Network


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

Hotspot


Dive into the research topics where Salvatore Piscuoglio is active.

Publication


Featured researches published by Salvatore Piscuoglio.


Annals of Oncology | 2014

Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle

L. De Mattos-Arruda; Britta Weigelt; Javier Cortes; Helen H. Won; Charlotte K.Y. Ng; Paolo Nuciforo; François-Clément Bidard; Claudia Aura; Cristina Saura; Vicente Peg; Salvatore Piscuoglio; Mafalda Oliveira; Y. Smolders; P. Patel; Larry Norton; Josep Tabernero; Michael F. Berger; Joan Seoane; Jorge S. Reis-Filho

BACKGROUND Plasma-derived cell-free tumor DNA (ctDNA) constitutes a potential surrogate for tumor DNA obtained from tissue biopsies. We posit that massively parallel sequencing (MPS) analysis of ctDNA may help define the repertoire of mutations in breast cancer and monitor tumor somatic alterations during the course of targeted therapy. PATIENT AND METHODS A 66-year-old patient presented with synchronous estrogen receptor-positive/HER2-negative, highly proliferative, grade 2, mixed invasive ductal-lobular carcinoma with bone and liver metastases at diagnosis. DNA extracted from archival tumor material, plasma and peripheral blood leukocytes was subjected to targeted MPS using a platform comprising 300 cancer genes known to harbor actionable mutations. Multiple plasma samples were collected during the fourth line of treatment with an AKT inhibitor. RESULTS Average read depths of 287x were obtained from the archival primary tumor, 139x from the liver metastasis and between 200x and 900x from ctDNA samples. Sixteen somatic non-synonymous mutations were detected in the liver metastasis, of which 9 (CDKN2A, AKT1, TP53, JAK3, TSC1, NF1, CDH1, MML3 and CTNNB1) were also detected in >5% of the alleles found in the primary tumor sample. Not all mutations identified in the metastasis were reliably identified in the primary tumor (e.g. FLT4). Analysis of ctDNA, nevertheless, captured all mutations present in the primary tumor and/or liver metastasis. In the longitudinal monitoring of the patient, the mutant allele fractions identified in ctDNA samples varied over time and mirrored the pharmacodynamic response to the targeted therapy as assessed by positron emission tomography-computed tomography. CONCLUSIONS This proof-of-principle study is one of the first to demonstrate that high-depth targeted MPS of plasma-derived ctDNA constitutes a potential tool for de novo mutation identification and monitoring of somatic genetic alterations during the course of targeted therapy, and may be employed to overcome the challenges posed by intra-tumor genetic heterogeneity. REGISTERED CLINICAL TRIAL: www.clinicaltrials.gov, NCT01090960.BACKGROUND Plasma-derived cell-free tumor DNA (ctDNA) constitutes a potential surrogate for tumor DNA obtained from tissue biopsies. We posit that massively parallel sequencing (MPS) analysis of ctDNA may help define the repertoire of mutations in breast cancer and monitor tumor somatic alterations during the course of targeted therapy. PATIENT AND METHODS A 66-year-old patient presented with synchronous estrogen receptor-positive/HER2-negative, highly proliferative, grade 2, mixed invasive ductal-lobular carcinoma with bone and liver metastases at diagnosis. DNA extracted from archival tumor material, plasma and peripheral blood leukocytes was subjected to targeted MPS using a platform comprising 300 cancer genes known to harbor actionable mutations. Multiple plasma samples were collected during the fourth line of treatment with an AKT inhibitor. RESULTS Average read depths of 287x were obtained from the archival primary tumor, 139x from the liver metastasis and between 200x and 900x from ctDNA samples. Sixteen somatic non-synonymous mutations were detected in the liver metastasis, of which 9 (CDKN2A, AKT1, TP53, JAK3, TSC1, NF1, CDH1, MML3 and CTNNB1) were also detected in >5% of the alleles found in the primary tumor sample. Not all mutations identified in the metastasis were reliably identified in the primary tumor (e.g. FLT4). Analysis of ctDNA, nevertheless, captured all mutations present in the primary tumor and/or liver metastasis. In the longitudinal monitoring of the patient, the mutant allele fractions identified in ctDNA samples varied over time and mirrored the pharmacodynamic response to the targeted therapy as assessed by positron emission tomography-computed tomography. CONCLUSIONS This proof-of-principle study is one of the first to demonstrate that high-depth targeted MPS of plasma-derived ctDNA constitutes a potential tool for de novo mutation identification and monitoring of somatic genetic alterations during the course of targeted therapy, and may be employed to overcome the challenges posed by intra-tumor genetic heterogeneity. REGISTERED CLINICAL TRIAL www.clinicaltrials.gov, NCT01090960.


Nature Communications | 2015

Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma

Leticia De Mattos-Arruda; Regina Mayor; Charlotte K.Y. Ng; Britta Weigelt; Francisco Martinez-Ricarte; D. Torrejon; Mafalda Oliveira; Alexandra Arias; Carolina Raventós; Jiabin Tang; Elena Guerini-Rocco; Elena Martinez-Saez; Sergio Lois; Oscar Marín; Xavier de la Cruz; Salvatore Piscuoglio; Russel Towers; Ana Vivancos; Vicente Peg; Santiago Ramón y Cajal; Joan Carles; Jordi Rodon; María González-Cao; Josep Tabernero; Enriqueta Felip; Joan Sahuquillo; Michael F. Berger; Javier Cortes; Jorge S. Reis-Filho; Joan Seoane

Cell-free circulating tumour DNA (ctDNA) in plasma has been shown to be informative of the genomic alterations present in tumours and has been used to monitor tumour progression and response to treatments. However, patients with brain tumours do not present with or present with low amounts of ctDNA in plasma precluding the genomic characterization of brain cancer through plasma ctDNA. Here we show that ctDNA derived from central nervous system tumours is more abundantly present in the cerebrospinal fluid (CSF) than in plasma. Massively parallel sequencing of CSF ctDNA more comprehensively characterizes the genomic alterations of brain tumours than plasma, allowing the identification of actionable brain tumour somatic mutations. We show that CSF ctDNA levels longitudinally fluctuate in time and follow the changes in brain tumour burden providing biomarkers to monitor brain malignancies. Moreover, CSF ctDNA is shown to facilitate and complement the diagnosis of leptomeningeal carcinomatosis.


Breast Cancer Research | 2014

Breast cancer intra-tumor heterogeneity

Luciano G. Martelotto; Charlotte K.Y. Ng; Salvatore Piscuoglio; Britta Weigelt; Jorge S. Reis-Filho

In recent years it has become clear that cancer cells within a single tumor can display striking morphological, genetic and behavioral variability. Burgeoning genetic, epigenetic and phenomenological data support the existence of intra-tumor genetic heterogeneity in breast cancers; however, its basis is yet to be fully defined. Two of the most widely evoked concepts to explain the origin of heterogeneity within tumors are the cancer stem cell hypothesis and the clonal evolution model. Although the cancer stem cell model appeared to provide an explanation for the variability among the neoplastic cells within a given cancer, advances in massively parallel sequencing have provided several lines of evidence to suggest that intra-tumor genetic heterogeneity likely plays a fundamental role in the phenotypic heterogeneity observed in cancers. Many challenges remain, however, in the interpretation of the next generation sequencing results obtained so far. Here we review the models that explain tumor heterogeneity, the causes of intra-tumor genetic diversity and their impact on our understanding and management of breast cancer, methods to study intra-tumor heterogeneity and the assessment of intra-tumor genetic heterogeneity in the clinic.


Nature Genetics | 2014

Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands

Ilan Weinreb; Salvatore Piscuoglio; Luciano G. Martelotto; Daryl Waggott; Charlotte K.Y. Ng; Bayardo Perez-Ordonez; Nicholas J. Harding; Javier A. Alfaro; Kenneth C. Chu; Agnes Viale; Nicola Fusco; Arnaud Da Cruz Paula; Caterina Marchiò; Rita A. Sakr; Raymond S. Lim; Lester D R Thompson; Simion I. Chiosea; Raja R. Seethala; Alena Skalova; Edward B. Stelow; Isabel Fonseca; Adel Assaad; Christine How; Jianxin Wang; Richard de Borja; Michelle Chan-Seng-Yue; Christopher J. Howlett; Anthony C. Nichols; Y Hannah Wen; Nora Katabi

Polymorphous low-grade adenocarcinoma (PLGA) is the second most frequent type of malignant tumor of the minor salivary glands. We identified PRKD1 hotspot mutations encoding p.Glu710Asp in 72.9% of PLGAs but not in other salivary gland tumors. Functional studies demonstrated that this kinase-activating alteration likely constitutes a driver of PLGA.


Genome Biology | 2014

Benchmarking mutation effect prediction algorithms using functionally validated cancer-related missense mutations

Luciano G. Martelotto; Charlotte K.Y. Ng; Maria Rosaria De Filippo; Yan Zhang; Salvatore Piscuoglio; Raymond S. Lim; Ronglai Shen; Larry Norton; Jorge S. Reis-Filho; Britta Weigelt

BackgroundMassively parallel sequencing studies have led to the identification of a large number of mutations present in a minority of cancers of a given site. Hence, methods to identify the likely pathogenic mutations that are worth exploring experimentally and clinically are required. We sought to compare the performance of 15 mutation effect prediction algorithms and their agreement. As a hypothesis-generating aim, we sought to define whether combinations of prediction algorithms would improve the functional effect predictions of specific mutations.ResultsLiterature and database mining of single nucleotide variants (SNVs) affecting 15 cancer genes was performed to identify mutations supported by functional evidence or hereditary disease association to be classified either as non-neutral (n = 849) or neutral (n = 140) with respect to their impact on protein function. These SNVs were employed to test the performance of 15 mutation effect prediction algorithms. The accuracy of the prediction algorithms varies considerably. Although all algorithms perform consistently well in terms of positive predictive value, their negative predictive value varies substantially. Cancer-specific mutation effect predictors display no-to-almost perfect agreement in their predictions of these SNVs, whereas the non-cancer-specific predictors showed no-to-moderate agreement. Combinations of predictors modestly improve accuracy and significantly improve negative predictive values.ConclusionsThe information provided by mutation effect predictors is not equivalent. No algorithm is able to predict sufficiently accurately SNVs that should be taken forward for experimental or clinical testing. Combining algorithms aggregates orthogonal information and may result in improvements in the negative predictive value of mutation effect predictions.


The Journal of Pathology | 2014

Characterization of the genomic features and expressed fusion genes in micropapillary carcinomas of the breast

Rachael Natrajan; Paul M. Wilkerson; Caterina Marchiò; Salvatore Piscuoglio; Charlotte K.Y. Ng; Patty Wai; Maryou B. Lambros; Eleftherios P. Samartzis; Konstantin J. Dedes; Jessica Frankum; Ilirjana Bajrami; Alicja Kopec; Alan Mackay; Roger A'Hern; Kerry Fenwick; Iwanka Kozarewa; Jarle Hakas; Costas Mitsopoulos; David Hardisson; Christopher J. Lord; Chandan Kumar-Sinha; Alan Ashworth; Britta Weigelt; Anna Sapino; Arul M. Chinnaiyan; Christopher A. Maher; Jorge S. Reis-Filho

Micropapillary carcinoma (MPC) is a rare histological special type of breast cancer, characterized by an aggressive clinical behaviour and a pattern of copy number aberrations (CNAs) distinct from that of grade‐ and oestrogen receptor (ER)‐matched invasive carcinomas of no special type (IC‐NSTs). The aims of this study were to determine whether MPCs are underpinned by a recurrent fusion gene(s) or mutations in 273 genes recurrently mutated in breast cancer. Sixteen MPCs were subjected to microarray‐based comparative genomic hybridization (aCGH) analysis and Sequenom OncoCarta mutation analysis. Eight and five MPCs were subjected to targeted capture and RNA sequencing, respectively. aCGH analysis confirmed our previous observations about the repertoire of CNAs of MPCs. Sequencing analysis revealed a spectrum of mutations similar to those of luminal B IC‐NSTs, and recurrent mutations affecting mitogen‐activated protein kinase family genes and NBPF10. RNA‐sequencing analysis identified 17 high‐confidence fusion genes, eight of which were validated and two of which were in‐frame. No recurrent fusions were identified in an independent series of MPCs and IC‐NSTs. Forced expression of in‐frame fusion genes (SLC2A1–FAF1 and BCAS4–AURKA) resulted in increased viability of breast cancer cells. In addition, genomic disruption of CDK12 caused by out‐of‐frame rearrangements was found in one MPC and in 13% of HER2‐positive breast cancers, identified through a re‐analysis of publicly available massively parallel sequencing data. In vitro analyses revealed that CDK12 gene disruption results in sensitivity to PARP inhibition, and forced expression of wild‐type CDK12 in a CDK12‐null cell line model resulted in relative resistance to PARP inhibition. Our findings demonstrate that MPCs are neither defined by highly recurrent mutations in the 273 genes tested, nor underpinned by a recurrent fusion gene. Although seemingly private genetic events, some of the fusion transcripts found in MPCs may play a role in maintenance of a malignant phenotype and potentially offer therapeutic opportunities.


The Journal of Pathology | 2015

Genomic landscape of adenoid cystic carcinoma of the breast.

Luciano G. Martelotto; Maria Rosaria De Filippo; Charlotte K.Y. Ng; Rachael Natrajan; Laetitia Fuhrmann; Joanna Cyrta; Salvatore Piscuoglio; Huei-Chi Wen; Raymond S. Lim; Ronglai Shen; Anne M. Schultheis; Y Hannah Wen; Marcia Edelweiss; Odette Mariani; Göran Stenman; Timothy A. Chan; Pierre-Emmanuel Colombo; Larry Norton; Anne Vincent-Salomon; Jorge S. Reis-Filho; Britta Weigelt

Adenoid cystic carcinoma (AdCC) is a rare type of triple‐negative breast cancer (TNBC) characterized by the presence of the MYB–NFIB fusion gene. The molecular underpinning of breast AdCCs other than the MYB–NFIB fusion gene remains largely unexplored. Here we sought to define the repertoire of somatic genetic alterations of breast AdCCs. We performed whole‐exome sequencing, followed by orthogonal validation, of 12 breast AdCCs to determine the landscape of somatic mutations and gene copy number alterations. Fluorescence in situ hybridization and reverse‐transcription PCR were used to define the presence of MYB gene rearrangements and MYB–NFIB chimeric transcripts. Unlike common forms of TNBC, we found that AdCCs have a low mutation rate (0.27 non‐silent mutations/Mb), lack mutations in TP53 and PIK3CA and display a heterogeneous constellation of known cancer genes affected by somatic mutations, including MYB, BRAF, FBXW7, SMARCA5, SF3B1 and FGFR2. MYB and TLN2 were affected by somatic mutations in two cases each. Akin to salivary gland AdCCs, breast AdCCs were found to harbour mutations targeting chromatin remodelling, cell adhesion, RNA biology, ubiquitination and canonical signalling pathway genes. We observed that, although breast AdCCs had rather simple genomes, they likely display intra‐tumour genetic heterogeneity at diagnosis. Taken together, these findings demonstrate that the mutational burden and mutational repertoire of breast AdCCs are more similar to those of salivary gland AdCCs than to those of other types of TNBCs, emphasizing the importance of histological subtyping of TNBCs. Furthermore, our data provide direct evidence that AdCCs harbour a distinctive mutational landscape and genomic structure, irrespective of the disease site of origin. Copyright


Histopathology | 2012

HMGA1 and HMGA2 protein expression correlates with advanced tumour grade and lymph node metastasis in pancreatic adenocarcinoma

Salvatore Piscuoglio; Inti Zlobec; Pierlorenzo Pallante; Romina Sepe; Arthur Zimmermann; Ioannis Diamantis; Luigi Terracciano; Alfredo Fusco; Eva Karamitopoulou

Piscuoglio S, Zlobec I, Pallante P, Sepe R, Esposito F, Zimmermann A, Diamantis I, Terracciano L, Fusco A & Karamitopoulou E 
(2012) Histopathology 60, 397–404 
HMGA1 and HMGA2 protein expression correlates with advanced tumour grade and lymph node metastasis in pancreatic adenocarcinoma


Genome Biology | 2015

Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification

Charlotte K.Y. Ng; Luciano G. Martelotto; Arnaud Gauthier; Huei-Chi Wen; Salvatore Piscuoglio; Raymond S. Lim; Catherine F. Cowell; Paul M. Wilkerson; Patty Wai; Daniel Nava Rodrigues; Laurent Arnould; Felipe C. Geyer; Silvio E Bromberg; Magali Lacroix-Triki; Frédérique Penault-Llorca; Sylvia Giard; Xavier Sastre-Garau; Rachael Natrajan; Larry Norton; Paul Cottu; Britta Weigelt; Anne Vincent-Salomon; Jorge S. Reis-Filho

BackgroundHER2 is overexpressed and amplified in approximately 15% of invasive breast cancers, and is the molecular target and predictive marker of response to anti-HER2 agents. In a subset of these cases, heterogeneous distribution of HER2 gene amplification can be found, which creates clinically challenging scenarios. Currently, breast cancers with HER2 amplification/overexpression in just over 10% of cancer cells are considered HER2-positive for clinical purposes; however, it is unclear as to whether the HER2-negative components of such tumors would be driven by distinct genetic alterations. Here we sought to characterize the pathologic and genetic features of the HER2-positive and HER2-negative components of breast cancers with heterogeneous HER2 gene amplification and to define the repertoire of potential driver genetic alterations in the HER2-negative components of these cases.ResultsWe separately analyzed the HER2-negative and HER2-positive components of 12 HER2 heterogeneous breast cancers using gene copy number profiling and massively parallel sequencing, and identified potential driver genetic alterations restricted to the HER2-negative cells in each case. In vitro experiments provided functional evidence to suggest that BRF2 and DSN1 overexpression/amplification, and the HER2 I767M mutation may be alterations that compensate for the lack of HER2 amplification in the HER2-negative components of HER2 heterogeneous breast cancers.ConclusionsOur results indicate that even driver genetic alterations, such as HER2 gene amplification, can be heterogeneously distributed within a cancer, and that the HER2-negative components are likely driven by genetic alterations not present in the HER2-positive components, including BRF2 and DSN1 amplification and HER2 somatic mutations.


Journal of the National Cancer Institute | 2015

Massively Parallel Sequencing-Based Clonality Analysis of Synchronous Endometrioid Endometrial and Ovarian Carcinomas

Anne M. Schultheis; Charlotte K.Y. Ng; Maria Rosaria De Filippo; Salvatore Piscuoglio; Gabriel S. Macedo; Sonia Gatius; Belen Perez Mies; Robert A. Soslow; Raymond S. Lim; Agnes Viale; Kety Huberman; Jose C. Palacios; Jorge S. Reis-Filho; Xavier Matias-Guiu; Britta Weigelt

Synchronous early-stage endometrioid endometrial carcinomas (EECs) and endometrioid ovarian carcinomas (EOCs) are associated with a favorable prognosis and have been suggested to represent independent primary tumors rather than metastatic disease. We subjected sporadic synchronous EECs/EOCs from five patients to whole-exome massively parallel sequencing, which revealed that the EEC and EOC of each case displayed strikingly similar repertoires of somatic mutations and gene copy number alterations. Despite the presence of mutations restricted to the EEC or EOC in each case, we observed that the mutational processes that shaped their respective genomes were consistent. High-depth targeted massively parallel sequencing of sporadic synchronous EECs/EOCs from 17 additional patients confirmed that these lesions are clonally related. In an additional Lynch Syndrome case, however, the EEC and EOC were found to constitute independent cancers lacking somatic mutations in common. Taken together, sporadic synchronous EECs/EOCs are clonally related and likely constitute dissemination from one site to the other.

Collaboration


Dive into the Salvatore Piscuoglio's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Britta Weigelt

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge S. Reis-Filho

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Luciano G. Martelotto

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Felipe C. Geyer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Raymond S. Lim

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kathleen A. Burke

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge