Network


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

Hotspot


Dive into the research topics where Francesca Avogadri is active.

Publication


Featured researches published by Francesca Avogadri.


Journal of Experimental Medicine | 2009

OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis

Daniel Hirschhorn-Cymerman; Gabrielle Rizzuto; Taha Merghoub; Adam D. Cohen; Francesca Avogadri; Alexander M. Lesokhin; Andrew D. Weinberg; Jedd D. Wolchok; Alan N. Houghton

Expansion and recruitment of CD4+ Foxp3+ regulatory T (T reg) cells are mechanisms used by growing tumors to evade immune elimination. In addition to expansion of effector T cells, successful therapeutic interventions may require reduction of T reg cells within the tumor microenvironment. We report that the combined use of the alkylating agent cyclophosphamide (CTX) and an agonist antibody targeting the co-stimulatory receptor OX40 (OX86) provides potent antitumor immunity capable of regressing established, poorly immunogenic B16 melanoma tumors. CTX administration resulted in tumor antigen release, which after OX86 treatment significantly enhanced the antitumor T cell response. We demonstrated that T reg cells are an important cellular target of the combination therapy. Paradoxically, the combination therapy led to an expansion of T reg cells in the periphery. In the tumor, however, the combination therapy induced a profound T reg cell depletion that was accompanied by an influx of effector CD8+ T cells leading to a favorable T effector/T reg cell ratio. Closer examination revealed that diminished intratumoral T reg cell levels resulted from hyperactivation and T reg cell–specific apoptosis. Thus, we propose that CTX and OX40 engagement represents a novel and rational chemoimmunotherapy.


Cancer Research | 2012

Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment.

Alexander M. Lesokhin; Tobias M. Hohl; Shigehisa Kitano; Czrina Cortez; Daniel Hirschhorn-Cymerman; Francesca Avogadri; Gabrielle Rizzuto; John J. Lazarus; Eric G. Pamer; Alan N. Houghton; Taha Merghoub; Jedd D. Wolchok

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that accumulate during tumor formation, facilitate immune escape, and enable tumor progression. MDSCs are important contributors to the development of an immunosuppressive tumor microenvironment that blocks the action of cytotoxic antitumor T effector cells. Heterogeneity in these cells poses a significant barrier to studying the in vivo contributions of individual MDSC subtypes. Herein, we show that granulocyte-macrophage colony stimulating factor, a cytokine critical for the numeric and functional development of MDSC populations, promotes expansion of a monocyte-derived MDSC population characterized by expression of CD11b and the chemokine receptor CCR2. Using a toxin-mediated ablation strategy to target CCR2-expressing cells, we show that these monocytic MDSCs regulate entry of activated CD8 T cells into the tumor site, thereby limiting the efficacy of immunotherapy. Our results argue that therapeutic targeting of monocytic MDSCs would enhance outcomes in immunotherapy.


Cancer Research | 2005

Cancer immunotherapy based on killing of Salmonella-infected tumor cells

Francesca Avogadri; Chiara Martinoli; Liljana Petrovska; Claudia Chiodoni; Pietro Transidico; Vincenzo Bronte; Renato Longhi; Mario P. Colombo; Gordon Dougan; Maria Rescigno

A major obstacle for the development of effective immunotherapy is the ability of tumors to escape the immune system. The possibility to kill tumor cells because they are recognized as infected rather than as malignant could help overcome immune escape mechanisms. Here we report a conceptually new approach of cancer immunotherapy based on in vivo infection of tumors and killing of infected tumor cells. Attenuated but still invasive, Salmonella typhimurium can be successfully exploited to invade melanoma cells that can present antigenic determinants of bacterial origin and become targets for anti-Salmonella-specific T cells. However, to fully appreciate the anticancer therapeutic properties of S. typhimurium, tumor-bearing mice need to be vaccinated against S. typhimurium before intratumoral Salmonella injection. Tumor infection when coupled to anti-Salmonella vaccination leads to 50% to 100% tumor-free mice with a better outcome on larger tumors. Invasive Salmonella also exert an indirect toxic effect on tumor cells through the recruitment of inflammatory cells and the cross-presentation of tumor antigens, which allow induction of tumor-specific immune response. This is effective in retarding the growth of untreated established distant tumors and in protecting the mice from subsequent tumor challenges.


Science Translational Medicine | 2010

Bacteria-Induced Gap Junctions in Tumors Favor Antigen Cross-Presentation and Antitumor Immunity

Fabiana Saccheri; Chiara Pozzi; Francesca Avogadri; Sara Barozzi; Mario Faretta; Paola Fusi; Maria Rescigno

Bacterial infection induces extra gap junctions in tumors, through which tumor antigens travel to antigen-presenting cells, triggering an effective antitumor immune response. Special Delivery: Immune Cells Receive a Package of Tumor-Specific Peptides Immune cells on patrol often recognize cancer cells as abnormal and eliminate them. But as tumors progress and proliferate, they can become invisible to these immune guardians of order. An injection of bacteria into the tumor can render them again visible to the immune system, thus promoting tumor-directed immune responses. If we understood how this reappearing act worked, we might be able to exploit it for cancer treatment. Saccheri et al. have discovered that the injected bacteria perform a key function: They reactivate connexin 43, a protein often suppressed in cancer cells that forms tiny communication channels—gap junctions—between cells. Tumor peptides escape through these channels and enter immune cells, which display the peptides on their surfaces, successfully triggering a specific immune response against the cancer. The authors showed that the bacteria Salmonella or its components elicited increased amounts of connexin 43 in melanoma cell lines from mice or humans. This connexin was used by the cells to form new gap junctions, allowing small molecules of the dye Lucifer yellow to pass between tumor cells or from tumor cells into antigen-presenting dendritic cells, which could also transfer dye among themselves. When the authors artificially inserted the protein ovalbumin into the melanoma cells in culture, it was broken down into peptides by the tumor proteasome, and these peptides passed through the gap junctions into cocultured dendritic cells, where they were displayed on the surface. Ovalbumin-specific T cells could be activated in response, and this activation depended on connexin 43. The authors then tested in mice whether this gap junction route for getting tumor peptides to dendritic cells operates in living animals. Although bacteria injected into tumors caused the eradication of that particular tumor independent of connexin and gap junctions, the regression of distant metastases, mediated by cytotoxic T cells, required the infection-induced gap junction mechanism. The delivery of tumor peptides to dendritic cells through gap junctions can be harnessed to generate tumor-specific dendritic cells ex vivo, a way to create tumor-specific immune cells for infusion into patients. Incubation of bacteria-infected melanoma cells with dendritic cells in vitro allowed loading of the immune cells with tumor peptides, conferring an effective antitumor response when the dendritic cells were injected into tumor-bearing mice. And this approach could have even more practical potential: These in vitro–loaded immune cells protected mice against developing tumors from seeded cancer cells, a “vaccination”-style preventive strategy. Antigen-presenting dendritic cells (DCs) trigger the activation of cytotoxic CD8 T cells that target and eliminate cells with the antigen on their surface. Although DCs usually pick up and process antigens themselves, they can also receive peptide antigens from other cells via gap junctions. We demonstrate here that infection with Salmonella can induce, in both human and murine melanoma cells, the up-regulation of connexin 43 (Cx43), a ubiquitous protein that forms gap junctions and that is normally lost during melanoma progression. Bacteria-treated melanoma cells can establish functional gap junctions with adjacent DCs. After bacterial infection, these gap junctions transferred preprocessed antigenic peptides from the tumor cells to the DCs, which then presented those peptides on their surface. These peptides activated cytotoxic T cells against the tumor antigen, which could control the growth of distant uninfected tumors. Melanoma cells in which Cx43 had been silenced, when infected in vivo with bacteria, failed to elicit a cytotoxic antitumor response, indicating that this Cx43 mechanism is the principal one used in vivo for the generation of antitumor responses. The Cx43-dependent cross-presentation pathway is more effective than standard protocols of DC loading (peptide, tumor lysates, or apoptotic bodies) for generating DC-based tumor vaccines that both inhibit existing tumors and prevent tumor establishment. In conclusion, we exploited an antimicrobial response present in tumor cells to activate cytotoxic CD8 T cells specific for tumor-generated peptides that could directly recognize and kill tumor cells.


Blood | 2010

Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs.

Takeshi Nakahara; Hiroshi Uchi; Alexander M. Lesokhin; Francesca Avogadri; Gabrielle Rizzuto; Daniel Hirschhorn-Cymerman; Katherine S. Panageas; Taha Merghoub; Jedd D. Wolchok; Alan N. Houghton

Cyclophosphamide (CTX), a commonly used chemotherapeutic agent can enhance immune responses. The ability of CTX to promote the proliferation of effector T cells and abrogate the function of regulatory T cells (Tregs) has been described. In this study, we examined the effects of CTX treatment on dendritic cell (DC) subsets and the subsequent outcome on the effector and suppressive arms of adaptive immunity. In secondary lymphoid tissues, tissue-derived migratory DCs (migratory DCs), lymphoid tissue-resident DCs (resident DCs), and plasmacytoid DCs (pDCs) are well described. CTX has profound and selective cytotoxic effects on CD8(+) resident DCs, but not skin-derived migratory DCs or pDCs in lymph nodes (LNs) and spleen, causing an imbalance among these DC subsets. CTX treatment increases the potency of DCs in antigen presentation and cytokine secretion, and partially inhibits the suppressor activity of Tregs. Adoptive transfer of CD8(+) DCs can reconstitute this population in regional draining LNs and abrogate the immune-enhancing effects of CTX in vivo. These findings demonstrate that CTX may improve immune responses by preferentially depleting CD8(+) lymphoid-resident DCs, which leads to diminished Treg suppression and enhanced effector T-cell function in vivo.


PLOS Pathogens | 2014

Modified Vaccinia Virus Ankara Triggers Type I IFN Production in Murine Conventional Dendritic Cells via a cGAS/STING-Mediated Cytosolic DNA-Sensing Pathway

Peihong Dai; Weiyi Wang; Hua Cao; Francesca Avogadri; Lianpan Dai; Ingo Drexler; Johanna A. Joyce; Xiao Dong Li; Zhijian J. Chen; Taha Merghoub; Stewart Shuman; Liang Deng

Modified vaccinia virus Ankara (MVA) is an attenuated poxvirus that has been engineered as a vaccine against infectious agents and cancers. Our goal is to understand how MVA modulates innate immunity in dendritic cells (DCs), which can provide insights to vaccine design. In this study, using murine bone marrow-derived dendritic cells, we assessed type I interferon (IFN) gene induction and protein secretion in response to MVA infection. We report that MVA infection elicits the production of type I IFN in murine conventional dendritic cells (cDCs), but not in plasmacytoid dendritic cells (pDCs). Transcription factors IRF3 (IFN regulatory factor 3) and IRF7, and the positive feedback loop mediated by IFNAR1 (IFN alpha/beta receptor 1), are required for the induction. MVA induction of type I IFN is fully dependent on STING (stimulator of IFN genes) and the newly discovered cytosolic DNA sensor cGAS (cyclic guanosine monophosphate-adenosine monophosphate synthase). MVA infection of cDCs triggers phosphorylation of TBK1 (Tank-binding kinase 1) and IRF3, which is abolished in the absence of cGAS and STING. Furthermore, intravenous delivery of MVA induces type I IFN in wild-type mice, but not in mice lacking STING or IRF3. Treatment of cDCs with inhibitors of endosomal and lysosomal acidification or the lysosomal enzyme Cathepsin B attenuated MVA-induced type I IFN production, indicating that lysosomal enzymatic processing of virions is important for MVA sensing. Taken together, our results demonstrate a critical role of the cGAS/STING-mediated cytosolic DNA-sensing pathway for type I IFN induction in cDCs by MVA. We present evidence that vaccinia virulence factors E3 and N1 inhibit the activation of IRF3 and the induction of IFNB gene in MVA-infected cDCs.


PLOS ONE | 2010

Alphavirus Replicon Particles Expressing TRP-2 Provide Potent Therapeutic Effect on Melanoma through Activation of Humoral and Cellular Immunity

Francesca Avogadri; Taha Merghoub; Maureen Maughan; Daniel Hirschhorn-Cymerman; John Morris; Erika Ritter; Robert A. Olmsted; Alan N. Houghton; Jedd D. Wolchok

Background Malignant melanoma is the deadliest form of skin cancer and is refractory to conventional chemotherapy and radiotherapy. Therefore alternative approaches to treat this disease, such as immunotherapy, are needed. Melanoma vaccine design has mainly focused on targeting CD8+ T cells. Activation of effector CD8+ T cells has been achieved in patients, but provided limited clinical benefit, due to immune-escape mechanisms established by advanced tumors. We have previously shown that alphavirus-based virus-like replicon particles (VRP) simultaneously activate strong cellular and humoral immunity against the weakly immunogenic melanoma differentiation antigen (MDA) tyrosinase. Here we further investigate the antitumor effect and the immune mechanisms of VRP encoding different MDAs. Methodology/Principal Findings VRP encoding different MDAs were screened for their ability to prevent the growth of the B16 mouse transplantable melanoma. The immunologic mechanisms of efficacy were investigated for the most effective vaccine identified, focusing on CD8+ T cells and humoral responses. To this end, ex vivo immune assays and transgenic mice lacking specific immune effector functions were used. The studies identified a potent therapeutic VRP vaccine, encoding tyrosinase related protein 2 (TRP-2), which provided a durable anti-tumor effect. The efficacy of VRP-TRP2 relies on a novel immune mechanism of action requiring the activation of both IgG and CD8+ T cell effector responses, and depends on signaling through activating Fcγ receptors. Conclusions/Significance This study identifies a VRP-based vaccine able to elicit humoral immunity against TRP-2, which plays a role in melanoma immunotherapy and synergizes with tumor-specific CD8+ T cell responses. These findings will aid in the rational design of future immunotherapy clinical trials.


European Journal of Immunology | 2008

Intra‐tumoral Salmonella typhimurium induces a systemic anti‐tumor immune response that is directed by low‐dose radiation to treat distal disease

Francesca Avogadri; Deepak Mittal; Fabiana Saccheri; Massimo Sarrafiore; Mario Ciocca; Paola Larghi; Roberto Orecchia; Maria Rescigno

Salmonella typhimurium is a facultative anaerobic bacterium able to multiply preferentially in tumors and inhibit their growth. The mechanisms through which Salmonella exerts its anti‐cancer properties are not fully understood. We recently showed that intra‐tumoral Salmonella injection results not only in the regression of even bulky tumor masses, but also impacts on the growth of distant untreated lesions. Here we describe how Salmonella exerts its systemic anti‐cancer effects and means to potentiate them. The outburst of an early inflammatory reaction in the treated tumor promotes the development of an immunostimulatory cytokine environment both locally and in the draining lymph node. Within the next 10 days, an efficient cross‐presentation of endogenous tumor antigens by dendritic cells at the tumor‐draining lymph node leads to the priming of effective anti‐tumor CD8+ T cell responses. This potentially broadly reactive T cell repertoire can be directed to other pre‐established melanomas by low‐dose radiotherapy enhancing the Salmonella anti‐cancer effect. We demonstrate that Salmonella‐based therapy coupled to low‐dose radiotherapy dampens tumor immune escape mechanisms at different levels and allows controlling systemic disease in a CD8+ T cell‐dependent manner.


PLOS ONE | 2011

Detection of Intra-Tumor Self Antigen Recognition during Melanoma Tumor Progression in Mice Using Advanced Multimode Confocal/Two Photon Microscope

David Schaer; Yongbiao Li; Taha Merghoub; Gabrielle Rizzuto; Amos Shemesh; Adam D. Cohen; Yanyun Li; Francesca Avogadri; Ricardo Toledo-Crow; Alan N. Houghton; Jedd D. Wolchok

Determining how tumor immunity is regulated requires understanding the extent to which the anti-tumor immune response “functions” in vivo without therapeutic intervention. To better understand this question, we developed advanced multimodal reflectance confocal/two photon fluorescence intra-vital imaging techniques to use in combination with traditional ex vivo analysis of tumor specific T cells. By transferring small numbers of melanoma-specific CD8+ T cells (Pmel-1), in an attempt to mimic physiologic conditions, we found that B16 tumor growth alone was sufficient to induce naive Pmel-1 T cell proliferation and acquisition of effector phenotype. Tumor -primed Pmel-1 T cells, are capable of killing target cells in the periphery and secrete IFNγ, but are unable to mediate tumor regression. Within the tumor, Pmel-1 T cells have highly confined mobility, displaying long term interactions with tumor cells. In contrast, adoptively transferred non tumor-specific OT-I T cells show neither confined mobility, nor long term interaction with B16 tumor cells, suggesting that intra-tumor recognition of cognate self antigen by Pmel-1 T cells occurs during tumor growth. Together, these data indicate that lack of anti-tumor efficacy is not solely due to ignorance of self antigen in the tumor microenvironment but rather to active immunosuppressive influences preventing a protective immune response.


Cancer immunology research | 2014

Combination of Alphavirus Replicon Particle–Based Vaccination with Immunomodulatory Antibodies: Therapeutic Activity in the B16 Melanoma Mouse Model and Immune Correlates

Francesca Avogadri; Roberta Zappasodi; Arvin Yang; Sadna Budhu; Nicole Malandro; Daniel Hirschhorn-Cymerman; Shakuntala Tiwari; Maureen Maughan; Robert A. Olmsted; Jedd D. Wolchok; Taha Merghoub

Avogadri and colleagues show that anti-CTLA-4 or anti-GITR immunomodulatory antibody improves the efficacy of a nonpathogenic viral vector–based vaccine (VRP-TRP-2) in the B16F10 melanoma mouse model. Superior antitumor protection conferred by anti-GITR was associated with enhanced humoral response and reduced CD4+PD-1+ T-cell intratumoral accumulation. Induction of potent immune responses to self-antigens remains a major challenge in tumor immunology. We have shown that a vaccine based on alphavirus replicon particles (VRP) activates strong cellular and humoral immunity to tyrosinase-related protein-2 (TRP2) melanoma antigen, providing prophylactic and therapeutic effects in stringent mouse models. Here, we report that the immunogenicity and efficacy of this vaccine is increased in combination with either antagonist anti-CTL antigen-4 (CTLA-4) or agonist anti-glucocorticoid-induced TNF family–related gene (GITR) immunomodulatory monoclonal antibodies (mAb). In the challenging therapeutic setting, VRP–TRP2 plus anti-GITR or anti–CTLA-4 mAb induced complete tumor regression in 90% and 50% of mice, respectively. These mAbs had similar adjuvant effects in priming an adaptive immune response against the vaccine-encoded antigen, augmenting, respectively, approximately 4- and 2-fold the TRP2-specific CD8+ T-cell response and circulating Abs, compared with the vaccine alone. Furthermore, while both mAbs increased the frequency of tumor-infiltrating CD8+ T cells, anti–CTLA-4 mAb also increased the quantity of intratumor CD4+Foxp3− T cells expressing the negative costimulatory molecule programmed death-1 (PD-1). Concurrent GITR expression on these cells suggests that they might be controlled by anti-GITR mAbs, thus potentially explaining their differential accumulation under the two treatment conditions. These findings indicate that combining immunomodulatory mAbs with alphavirus-based anticancer vaccines can provide therapeutic antitumor immune responses in a stringent mouse model, suggesting potential utility in clinical trials. They also indicate that tumor-infiltrating CD4+Foxp3−PD-1+ T cells may affect the outcome of immunomodulatory treatments. Cancer Immunol Res; 2(5); 448–58. ©2014 AACR.

Collaboration


Dive into the Francesca Avogadri's collaboration.

Top Co-Authors

Avatar

Taha Merghoub

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jedd D. Wolchok

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Maria Rescigno

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar

Alan N. Houghton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Hirschhorn-Cymerman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabrielle Rizzuto

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Angelica Sonzogni

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppe Viale

European Institute of Oncology

View shared research outputs
Researchain Logo
Decentralizing Knowledge