Network


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

Hotspot


Dive into the research topics where Angela M. Savino is active.

Publication


Featured researches published by Angela M. Savino.


Haematologica | 2013

Impact of IKZF1 deletions on IKZF1 expression and outcome in Philadelphia chromosome negative childhood BCP-ALL. Reply to “Incidence and biological significance of IKZF1/Ikaros gene deletions in pediatric Philadelphia chromosome negative and Philadelphia chromosome positive B-cell precursor acute lymphoblastic leukemia”

Chiara Palmi; Tobia Lana; Daniela Silvestri; Angela M. Savino; Geertruy te Kronnie; Conter; G Basso; Andrea Biondi; Mg Valsecchi; Gianni Cazzaniga

We thank Dr. Qazi and Dr. Uckun for their letter that focused on the biological relevance of IKZF1 deletions in pediatric acute lymphoblastic leukemia (ALL), and in particular on the lack of correlation between IKZF1 deletions and the expected deregulation of IKZF1 expression. This information


Proceedings of the National Academy of Sciences of the United States of America | 2017

Suppressors and activators of JAK-STAT signaling at diagnosis and relapse of acute lymphoblastic leukemia in Down syndrome

Omer Schwartzman; Angela M. Savino; Michael Gombert; Chiara Palmi; Gunnar Cario; Martin Schrappe; Cornelia Eckert; Arend von Stackelberg; Jin Yan Huang; Michal Hameiri-Grossman; Smadar Avigad; Geertruy te Kronnie; Ifat Geron; Yehudit Birger; Avigail Rein; Giulia Zarfati; Ute Fischer; Zohar Mukamel; Martin Stanulla; Andrea Biondi; Giovanni Cazzaniga; Amedeo Vetere; Bridget K. Wagner; Zhu Chen; Sai-Juan Chen; Amos Tanay; Arndt Borkhardt; Shai Izraeli

Significance Children with Down syndrome are at increased risk for B-cell acute lymphoblastic leukemia (DS-ALL), often expressing cytokine receptor-like factor 2 (CRLF2). Here we studied matched diagnosis and relapse DS-ALLs to understand the pathogenesis of relapse. We confirm that enhanced JAK-STAT signaling frequently “drives” CRLF2pos DS-ALL at diagnosis, but discovered that clones with JAK mutations are unstable, suggesting that they also endowed the transformed cells with vulnerabilities. We find USP9X loss in up to 25% of CRLF2pos ALLs, and demonstrate that its ablation decreases the toxic effect of JAK2 hypersignaling. Thus, in CRLF2pos ALLs JAK-STAT signaling is often buffered by loss of USP9X. These results have therapeutic implications because they suggest that ALL cells can tolerate a limited range of JAK-STAT signaling. Children with Down syndrome (DS) are prone to development of high-risk B-cell precursor ALL (DS-ALL), which differs genetically from most sporadic pediatric ALLs. Increased expression of cytokine receptor-like factor 2 (CRLF2), the receptor to thymic stromal lymphopoietin (TSLP), characterizes about half of DS-ALLs and also a subgroup of sporadic “Philadelphia-like” ALLs. To understand the pathogenesis of relapsed DS-ALL, we performed integrative genomic analysis of 25 matched diagnosis-remission and -relapse DS-ALLs. We found that the CRLF2 rearrangements are early events during DS-ALL evolution and generally stable between diagnoses and relapse. Secondary activating signaling events in the JAK-STAT/RAS pathway were ubiquitous but highly redundant between diagnosis and relapse, suggesting that signaling is essential but that no specific mutations are “relapse driving.” We further found that activated JAK2 may be naturally suppressed in 25% of CRLF2pos DS-ALLs by loss-of-function aberrations in USP9X, a deubiquitinase previously shown to stabilize the activated phosphorylated JAK2. Interrogation of large ALL genomic databases extended our findings up to 25% of CRLF2pos, Philadelphia-like ALLs. Pharmacological or genetic inhibition of USP9X, as well as treatment with low-dose ruxolitinib, enhanced the survival of pre-B ALL cells overexpressing mutated JAK2. Thus, somehow counterintuitive, we found that suppression of JAK-STAT “hypersignaling” may be beneficial to leukemic B-cell precursors. This finding and the reduction of JAK mutated clones at relapse suggest that the therapeutic effect of JAK specific inhibitors may be limited. Rather, combined signaling inhibitors or direct targeting of the TSLP receptor may be a useful therapeutic strategy for DS-ALL.


Leukemia | 2017

The histone deacetylase inhibitor givinostat (ITF2357) exhibits potent anti-tumor activity against CRLF2 -rearranged BCP-ALL

Angela M. Savino; Jolanda Sarno; Luca Trentin; Margherita Vieri; Grazia Fazio; Michela Bardini; Cristina Bugarin; Gianluca Fossati; Kara L. Davis; Giuseppe Gaipa; Shai Izraeli; Lueder H. Meyer; Garry P. Nolan; Andrea Biondi; G te Kronnie; Chiara Palmi; Gianni Cazzaniga

Leukemias bearing CRLF2 and JAK2 gene alterations are characterized by aberrant JAK/STAT signaling and poor prognosis. The HDAC inhibitor givinostat/ITF2357 has been shown to exert anti-neoplastic activity against both systemic juvenile idiopathic arthritis and myeloproliferative neoplasms through inhibition of the JAK/STAT pathway. These findings led us to hypothesize that givinostat might also act against CRLF2-rearranged BCP-ALL, which lack effective therapies. Here, we found that givinostat inhibited proliferation and induced apoptosis of BCP-ALL CRLF2-rearranged cell lines, positive for exon 16 JAK2 mutations. Likewise, givinostat killed primary cells, but not their normal hematopoietic counterparts, from patients carrying CRLF2 rearrangements. At low doses, givinostat downregulated the expression of genes belonging to the JAK/STAT pathway and inhibited STAT5 phosphorylation. In vivo, givinostat significantly reduced engraftment of human blasts in patient-derived xenograft models of CRLF2-positive BCP-ALL. Importantly, givinostat killed ruxolitinib-resistant cells and potentiated the effect of current chemotherapy. Thus, givinostat in combination with conventional chemotherapy may represent an effective therapeutic option for these difficult–to-treat subsets of ALL. Lastly, the selective killing of cancer cells by givinostat may allow the design of reduced intensity regimens in CRLF2-rearranged Down syndrome-associated BCP-ALL patients with an overall benefit in terms of both toxicity and related complications.


Haematologica | 2015

Fine tuning of surface CRLF2 expression and its associated signaling profile in childhood B-cell precursor acute lymphoblastic leukemia

Cristina Bugarin; Jolanda Sarno; Chiara Palmi; Angela M. Savino; Geertruy te Kronnie; Michael Dworzak; Angela Shumich; Barbara Buldini; Oscar Maglia; Simona Sala; Ilaria Bronzini; Jean Pierre Bourquin; Ester Mejstrikova; Ondrej Hrusak; Drorit Luria; Giuseppe Basso; Shai Izraeli; Andrea Biondi; Giovanni Cazzaniga; Giuseppe Gaipa

Genomic rearrangements of the cytokine receptor-like factor 2 (CRLF2) gene,[1][1],[2][2] which is part of the thymic stromal lymphopoietin receptor (TSLPR), result in overexpression of CRLF2 itself leading to JAK2-mediated activation of STAT5, which regulates cell proliferation, survival, and


Oncotarget | 2016

CRLF2 over-expression is a poor prognostic marker in children with high risk T-cell acute lymphoblastic leukemia

Chiara Palmi; Angela M. Savino; Daniela Silvestri; Ilaria Bronzini; Gunnar Cario; Maddalena Paganin; Barbara Buldini; Marta Galbiati; Martina U. Muckenthaler; Cristina Bugarin; Pamela Della Mina; Stefan Nagel; Elena Barisone; Fiorina Casale; Franco Locatelli; Luca Lo Nigro; Concetta Micalizzi; Rosanna Parasole; Andrea Pession; Maria Caterina Putti; Nicola Santoro; Anna Maria Testi; Ottavio Ziino; Andreas E. Kulozik; Martin Zimmermann; Martin Schrappe; Antonello Villa; Giuseppe Gaipa; Giuseppe Basso; Andrea Biondi

