposes HAUSP-mediated deubiquitination and this promotes the shuttle of PTEN to the cytoplasm. In the cytoplasm, NPMc+ prevents HAUSP from deubiquitinating PTEN, causing the latter to stay in the cytoplasm where it is polyubiquitinated and degraded. Our findings delineate a new NPM1–HAUSP molecular interaction controlling PTEN deubiquitination and trafficking.


Experimental Hematology | 2011

Sphingosine kinase 1 overexpression is regulated by signaling through PI3K, AKT2, and mTOR in imatinib-resistant chronic myeloid leukemia cells

Gabriella Marfe; Carla Di Stefano; Alessandra Gambacurta; Tiziana Ottone; Valentina Martini; Elisabetta Abruzzese; Luca Mologni; Paola Sinibaldi-Salimei; Paolo de Fabritis; Carlo Gambacorti-Passerini; S. Amadori; Raymond B. Birge

OBJECTIVE As a better understanding of the molecular basis of carcinogenesis has emerged, oncogene-specific cell-signaling pathways have been successfully targeted to treat human malignances. Despite impressive advances in oncogene-directed therapeutics, genetic instability in cancer cells often manifest acquired resistance. This is particularly noted in the use of tyrosine kinase inhibitors therapies and not more evident than for chronic myeloid leukemia. Therefore, it is of great importance to understand the molecular mechanisms affecting cancer cell sensitivity and resistance to tyrosine kinase inhibitors. MATERIALS AND METHODS In this study, we used continuous exposure to stepwise increasing concentrations of imatinib (0.6-1 μM) to select imatinib-resistant K562 cells. RESULTS Expression of BCR-ABL increased both at RNA and protein levels in imatinib-resistant cell lines. Furthermore, expression levels of sphingosine kinase 1 (SphK1) were increased significantly in resistant cells, channeling sphingoid bases to the SphK1 pathway and activating sphingosine-1-phosphate-dependent tyrosine phosphorylation pathways that include the adaptor protein Crk. The partial inhibition of SphK1 activity by N,N-dimethylsphingosine or expression by small interfering RNA increased sensitivity to imatinib-induced apoptosis in resistant cells and returned BCR-ABL to baseline levels. To determine the resistance mechanism-induced SphK1 upregulation, we used pharmacological inhibitors of the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin signaling pathway and observed robust downmodulation of SphK1 expression and activity when AKT2, but not AKT1 or AKT3, was suppressed. CONCLUSIONS These results demonstrate that SphK1 is upregulated in imatinib-resistant K562 cells by a pathway contingent on a phosphoinositide 3-kinase/AKT2/mammalian target of rapamycin signaling pathway. We propose that SphK1 plays an important role in development of acquired resistance to imatinib in chronic myeloid leukemia cell lines.


Biochimica et Biophysica Acta | 2015

BRCA1, PARP1 and γH2AX in acute myeloid leukemia: Role as biomarkers of response to the PARP inhibitor olaparib

Isabella Faraoni; Mirco Compagnone; Serena Lavorgna; Daniela F. Angelini; Maria Teresa Cencioni; Eleonora Piras; Paola Panetta; Tiziana Ottone; Susanna Dolci; Adriano Venditti; Grazia Graziani; Francesco Lo-Coco

Olaparib (AZD-2281, Ku-0059436) is an orally bioavailable and well-tolerated poly(ADP-ribose) polymerase (PARP) inhibitor currently under investigation in patients with solid tumors. To study the clinical potential of olaparib as a single-agent for the treatment of acute myeloid leukemia (AML) patients, we analyzed the in vitro sensitivity of AML cell lines and primary blasts. Clinically achievable concentrations of olaparib were able to induce cell death in the majority of primary AML case samples (88%) and tested cell lines. At these concentrations, olaparib preferentially killed leukemic blasts sparing normal lymphocytes derived from the same patient and did not substantially affect the viability of normal bone marrow and CD34-enriched peripheral blood cells obtained from healthy donors. Most primary AML analyzed were characterized by low BRCA1 mRNA level and undetectable protein expression that likely contributed to explain their sensitivity to olaparib. Noteworthy, while PARP1 over-expression was detected in blasts not responsive to olaparib, phosphorylation of the histone H2AFX (γH2AX) was associated with drug sensitivity. As to genetic features of tested cases the highest sensitivity was shown by a patient carrying a 11q23 deletion. The high sensitivity of AML blasts and the identification of biomarkers potentially able to predict response and/or resistance may foster further investigation of olaparib monotherapy for AML patients unfit to conventional chemotherapy.


Blood | 2010

A monoclonal antibody against mutated nucleophosmin 1 for the molecular diagnosis of acute myeloid leukemias.

Alicja M. Gruszka; Serena Lavorgna; Maria Irno Consalvo; Tiziana Ottone; Chiara Martinelli; Mario Cinquanta; Giuseppe Ossolengo; Giancarlo Pruneri; Francesco Buccisano; Mariadomenica Divona; Michele Cedrone; Emanuele Ammatuna; Adriano Venditti; Ario de Marco; Francesco Lo-Coco; Pier Giuseppe Pelicci

Mutations in the nucleophosmin 1 (NPM1) gene are the most frequent genetic aberrations of acute myeloid leukemia (AML) and define a clinically distinct subset of AML. A monoclonal antibody (T26) was raised against a 19-amino acid polypeptide containing the unique C-terminus of the type A NPM1 mutant protein. T26 recognized 10 of the 21 known NPM1 mutants, including the A, B, and D types, which cover approximately 95% of all cases, and did not cross-react with wild-type NPM1 or unrelated cellular proteins. It performed efficiently with different detection technologies, including immunofluorescence, immunohistochemistry, and flow cytometry. Within a series of consecutive de novo AML patients, 44 of 110 (40%) and 15 of 39 (38%) cases scored positive using the T26 antibody in immunofluorescence and flow cytometry assays, respectively. T26-positive cases were found to be all carrying mutations of NPM1 exclusively, as determined by molecular analysis. T26 is the first antibody that specifically recognizes a leukemia-associated mutant protein. Immunofluorescence or flow cytometry using T26 may thus become a new tool for a rapid, simple, and cost-effective molecular diagnosis of AMLs.


Cell Death and Disease | 2013

Argonaute 2 sustains the gene expression program driving human monocytic differentiation of acute myeloid leukemia cells

I Iosue; Roberta Quaranta; Silvia Masciarelli; Giulia Fontemaggi; E M Batassa; C Bertolami; Tiziana Ottone; Mariadomenica Divona; Beatrice Salvatori; Fabrizio Padula; Alessandro Fatica; Francesco Lo-Coco; Clara Nervi; Francesco Fazi

MicroRNAs are key regulators of many biological processes, including cell differentiation. These small RNAs exert their function assembled in the RNA-induced silencing complexes (RISCs), where members of Argonaute (Ago) family of proteins provide a unique platform for target recognition and gene silencing. Here, by using myeloid cell lines and primary blasts, we show that Ago2 has a key role in human monocytic cell fate determination and in LPS-induced inflammatory response of 1,25-dihydroxyvitamin D3 (D3)-treated myeloid cells. The silencing of Ago2 impairs the D3-dependent miR-17-5p/20a/106a, miR-125b and miR-155 downregulation, the accumulation of their translational targets AML1, VDR and C/EBPβ and monocytic cell differentiation. Moreover, we show that Ago2 is recruited on miR-155 host gene promoter and on the upstream region of an overlapping antisense lncRNA, determining their epigenetic silencing, and miR-155 downregulation. These findings highlight Ago2 as a new factor in myeloid cell fate determination in acute myeloid leukemia cells.


Leukemia | 2016

PML–RARα kinetics and impact of FLT3 –ITD mutations in newly diagnosed acute promyelocytic leukaemia treated with ATRA and ATO or ATRA and chemotherapy

Laura Cicconi; Mariadomenica Divona; C Ciardi; Tiziana Ottone; A Ferrantini; S Lavorgna; V Alfonso; Francesca Paoloni; Alfonso Piciocchi; Giuseppe Avvisati; F Ferrara; E Di Bona; Francesco Albano; Massimo Breccia; Elisa Cerqui; Marco Sborgia; Mariagrazia Kropp; Armando Santoro; Alessandro Levis; Simona Sica; S Amadori; Maria Teresa Voso; Franco Mandelli; Francesco Lo-Coco

The APL0406 study showed that arsenic trioxide (ATO) and all-trans retinoic acid (ATRA) are not inferior to standard ATRA and chemotherapy (CHT) in newly diagnosed, low–intermediaterisk acute promyelocytic leukaemia (APL). We analysed the kinetics of promyelocytic leukaemia–retinoic acid receptor-α (PML–RARα) transcripts by real-time quantitative PCR (RQ-PCR) in bone marrow samples from 184 patients and assessed the prognostic impact of fms-related tyrosine kinase 3–internal tandem duplication (FLT3–ITD) in 159 patients enrolled in this trial in Italy. After induction therapy, the reduction of PML–RARα transcripts was significantly greater in patients receiving ATRA-CHT as compared with those treated with ATRA–ATO (3.4 vs 2.9 logs; P=0.0182). Conversely, at the end of consolidation, a greater log reduction of PML–RARα transcripts was detected in the ATRA–ATO as compared with the ATRA–CHT group (6.3 vs 5.3 logs; P=0.0024). FLT3–ITD mutations had no significant impact on either event-free survival (EFS) or cumulative incidence of relapse in patients receiving ATRA–ATO, whereas a trend for inferior EFS was observed in FLT3–ITD-positive patients receiving ATRA-CHT. Our study shows at the molecular level that ATRA–ATO exerts at least equal and probably superior antileukaemic efficacy compared with ATRA–CHT in low–intermediaterisk APL. The data also suggest that ATRA–ATO may abrogate the negative prognostic impact of FLT3–ITD.

Collaboration


Dive into the Tiziana Ottone's collaboration.

Top Co-Authors

Avatar

Francesco Lo-Coco

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Serena Lavorgna

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emanuele Ammatuna

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Adriano Venditti

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Laura Cicconi

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Maria Teresa Voso

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Syed Khizer Hasan

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Francesco Buccisano

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Paola Panetta

University of Rome Tor Vergata

View shared research outputs
Researchain Logo
Decentralizing Knowledge