Network


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

Hotspot


Dive into the research topics where Roberta Fasani is active.

Publication


Featured researches published by Roberta Fasani.


Breast Cancer Research and Treatment | 2007

Metaplastic carcinoma of the breast, an unusual disease with worse prognosis: the experience of the European Institute of Oncology and review of the literature.

A. Luini; Marisel Aguilar; Giovanna Gatti; Roberta Fasani; Edoardo Botteri; Jack Antonio Diaz Brito; Patrick Maisonneuve; Anna Rita Vento; Giuseppe Viale

BackgroundMetaplastic carcinoma of the breast is a rare form of breast cancer and has an uncertain prognostic significance. The purpose of the present study was to compare the clinical course, and prognosis, between this type of tumor and poorly differentiated ductal carcinoma.Patients and methodsWe analyzed 37 cases of metaplastic carcinoma of the breast treated at our institution (European Institute of Oncology in Milan, Italy) between 1997 and 2004, comparing them with 72 cases (control group) of poorly differentiated ductal carcinoma. All 109 patients had negative receptors and were G3 at final histology. The control cases were matched according to year of surgery, pT (pT1 vs. pT2/3/4), and pN (absent vs. present).ResultsOf the 37 patients, eleven died from disease progression, eight developed metastatic disease and two experienced local recurrence. In the control group (72 patients) we observed three deaths due to disease progression, 13 distant metastases, and two local recurrences.ConclusionThe overall survival in the metaplastic carcinoma group was significantly worse than in the control group. As regards to disease-free survival, there was no statistically significant difference between the two groups.


Cancer Research | 2008

The Differential Role of L1 in Ovarian Carcinoma and Normal Ovarian Surface Epithelium

Silvia Zecchini; Marco Bianchi; Nicoletta Colombo; Roberta Fasani; Giovanni Goisis; Chiara Casadio; Giuseppe Viale; Jinsong Liu; Meenhard Herlyn; Andrew K. Godwin; Paolo Nuciforo; Ugo Cavallaro

Epithelial ovarian carcinoma (EOC) arises from the ovarian surface epithelium (OSE), a monolayer of poorly differentiated epithelial cells that lines the ovary. The molecular mechanisms underlying EOC invasion into the surrounding stroma and dissemination to the peritoneum and to retroperitoneal lymph nodes are still unclear. Here, we analyzed the expression and the functional role of the cell adhesion molecule L1 during EOC development. In patient-derived samples, L1 was expressed both in OSE and in a subset of EOC, in the latter being mostly restricted to the invasive areas of the tumors. The expression of L1 correlated significantly with poor outcome and with unfavorable clinicopathologic features of the disease. The peculiar expression pattern of L1 in normal OSE and invasive EOC raised the possibility that this adhesion molecule serves a different function in nontransformed versus neoplastic ovarian epithelial cells. Indeed, we showed that in OSE cells L1 supports cell-cell adhesion and enhances apoptosis, whereas it has no effect on cell proliferation and invasion. In contrast, L1 inhibits cell-cell adhesion and apoptosis in ovarian carcinoma cells, where it promotes malignancy-related properties, such as cell proliferation, Erk1/2-dependent and phosphoinositide 3-kinase-dependent invasion, and transendothelial migration. Interestingly, a crosstalk with the fibroblast growth factor receptor signaling is implicated in the promalignant function of L1 in tumor cells. Our findings point to L1 as an EOC biomarker correlating with poor prognosis, and highlight a switch in L1 function associated to the neoplastic transformation of ovarian epithelial cells, thus implicating L1 as a potential therapeutic target.


Human Pathology | 2003

Molecular and immunohistochemical analysis of HER2/neu oncogene in synovial sarcoma

Paolo Nuciforo; Caterina Pellegrini; Roberta Fasani; Marco Maggioni; Guido Coggi; Antonina Parafioriti; Silvano Bosari

Amplification and/or overexpression of HER2/neu have been documented in many types of epithelial tumor and recently has been reported in sarcomas, particularly in osteosarcomas. But the role of HER2/neu alterations in soft tissue tumors remains poorly understood. Thus the present study investigates the expression of HER2/neu in 13 patients with synovial sarcoma (SS). In this study, HER2/neu mRNA levels were measured in frozen tissue samples using a real-time reverse transcription-polymerase chain reaction assay; protein expression was assessed by immunohistochemistry using an anti-HER2/neu polyclonal antibody. Six normal skeletal muscle specimens were used to establish basal levels of HER2/neu mRNA. HER2/neu transcripts were detected in all normal tissues and SSs. Four of 13 sarcomas (31%) demonstrated HER2/neu mRNA levels above the mean value, whereas 3 tumors (23%) displayed HER2/neu protein overexpression. Both membranous and cytoplasmic patterns of immunostaining were observed, and a strong correlation was found between protein expression and mRNA level (P = 0.01). Increased HER2/neu mRNA levels were significantly associated with a lower risk of developing recurrences (P = 0.02). Moreover, none of the patients with HER2/neu overexpression developed metastasis. Our data demonstrate that HER2/neu is expressed in SSs and that both membrane and cytoplasmic HER2/neu expression correlate with mRNA levels. Our results show that the presence of increased levels of HER2/neu in SSs is associated with a more favorable clinical course. Further studies are needed to assess the role of this oncogene in SSs and to evaluate the application of inhibitory humanized monoclonal antibodies in the treatment regimens for this malignancy.


The Journal of Pathology | 2005

The transactivating isoforms of p63 are overexpressed in high-grade follicular lymphomas independent of the occurrence of p63 gene amplification

Giancarlo Pruneri; Sonia Fabris; Patrizia Dell'Orto; Maria Olivia Biasi; Stefano Valentini; Barbara Del Curto; Daniele Laszlo; Laura Cattaneo; Roberta Fasani; Laura Rossini; Michela Manzotti; Francesco Bertolini; Giovanni Martinelli; Antonino Neri; Giuseppe Viale

p63 is a p53‐related gene mapping to 3q28 that codes for multiple mRNA transcripts with (TA‐p63) or without (ΔN‐p63) transactivating effects on genes that promote cell differentiation and apoptosis. We analysed p63 alterations by immunohistochemistry, quantitative real‐time RT‐PCR and FISH in a series of 45 follicular lymphomas (FL). None of the tumours showed immunoreactivity for the p40 antibody, which recognizes only the truncated isoforms of p63, or ΔN‐p63 mRNA expression. Immunoreactivity for the 4A4 antibody, which recognizes both the transactivating and the truncated p63 isoforms, was found in 5 ± 5.5%, 6.85 ± 4.88% and 33.2 ± 22.31% of grade I, II and III FL cells, respectively (p < 0.0001). Quantitative RT‐PCR analysis showed that all cases but one had TA‐p63 mRNA levels higher than non‐neoplastic lymphocytes, and that TA‐p63 mRNA expression correlated significantly (r = 0.9194, p < 0.0001) with the prevalence of p63 immunoreactivity. FISH extra signals for the p63 gene were found in seven (23.3%) of the 30 cases analysed (0/6 grade I, 2/15 grade II and 5/9 grade III; p = 0.01937). Further hybridizations showed a pattern highly suggestive of chromosome 3 polysomy in six cases. One of these cases also bore extra copies of the p63 and bcl‐6 genes. Co‐localization of p63 and IgH signals was found in one case. No association between the prevalence of p63 immunoreactivity and extra p63 gene signals was detectable when the cases were dichotomized according to a p63 immunoreactivity threshold of 10%. Our data suggest that TA‐p63 is overexpressed in high‐grade FL, possibly independent of the occurrence of gene abnormalities, and that it may be involved in the highly complex mechanism of regulation of apoptosis of FL cells. Copyright


The Journal of Pathology | 2003

Immunoreactivity for cyclin D3 is frequently detectable in high-grade primary gastric lymphomas in the absence of the t(6;14)(p21.1;q32.3) chromosomal translocation

Giancarlo Pruneri; Sonia Fabris; Roberta Fasani; Barbara Del Curto; Carlo Capella; Barbara Pozzi; Teresio Motta; Salvatore Andreola; Andres Jm Ferreri; Maurilio Ponzoni; Giuseppe Viale; Antonino Neri