Pediatric T-ALL patients have a worse outcome compared to BCP-ALL patients and they could benefit from new prognostic marker identification. Alteration of CRLF2 gene, a hallmark correlated with poor outcome in BCP-ALL, has not been reported in T-ALL. We analyzed CRLF2 expression in 212 T-ALL pediatric patients enrolled in AIEOP-BFM ALL2000 study in Italian and German centers. Seventeen out of 120 (14.2%) Italian patients presented CRLF2 mRNA expression 5 times higher than the median (CRLF2-high); they had a significantly inferior event-free survival (41.2%±11.9 vs. 68.9%±4.6, p=0.006) and overall survival (47.1%±12.1 vs. 73.8%±4.3, p=0.009) and an increased cumulative incidence of relapse/resistance (52.9%±12.1 vs. 26.2%±4.3, p=0.007) compared to CRLF2-low patients. The prognostic value of CRLF2 over-expression was validated in the German cohort. Of note, CRLF2 over-expression was associated with poor prognosis in the high risk (HR) subgroup where CRLF2-high patients were more frequently allocated. Interestingly, although in T-ALL CRLF2 protein was localized mainly in the cytoplasm, in CRLF2-high blasts we found a trend towards a stronger TSLP-induced pSTAT5 response, sensitive to the JAK inhibitor Ruxolitinib. In conclusion, CRLF2 over-expression is a poor prognostic marker identifying a subset of HR T-ALL patients that could benefit from alternative therapy, potentially targeting the CRLF2 pathway.


Molecular Cancer Research | 2014

Cytoskeletal Regulatory Gene Expression and Migratory Properties of B-cell Progenitors Are Affected by the ETV6–RUNX1 Rearrangement

Chiara Palmi; Grazia Fazio; Angela M. Savino; Julia Procter; Louise Howell; Valeria Cazzaniga; Margherita Vieri; Giulia Longinotti; Ilaria Brunati; Valentina Andre; Pamela Della Mina; Antonello Villa; Mel Greaves; Andrea Biondi; Giovanna D'Amico; Anthony M. Ford; Giovanni Cazzaniga

Although the ETV6–RUNX1 fusion is a frequent initiating event in childhood leukemia, its role in leukemogenesis is only partly understood. The main impact of the fusion itself is to generate and sustain a clone of clinically silent preleukemic B-cell progenitors (BCP). Additional oncogenic hits, occurring even several years later, are required for overt disease. The understanding of the features and interactions of ETV6–RUNX1–positive cells during this “latency” period may explain how these silent cells can persist and whether they could be prone to additional genetic changes. In this study, two in vitro murine models were used to investigate whether ETV6–RUNX1 alters the cellular adhesion and migration properties of BCP. ETV6–RUNX1–expressing cells showed a significant defect in the chemotactic response to CXCL12, caused by a block in CXCR4 signaling, as demonstrated by inhibition of CXCL12-associated calcium flux and lack of ERK phosphorylation. Moreover, the induction of ETV6–RUNX1 caused changes in the expression of cell-surface adhesion molecules. The expression of genes regulating the cytoskeleton was also affected, resulting in a block of CDC42 signaling. The abnormalities described here could alter the interaction of ETV6–RUNX1 preleukemic BCP with the microenvironment and contribute to the pathogenesis of the disease. Implications: Alterations in the expression of cytoskeletal regulatory genes and migration properties of BCP represent early events in the evolution of the disease, from the preleukemic phase to the clinical onset, and suggest new strategies for effective eradication of leukemia. Mol Cancer Res; 12(12); 1796–806. ©2014 AACR.


Molecular Cancer Therapeutics | 2018

Antileukemic Efficacy of BET Inhibitor in a Preclinical Mouse Model of MLL-AF4+ Infant ALL

Michela Bardini; Luca Trentin; Francesca Rizzo; Margherita Vieri; Angela M. Savino; Patricia Garrido Castro; Grazia Fazio; Eddy Hj van Roon; Mark Kerstjens; Nicholas Smithers; Rab K. Prinjha; Geertruy te Kronnie; Giuseppe Basso; Ronald W. Stam; Rob Pieters; Andrea Biondi; Giovanni Cazzaniga

MLL-rearranged acute lymphoblastic leukemia (ALL) occurring in infants is a rare but very aggressive leukemia, typically associated with a dismal prognosis. Despite the development of specific therapeutic protocols, infant patients with MLL-rearranged ALL still suffer from a low cure rate. At present, novel therapeutic approaches are urgently needed. Recently, the use of small molecule inhibitors targeting the epigenetic regulators of the MLL complex emerged as a promising strategy for the development of a targeted therapy. Herein, we have investigated the effects of bromodomain and extra-terminal (BET) function abrogation in a preclinical mouse model of MLL-AF4+ infant ALL using the BET inhibitor I-BET151. We reported that I-BET151 is able to arrest the growth of MLL-AF4+ leukemic cells in vitro, by blocking cell division and rapidly inducing apoptosis. Treatment with I-BET151 in vivo impairs the leukemic engraftment of patient-derived primary samples and lower the disease burden in mice. I-BET151 affects the transcriptional profile of MLL-rearranged ALL through the deregulation of BRD4, HOXA7/HOXA9, and RUNX1 gene networks. Moreover, I-BET151 treatment sensitizes glucocorticoid-resistant MLL-rearranged cells to prednisolone in vitro and is more efficient when used in combination with HDAC inhibitors, both in vitro and in vivo. Given the aggressiveness of the disease, the failure of the current therapies and the lack of an ultimate cure, this study paves the way for the use of BET inhibitors to treat MLL-rearranged infant ALL for future clinical applications. Mol Cancer Ther; 17(8); 1705–16. ©2018 AACR.


Oncotarget | 2018

SRC/ABL inhibition disrupts CRLF2-driven signaling to induce cell death in B-cell acute lymphoblastic leukemia

Jolanda Sarno; Angela M. Savino; Chiara Buracchi; Chiara Palmi; Stefania Pinto; Cristina Bugarin; Astraea Jager; Silvia Bresolin; Ruth C. Barber; Daniela Silvestri; Shai Israeli; Martin J. S. Dyer; Giovanni Cazzaniga; Garry P. Nolan; Andrea Biondi; Kara L. Davis; Giuseppe Gaipa

Children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) overexpressing the CRLF2 gene (hiCRLF2) have poor prognosis. CRLF2 protein overexpression leads to activated JAK/STAT signaling and trials are underway using JAK inhibitors to overcome treatment failure. Pre-clinical studies indicated limited efficacy of single JAK inhibitors, thus additional pathways must be targeted in hiCRLF2 cells. To identify additional activated networks, we used single-cell mass cytometry to examine 15 BCP-ALL primary patient samples. We uncovered a coordinated signaling network downstream of CRLF2 characterized by co-activation of JAK/STAT, PI3K, and CREB pathways. This CRLF2-driven network could be more effectively disrupted by SRC/ABL inhibition than single-agent JAK or PI3K inhibition, and this could be demonstrated even in primary minimal residual disease (MRD) cells. Our study suggests SCR/ABL inhibition as effective in disrupting the cooperative functional networks present in hiCRLF2 BCP-ALL patients, supporting further investigation of this strategy in pre-clinical studies.


Haematologica | 2016

On mice and humans: the role of thymic stromal lymphopoietin in human B-cell development and leukemia.

Angela M. Savino; Shai Izraeli

The development of immunodeficient mouse models has revolutionized the ability to study human disease in vivo . One of the difficulties is that many mouse and human cytokines are not cross-reactive. For this reason, the in vivo models currently used are not optimized enough to recapitulate the


Experimental Hematology | 2018

SCD Takes Leukemia to the CNS

Angela M. Savino; Orianne Olivares; Shani Barel; Lev Yakimov; Ifat Geron; Hila Fishman; Christina Halsey; Eyal Gottlieb; Shai Izraeli

Collaboration


Dive into the Angela M. Savino's collaboration.

Top Co-Authors

Avatar

Andrea Biondi

University of Milano-Bicocca

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppe Gaipa

University of Milano-Bicocca

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge