Network


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

Hotspot


Dive into the research topics where Annamaria la Torre is active.

Publication


Featured researches published by Annamaria la Torre.


Nature | 2015

Comprehensive genomic profiles of small cell lung cancer

Julie George; Jing Shan Lim; Se Jin Jang; Yupeng Cun; Luka Ozretić; Gu Kong; Frauke Leenders; Xin Lu; Lynnette Fernandez-Cuesta; Graziella Bosco; Christian Müller; Ilona Dahmen; Nadine S. Jahchan; Kwon-Sik Park; Dian Yang; Anthony N. Karnezis; Dedeepya Vaka; Angela Torres; Maia Segura Wang; Jan O. Korbel; Roopika Menon; Sung-Min Chun; Deokhoon Kim; Matt Wilkerson; Neil Hayes; David Engelmann; Brigitte M. Pützer; Marc Bos; Sebastian Michels; Ignacija Vlasic

We have sequenced the genomes of 110 small cell lung cancers (SCLC), one of the deadliest human cancers. In nearly all the tumours analysed we found bi-allelic inactivation of TP53 and RB1, sometimes by complex genomic rearrangements. Two tumours with wild-type RB1 had evidence of chromothripsis leading to overexpression of cyclin D1 (encoded by the CCND1 gene), revealing an alternative mechanism of Rb1 deregulation. Thus, loss of the tumour suppressors TP53 and RB1 is obligatory in SCLC. We discovered somatic genomic rearrangements of TP73 that create an oncogenic version of this gene, TP73Δex2/3. In rare cases, SCLC tumours exhibited kinase gene mutations, providing a possible therapeutic opportunity for individual patients. Finally, we observed inactivating mutations in NOTCH family genes in 25% of human SCLC. Accordingly, activation of Notch signalling in a pre-clinical SCLC mouse model strikingly reduced the number of tumours and extended the survival of the mutant mice. Furthermore, neuroendocrine gene expression was abrogated by Notch activity in SCLC cells. This first comprehensive study of somatic genome alterations in SCLC uncovers several key biological processes and identifies candidate therapeutic targets in this highly lethal form of cancer.


Epigenetics | 2011

Frequent epigenetics inactivation of KEAP1 gene in non-small cell lung cancer

Lucia Anna Muscarella; Paola Parrella; Vito D’Alessandro; Annamaria la Torre; Raffaela Barbano; Andrea Fontana; Antonio Tancredi; Vito Guarnieri; Teresa Balsamo; Michelina Coco; Massimiliano Copetti; Fabio Pellegrini; Patrizia De Bonis; Michele Bisceglia; Gerardo Scaramuzzi; Evaristo Maiello; Vanna Maria Valori; Giuseppe Merla; Gianluigi Vendemiale; Vito Michele Fazio

The KEAP1/Nrf2 pathway is a master regulator of several redox-sensitive genes implicated in resistance of tumor cells against chemotherapeutic drugs. Recent data suggest that epigenetic mechanisms may play a pivotal role in the regulation of KEAP1 expression. We performed a comprehensive genetic and epigenetic analysis of the KEAP1 gene in 47 non-small cell lung cancer tissues and normal specimens. Promoter methylation analysis was performed using a quantitative methylation specific PCR assay in real time. Methylation at the KEAP1 promoter region was detected in 22 out of the 47 NSCLCs (47%) and in none of the normal tissues analyzed. Somatic mutations were detected in 7 out of the 47 tumors (15%) and loss of heterozygosity (LOH) in 10 out of the 47 (21%) of the cases. Overall, we found at least one molecular alteration in 57% of the cases. Approximately one third of the tumors had two alterations and this feature was associated with higher risk of disease progression in univariate COX regression analysis (HR = 3.62; 95% CI 1.24–10.65, p = 0.02). This result was confirmed by Kaplan-Meier analysis, which demonstrated an association between worst outcome and KEAP1 double alterations (p = 0.01, Log rank test). Our results further suggest that deregulation of the NRF2/KEAP1 system could play a pivotal role in the cancerogenesis of NSCLC. In addition identifying patients with KEAP1 genetic and epigenetic abnormalities may contribute to disease progression prediction and response to therapy in lung cancer patients.


Epigenetics | 2013

Aberrant Keap1 methylation in breast cancer and association with clinicopathological features

Raffaela Barbano; Lucia Anna Muscarella; Barbara Pasculli; Vanna Maria Valori; Andrea Fontana; Michelina Coco; Annamaria la Torre; Teresa Balsamo; Maria Luana Poeta; Giovanni Francesco Marangi; Evaristo Maiello; Marina Castelvetere; Fabio Pellegrini; Roberto Murgo; Vito Michele Fazio; Paola Parrella

Keap1 (Kelch-like ECH-associated protein 1) is an adaptor protein that mediates the ubiquitination/degradation of genes regulating cell survival and apoptosis under oxidative stress conditions. We determined methylation status of the KEAP1 promoter in 102 primary breast cancers, 14 pre-invasive lesions, 38 paired normal breast tissues and 6 normal breast from reductive mammoplasty by quantitative methylation specific PCR (QMSP). Aberrant promoter methylation was detected in 52 out of the 102 primary breast cancer cases (51%) and 10 out of 14 pre-invasive lesions (71%). No mutations of the KEAP1 gene were identified in the 20 breast cancer cases analyzed by fluorescence based direct sequencing. Methylation was more frequent in the subgroup of patients identified as ER positive-HER2 negative tumors (66.7%) as compared with triple-negative breast cancers (35%) (p = 0.05, Chi-square test). The impact of the interactions between Er, PgR, Her2 expression and KEAP1 methylation on mortality was investigated by RECPAM multivariable statistical analysis, identifying four prognostic classes at different mortality risks. Triple-negative breast cancer patients with KEAP1 methylation had higher mortality risk than patients without triple-negative breast cancer (HR = 14.73, 95%CI: 3.65–59.37). Both univariable and multivariable COX regressions analyses showed that KEAP1 methylation was associated with a better progression free survival in patients treated with epirubicin/cyclophosfamide and docetaxel as sequential chemotherapy (HR = 0.082; 95%CI: 0.007–0.934). These results indicate that aberrant promoter methylation of the KEAP1 gene is involved in breast cancerogenesis. In addition, identifying patients with KEAP1 epigenetic abnormalities may contribute to disease progression prediction in breast cancer patients.


Lung Cancer | 2010

Molecular analysis of the HuD gene in neuroendocrine lung cancers

Vito D'Alessandro; Lucia Anna Muscarella; Annamaria la Torre; Michele Bisceglia; Paola Parrella; Gerardo Scaramuzzi; Clelia Tiziana Storlazzi; Domenico Trombetta; Klaas Kok; Angelo De Cata; Marco Sperandeo; Leopoldo Zelante; Massimo Carella; Gianluigi Vendemiale

n-ELAV (neuronal-Embryonic Lethal, Abnormal Vision)-like genes belong to a family codifying for onconeural RNA-binding proteins, also called Hu antigens. Anti-Hu-antibodies (anti-Hu-Ab) are typically associated with paraneoplastic encephalomyelitis/sensory neuropathy (PEM/PSN), and low titres of anti-Hu-Ab were found in neural/neuroendocrine neoplasms, especially small cell lung cancer (SCLC). To date, few studies have been published focused on the genetic causes of their involvement in the pathogenesis of neuroendocrine tumors (NE). Here we analyzed 20 primary human neuroendocrine lung tumor tissues for somatic mutations in the HuD gene. Two inactivating mutations (a frameshift and a stop codon mutation) and 11 nucleotide changes were detected in the coding sequence of HuD gene in 7 different lung tumors. Our results on SCLC and carcinoid tissues support the hypothesis that alterations of nELAV genes could be involved in the onset and/or progression of a subset of neuroendocrine lung tumors.


Cellular Oncology | 2011

Gene expression of somatostatin receptor subtypes SSTR2a, SSTR3 and SSTR5 in peripheral blood of neuroendocrine lung cancer affected patients

Lucia Anna Muscarella; Vito D’Alessandro; Annamaria la Torre; Massimiliano Copetti; Angelo De Cata; Paola Parrella; Marco Sperandeo; Fabio Pellegrini; Vincenzo Frusciante; Evaristo Maiello; Giuseppe Merla; Vito Michele Fazio; Gianluigi Vendemiale

BackgroundSomatostatin (SS) acts as a universal endocrine off-switch, and also inhibits the growth of neuroendocrine tumours through its specific receptors (SSTRs). Somatostatin receptors are G-protein-coupled receptors, which are encoded by five separate genes (SSTR1-5). Short peptide analogues demonstrate specific binding only for the subgroup consisting of SSTR2a, SSTR3 and SSTR5. Moreover, previous studies reported that expression of mRNA for SSTR2a correlated with therapeutic outcome in patients with carcinoid tumours treated with somatostatin analogs.PurposeTo develop and apply a Real Time Quantitative PCR technique (RT-qPCR) to compare and contrast the mRNA levels of SSTR2a, SSTR3 and SSTR5 in Neuroendocrine Lung Cancer affected patients.MethodsPeripheral blood samples from 21 neuroendocrine lung cancer affected patients (14 SCLC, 6 LC and 1 LCNEC) subjected to scintigraphy with 111In-DTPA-D-Phe1-octreotide (OctreoScan) and 24 healthy blood donors were investigated by RT-qPCR. mRNA levels for SSTR2a, SSTR3 and SSTR5 were measured in peripheral blood samples with a relative quantification method using plasmid dilutions as calibration curves and GAPDH as reference gene.ResultsA statistically significant increase in target genes/GAPDH copy number ratio was found for SSTR2a (median 38; IQR 22–141) and SSTR5 (median 51; IQR 19–499) in neuroendocrine lung cancer affected patients as compared with samples from healthy blood donors (P ≤ 0.0003 and P ≤ 0.0005). Since low levels of expression were detected in the control group for all three genes, optimal cut-off values were assessed using ROC curve analyses and were equal to 9.05 for SSTR2a and 16.97 for SSTR5. These cut off values resulted in a sensitivity of 86% (95%IC 65–95) for both markers and a specificity of 83% (95%IC 64–93%) and 79% (95%IC 60–91%) for SSTR2a and SSTR5 respectively. Comparison between OctreoScan results and RT-qPCR analysis demonstrated agreement in 76% of the cases.ConclusionsOur results suggest that SSTR2a and SSTR5 mRNAs are detectable in peripheral blood of neuroendocrine lung cancer affected patients using real-time quantitative PCR, with a good agreement with OctreoScan. The high sensitivity of this non-invasive molecular technique suggests that this method could represent a useful tool in the clinical management of neuroendocrine lung cancers.


Oncotarget | 2017

Keap1/Nrf2 pathway in kidney cancer: frequent methylation of KEAP1 gene promoter in clear renal cell carcinoma

Federico Pio Fabrizio; Manuela Costantini; Massimiliano Copetti; Annamaria la Torre; Angelo Sparaneo; Andrea Fontana; Luana Poeta; Michele Gallucci; Steno Sentinelli; Paolo Graziano; Paola Parente; Vincenzo Pompeo; Laura De Salvo; Giuseppe Simone; Rocco Papalia; Francesco Picardo; Teresa Balsamo; Gerardo Flammia; Domenico Trombetta; Angela Pantalone; Klaas Kok; Ferronika Paranita; Lucia Anna Muscarella; Vito Michele Fazio

The Keap1/Nrf2 pathway is a master regulator of the cellular redox state through the induction of several antioxidant defence genes implicated in chemotherapeutic drugs resistance of tumor cells. An increasing body of evidence supports a key role for Keap1/Nrf2 pathway in kidney diseases and renal cell carcinoma (RCC), but data concerning the molecular basis and the clinical effect of its deregulation remain incomplete. Here we present a molecular profiling of the KEAP1 and NFE2L2 genes in five different Renal Cell Carcinoma histotypes by analysing 89 tumor/normal paired tissues (clear cell Renal Carcinoma, ccRCCs; Oncocytomas; Papillary Renal Cell Carcinoma Type 1, PRCC1; Papillary Renal Cell Carcinoma Type 2, PRCC2; and Chromophobe Cell Carcinoma). A tumor-specific DNA methylation of the KEAP1 gene promoter region was found as a specific feature of the ccRCC subtype (18/37, 48.6%) and a direct correlation with mRNA levels was confirmed by in vitro 5-azacytidine treatment. Analysis of an independent data set of 481 ccRCC and 265 PRCC tumors corroborates our results and multivariate analysis reveals a significant correlation among ccRCCs epigenetic KEAP1 silencing and staging, grading and overall survival. Our molecular results show for the the first time the epigenetic silencing of KEAP1 promoter as the leading mechanism for modulation of KEAP1 expression in ccRCCs and corroborate the driver role of Keap1/Nrf2 axis deregulation with potential new function as independent epigenetic prognostic marker in renal cell carcinoma.


Journal of Neuropathology and Experimental Neurology | 2014

Molecular Dissection of the VHL Gene in Solitary Capillary Hemangioblastoma of the Central Nervous System

Lucia Anna Muscarella; Annamaria la Torre; Augusta Faienza; Demenico Catapano; Michele Bisceglia; Vincenzo D'Angelo; Paola Parrella; Michelina Coco; Grazia Fini; Angelo Tancredi; Leopoldo Zelante; Vito Michele Fazio; Leonardo D'Agruma

Capillary hemangioblastomas (HGBs) of the CNS occur either sporadically or as part of the von Hippel-Lindau (VHL) syndrome. Molecular characterizations of the VHL gene in sporadic HGBs at the somatic level have been limited to date. We investigated the VHL gene in 57 patients most of whom (55 [96%] of 57) had a solitary CNS HGB at the time of surgery. Tissues from 23 HGBs of these patients (2 VHL related and 21 unrelated) were also investigated at genetic and epigenetic levels. Two of the 51 patients with apparently sporadic HGBs and no additional evidence of VHL (∼4%) were found to have a germline VHL gene mutation; both of these patients subsequently developed evidence of VHL syndrome. Somatic VHL gene mutations were found in 11 (52%) of the 21 non-VHL-related cases. A germline mutation was identified in 5 (84%) of 6 VHL-associated HGBs; double gene inactivation was observed in tumor tissue from VHL syndrome patients. Seven different previously unreported VHL gene alterations (6 somatic and 1 germline) were identified; double hits were identified in 7 (12%) of 57 cases. Our findings confirm the usefulness of VHL gene analysis at the germline level in patients who present with apparently solitary HGB. Moreover, the genetic and epigenetic VHL gene investigations performed support a key role for functional alterations of the VHL gene in sporadic neuraxial HGB.


PLOS ONE | 2013

Identification and Functional Characterization of Three NoLS (Nucleolar Localisation Signals) Mutations of the CDC73 Gene

Valerio Pazienza; Annamaria la Torre; Filomena Baorda; Michela Alfarano; Massimiliano Chetta; Lucia Anna Muscarella; Claudia Battista; Massimiliano Copetti; Dieter Kotzot; Klaus Kapelari; Dalia Al-Abdulrazzaq; Kusiel Perlman; Etienne Sochett; David E. C. Cole; Fabio Pellegrini; Lucie Canaff; Geoffrey N. Hendy; Leonardo D’Agruma; Leopoldo Zelante; Massimo Carella; Alfredo Scillitani; Vito Guarnieri

Hyperparathyroidism Jaw-Tumour Syndrome (HPT-JT) is characterized by primary hyperparathyroidism (PHPT), maxillary/mandible ossifying fibromas and by parathyroid carcinoma in 15% of cases. Inactivating mutations of the tumour suppressor CDC73/HRPT2 gene have been found in HPT-JT patients and also as genetic determinants of sporadic parathyroid carcinoma/atypical adenomas and, rarely, typical adenomas, in familial PHPT. Here we report the genetic and molecular analysis of the CDC73/HRPT2 gene in three patients affected by PHPT due to atypical and typical parathyroid adenomas, in one case belonging to familial PHPT. Flag-tagged WT and mutant CDC73/HRPT2 proteins were transiently transfected in HEK293 cells and functional assays were performed in order to investigate the effect of the variants on the whole protein expression, nuclear localization and cell overgrowth induction. We identified four CDC73/HRPT2 gene mutations, three germline (c.679_680delAG, p.Val85_Val86del and p.Glu81_Pro84del), one somatic (p.Arg77Pro). In three cases the mutation was located within the Nucleolar Localisation Signals (NoLS). The three NoLS variants led to instability either of the corresponding mutated protein or mRNA or both. When transfected in HEK293 cells, NoLS mutated proteins mislocalized with a predeliction for cytoplasmic or nucleo-cytoplasmic localization and, finally, they resulted in overgrowth, consistent with a dominant negative interfering effect in the presence of the endogenous protein.


Advances in Anatomic Pathology | 2017

Extraneuraxial Hemangioblastoma: Clinicopathologic Features and Review of the Literature

Michele Bisceglia; Lucia Anna Muscarella; Carlos A. Galliani; Nina Zidar; David Ben-Dor; Gianandrea Pasquinelli; Annamaria la Torre; Angelo Sparaneo; Julie C. Fanburg-Smith; Janez Lamovec; Michal Michal; Carlos E. Bacchi

Extraneuraxial hemangioblastoma occurs in nervous paraneuraxial structures, somatic tissues, and visceral organs, as part of von Hippel-Lindau disease (VHLD) or in sporadic cases. The VHL gene plausibly plays a key role in the initiation and tumorigenesis of both central nervous system and extraneuraxial hemangioblastoma, therefore, the underlying molecular and genetic mechanisms of the tumor growth are initially reviewed. The clinical criteria for the diagnosis of VHLD are summarized, with emphasis on the distinction of sporadic hemangioblastoma from the form fruste of VHLD (eg, hemangioblastoma-only VHLD). The world literature on the topic of extraneuraxial hemangioblastomas has been comprehensively reviewed with ∼200 cases reported to date: up to 140 paraneuraxial, mostly of proximal spinal nerve roots, and 65 peripheral, 15 of soft tissue, 6 peripheral nerve, 5 bone, and 39 of internal viscera, including 26 renal and 13 nonrenal. A handful of possible yet uncertain cases from older literature are not included in this review. The clinicopathologic features of extraneuraxial hemangioblastoma are selectively presented by anatomic site of origin, and the differential diagnosis is emphasized in these subsets. Reference is made also to 10 of the authors’ personal cases of extraneuraxial hemangioblastomas, which include 4 paraneuraxial and 6 peripheral (2 soft tissue hemangioblastoma and 4 renal).


Oncotarget | 2018

Large deletion at the CDC73 gene locus and search for predictive markers of the presence of a CDC73 genetic lesion

Lucia Anna Muscarella; Daniela Turchetti; Andrea Fontana; Filomena Baorda; Orazio Palumbo; Annamaria la Torre; Danilo de Martino; Renato Franco; Nunzia Simona Losito; Andrea Repaci; Uberto Pagotto; Luigia Cinque; Massimiliano Copetti; Maria Grazia Chiofalo; Luciano Pezzullo; Paolo Graziano; Alfredo Scillitani; Vito Guarnieri

The Hyperparathyroidism with Jaw-Tumours syndrome is caused by mutations of the CDC73 gene: it has been suggested that early onset of the disease and high Ca2+ levels may predict the presence of a CDC73 mutation. We searched for large deletions at the CDC73 locus in patients with: HPT-JT (nr 2), atypical adenoma (nr 7) or sporadic parathyroid carcinoma (nr 11) with a specific MLPA and qRT-PCR assays applied on DNA extracted from whole blood. A Medline search in database for all the papers reporting a CDC73 gene mutation, clinical/histological diagnosis, age at onset, Ca2+, PTH levels for familial/sporadic cases was conducted with the aim to possibly identify biochemical/clinical markers predictive, in first diagnosis, of the presence of a CDC73 gene mutation. A novel genomic deletion of the first 10 exons of the CDC73 gene was found in a 3-generation HPT-JT family, confirmed by SNP array analysis. A classification tree built on the published data, showed the highest probability of having a CDC73 mutation in subjects with age at the onset < 41.5 years (44/47 subjects, 93.6%, had the mutation). Whereas the lowest probability was found in subjects with age at the onset ≥ 41.5 years and Ca2+ levels <13.96 mg/dL (7/20 subjects, 35.0%, had the mutation, odds ratio = 27.1, p < 0.001). We report a novel large genomic CDC73 gene deletion identified in an Italian HPT-JT family. Age at onset < 41.5 ys and Ca2+ > 13.96 mg/dL are predictive for the presence of a CDC73 genetic lesion.

Collaboration


Dive into the Annamaria la Torre's collaboration.

Top Co-Authors

Avatar

Lucia Anna Muscarella

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Vito Michele Fazio

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Paola Parrella

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Teresa Balsamo

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Evaristo Maiello

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Massimiliano Copetti

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Raffaela Barbano

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michele Bisceglia

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Michelina Coco

Casa Sollievo della Sofferenza

View shared research outputs
Researchain Logo
Decentralizing Knowledge