Cyclin D3 plays a pivotal role in controlling the physiological progression from the G1 to the S phase of the cell cycle. Recent data suggest that cyclin D3 may be deregulated in extranodal non‐Hodgkins lymphomas (NHLs) as a consequence of the t(6;14)(p21.1;q32.3) translocation. The present study investigated for the first time by dual‐colour fluorescence in situ hybridization (FISH) on interphase nuclei and immunohistochemistry the prevalence of the t(6;14) translocation and cyclin D3 immunoreactivity (IR) in a series of 29 stage I–IIE primary gastric NHLs (PGLs). No case showed the t(6;14) translocation. However, in five (17.2%) cases (two extranodal marginal zone lymphomas of MALT type, LGM; one diffuse large‐cell lymphoma with a MALT component, DLCLM; and two diffuse large‐cell lymphomas without a MALT component, DLCL), three to four cyclin D3 signals were detected by FISH. Co‐hybridization with probes specific for the centromeric region and long arm of chromosome 6 indicated trisomy in one case (DLCL), whereas in the remaining four cases the pattern was highly suggestive of the presence of an isochromosome 6p. One (12.5%) case of LGM, six (75%) cases of DLCLM, and seven (53.8%) cases of DLCL (p = 0.0378) were immunoreactive for cyclin D3. Cyclin D3 IR was detected in two (40%) of the five cases with extra cyclin D3 signals and in 12 of the remaining 24 cases (50%, p = 1.000). These results suggest that the t(6;14) may represent a rare event in the pathogenesis of PGL and that cyclin D3 deregulation is most likely the result of epigenetic mechanisms. Copyright


Tumori | 2009

MRI features of cotyledonoid dissecting leiomyoma of the uterus

Lorenzo Preda; Stefania Rizzo; Maria Sole Prevedoni Gorone; Roberta Fasani; Angelo Maggioni; Massimo Bellomi

Imaging evaluation of uterine masses is important to assess the type of lesion and to target surgery, if surgical excision is necessary. This can be decisive in fertile women with benign masses resembling malignancies, in order to avoid overtreatment. In this study, the magnetic resonance imaging (MRI) appearance of cotyledonoid dissecting leiomyoma of the uterus, a rare benign variant of leiomyoma mimicking malignancy, is presented.


Annals of Oncology | 2018

RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer

Cristina Cruz; M Castroviejo-Bermejo; S Gutiérrez-Enríquez; A Llop-Guevara; Yasir H. Ibrahim; Albert Gris-Oliver; S Bonache; Beatriz Morancho; Alejandra Bruna; O M Rueda; Z Lai; U M Polanska; G N Jones; P Kristel; L de Bustos; Mario Guzmán; Olga Rodriguez; Judit Grueso; G Montalban; Ginevra Caratú; F. Mancuso; Roberta Fasani; J. Jimenez; W J Howat; B Dougherty; Ana Vivancos; Paolo Nuciforo; X Serres-Créixams; Isabel T. Rubio; E Cadogan

Abstract Background BRCA1 and BRCA2 (BRCA1/2)-deficient tumors display impaired homologous recombination repair (HRR) and enhanced sensitivity to DNA damaging agents or to poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi). Their efficacy in germline BRCA1/2 (gBRCA1/2)-mutated metastatic breast cancers has been recently confirmed in clinical trials. Numerous mechanisms of PARPi resistance have been described, whose clinical relevance in gBRCA-mutated breast cancer is unknown. This highlights the need to identify functional biomarkers to better predict PARPi sensitivity. Patients and methods We investigated the in vivo mechanisms of PARPi resistance in gBRCA1 patient-derived tumor xenografts (PDXs) exhibiting differential response to PARPi. Analysis included exome sequencing and immunostaining of DNA damage response proteins to functionally evaluate HRR. Findings were validated in a retrospective sample set from gBRCA1/2-cancer patients treated with PARPi. Results RAD51 nuclear foci, a surrogate marker of HRR functionality, were the only common feature in PDX and patient samples with primary or acquired PARPi resistance. Consistently, low RAD51 was associated with objective response to PARPi. Evaluation of the RAD51 biomarker in untreated tumors was feasible due to endogenous DNA damage. In PARPi-resistant gBRCA1 PDXs, genetic analysis found no in-frame secondary mutations, but BRCA1 hypomorphic proteins in 60% of the models, TP53BP1-loss in 20% and RAD51-amplification in one sample, none mutually exclusive. Conversely, one of three PARPi-resistant gBRCA2 tumors displayed BRCA2 restoration by exome sequencing. In PDXs, PARPi resistance could be reverted upon combination of a PARPi with an ataxia-telangiectasia mutated (ATM) inhibitor. Conclusion Detection of RAD51 foci in gBRCA tumors correlates with PARPi resistance regardless of the underlying mechanism restoring HRR function. This is a promising biomarker to be used in the clinic to better select patients for PARPi therapy. Our study also supports the clinical development of PARPi combinations such as those with ATM inhibitors.


Science Translational Medicine | 2018

p95HER2–T cell bispecific antibody for breast cancer treatment

Irene Rius Ruiz; Rocio Vicario; Beatriz Morancho; Cristina Morales; Enrique J. Arenas; Sylvia Herter; Anne Freimoser-Grundschober; Jitka Somandin; Johannes Sam; Oliver Ast; Águeda Martinez Barriocanal; Antonio Luque; Marta Escorihuela; Ismael Varela; Isabel Cuartas; Paolo Nuciforo; Roberta Fasani; Vicente Peg; Isabel T. Rubio; Javier Cortes; Violeta Serra; Santiago Escrivá-de-Romaní; Jeff Sperinde; Ahmed Chenna; Weidong Huang; John Winslow; Joan Albanell; Joan Seoane; Maurizio Scaltriti; José Baselga

T cell bispecific antibodies against an isoform of HER2 effectively target many HER2-expressing tumors but not normal tissues. Fine-tuning HER2 targeting HER2 receptor tyrosine kinase is frequently overexpressed in breast and gastric cancer. HER2-overexpressing tumors can be treated with trastuzumab, an antibody against this receptor, and additional methods of targeting HER2 are also being developed. Unfortunately, HER2 is also expressed in normal tissues, resulting in unacceptable toxicities when HER2-targeting therapies damage healthy organs. Rius Ruiz et al. now propose targeting p95HER2, a carboxyl-terminal fragment of HER2 that is expressed in almost half of HER2-positive tumors. The authors demonstrate the effectiveness of this approach, as well as its safety due to the lack of p95HER2 expression in nontumor tissues. T cell bispecific antibodies (TCBs) are engineered molecules that include, within a single entity, binding sites to the T cell receptor and to tumor-associated or tumor-specific antigens. The receptor tyrosine kinase HER2 is a tumor-associated antigen in ~25% of breast cancers. TCBs targeting HER2 may result in severe toxicities, likely due to the expression of HER2 in normal epithelia. About 40% of HER2-positive tumors express p95HER2, a carboxyl-terminal fragment of HER2. Using specific antibodies, here, we show that p95HER2 is not expressed in normal tissues. We describe the development of p95HER2-TCB and show that it has a potent antitumor effect on p95HER2-expressing breast primary cancers and brain lesions. In contrast with a TCB targeting HER2, p95HER2-TCB has no effect on nontransformed cells that do not overexpress HER2. These data pave the way for the safe treatment of a subgroup of HER2-positive tumors by targeting a tumor-specific antigen.


Annals of Oncology | 2018

A predictive model of pathologic response based on tumor cellularity and tumor-infiltrating lymphocytes (CelTIL) in HER2-positive breast cancer treated with chemo-free dual HER2 blockade

Paolo Nuciforo; T Pascual; Javier Cortes; Antonio Llombart-Cussac; Roberta Fasani; Laia Paré; M.M. Oliveira; Patricia Galván; Noelia Martínez; Begoña Bermejo; Marcela A. Vidal; Sonia Pernas; Ramon Lopez; Montserrat Muñoz; I Garau; Luis Manso; J Alarcón; E Martínez; V Rodrik-Outmezguine; Jan C. Brase; P Villagrasa; Aleix Prat; E Holgado

Background The presence of stromal tumor-infiltrating lymphocytes (TILs) is associated with increased pathologic complete response (pCR) and improved outcomes in HER2-positive early-breast cancer (BC) treated with anti-HER2-based chemotherapy. In the absence of chemotherapy, the association of TILs with pCR following anti-HER2 therapy-only is largely unknown. Patients and methods The PAMELA neoadjuvant trial treated 151 women with HER2-positive BC with lapatinib and trastuzumab [and hormonal therapy if hormone receptor (HR)-positive] for 18 weeks. Percentage of TILs and tumor cellularity were determined at baseline (N = 148) and at day 15 (D15) of treatment (N = 134). Associations of TILs and tumor cellularity with pCR in the breast were evaluated. A combined score based on tumor cellularity and TILs (CelTIL) measured at D15 was derived in PAMELA, and validated in D15 samples from 65 patients with HER2-positive disease recruited in the LPT109096 neoadjuvant trial, where anti-HER2 therapy-only was administer for 2 weeks, then standard chemotherapy was added for 24 weeks. Results In PAMELA, baseline and D15 TILs were significantly associated with pCR in univariate analysis. In multivariable analysis, D15 TILs, but not baseline TILs, were significantly associated with pCR. At D15, TILs and tumor cellularity were found independently associated with pCR. A combined score (CelTIL) taking into account both variables was derived. CelTIL at D15 as a continuous variable was significantly associated with pCR, and patients with CelTIL-low and CelTIL-high scores had a pCR rate of 0% and 33%, respectively. In LPT109096, CelTIL at D15 was found associated with pCR both as a continuous variable and as group categories using a pre-defined cut-off (75.0% versus 33.3%). Conclusions On-treatment TILs, but not baseline TILs, are independently associated with response following anti-HER2 therapy-only. A combined score of TILs and tumor cellularity measured at D15 provides independent predictive information upon completion of neoadjuvant anti-HER2-based therapy. Clinical trial number NCT01973660.


