Network


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

Hotspot


Dive into the research topics where Valentina Gianfelici is active.

Publication


Featured researches published by Valentina Gianfelici.


Nature Genetics | 2013

Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia

Kim De Keersmaecker; Zeynep Kalender Atak; Ning Li; Carmen Vicente; Stephanie Patchett; Tiziana Girardi; Valentina Gianfelici; Ellen Geerdens; Emmanuelle Clappier; Michaël Porcu; Idoya Lahortiga; Rossella Luca; Jiekun Yan; Gert Hulselmans; Hilde Vranckx; Roel Vandepoel; Bram Sweron; Kris Jacobs; Nicole Mentens; Iwona Wlodarska; Barbara Cauwelier; Jacqueline Cloos; Jean Soulier; Anne Uyttebroeck; Claudia Bagni; Bassem A. Hassan; Peter Vandenberghe; Arlen W. Johnson; Stein Aerts; Jan Cools

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


PLOS Genetics | 2013

Comprehensive analysis of transcriptome variation uncovers known and novel driver events in T-cell acute lymphoblastic leukemia

Zeynep Kalender Atak; Valentina Gianfelici; Gert Hulselmans; Kim De Keersmaecker; Arun George Devasia; Ellen Geerdens; Nicole Mentens; Sabina Chiaretti; Kaat Durinck; Anne Uyttebroeck; Peter Vandenberghe; Iwona Wlodarska; Jacqueline Cloos; Robin Foà; Franki Speleman; Jan Cools; Stein Aerts

RNA-seq is a promising technology to re-sequence protein coding genes for the identification of single nucleotide variants (SNV), while simultaneously obtaining information on structural variations and gene expression perturbations. We asked whether RNA-seq is suitable for the detection of driver mutations in T-cell acute lymphoblastic leukemia (T-ALL). These leukemias are caused by a combination of gene fusions, over-expression of transcription factors and cooperative point mutations in oncogenes and tumor suppressor genes. We analyzed 31 T-ALL patient samples and 18 T-ALL cell lines by high-coverage paired-end RNA-seq. First, we optimized the detection of SNVs in RNA-seq data by comparing the results with exome re-sequencing data. We identified known driver genes with recurrent protein altering variations, as well as several new candidates including H3F3A, PTK2B, and STAT5B. Next, we determined accurate gene expression levels from the RNA-seq data through normalizations and batch effect removal, and used these to classify patients into T-ALL subtypes. Finally, we detected gene fusions, of which several can explain the over-expression of key driver genes such as TLX1, PLAG1, LMO1, or NKX2-1; and others result in novel fusion transcripts encoding activated kinases (SSBP2-FER and TPM3-JAK2) or involving MLLT10. In conclusion, we present novel analysis pipelines for variant calling, variant filtering, and expression normalization on RNA-seq data, and successfully applied these for the detection of translocations, point mutations, INDELs, exon-skipping events, and expression perturbations in T-ALL.


Blood | 2014

JAK3 mutants transform hematopoietic cells through JAK1 activation, causing T-cell acute lymphoblastic leukemia in a mouse model

Sandrine Degryse; Charles E. de Bock; Luk Cox; Sofie Demeyer; Olga Gielen; Nicole Mentens; Kris Jacobs; Ellen Geerdens; Valentina Gianfelici; Gert Hulselmans; Mark Fiers; Stein Aerts; J Meijerink; Thomas Tousseyn; Jan Cools

JAK3 is a tyrosine kinase that associates with the common γ chain of cytokine receptors and is recurrently mutated in T-cell acute lymphoblastic leukemia (T-ALL). We tested the transforming properties of JAK3 pseudokinase and kinase domain mutants using in vitro and in vivo assays. Most, but not all, JAK3 mutants transformed cytokine-dependent Ba/F3 or MOHITO cell lines to cytokine-independent proliferation. JAK3 pseudokinase mutants were dependent on Jak1 kinase activity for cellular transformation, whereas the JAK3 kinase domain mutant could transform cells in a Jak1 kinase-independent manner. Reconstitution of the IL7 receptor signaling complex in 293T cells showed that JAK3 mutants required receptor binding to mediate downstream STAT5 phosphorylation. Mice transplanted with bone marrow progenitor cells expressing JAK3 mutants developed a long-latency transplantable T-ALL-like disease, characterized by an accumulation of immature CD8(+) T cells. In vivo treatment of leukemic mice with the JAK3 selective inhibitor tofacitinib reduced the white blood cell count and caused leukemic cell apoptosis. Our data show that JAK3 mutations are drivers of T-ALL and require the cytokine receptor complex for transformation. These results warrant further investigation of JAK1/JAK3 inhibitors for the treatment of T-ALL.


Blood | 2012

Mutation of the receptor tyrosine phosphatase PTPRC (CD45) in T-cell acute lymphoblastic leukemia

Michaël Porcu; Maria Kleppe; Valentina Gianfelici; Ellen Geerdens; Kim De Keersmaecker; Marco Tartaglia; Robin Foà; Jean Soulier; Barbara Cauwelier; Anne Uyttebroeck; Elizabeth Macintyre; Peter Vandenberghe; Vahid Asnafi; Jan Cools

The protein tyrosine phosphatase CD45, encoded by the PTPRC gene, is well known as a regulator of B- and T-cell receptor signaling. In addition, CD45 negatively regulates JAK family kinases downstream of cytokine receptors. Here, we report the presence of CD45 inactivating mutations in T-cell acute lymphoblastic leukemia. Loss-of-function mutations of CD45 were detected in combination with activating mutations in IL-7R, JAK1, or LCK, and down-regulation of CD45 expression caused increased signaling downstream of these oncoproteins. Furthermore, we demonstrate that down-regulation of CD45 expression sensitizes T cells to cytokine stimulation, as observed by increased JAK/STAT signaling, whereas overexpression of CD45 decreases cytokine-induced signaling. Taken together, our data identify a tumor suppressor role for CD45 in T-cell acute lymphoblastic leukemia.


PLOS ONE | 2012

High Accuracy Mutation Detection in Leukemia on a Selected Panel of Cancer Genes

Zeynep Kalender Atak; Kim De Keersmaecker; Valentina Gianfelici; Ellen Geerdens; Roel Vandepoel; Daphnie Pauwels; Michaël Porcu; Idoya Lahortiga; Vanessa Brys; Willy G. Dirks; Hilmar Quentmeier; Jacqueline Cloos; Harry Cuppens; Anne Uyttebroeck; Peter Vandenberghe; Jan Cools; Stein Aerts

With the advent of whole-genome and whole-exome sequencing, high-quality catalogs of recurrently mutated cancer genes are becoming available for many cancer types. Increasing access to sequencing technology, including bench-top sequencers, provide the opportunity to re-sequence a limited set of cancer genes across a patient cohort with limited processing time. Here, we re-sequenced a set of cancer genes in T-cell acute lymphoblastic leukemia (T-ALL) using Nimblegen sequence capture coupled with Roche/454 technology. First, we investigated how a maximal sensitivity and specificity of mutation detection can be achieved through a benchmark study. We tested nine combinations of different mapping and variant-calling methods, varied the variant calling parameters, and compared the predicted mutations with a large independent validation set obtained by capillary re-sequencing. We found that the combination of two mapping algorithms, namely BWA-SW and SSAHA2, coupled with the variant calling algorithm Atlas-SNP2 yields the highest sensitivity (95%) and the highest specificity (93%). Next, we applied this analysis pipeline to identify mutations in a set of 58 cancer genes, in a panel of 18 T-ALL cell lines and 15 T-ALL patient samples. We confirmed mutations in known T-ALL drivers, including PHF6, NF1, FBXW7, NOTCH1, KRAS, NRAS, PIK3CA, and PTEN. Interestingly, we also found mutations in several cancer genes that had not been linked to T-ALL before, including JAK3. Finally, we re-sequenced a small set of 39 candidate genes and identified recurrent mutations in TET1, SPRY3 and SPRY4. In conclusion, we established an optimized analysis pipeline for Roche/454 data that can be applied to accurately detect gene mutations in cancer, which led to the identification of several new candidate T-ALL driver mutations.


International Journal of Cancer | 2014

Identification of a novel, recurrent MBTD1-CXorf67 fusion in low-grade endometrial stromal sarcoma.

Barbara Dewaele; Joanna Przybyl; Anna Quattrone; Julio Finalet Ferreiro; Vanessa Vanspauwen; Ellen Geerdens; Valentina Gianfelici; Zeynep Kalender; Agnieszka Wozniak; Philippe Moerman; Raf Sciot; Sabrina Croce; Frédéric Amant; Peter Vandenberghe; Jan Cools; Maria Debiec-Rychter

Endometrial stromal sarcomas (ESSs) are a genetically heterogeneous group of rare uterine neoplasms that are commonly driven by recurrent gene rearrangements. In conventional low‐grade ESS, JAZF1‐SUZ12, PHF1‐JAZF1, EPC1‐PHF1 and MEAF6‐PHF1, and recently described ZC3H7‐BCOR chimeric fusions have been reported in > 50% of cases. Conversely, oncogenic t(10;17)(q22;p13) translocation yields YWHAE‐FAM22A/B chimeric proteins that are associated with histologically high‐grade and clinically more aggressive ESS. Integrating whole‐transcriptome paired‐end RNA sequencing with fluorescence in situ hybridization (FISH) and banding cytogenetics, we identified MBTD1 (malignant brain tumor domain‐containing 1) and CXorf67 (chromosome X open reading frame 67) as the genes involved in the novel reciprocal t(X;17)(p11.2;q21.33) translocation in two independent low‐grade ESS of classical histology. The presence of the MBTD1‐CXorf67 fusion transcript was validated in both cases using reverse‐transcription polymerase chain reaction followed by Sanger sequencing. A specific FISH assay was developed to detect the novel t(X;17) translocation in formalin‐fixed paraffin‐embedded material, and resulted in identification of an additional low‐grade ESS case positive for the MBTD1‐CXorf67 fusion among 25 uterine stromal tumors [14 ESS and 11 undifferentiated endometrial sarcomas (UESs)] that were negative for JAZF1 and YWHAE rearrangements. Gene expression profiles of seven ESS (including three with YWHAE and two with JAZF1 rearrangements) and four UES without specific chromosomal aberrations indicated clustering of tumors with MBTD1‐CXorf67 fusion together with low‐grade JAZF1‐associated ESS. The chimeric MBTD1‐CXorf67 fusion identifies yet another cytogenetically distinct subgroup of low‐grade ESS and offers the opportunity to shed light on the functions of two poorly characterized genes.


Haematologica | 2009

Late relapse of acute myeloid leukemia with mutated NPM1 after eight years: evidence of NPM1 mutation stability

Giovanna Meloni; Marco Mancini; Valentina Gianfelici; Maria Paola Martelli; Robin Foà; Brunangelo Falini

Late relapse (>5 years) of acute myeloid leukemia (AML) is rare.[1][1] Detecting at the time of late relapse the same genetic alteration as at diagnosis strongly suggests it may play a critical role in leukemogenesis. We previously reported in this journal that in AML with mutated nucleophosmin (


Haematologica | 2012

Activation of the NOTCH1 pathway in chronic lymphocytic leukemia

Valentina Gianfelici

Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults and is characterized by the monoclonal expansion of CD5+ B cells. CLL shows clinical heterogeneity, from patients with very stable disease to patients with a rapidly progressive disease that is refractory to therapy. In


Haematologica | 2016

RNA sequencing unravels the genetics of refractory/relapsed T-cell acute lymphoblastic leukemia. Prognostic and therapeutic implications.

Valentina Gianfelici; Sabina Chiaretti; Sofie Demeyer; Filomena Di Giacomo; Monica Messina; Roberta La Starza; Nadia Peragine; Francesca Paoloni; Ellen Geerdens; Valentina Pierini; Loredana Elia; Marco Mancini; Maria Stefania De Propris; Valerio Apicella; Gianluca Gaidano; Anna Maria Testi; Antonella Vitale; Marco Vignetti; Cristina Mecucci; Anna Guarini; Jan Cools; Robin Foà

Despite therapeutic improvements, a sizable number of patients with T-cell acute lymphoblastic leukemia still have a poor outcome. To unravel the genomic background associated with refractoriness, we evaluated the transcriptome of 19 cases of refractory/early relapsed T-cell acute lymphoblastic leukemia (discovery cohort) by performing RNA-sequencing on diagnostic material. The incidence and prognostic impact of the most frequently mutated pathways were validated by Sanger sequencing on genomic DNA from diagnostic samples of an independent cohort of 49 cases (validation cohort), including refractory, relapsed and responsive cases. Combined gene expression and fusion transcript analyses in the discovery cohort revealed the presence of known oncogenes and identified novel rearrangements inducing overexpression, as well as inactivation of tumor suppressor genes. Mutation analysis identified JAK/STAT and RAS/PTEN as the most commonly disrupted pathways in patients with chemorefractory disease or early relapse, frequently in association with NOTCH1/FBXW7 mutations. The analysis on the validation cohort documented a significantly higher risk of relapse, inferior overall survival, disease-free survival and event-free survival in patients with JAK/STAT or RAS/PTEN alterations. Conversely, a significantly better survival was observed in patients harboring only NOTCH1/FBXW7 mutations: this favorable prognostic effect was abrogated by the presence of concomitant mutations. Preliminary in vitro assays on primary cells demonstrated sensitivity to specific inhibitors. These data document the negative prognostic impact of JAK/STAT and RAS/PTEN mutations in T-cell acute lymphoblastic leukemia and suggest the potential clinical application of JAK and PI3K/mTOR inhibitors in patients harboring mutations in these pathways.


Medical Principles and Practice | 2014

Genomic Characterization of Acute Leukemias

Sabina Chiaretti; Valentina Gianfelici; Giulia Ceglie; Robin Foà

Over the past two decades, hematologic malignancies have been extensively evaluated due to the introduction of powerful technologies, such as conventional karyotyping, FISH analysis, gene and microRNA expression profiling, array comparative genomic hybridization and SNP arrays, and next-generation sequencing (including whole-exome sequencing and RNA-seq). These analyses have allowed for the refinement of the mechanisms underlying the leukemic transformation in several oncohematologic disorders and, more importantly, they have permitted the definition of novel prognostic algorithms aimed at stratifying patients at the onset of disease and, consequently, treating them in the most appropriate manner. Furthermore, the identification of specific molecular markers is opening the door to targeted and personalized medicine. The most important findings on novel acquisitions in the context of acute lymphoblastic leukemia of both B and T lineage and de novo acute myeloid leukemia are described in this review.

Collaboration


Dive into the Valentina Gianfelici's collaboration.

Top Co-Authors

Avatar

Robin Foà

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Sabina Chiaretti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Jan Cools

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Anna Guarini

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Antonella Vitale

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Ellen Geerdens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Monica Messina

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Peter Vandenberghe

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Stein Aerts

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Anna Lucia Fedullo

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge