Network


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

Hotspot


Dive into the research topics where Erika Zonari is active.

Publication


Featured researches published by Erika Zonari.


Cancer Cell | 2011

Targeting the ANG2/TIE2 Axis Inhibits Tumor Growth and Metastasis by Impairing Angiogenesis and Disabling Rebounds of Proangiogenic Myeloid Cells

Roberta Mazzieri; Ferdinando Pucci; Davide Moi; Erika Zonari; Anna Ranghetti; Alvise Berti; Letterio S. Politi; Bernhard Gentner; Jeffrey L. Brown; Luigi Naldini; Michele De Palma

Tumor-infiltrating myeloid cells convey proangiogenic programs that counteract the efficacy of antiangiogenic therapy. Here, we show that blocking angiopoietin-2 (ANG2), a TIE2 ligand and angiogenic factor expressed by activated endothelial cells (ECs), regresses the tumor vasculature and inhibits progression of late-stage, metastatic MMTV-PyMT mammary carcinomas and RIP1-Tag2 pancreatic insulinomas. ANG2 blockade did not inhibit recruitment of MRC1(+) TIE2-expressing macrophages (TEMs) but impeded their upregulation of Tie2, association with blood vessels, and ability to restore angiogenesis in tumors. Conditional Tie2 gene knockdown in TEMs was sufficient to decrease tumor angiogenesis. Our findings support a model wherein the ANG2-TIE2 axis mediates cell-to-cell interactions between TEMs and ECs that are important for tumor angiogenesis and can be targeted to induce effective antitumor responses.


Cancer Cell | 2008

Tumor-Targeted Interferon-α Delivery by Tie2-Expressing Monocytes Inhibits Tumor Growth and Metastasis

Michele De Palma; Roberta Mazzieri; Letterio S. Politi; Ferdinando Pucci; Erika Zonari; Giovanni Sitia; Stefania Mazzoleni; Davide Moi; Mary Anna Venneri; Stefano Indraccolo; Andrea Falini; Luca G. Guidotti; Rossella Galli; Luigi Naldini

The use of type I interferons (IFNs) in cancer therapy has been limited by ineffective dosing and significant toxicity. Here, we exploited the tumor-homing ability of proangiogenic Tie2-expressing monocytes (TEMs) to deliver IFN-alpha to tumors. By transplanting hematopoietic progenitors transduced with a Tie2 promoter/enhancer-driven Ifna1 gene, we turned TEMs into IFN-alpha cell vehicles that efficiently targeted the IFN response to orthotopic human gliomas and spontaneous mouse mammary carcinomas and obtained significant antitumor responses and near complete abrogation of metastasis. TEM-mediated IFN-alpha delivery inhibited tumor angiogenesis and activated innate and adaptive immune cells but did not impair myelopoiesis and wound healing detectably. These results illustrate the therapeutic potential of gene- and cell-based IFN-alpha delivery and should allow the development of IFN treatments that more effectively treat cancer.


Blood | 2013

A role for miR-155 in enabling tumor-infiltrating innate immune cells to mount effective antitumor responses in mice

Erika Zonari; Ferdinando Pucci; Massimo Saini; Roberta Mazzieri; Letterio S. Politi; Bernhard Gentner; Luigi Naldini

A productive immune response requires transient upregulation of the microRNA miR-155 in hematopoietic cells mediating innate and adaptive immunity. In order to investigate miR-155 in the context of tumor-associated immune responses, we stably knocked down (KD) miR-155 in the myeloid compartment of MMTV-PyMT mice, a mouse model of spontaneous breast carcinogenesis that closely mimics tumor-host interactions seen in humans. Notably, miR-155/KD significantly accelerated tumor growth by impairing classic activation of tumor-associated macrophages (TAMs). This created an imbalance toward a protumoral microenvironment as evidenced by a lower proportion of CD11c(+) TAMs, reduced expression of activation markers, and the skewing of immune cells within the tumor toward an macrophage type 2/T helper 2 response. This study highlights the importance of tumor-infiltrating hematopoietic cells in constraining carcinogenesis and establishes an antitumoral function of a prototypical oncomiR.


Molecular Therapy | 2015

Dual-regulated Lentiviral Vector for Gene Therapy of X-linked Chronic Granulomatosis

Maria Chiriaco; Giada Farinelli; Valentina Capo; Erika Zonari; Samantha Scaramuzza; Gigliola Di Matteo; Lucia Sergi Sergi; Maddalena Migliavacca; Raisa Jofra Hernandez; Ferdinando Bombelli; Ezio Giorda; Anna Kajaste-Rudnitski; Didier Trono; Manuel Grez; Paolo Rossi; Andrea Finocchi; Luigi Naldini; Bernhard Gentner; Alessandro Aiuti

Regulated transgene expression may improve the safety and efficacy of hematopoietic stem cell (HSC) gene therapy. Clinical trials for X-linked chronic granulomatous disease (X-CGD) employing gammaretroviral vectors were limited by insertional oncogenesis or lack of persistent engraftment. Our novel strategy, based on regulated lentiviral vectors (LV), targets gp91(phox) expression to the differentiated myeloid compartment while sparing HSC, to reduce the risk of genotoxicity and potential perturbation of reactive oxygen species levels. Targeting was obtained by a myeloid-specific promoter (MSP) and posttranscriptional, microRNA-mediated regulation. We optimized both components in human bone marrow (BM) HSC and their differentiated progeny in vitro and in a xenotransplantation model, and generated therapeutic gp91(phox) expressing LVs for CGD gene therapy. All vectors restored gp91(phox) expression and function in human X-CGD myeloid cell lines, primary monocytes, and differentiated myeloid cells. While unregulated LVs ectopically expressed gp91(phox) in CD34(+) cells, transcriptionally and posttranscriptionally regulated LVs substantially reduced this off-target expression. X-CGD mice transplanted with transduced HSC restored gp91(phox) expression, and MSP-driven vectors maintained regulation during BM development. Combining transcriptional (SP146.gp91-driven) and posttranscriptional (miR-126-restricted) targeting, we achieved high levels of myeloid-specific transgene expression, entirely sparing the CD34(+) HSC compartment. This dual-targeted LV construct represents a promising candidate for further clinical development.


Stem cell reports | 2017

Efficient Ex Vivo Engineering and Expansion of Highly Purified Human Hematopoietic Stem and Progenitor Cell Populations for Gene Therapy

Erika Zonari; Giacomo Desantis; Carolina Petrillo; Francesco Boccalatte; Maria Rosa Lidonnici; Anna Kajaste-Rudnitski; Alessandro Aiuti; Giuliana Ferrari; Luigi Naldini; Bernhard Gentner

Summary Ex vivo gene therapy based on CD34+ hematopoietic stem cells (HSCs) has shown promising results in clinical trials, but genetic engineering to high levels and in large scale remains challenging. We devised a sorting strategy that captures more than 90% of HSC activity in less than 10% of mobilized peripheral blood (mPB) CD34+ cells, and modeled a transplantation protocol based on highly purified, genetically engineered HSCs co-infused with uncultured progenitor cells. Prostaglandin E2 stimulation allowed near-complete transduction of HSCs with lentiviral vectors during a culture time of less than 38 hr, mitigating the negative impact of standard culture on progenitor cell function. Exploiting the pyrimidoindole derivative UM171, we show that transduced mPB CD34+CD38− cells with repopulating potential could be expanded ex vivo. Implementing these findings in clinical gene therapy protocols will improve the efficacy, safety, and sustainability of gene therapy and generate new opportunities in the field of gene editing.


Oncogene | 2007

A direct link between expression of urokinase plasminogen activator receptor, growth rate and oncogenic transformation in mouse embryonic fibroblasts.

Roberta Mazzieri; Federico Furlan; Silvia D'Alessio; Erika Zonari; Francesco Talotta; Pasquale Verde; Francesco Blasi

In addition to its role in invasion and metastasis of several tumors, the multifunctional urokinase receptor uPAR (urokinase plasminogen activator receptor) is directly involved in the growth of several cancer cells in vitro and in vivo. We have compared growth rate and oncogenic transformation in wild-type (wt) or uPAR−/− mouse embryonic fibroblasts (MEFs). Surprisingly, uPAR−/− MEFs grew faster than wt MEFs. This agreed with elevated levels of cell cycle mediators like extracellular signal-regulated protein kinase, p38, AP1 and Cyclin D1. Infection with a uPAR retrovirus reverted the effect, decreasing the growth rate.When MEFs were transformed with H-RasV12 and E1A oncogenes, the efficiency of transformation in uPAR−/− MEFs was higher than in wt. UPAR−/− MEFs grew faster at low serum, produced more colonies in agar and produced tumors in vivo in nude mice with a lower latency period. The properties of the heterozygous uPAR+/− MEFs were always intermediate. We conclude therefore that in MEFs uPAR concentration controls cell proliferation and the transforming activity of some oncogenes.


Molecular Therapy | 2016

235. Improved Ex Vivo Gene Therapy Using Highly Purified Hematopoietic Stem and Progenitor Cells

Erika Zonari; Oriana Meo; Samantha Scaramuzza; Eugenio Montini; Giuliana Ferrari; Luigi Naldini; Bernhard Gentner

Ex vivo gene addition into CD34+ hematopoietic stem and progenitor cells (HSPC) followed by autologous transplantation has proven a safe and efficacious therapy for immunodeficiencies, storage diseases and hemoglobinopathies. According to xenograft models and population size estimates of vector-marked cells in gene therapy-treated patients, less than 0.01% of infused CD34+ cells drive long-term (LT) repopulation. Advances in clinical-grade cell sorting technology may make HSC-enriched CD34+ subpopulations accessible for gene therapy, with advantages in terms of lentiviral vector (LV) cost, safety (lowering of integration load) and, potentially, efficacy. By differentially marking mobilized peripheral blood (mPB) CD34 subpopulations distinguished by increasing levels of CD38 expression in order to quantitatively assess their hematopoietic output in an NSG xenograft model over 6 months, we previously mapped most (>90%) LT repopulating capacity to CD34+CD38- cells (lowest 10% CD38 staining), while CD34+CD38int cells drove short-term (ST) reconstitution during the first 2 months after transplantation. We now characterize these subpopulations in terms of CD90 expression, a marker, which has been used in conjunction with CD34 for HSC purification in past clinical trials. CD34+ mPB cells were sorted into CD38-90+ (5% of CD34+), CD38-90- (5%), CD38+90+ (30%) and CD38+90- (60%) fractions and exhaustively transduced with GFP-, OFP-, BFP- and mCherry-expressing LVs, respectively. Differentially marked subfractions were pooled maintaining their original proportions and transplanted into NSG mice. ST engraftment mainly came from CD38+ cells, with equal contribution from the CD90+ and CD90- compartment. LT engraftment was almost exclusively derived from CD34+CD38- cells, of which 70% came from CD90+ and 30% from CD90- cells. Hence, CD34+CD38- is a more sensitive and specific marker combination than CD34+CD90+ to purify LT-HSC. CD34+CD38- cells can be purified by a sequential bead-based selection (CD34 selection of CD38-depleted cells) potentially applicable to clinical practice. We show that CD34+CD38- cells can be efficiently transduced with clinical grade LVs using shortened ex vivo manipulation protocols, reaching similar gene marking levels as with the standard protocol currently used in clinical trials that comprises a double dose of LV. Transduction was stable for at least 5 months when serially measured in xenotransplanted mice, and mice showed multi-lineage hematopoiesis indistinguishable from CD34+ grafts. Based on these results, we are aiming towards clinical development of a new gene therapy protocol based on CD34+CD38-HSPC efficiently transduced with minimum ex vivo culture time (<36h). Our platform will substantially improve the efficacy, safety and feasibility of future ex vivo gene therapy studies.


Molecular Therapy | 2015

295. Hematopoietic Stem Cell Gene Therapy (2.0) Based on Purified CD34+CD38- Cells

Erika Zonari; Francesco Boccalatte; Tiziana Plati; Alessandro Aiuti; Giuliana Ferrari; Eugenio Montini; Luigi Naldini; Bernhard Gentner

Lentiviral (LV)-based hematopoietic stem and progenitor cell (HSPC) gene therapy is becoming a promising alternative to allogeneic stem cell transplantation for curing genetic diseases. To potentially improve the efficacy, safety and economic sustainability of HSPC transduction, we reasoned to genetically manipulate only the more potent CD34+CD38- HSPC, thereby improving HSPC maintenance in culture in the absence of differentiating cells and downscaling the cell therapy product by a factor of ten without compromising long-term engraftment. This approach would also decrease the total load of vector integration infused in the patients, thus improving its overall safety.First, we determined the engraftment kinetics of CD34+ mobilized peripheral blood (mPB) subpopulations over a 24wk xenotransplantation period. We sorted CD34+ mPB into 4 fractions with increasing expression levels of CD38 and marked each fraction with a specific fluorescent protein allowing to track the population of origin driving hematopoietic reconstitution. Differentially labeled fractions were mixed, and various combinations of CD38-, CD38int and CD38hi HSPC were injected into NSG mice (2 exp, n=30). Almost all long-term repopulating capacity (>90%) was contained within CD34+CD38- cells, and these cells took over hematopoiesis by 9wks. Instead, early reconstitution was mainly driven by CD34+CD38int progenitor cells.In the prospect of a clinical translation, we then modeled the co-administration of gene-modified CD34+CD38- mPB cells with uncultured CD34+CD38int/+ supporter cells aimed to drive fast hematopoietic recovery (3 exp, n=38). Repopulation by gene-modified CD34+CD38- cells was slower (15wks) and incomplete ( 5 wks and re-established long-term (24wks) gene marking up to 85%, thus allowing to benefit from prompt hematopoietic recovery driven by transiently repopulating CD38int/+ supporter cells.Last, we optimized LV transduction in the framework of an improved culture protocol. Exposing CD34+ or CD34+CD38- mPB cells to prostaglandin E2 (PGE2) increased transduction efficiency 1.5-2.5x, allowing to markedly reduce pre-stimulation and LV exposure times better preserving HSC functions. Importantly, the higher gene-transfer efficiency was maintained for up to 24 wks following xenotransplantation (n=33), suggesting that PGE2 facilitated LV transduction in long-term HSC.In summary, these results support the clinical development of novel HSPC gene therapy protocols based on the modification of highly purified HSC subsets, with the prospect to improve the efficacy, safety and feasibility of future ex vivo gene therapy studies.


Molecular Therapy | 2015

281. Engineering Hematopoiesis for Tumor-Targeted Interferon-alpha Delivery Inhibits Multuple Myeloma and B Cell Malignancies

Giulia Escobar; Anna Ranghetti; Luigi Barbarossa; Erika Zonari; Silvia Nucera; Fabio Ciceri; Bernhard Gentner; Luigi Naldini

A protective and immunosuppressive tumor microenvironment is considered a key factor for the failure of anti-cancer treatments. We developed a strategy to turn a pro-tumoral into an anti-tumoral microenvironment by transplanting genetically engineered hematopoietic stem/progenitor cells (HSPC). We exploit a transcriptionally and post-transcriptionally regulated Interferon-alpha (IFNa) cassette to drive specific expression of this pleiotrophic antitumor cytokine in tumor-infiltrating monocytes/macrophages (IFNa gene therapy). When applied to spontaneous mouse or orthotopic human breast cancer models, IFNa gene therapy with mouse or human HSPC, respectively, inhibited tumor and metastases growth by activating innate and adaptive immune cells in the tumor (Escobar et al, Sci Transl Med 2014). To test this approach in clinics, we aim for a first-in-human trial of IFNa gene therapy in patients undergoing autologous transplantation, a procedure widely practiced for Multiple Myeloma (MM) and Lymphoma. To detect potential untoward effects of chronic IFNa exposure on human hematopoiesis, we transduced mobilized peripheral blood CD34+ cells with the human IFNa lentiviral vector (LV). A single round of transduction yielded vector copy numbers (VCN) of 0.4 to 4 according to the protocol used, with 30-90% transduction efficiency and 1-4 VCN per cell. This IFNa-engineered graft was mixed with mock-transduced CD34+ cells in various proportions and transplanted into NSG mice. All mice engrafted and showed long-term, multilineage hematopoietic output, with a dose-dependent decrease in the human graft at high, supratherapeutic VCN. To explore the efficacy of IFNa gene therapy on MM, we intravenously injected NSG mice reconstituted with IFNa LV-transduced CD34+ cells (VCN:1-3; chimerism 20-50%) with the human MM.1S cell line and followed MM growth by bioluminescence imaging and serial blood and BM exams. IFNa gene therapy strongly delayed myeloma bone disease and improved survival (9 wks vs. 13 wks median survival, p=0.03). Moreover, we tested the efficacy of IFNa gene therapy on human primary Ph+ B-ALL blasts as a model of high-grade B cell malignancies. Strikingly, IFNa gene therapy substantially reduced B-ALL growth in NSG and NSG3GS mice (p=0.002 and p=0.015), and Imatinib treatment, the standard of care for Ph+ALL, gave an additive effect when combined with IFNa gene therapy. These encouraging results pave the way for a phase I/II trial in patients with MM or B cell malignancies. To better understand the mechanism of IFNa gene therapy, we are setting up immuno-competent mouse models of MM and lymphoid malignancies. We developed a spontaneous model of high grade B cell leukemia/lymphoma based on ectopic expression of miR-126. IFNa gene therapy significantly reduced leukemic burden. Moreover, we derived more immunogenic leukemic subclones engineered with the ovalbumin or huCD20 antigen. These lines will help defining the immunomodulatory effect of IFNa gene therapy and allow testing combination with monoclonal antibodies or adoptive T cell transfer.


Blood | 2007

Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer

Mary Anna Venneri; Michele De Palma; Maurilio Ponzoni; Ferdinando Pucci; Cristina Scielzo; Erika Zonari; Roberta Mazzieri; Claudio Doglioni; Luigi Naldini

Collaboration


Dive into the Erika Zonari's collaboration.

Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Bernhard Gentner

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alessandro Aiuti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Ferdinando Pucci

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Giuliana Ferrari

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Anna Kajaste-Rudnitski

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Letterio S. Politi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Samantha Scaramuzza

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Michele De Palma

École Polytechnique Fédérale de Lausanne

View shared research outputs
Researchain Logo
Decentralizing Knowledge