Cancer Research | 2017

Abstract S3-03: PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial

A Prat Aparicio; J. Cortes Castan; Laia Paré; Patricia Galván; Begoña Bermejo; Noelia Martínez; María N. Vidal; Sonia Pernas; Rafael López; Montserrat Muñoz; Paolo Nuciforo; Roberta Fasani; Serafin Morales; Mafalda Oliveira; L de la Pena; A Peláez; Antonio Llombart

Background: Prior neoadjuvant studies in HER2+ breast cancer have shown that dual HER2 blockade without chemotherapy achieves pathological complete responses (pCR) rates of 6-36% (TBCRC006/TBCRC023/NeoSphere). However, a major challenge today is how to select prospectively patients who will derive the maximum benefit from dual anti-HER2 therapies without chemotherapy. In this context, we and others have previously shown that HER2+ disease is biologically heterogeneous and composed of all the intrinsic molecular subtypes (Luminal A, Luminal B, HER2-enriched [HER2-E] and Basal-like). Among them, the HER2-E subtype shows the highest activation of the EGFR/HER2 pathway. Methods: PAMELA (NCT01973660) is a non-randomized, open-label, multicentric, prospective translational research study in stage I-IIIA HER2+ breast cancer designed to evaluate the ability of the PAM50 intrinsic subtypes to predict pCR in the breast (pCRB; in situ allowed) following 18 weeks of neoadjuvant lapatinib and trastuzumab). Patients with HR+ disease received letrozole (if postmenopausal) or tamoxifen (if pre-menopausal). The primary objective was to compare the pCRB rates of the HER2-E versus the non-HER2-E subtypes in the intent-to-treat population. The study was planned with a power of 95% at a significance level of 0.05 to detect an absolute relative difference in pCRB rates between the two groups of 27% (i.e. 35% in HER2E and 8% in non-HER2-E). Day-15 formalin-fixed, paraffin-embedded tissue samples were prospectively collected and gene expression profiled using the nCounter platform. The intrinsic subtypes were identified using the research-based PAM50 predictor (Parker JCO 2009). Results: A total of 151 patients were recruited (n=77 HR+ and n=74 HR-). Patient characteristics were: mean age (55 years), mean tumor size (2.84 cm), negative axilla (63.5%) and postmenopausal (60.2%). At baseline, intrinsic subtype distribution was: HER2-E (n=101, 66.9%), Luminal A (n=22; 14.6%), Luminal B (n=16; 10.6%), Basal-like (n=9; 6%) and Normal-Like (n=3; 2%). The overall pCRB was 30.5% (46/151), 18.2% in HR+ disease and 43.2% in HR- disease. Five patients (3.3%) presented progressive disease. Rates of pCRB in HER2-E and non-HER2-E subtypes were 40.6% and 10.0% (p Conclusions: The PAMELA trial met its primary endpoint. PAM50 HER2-E subtype identifies patients with HER2+ disease likely to derive a large benefit from dual anti-HER2 therapies +/- endocrine therapy, especially in HER2+/HR+ disease. In addition, early changes in gene expression indicative of a reduction of tumor cellularity are predictive of pathological complete response at surgery. Citation Format: Prat Aparicio A, Cortes Castan J, Pare L, Galvan P, Bermejo B, Martinez N, Vidal M, Pernas S, Lopez R, Munoz M, Nuciforo P, Fasani R, Morales S, Oliveira M, de La Pena L, Pelaez A, Llombart A. PAM50 intrinsic subtype as a predictor of pathological complete response following neoadjuvant dual HER2 blockade without chemotherapy in HER2-positive breast cancer: First results of the PAMELA clinical trial [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr S3-03.

Collaboration


Dive into the Roberta Fasani's collaboration.

Top Co-Authors

Avatar

Isabel T. Rubio

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppe Viale

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aleix Prat

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Antonio Llombart

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Beatriz Morancho

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noelia Martínez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Maurizio Scaltriti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge