Network


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

Hotspot


Dive into the research topics where Davide Moi is active.

Publication


Featured researches published by Davide Moi.


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.


Nature | 2014

Targeted genome editing in human repopulating haematopoietic stem cells

Pietro Genovese; Giulia Schiroli; Giulia Escobar; Tiziano Di Tomaso; Claudia Firrito; Andrea Calabria; Davide Moi; Roberta Mazzieri; Chiara Bonini; Michael C. Holmes; Philip D. Gregory; Mirjam van der Burg; Bernhard Gentner; Eugenio Montini; Angelo Lombardo; Luigi Naldini

Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.


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.


Cell Reports | 2012

miR-511-3p Modulates Genetic Programs of Tumor-Associated Macrophages

Mario Leonardo Squadrito; Ferdinando Pucci; Laura Magri; Davide Moi; Gregor D. Gilfillan; Anna Ranghetti; Andrea Casazza; Massimiliano Mazzone; Robert Lyle; Luigi Naldini; Michele De Palma

Expression of the mannose receptor (MRC1/CD206) identifies macrophage subtypes, such as alternatively activated macrophages (AAMs) and M2-polarized tumor-associated macrophages (TAMs), which are endowed with tissue-remodeling, proangiogenic, and protumoral activity. However, the significance of MRC1 expression for TAMs protumoral activity is unclear. Here, we describe and characterize miR-511-3p, an intronic microRNA (miRNA) encoded by both mouse and human MRC1 genes. By using sensitive miRNA reporter vectors, we demonstrate robust expression and bioactivity of miR-511-3p in MRC1(+) AAMs and TAMs. Unexpectedly, enforced expression of miR-511-3p tuned down the protumoral gene signature of MRC1(+) TAMs and inhibited tumor growth. Our findings suggest that transcriptional activation of Mrc1 in TAMs evokes a genetic program orchestrated by miR-511-3p, which limits rather than enhances their protumoral functions. Besides uncovering a role for MRC1 as gatekeeper of TAMs protumoral genetic programs, these observations suggest that endogenous miRNAs may operate to establish thresholds for inflammatory cell activation in tumors.


Science Translational Medicine | 2014

Genetic engineering of hematopoiesis for targeted IFN-α delivery inhibits breast cancer progression.

Giulia Escobar; Davide Moi; Anna Ranghetti; Pinar Ozkal-Baydin; Mario Leonardo Squadrito; Anna Kajaste-Rudnitski; Attilio Bondanza; Bernhard Gentner; Michele De Palma; Roberta Mazzieri; Luigi Naldini

Inhibition of primary and metastatic breast cancer by targeted delivery of interferon-α by tumor-infiltrating monocytes. Toasting Tumor Immunotherapy Tumor immunotherapy is a promising new strategy for cancer treatment but is somewhat limited by the immunosuppressive nature of the tumor microenvironment. Type I interferons (IFNs) have been shown to promote tumor immunity, but systemic toxicity has limited their use. Genetic engineering of hematopoietic stem cells (HSCs) is one way to modulate the immune response in the tumor microenvironment. Now, Escobar et al. attempt to overcome this immunosuppression by introducing IFN-α into tumor-infiltrating macrophages. The authors developed a way to insert an IFN-α transgene into HSCs, but restrict IFN-α expression to differentiated monocytes. They then tested their cells in human hematochimeric mice. HSC engraftment and repopulation is inhibited if IFN-α is expressed in the less differentiated cells. However, here, the HSCs engraft and repopulate NSG mice. What’s more, these cells inhibit tumor progression and experimental metastasis in an autologous model of breast cancer, effectively reprogramming the tumor microenvironment. The immunosuppressive tumor microenvironment represents a major hurdle to cancer therapy. We developed a gene transfer strategy into hematopoietic stem cells (HSCs) to target transgene expression to tumor-infiltrating monocytes/macrophages. Using a combination of transcriptional and microRNA-mediated control, we achieved selective expression of an interferon-α (IFN-α) transgene in differentiated monocytes of human hematochimeric mice. We show that IFN-α transgene expression does not impair engraftment and long-term multilineage repopulation of NSG (NOD/LtSz-scidIL2Rγnull) mice by transplanted human HSCs. By providing a source of human cytokines in the mice, we improved the functional reconstitution of human myeloid, natural killer, and T cell lineages, and achieved enhanced immune-mediated clearance of transplanted human breast tumors when hematopoiesis was engineered for tumor-targeted IFN-α expression. By applying our strategy to mouse breast cancer models, we achieved inhibition of tumor progression and experimental metastases in an autologous setting, likely through enhanced generation of effector T cells and their recruitment to the neoplastic tissues. By forcing IFN-α expression in tumor-infiltrating macrophages, we blunted their innate protumoral activity and reprogrammed the tumor microenvironment toward more effective dendritic cell activation and immune effector cell cytotoxicity. Overall, our studies validate the feasibility, safety, and therapeutic potential of a new cancer gene therapy strategy, and open the way to test this approach as adjuvant therapy in advanced breast cancer patients.


Cancer Research | 2015

Urokinase receptor promotes skin tumor formation by preventing epithelial cell activation of Notch1

Roberta Mazzieri; Giovanni Pietrogrande; Laura Gerasi; Alessandro Gandelli; Piergiuseppe Colombo; Davide Moi; Chiara Brombin; Alessandro Ambrosi; Silvio Danese; Paolo Mignatti; Francesco Blasi; Silvia D'Alessio

The urokinase-type plasminogen activator receptor (uPAR) has a well-established role in cancer progression, but it has been little studied at earlier stages of cancer initiation. Here, we show that uPAR deficiency in the mouse dramatically reduces susceptibility to the classical two-stage protocol of inflammatory skin carcinogenesis. uPAR genetic deficiency decreased papilloma formation and accelerated keratinocyte differentiation, effects mediated by Notch1 hyperactivation. Notably, Notch1 inhibition in uPAR-deficient mice rescued their susceptibility to skin carcinogenesis. Clinically, we found that human differentiated keratoacanthomas expressed low levels of uPAR and high levels of activated Notch1, with opposite effects in proliferating tumors, confirming the relevance of the observations in mice. Furthermore, we found that TACE-dependent activation of Notch1 in basal kerantinocytes was modulated by uPAR. Mechanistically, uPAR sequestered TACE within lipid rafts to prevent Notch1 activation, thereby promoting cell proliferation and tumor formation. Given that uPAR signaling is nonessential for normal epidermal homeostasis, our results argue that uPAR may present a promising disease-specific target for preventing skin cancer development.


British Journal of Ophthalmology | 2016

Angiopoietin 2 expression in the cornea and its control of corneal neovascularisation.

Giulio Ferrari; Chiara Giacomini; Fabio Bignami; Davide Moi; Anna Ranghetti; Claudio Doglioni; Luigi Naldini; Paolo Rama; Roberta Mazzieri

Purpose To define proangiogenic angiopoietin 2 (ANG2) expression and role(s) in human and mouse vascularised corneas. Further, to evaluate the effect of ANG2 inhibition on corneal neovascularisation (CNV). Methods CNV was induced in FVB mice by means of intrastromal suture placement. One group of animals was sacrificed 10 days later; corneas were immunostained for ANG2 and compared with (i) mouse non-vascularised corneas and (ii) human vascularised and non-vascularised corneas. A second group of CNV animals was treated systemically with an anti-ANG2 antibody. After 10 days, the corneas were whole-mounted, stained for CD31 and LYVE1 and lymphatic/blood vessels quantified. In another set of experiments, the corneal basal Bowman membrane was either (i) removed or (ii) left in place. After 2 or 10 days the corneas were removed and immunostained for collagen IV, ANG2, CD31, LYVE1, CD11b and MRC1 markers. Results In human beings and mice, ANG2 is expressed only in the epithelium, and, mildly, in the endothelium, of the avascular cornea. Instead, it is expressed in the epithelium, endothelium and stroma of vascularised corneas. Disruption of the Bowman membrane is associated with a significant increase of (i) ANG2 stromal expression and (ii) proangiogenic macrophage infiltration in the corneal stroma. Finally, blocking ANG2 significantly reduced hemangiogenesis, lymphangiogenesis and macrophage infiltration. Conclusions Balancing proper healing and good vision is crucial in the cornea, constantly exposed to potential injuries. In this paper, we suggest the existence of a mechanism regulating the onset of inflammation (and associated CNV) depending on injury severity.


OncoImmunology | 2018

B cell lymphoma progression promotes the accumulation of circulating Ly6Clo monocytes with immunosuppressive activity

Sara J. McKee; Zewen K. Tuong; Takumi Kobayashi; Brianna L. Doff; Megan S.F. Soon; Michael Nissen; Pui Yeng Lam; Colm Keane; Frank Vari; Davide Moi; Roberta Mazzieri; Graham R. Leggatt; Maher K. Gandhi; Stephen R. Mattarollo

ABSTRACT Monocytosis is considered a poor prognostic factor for many cancers, including B cell lymphomas. The mechanisms by which different monocyte subsets support the growth of lymphoma is poorly understood. Using a pre-clinical mouse model of B cell non-Hodgkins lymphoma (B-NHL), we investigated the impact of tumor progression on circulating monocyte levels, subset distribution and their activity, with a focus on immune suppression. B-NHL development corresponded with significant expansion initially of classical (Ly6Chi) and non-classical (Ly6Clo) monocytes, with accumulation and eventual predominance of Ly6Clo cells. The lymphoma environment promoted the conversion, preferential survival and immune suppressive activity of Ly6Clo monocytes. Ly6Clo monocytes expressed higher levels of immunosuppressive genes including PD-L1/2, Arg1, IDO1 and CD163, compared to Ly6Chi monocytes. Both monocyte subsets suppressed CD8 T cell proliferation and IFN-γ production in vitro, but via different mechanisms. Ly6Chi monocyte suppression was contact dependent, while Ly6Clo monocytes suppressed via soluble mediators, including IDO and arginase. Ly6Clo monocytes could be selectively depleted in tumor-bearing hosts by liposomal doxorubicin treatment, further enhanced by co-administration of anti-4-1BB monoclonal antibody. This treatment led to a reduction in tumor growth, but failed to improve overall survival. Analogous immunosuppressive monocytes were observed in peripheral blood of diffuse large B cell lymphoma patients and actively suppressed human CD8 T cell proliferation. This study highlights a potential immune evasion strategy deployed by B cell lymphoma involving accumulation of circulating non-classical monocytes with immunosuppressive activity.


Journal of Clinical Investigation | 2018

Self-adjuvanting nanoemulsion targeting dendritic cell receptor Clec9A enables antigen-specific immunotherapy

Bijun Zeng; Anton P. J. Middelberg; Adrian Tandhyka Gemiarto; Kelli P. A. MacDonald; Alan G. Baxter; Meghna Talekar; Davide Moi; Kirsteen M. Tullett; Irene Caminschi; Mireille H. Lahoud; Roberta Mazzieri; Riccardo Dolcetti; Ranjeny Thomas

Non–antigen-specific stimulatory cancer immunotherapies are commonly complicated by off-target effects. Antigen-specific immunotherapy, combining viral tumor antigen or personalized neoepitopes with immune targeting, offers a solution. However, the lack of flexible systems targeting tumor antigens to cross-presenting dendritic cells (DCs) limits clinical development. Although antigen–anti-Clec9A mAb conjugates target cross-presenting DCs, adjuvant must be codelivered for cytotoxic T lymphocyte (CTL) induction. We functionalized tailored nanoemulsions encapsulating tumor antigens to target Clec9A (Clec9A-TNE). Clec9A-TNE encapsulating OVA antigen targeted and activated cross-presenting DCs without additional adjuvant, promoting antigen-specific CD4+ and CD8+ T cell proliferation and CTL and antibody responses. OVA-Clec9A-TNE–induced DC activation required CD4 and CD8 epitopes, CD40, and IFN-&agr;. Clec9A-TNE encapsulating HPV E6/E7 significantly suppressed HPV-associated tumor growth, while E6/E7–CpG did not. Clec9A-TNE loaded with pooled B16-F10 melanoma neoepitopes induced epitope-specific CD4+ and CD8+ T cell responses, permitting selection of immunogenic neoepitopes. Clec9A-TNE encapsulating 6 neoepitopes significantly suppressed B16-F10 melanoma growth in a CD4+ T cell–dependent manner. Thus, cross-presenting DCs targeted with antigen–Clec9A-TNE stimulate therapeutically effective tumor-specific immunity, dependent on T cell help.


International Journal of Cancer | 2018

Immune system augmentation via humanization using stem/progenitor cells and bioengineering in a breast cancer model study: Humanized mouse model for cancer study

Abbas Shafiee; Jacqui A. McGovern; Christoph A. Lahr; Christoph Meinert; Davide Moi; Ferdinand Wagner; Marietta Landgraf; Elena M. De-Juan-Pardo; Roberta Mazzieri; Dietmar W. Hutmacher

Despite significant advances, most current in vivo models fail to fully recapitulate the biological processes that occur in humans. Here we aimed to develop an advanced humanized model with features of an organ bone by providing different bone tissue cellular compartments including preosteoblasts, mesenchymal stem/stromal (MSCs), endothelial and hematopoietic cells in an engineered microenvironment. The bone compartment was generated by culturing the human MSCs, umbilical vein endothelial cells with gelatin methacryloyl hydrogels in the center of a melt‐electrospun polycaprolactone tubular scaffolds, which were seeded with human preosteoblasts. The tissue engineered bone (TEB) was subcutaneously implanted into the NSG mice and formed a morphologically and functionally organ bone. Mice were further humanized through the tail vein injection of human cord blood derived CD34+ cells, which then populated in the mouse bone marrow, spleen and humanized TEB (hTEB). 11 weeks after CD34+ transplantation, metastatic breast cancer cells (MDA‐MB‐231BO) were orthotopically injected. Cancer cell injection resulted in the formation of a primary tumor and metastasis to the hTEB and mouse organs. Less frequent metastasis and lower tumor burden were observed in hematochimeric mice, suggesting an immune‐mediated response against the breast cancer cells. Overall, our results demonstrate the efficacy of tissue engineering approaches to study species‐specific cancer‐bone interactions. Further studies using genetically modified hematopoietic stem cells and bioengineered microenvironments will enable us to address the specific roles of signaling molecules regulating hematopoietic niches and cancer metastasis in vivo.

Collaboration


Dive into the Davide Moi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luigi Naldini

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
Top Co-Authors

Avatar

Dietmar W. Hutmacher

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Jacqui A. McGovern

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ferdinando Pucci

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Giulia Escobar

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Abbas Shafiee

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Christoph A. Lahr

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Elena M. De-Juan-Pardo

Queensland University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge