173 results on '"Vincenzo Bronte"'
Search Results
2. The transcriptional response in human umbilical vein endothelial cells exposed to insulin: a dynamic gene expression approach.
- Author
-
Barbara Di Camillo, Tiziana Sanavia, Elisabetta Iori, Vincenzo Bronte, Enrica Roncaglia, Alberto Maran, Angelo Avogaro, Gianna Toffolo, and Claudio Cobelli
- Subjects
Medicine ,Science - Abstract
BACKGROUND: In diabetes chronic hyperinsulinemia contributes to the instability of the atherosclerotic plaque and stimulates cellular proliferation through the activation of the MAP kinases, which in turn regulate cellular proliferation. However, it is not known whether insulin itself could increase the transcription of specific genes for cellular proliferation in the endothelium. Hence, the characterization of transcriptional modifications in endothelium is an important step for a better understanding of the mechanism of insulin action and the relationship between endothelial cell dysfunction and insulin resistance. METHODOLOGY AND PRINCIPAL FINDINGS: The transcriptional response of endothelial cells in the 440 minutes following insulin stimulation was monitored using microarrays and compared to a control condition. About 1700 genes were selected as differentially expressed based on their treated minus control profile, thus allowing the detection of even small but systematic changes in gene expression. Genes were clustered in 7 groups according to their time expression profile and classified into 15 functional categories that can support the biological effects of insulin, based on Gene Ontology enrichment analysis. In terms of endothelial function, the most prominent processes affected were NADH dehydrogenase activity, N-terminal myristoylation domain binding, nitric-oxide synthase regulator activity and growth factor binding. Pathway-based enrichment analysis revealed "Electron Transport Chain" significantly enriched. Results were validated on genes belonging to "Electron Transport Chain" pathway, using quantitative RT-PCR. CONCLUSIONS: As far as we know, this is the first systematic study in the literature monitoring transcriptional response to insulin in endothelial cells, in a time series microarray experiment. Since chronic hyperinsulinemia contributes to the instability of the atherosclerotic plaque and stimulates cellular proliferation, some of the genes identified in the present work are potential novel candidates in diabetes complications related to endothelial dysfunction.
- Published
- 2010
- Full Text
- View/download PDF
3. Covid-19 Interstitial Pneumonia: Histological and Immunohistochemical Features on Cryobiopsies
- Author
-
Vincenzo Bronte, Venerino Poletti, Stefano Maitan, Alessandra Dubini, Marco Chilosi, Franco Stella, Athol U. Wells, Claudio Doglioni, Antonio Vizzuso, Silvia Puglisi, Giovanni Pizzolo, Vanni Agnoletti, Federica Pedica, Claudia Ravaglia, Giulio Rossi, Vittorio Sambri, Giovanni Poletti, Sara Piciucchi, and Doglioni C, Ravaglia C, Chilosi M, Rossi G, Dubini A, Pedica F, Piciucchi S, Vizzuso A, Stella F, Maitan S, Agnoletti V, Puglisi S, Poletti G, Sambri V, Pizzolo G, Bronte V, Wells AU, Poletti V.
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Coronaviru ,Lung biopsy ,Lung injury ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Cryobiopsy ,030212 general & internal medicine ,Diffuse alveolar damage ,Lung ,Acute respiratory distress syndrome ,SARS-CoV-2 ,business.industry ,CD68 ,pSTAT-3 ,Hyperplasia ,medicine.disease ,Coronavirus ,Endothelial stem cell ,Pneumonia ,medicine.anatomical_structure ,030228 respiratory system ,Indoleamine 2,3-dioxygenase-1 ,Covid-19 ,business - Abstract
Background: The pathogenetic steps leading to Covid-19 interstitial pneumonia remain to be clarified. Most postmortem studies to date reveal diffuse alveolar damage as the most relevant histologic pattern. Antemortem lung biopsy may however provide more precise data regarding the earlier stages of the disease, providing a basis for novel treatment approaches. Objectives: To ascertain the morphological and immunohistochemical features of lung samples obtained in patients with moderate Covid-19 pneumonia. Methods: Transbronchial lung cryobiopsy was carried out in 12 Covid-19 patients within 20 days of symptom onset. Results: Histopathologic changes included spots of patchy acute lung injury with alveolar type II cell hyperplasia, with no evidence of hyaline membranes. Strong nuclear expression of phosphorylated STAT3 was observed in >50% of AECII. Interalveolar capillaries showed enlarged lumen and were in part arranged in superposed rows. Pulmonary venules were characterized by luminal enlargement, thickened walls, and perivascular CD4+ T-cell infiltration. A strong nuclear expression of phosphorylated STAT3, associated with PD-L1 and IDO expression, was observed in endothelial cells of venules and interstitial capillaries. Alveolar spaces macrophages exhibited a peculiar phenotype (CD68, CD11c, CD14, CD205, CD206, CD123/IL3AR, and PD-L1). Conclusions: Morphologically distinct features were identified in early stages of Covid-19 pneumonia, with epithelial and endothelial cell abnormalities different from either classical interstitial lung diseases or diffuse alveolar damage. Alveolar type II cell hyperplasia was a prominent event in the majority of cases. Inflammatory cells expressed peculiar phenotypes. No evidence of hyaline membranes and endothelial changes characterized by IDO expression might in part explain the compliance and the characteristic pulmonary vasoplegia observed in less-advanced Covid-19 pneumonia.
- Published
- 2021
- Full Text
- View/download PDF
4. Baricitinib restrains the immune dysregulation in patients with severe COVID-19
- Author
-
Veronica Batani, Manuela Iezzi, Daniela Righetti, Stefano Ugel, Simone Caligola, Enrico Polati, Pier Giuseppe Pelicci, Walter Mosaner, Claudio Lunardi, Katia Donadello, Francesco De Sanctis, Alessandra Fiore, Varvara Petrova, Simonetta Friso, Federica Facciotti, Lorena Torroni, Andrea Claudio Comel, Paolo Bazzoni, Laura Pinton, Francesca Pizzolo, Mariaelisa Rampudda, Francesca Hofer, Elisa Tinazzi, Rosalinda Trovato, Vincenzo Bronte, Antonio Vella, Stefania Canè, Oliviero Olivieri, Chiara Musiu, and Roza Maria Barouni
- Subjects
Male ,0301 basic medicine ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,medicine.disease_cause ,Severity of Illness Index ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fraction of inspired oxygen ,Oxygen therapy ,medicine ,Humans ,Longitudinal Studies ,B cell ,Aged ,Aged, 80 and over ,B-Lymphocytes ,Sulfonamides ,biology ,SARS-CoV-2 ,business.industry ,COVID-19 ,Off-Label Use ,General Medicine ,Middle Aged ,Immune dysregulation ,COVID-19 Drug Treatment ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Purines ,030220 oncology & carcinogenesis ,Immunology ,Commentary ,biology.protein ,Azetidines ,Cytokines ,Pyrazoles ,Female ,Antibody ,business - Abstract
BACKGROUNDPatients with coronavirus disease 2019 (COVID-19) develop pneumonia generally associated with lymphopenia and a severe inflammatory response due to uncontrolled cytokine release. These mediators are transcriptionally regulated by the JAK/STAT signaling pathways, which can be disabled by small molecules.METHODSWe treated a group of patients (n = 20) with baricitinib according to an off-label use of the drug. The study was designed as an observational, longitudinal trial and approved by the local ethics committee. The patients were treated with 4 mg baricitinib twice daily for 2 days, followed by 4 mg per day for the remaining 7 days. Changes in the immune phenotype and expression of phosphorylated STAT3 (p-STAT3) in blood cells were evaluated and correlated with serum-derived cytokine levels and antibodies against severe acute respiratory syndrome-coronavirus 2 (anti-SARS-CoV-2). In a single treated patient, we also evaluated the alteration of myeloid cell functional activity.RESULTSWe provide evidence that patients treated with baricitinib had a marked reduction in serum levels of IL-6, IL-1β, and TNF-α, a rapid recovery of circulating T and B cell frequencies, and increased antibody production against the SARS-CoV-2 spike protein, all of which were clinically associated with a reduction in the need for oxygen therapy and a progressive increase in the P/F (PaO2, oxygen partial pressure/FiO2, fraction of inspired oxygen) ratio.CONCLUSIONThese data suggest that baricitinib prevented the progression to a severe, extreme form of the viral disease by modulating the patients' immune landscape and that these changes were associated with a safer, more favorable clinical outcome for patients with COVID-19 pneumonia.TRIAL REGISTRATIONClinicalTrials.gov NCT04438629.FUNDINGThis work was supported by the Fondazione Cariverona (ENACT Project) and the Fondazione TIM.
- Published
- 2020
- Full Text
- View/download PDF
5. Increased Arginase1 expression in tumor microenvironment promotes mammary carcinogenesis via multiple mechanisms
- Author
-
Michela Croce, Patrizia Damonte, Vincenzo Bronte, Laura Chiossone, Ottavia Barbieri, Paola Vacca, Simonetta Astigiano, Simona Pigozzi, and Monica Morini
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,Transgene ,Apoptosis ,Mice, Transgenic ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer stem cell ,Tumor Cells, Cultured ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Cell Proliferation ,Mammary tumor ,Tumor microenvironment ,Arginase ,Chemistry ,Cell growth ,Mammary Neoplasms, Experimental ,General Medicine ,Cell Transformation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis - Abstract
Arginine metabolism plays a significant role in regulating cell function, affecting tumor growth and metastatization. To study the effect of the arginine-catabolizing enzyme Arginase1 (ARG1) on tumor microenvironment, we generated a mouse model of mammary carcinogenesis by crossbreeding a transgenic mouse line overexpressing ARG1 in macrophages (FVBArg+/+) with the MMTV-Neu mouse line (FVBNeu+/+). This double transgenic line (FVBArg+/−;Neu+/+) showed a significant shortening in mammary tumor latency, and an increase in the number of mammary nodules. Transfer of tumor cells from FVBNeu+/+ into either FVB wild type or FVBArg+/+ mice resulted in increase regulatory T cells in the tumor infiltrate, suggestive of an impaired antitumor immune response. However, we also found increased frequency of tumor stem cells in tumors from FVBArg+/−;Neu+/+ transgenic compared with FVBNeu+/+ mice, suggesting that increased arginine metabolism in mammary tumor microenvironment may supports the cancer stem cells niche. We provide in vivo evidence of a novel, yet unexploited, mechanism through which ARG1 may contribute to tumor development.
- Published
- 2020
- Full Text
- View/download PDF
6. Tumor-Derived Prostaglandin E2 Promotes p50 NF-κB-Dependent Differentiation of Monocytic MDSCs
- Author
-
Viviana Piccolo, Augusto Bleve, Renato Ostuni, Sara Morlacchi, Monica Rimoldi, Chiara Porta, Silvia Tartari, Mario P. Colombo, Alessandro Ippolito, Mariangela Storto, Giulia Soldà, Paola Larghi, Sabina Sangaletti, Stefano Duga, Claudio Tripodo, Tiziana Pressiani, Gioacchino Natoli, Lorenza Rimassa, Laura Strauss, Antonio Sica, Emilio Hirsch, Andrea Doni, Vincenzo Bronte, Stefania Banfi, Fiorella Balzac, Francesca Maria Consonni, Maria Grazia Totaro, Emilia Turco, Porta, Chiara, Consonni, Francesca Maria, Morlacchi, Sara, Sangaletti, Sabina, Bleve, Augusto, Totaro, Maria Grazia, Larghi, Paola, Rimoldi, Monica, Tripodo, Claudio, Strauss, Laura, Banfi, Stefania, Storto, Mariangela, Pressiani, Tiziana, Rimassa, Lorenza, Tartari, Silvia, Ippolito, Alessandro, Doni, Andrea, Soldà, Giulia, Duga, Stefano, Piccolo, Viviana, Ostuni, Renato, Natoli, Gioacchino, Bronte, Vincenzo, Balzac, Fiorella, Turco, Emilia, Hirsch, Emilio, Colombo, Mario P, and Sica, Antonio
- Subjects
0301 basic medicine ,Cancer Research ,Cellular differentiation ,Prostaglandin E2 receptor ,medicine.medical_treatment ,Melanoma, Experimental ,Apoptosis ,Settore MED/08 - Anatomia Patologica ,Nitric Oxide ,Dinoprostone ,Monocytes ,Interferon-gamma ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Oxytocics ,Immune Tolerance ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Prostaglandin E2 ,Cell Proliferation ,Chemistry ,Myeloid-Derived Suppressor Cells ,NF-kappa B p50 Subunit ,Cell Differentiation ,Immunotherapy ,Acquired immune system ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,p50 NF-κB, differentiation of monocytic , MDSC ,030220 oncology & carcinogenesis ,Myeloid-derived Suppressor Cell ,Cancer research ,Tumor necrosis factor alpha ,Colorectal Neoplasms ,medicine.drug - Abstract
Myeloid-derived suppressor cells (MDSC) include immature monocytic (M-MDSC) and granulocytic (PMN-MDSC) cells that share the ability to suppress adaptive immunity and to hinder the effectiveness of anticancer treatments. Of note, in response to IFNγ, M-MDSCs release the tumor-promoting and immunosuppressive molecule nitric oxide (NO), whereas macrophages largely express antitumor properties. Investigating these opposing activities, we found that tumor-derived prostaglandin E2 (PGE2) induces nuclear accumulation of p50 NF-κB in M-MDSCs, diverting their response to IFNγ toward NO-mediated immunosuppression and reducing TNFα expression. At the genome level, p50 NF-κB promoted binding of STAT1 to regulatory regions of selected IFNγ-dependent genes, including inducible nitric oxide synthase (Nos2). In agreement, ablation of p50 as well as pharmacologic inhibition of either the PGE2 receptor EP2 or NO production reprogrammed M-MDSCs toward a NOS2low/TNFαhigh phenotype, restoring the in vivo antitumor activity of IFNγ. Our results indicate that inhibition of the PGE2/p50/NO axis prevents MDSC-suppressive functions and restores the efficacy of anticancer immunotherapy. Significance: Tumor-derived PGE2-mediated induction of nuclear p50 NF-κB epigenetically reprograms the response of monocytic cells to IFNγ toward an immunosuppressive phenotype, thus retrieving the anticancer properties of IFNγ.
- Published
- 2020
- Full Text
- View/download PDF
7. A liquid biopsy-based approach identifies myeloid cells, STAT3 and arginase-1 as predictors of glioma risk score and patients' survival
- Author
-
Elena Masetto, Stefania Canè, Vittorina Zagonel, Susanna Mandruzzato, Domenico D'Avella, Vincenzo Bronte, Paola Del Bianco, Laura Pinton, Sara Magri, Giuseppe Lombardi, Francesco Volpin, Marta Padovan, Daniela Basso, and Alessandro Della Puppa
- Subjects
Oncology ,medicine.medical_specialty ,Framingham Risk Score ,biology ,business.industry ,medicine.disease ,Arginase ,Internal medicine ,Glioma ,Myeloid cells ,medicine ,biology.protein ,Liquid biopsy ,business ,STAT3 - Abstract
Background Although gliomas are strictly confined to the central nervous system, their negative influence over the immune system can extend to peripheral circulation. The immune suppression exerted by myeloid cells is capable of affecting both response to therapy and disease outcome. Here we analyzed the expansion of several myeloid parameters in the blood of low- and high-grade gliomas and assessed their relevance as biomarkers of disease and clinical outcome. Methods Peripheral blood was obtained from 134 low- and high-grade glioma patients before surgery and treatment. Myeloid cell subsets such as total CD14+, CD14+/p-STAT3+, CD14+/PD-L1+, CD15+ cells and 4 myeloid derived suppressor cell (MDSC) subsets, were evaluated by multiparametric flow cytometry. Arginase-1 (ARG1) quantity and activity was determined in the plasma. Principal component analysis was performed to define correlations between myeloid markers. Multivariable logistic regression model was used to obtain a diagnostic score to discriminate glioma patients from healthy controls, and between each glioma grade. A glioblastoma prognostic model was determined by multiple Cox regression using clinical and myeloid parameters. Results In the blood of glioma patients, changes in myeloid parameters associated with immune suppression were identified and allowed us to define a diagnostic score calculating the risk of being a glioma patient, that included CD15+ cells, MDSC1, MDSC3, p-STAT3 and ARG1 activity. Of note, the same parameters, together with age, can also be used to calculate the risk score in differentiating each glioma grade. Finally, a prognostic model for glioblastoma patients stemmed out from a Cox multiple analysis, highlighting the role of MDSC, p-STAT3 and ARG1 activity together with clinical parameters in predicting the patient outcome. Conclusions This work emphasizes the role of systemic immune suppression carried out by myeloid cells in gliomas. The identification of biomarkers associated with immune landscape, diagnosis and outcome of glioblastoma patients lays the ground for their clinical use for stratification and follow up.
- Published
- 2021
- Full Text
- View/download PDF
8. Immunization practices and risk of anaphylaxis: a current update, comprehensive of COVID-19 vaccination data
- Author
-
Alessandra Arcolaci, Vincenzo Bronte, and Giovanna Zanoni
- Subjects
Male ,medicine.medical_specialty ,education.field_of_study ,Vaccines ,COVID-19 Vaccines ,business.industry ,Incidence (epidemiology) ,Immunology ,Population ,Postmarketing surveillance ,medicine.disease ,Vaccination ,Immunization ,Pharmacovigilance ,medicine ,Immunology and Allergy ,Humans ,Female ,Intensive care medicine ,business ,education ,Contraindication ,Anaphylaxis - Abstract
Purpose of review This review aims to provide an updated report in regards to the correlation between vaccines and anaphylaxis and the related risk in the population. Recent findings Initial reports showed higher incidence of anaphylaxis following messenger RNA COVID-19 vaccines compared with 'routine' vaccinations, likely influenced by the great attention paid to these 'new' vaccines. However, anaphylaxis has still to be considered quite rare and its incidence will be systematically reconsidered in the light of additional data collected. Summary Adverse reactions to vaccines are commonly reported but most of them are nonspecific mild events, whereas vaccine-related anaphylaxis is considered a rare event, with an incidence rate equal to 1.3 cases per million vaccine doses administered. As anaphylaxis reports usually start to be reported to passive pharmacovigilance during postmarketing surveillance, the first data are used to be influenced by under- and over-reporting and lack of denominators and following studies are needed to confirm the causal relationship. This might create an initial overcautiously approach to new immunization practices but, being anaphylaxis a potential life-threatening event, every suspected contraindication has to be deepened to maximize effectiveness and safety profile and constantly redefined not to exclude an overestimated population group who could receive the vaccine uneventfully.
- Published
- 2021
9. Nicotinamide Phosphoribosyltransferase Acts as a Metabolic Gate for Mobilization of Myeloid-Derived Suppressor Cells
- Author
-
Vincenzo Bronte, Gian Cesare Tron, Mario P. Colombo, Francesca Maria Consonni, Ubaldina Galli, Massimiliano Mazzone, Ambra A. Grolla, Tiziana Pressiani, Sabina Sangaletti, Claudio Tripodo, Sara Morlacchi, Mariangela Storto, Rosalinda Trovato, Cristina Travelli, Antonio Sica, Paola Portararo, Stefano Ugel, Armando A. Genazzani, Lorenza Rimassa, Travelli C., Consonni F.M., Sangaletti S., Storto M., Morlacchi S., Grolla A.A., Galli U., Tron G.C., Portararo P., Rimassa L., Pressiani T., Mazzone M., Trovato R., Ugel S., Bronte V., Tripodo C., Colombo M.P., Genazzani A.A., and Sica A.
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,medicine.medical_treatment ,Nude ,Nicotinamide phosphoribosyltransferase ,Apoptosis ,Colorectal Neoplasm ,Inbred C57BL ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Tumor Cells, Cultured ,Hematopoiesi ,Nicotinamide Phosphoribosyltransferase ,Inbred BALB C ,Mice, Inbred BALB C ,Cultured ,biology ,Sarcoma ,Tumor Cells ,Haematopoiesis ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Sirtuin ,Female ,Sarcoma, Experimental ,Colorectal Neoplasms ,Animals ,Cell Proliferation ,Hematopoiesis ,Humans ,Mammary Neoplasms, Experimental ,Mice, Inbred C57BL ,Mice, Nude ,Myeloid-Derived Suppressor Cells ,NAD ,Signal Transduction ,Xenograft Model Antitumor Assays ,Human ,Experimental ,03 medical and health sciences ,medicine ,Myeloid-Derived Suppressor Cell ,Animal ,Mammary Neoplasms ,Apoptosi ,Immunotherapy ,030104 developmental biology ,chemistry ,biology.protein ,Cancer research ,Myeloid-derived Suppressor Cell ,NAD+ kinase ,Bone marrow - Abstract
Cancer induces alteration of hematopoiesis to fuel disease progression. We report that in tumor-bearing mice the macrophage colony-stimulating factor elevates the myeloid cell levels of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD salvage pathway, which acts as negative regulator of the CXCR4 retention axis of hematopoietic cells in the bone marrow. NAMPT inhibits CXCR4 through a NAD/Sirtuin 1–mediated inactivation of HIF1α-driven CXCR4 gene transcription, leading to mobilization of immature myeloid-derived suppressor cells (MDSC) and enhancing their production of suppressive nitric oxide. Pharmacologic inhibition or myeloid-specific ablation of NAMPT prevented MDSC mobilization, reactivated specific antitumor immunity, and enhanced the antitumor activity of immune checkpoint inhibitors. Our findings identify NAMPT as a metabolic gate of MDSC precursor function, providing new opportunities to reverse tumor immunosuppression and to restore clinical efficacy of immunotherapy in patients with cancer. Significance: These findings identify NAMPT as a metabolic gate of MDSC precursor function, providing new opportunities to reverse tumor immunosuppression and to restore clinical efficacy of immunotherapy in cancer patients.
- Published
- 2019
- Full Text
- View/download PDF
10. Fatal cytokine release syndrome by an aberrant FLIP/STAT3 axis
- Author
-
Manuela Iezzi, Katia Donadello, Zheng-Li Shi, Stefano Ugel, Stefania Canè, Ying Chen, Marco Chilosi, Ido Amit, Peter J. Murray, Vincenzo Bronte, Francesca Hofer, Enrico Polati, Evelina Tacconelli, Piera Amelia Iezzi, Andrea Capece, Francesco De Sanctis, Alessandra Fiore, Francesco Domenico Del Pizzo, Leonardo Gottin, Serena Zilio, Silvio Bicciato, Chiara Musiu, Simone Caligola, Roza Maria Barouni, Alessia Lamolinara, Domenico Angelucci, Ildo Polidoro, Paolo Serafini, Cristina Frusteri, Andrea Grilli, Annalisa Adamo, and Domenico Girelli
- Subjects
Male ,medicine.medical_treatment ,C-FLIP ,Inbred C57BL ,Mice ,80 and over ,Medicine ,FLICE-INHIBITORY PROTEINS ,STAT3 ,Aged, 80 and over ,Caspase 8 ,biology ,DEATH ,Middle Aged ,Pathophysiology ,APOPTOSIS ,Cytokine release syndrome ,Cytokine ,Cytokines ,Female ,medicine.symptom ,Cytokine Release Syndrome ,Signal Transduction ,EXPRESSION ,STAT3 Transcription Factor ,Inflammation ,Lung injury ,Article ,Immune system ,Animals ,Humans ,Interleukin 6 ,Molecular Biology ,Aged ,TRANSPLANTATION ,business.industry ,SARS-CoV-2 ,FLICE-INHIBITORY PROTEINS, C-FLIP, NUCLEAR-LOCALIZATION, APOPTOSIS, INFLAMMATION, MACROPHAGE, EXPRESSION, COVID-19, DEATH, TRANSPLANTATION ,NUCLEAR-LOCALIZATION ,COVID-19 ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,Flip ,Immunology ,biology.protein ,MACROPHAGE ,business - Abstract
Inflammatory responses rapidly detect pathogen invasion and mount a regulated reaction. However, dysregulated anti-pathogen immune responses can provoke life-threatening inflammatory pathologies collectively known as cytokine release syndrome (CRS), exemplified by key clinical phenotypes unearthed during the SARS-Cov-2 pandemic. The underlying pathophysiology of CRS remains elusive. We found that FLIP, a protein that controls caspase-8 death pathways, was highly expressed in myeloid cells of COVID-19 lungs. FLIP controlled CRS by fueling a STAT3-dependent inflammatory program. Indeed, constitutive expression of a viral FLIP homologue in myeloid cells triggered a STAT3-linked, progressive and fatal inflammatory syndrome in mice, characterized by elevated cytokine output, lymphopenia, lung injury and multiple organ dysfunctions that mimicked human CRS. As STAT3-targeting approaches relieved inflammation, immune disorders, and organ failures in these mice, targeted intervention towards this pathway could suppress the lethal CRS inflammatory state.One sentence summaryFLIP-expressing myeloid cells are key drivers of CRS through aberrant overexpression of STAT3 pathway. STAT3-targeting is effective in mitigating CRS like severe COVID-19.
- Published
- 2021
11. Arginase 1-Based Immune Modulatory Vaccines Induce Anticancer Immunity and Synergize with Anti-PD-1 Checkpoint Blockade
- Author
-
Evelina Martinenaite, Annalisa Adamo, Stine Emilie Weis-Banke, Katharina L. Kopp, Maria Perez-Penco, Stefano Ugel, Marion Chapellier, Vincenzo Bronte, Mie Linder Hübbe, Cristina Frusteri, Marco Carretta, Mia Aaboe Jørgensen, Mai-Britt Zocca, Francesco De Sanctis, Ayako Wakatsuki Pedersen, Manuela Iezzi, Mads Hald Andersen, and Daniel Hargbøll Madsen
- Subjects
Cancer Research ,medicine.medical_treatment ,Immunology ,complex mixtures ,Proinflammatory cytokine ,Mice ,Immune system ,Immunity ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,Medicine ,Animals ,Humans ,Myeloid Cells ,Tumor microenvironment ,Vaccines ,Arginase ,business.industry ,ELISPOT ,Immunotherapy ,Xenograft Model Antitumor Assays ,Blockade ,Immunosurveillance ,Cancer research ,Female ,business - Abstract
Expression of the L-arginine catabolizing enzyme arginase 1 (ARG1) is a central immunosuppressive mechanism mediated by tumor-educated myeloid cells. Increased activity of ARG1 promotes the formation of an immunosuppressive microenvironment and leads to a more aggressive phenotype in many cancers. Intrinsic T-cell immunity against ARG1-derived epitopes in the peripheral blood of cancer patients and healthy subjects has previously been demonstrated. To evaluate the antitumor efficacy of ARG1-derived peptide vaccines as a monotherapy and as a combinational therapy with checkpoint blockade, different in vivo syngeneic mouse tumor models were utilized. To evaluate the antitumor effects, flow cytometry analysis and IHC were performed on tumors, and ELISPOT assays were performed to characterize immune responses. We show that ARG1-targeting therapeutic vaccines were able to activate endogenous antitumor immunity in several in vivo syngeneic mouse tumor models and to modulate the cell composition of the tumor microenvironment without causing any associated side effects or systemic toxicity. ARG1-targeting vaccines in combination with anti–PD-1 also resulted in increased T-cell infiltration, decreased ARG1 expression, reduced suppressive function of tumor-educated myeloid cells, and a shift in the M1/M2 ratio of tumor-infiltrating macrophages. These results indicated that the induced shift toward a more proinflammatory microenvironment by ARG1-targeting immunotherapy favors effective tumor control when combined with anti–PD-1 checkpoint blockade. Our data illustrate the ability of ARG1-based immune modulatory vaccination to elicit antigen-specific immunosurveillance and imply the feasibility of this novel immunotherapeutic approach for clinical translation.
- Published
- 2021
12. Monocytes in the Tumor Microenvironment
- Author
-
Stefania Canè, Vincenzo Bronte, Stefano Ugel, and Francesco De Sanctis
- Subjects
0301 basic medicine ,Angiogenesis ,medicine.medical_treatment ,Biology ,Monocytes ,Pathology and Forensic Medicine ,Immune tolerance ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,medicine ,Tumor Microenvironment ,cancer ,Animals ,Humans ,innate immunity ,Tumor microenvironment ,Innate immune system ,immunotherapy ,monocytes ,tumor microenvironment ,Effector ,Cancer ,Immunotherapy ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Tumor Escape - Abstract
Immunotherapy has revolutionized cancer treatment over the past decade. Nonetheless, prolonged survival is limited to relatively few patients. Cancers enforce a multifaceted immune-suppressive network whose nature is progressively shaped by systemic and local cues during tumor development. Monocytes bridge innate and adaptive immune responses and can affect the tumor microenvironment through various mechanisms that induce immune tolerance, angiogenesis, and increased dissemination of tumor cells. Yet monocytes can also give rise to antitumor effectors and activate antigen-presenting cells. This yin-yang activity relies on the plasticity of monocytes in response to environmental stimuli. In this review, we summarize current knowledge of the ontogeny, heterogeneity, and functions of monocytes and monocyte-derived cells in cancer, pinpointing the main pathways that are important for modeling the immunosuppressive tumor microenvironment.
- Published
- 2021
13. GM-CSF Nitration Is a New Driver of Myeloid Suppressor Cell Activity in Tumors
- Author
-
Bianca Calì, Andrielly H. R. Agnellini, Chiara Cioccarelli, Ricardo Sanchez-Rodriguez, Andrea Predonzani, Giulia Ilaria Toffolo, Antonella Viola, Vincenzo Bronte, Giorgio Arrigoni, Francesco Zonta, Laura Albertoni, Claudia Mescoli, Ilaria Marigo, and Barbara Molon
- Subjects
Myeloid ,Colorectal cancer ,T cell ,Immunology ,Biology ,Immunomodulation ,Mice ,Immune system ,Immunity ,Cell Line, Tumor ,Neoplasms ,medicine ,Immunology and Allergy ,Animals ,tumor microenvironment ,Original Research ,Tumor microenvironment ,immunosuppression ,Myeloid-Derived Suppressor Cells ,Cancer ,Granulocyte-Macrophage Colony-Stimulating Factor ,Cell Differentiation ,RC581-607 ,medicine.disease ,cytokines ,post-translational modification ,reactive nitrogen species ,Disease Models, Animal ,medicine.anatomical_structure ,Tumor progression ,Cancer research ,Disease Susceptibility ,Immunologic diseases. Allergy ,Protein Processing, Post-Translational ,Biomarkers ,Signal Transduction - Abstract
Reactive oxygen species, including RNS, contribute to the control of multiple immune cell functions within the tumor microenvironment (TME). Tumor-infiltrating myeloid cells (TIMs) represent the archetype of tolerogenic cells that actively contribute to dismantle effective immunity against cancer. TIMs inhibit T cell functions and promote tumor progression by several mechanisms including the amplification of the oxidative/nitrosative stress within the TME. In tumors, TIM expansion and differentiation is regulated by the granulocyte-macrophage colony-stimulating factor (GM-CSF), which is produced by cancer and immune cells. Nevertheless, the role of GM-CSF in tumors has not yet been fully elucidated. In this study, we show that GM-CSF activity is significantly affected by RNS-triggered post-translational modifications. The nitration of a single tryptophan residue in the sequence of GM-CSF nourishes the expansion of highly immunosuppressive myeloid subsets in tumor-bearing hosts. Importantly, tumors from colorectal cancer patients express higher levels of nitrated tryptophan compared to non-neoplastic tissues. Collectively, our data identify a novel and selective target that can be exploited to remodel the TME and foster protective immunity against cancer.
- Published
- 2021
14. The pathogenic role of epithelial and endothelial cells in early-phase COVID-19 pneumonia: victims and partners in crime
- Author
-
Alessandra Dubini, Vincenzo Bronte, Giulio Rossi, Guido Martignoni, Claudia Ravaglia, Venerino Poletti, Claudio Doglioni, Federica Pedica, Sara Piciucchi, Marco Chilosi, and Giovanni Pizzolo
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Biopsy ,Cell Communication ,Review Article ,Pulmonary compliance ,B7-H1 Antigen ,Pathology and Forensic Medicine ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,COVID-19 pneumonia ,Phosphorylation ,Diffuse alveolar damage ,Lung ,medicine.diagnostic_test ,business.industry ,COVID-19 ,Endothelial Cells ,Epithelial Cells ,Hyperplasia ,Prognosis ,medicine.disease ,Pneumonia ,030104 developmental biology ,medicine.anatomical_structure ,Respiratory failure ,030220 oncology & carcinogenesis ,Cytokines ,Infection ,business ,Signal Transduction - Abstract
Current understanding of the complex pathogenesis of COVID-19 interstitial pneumonia pathogenesis in the light of biopsies carried out in early/moderate phase and histology data obtained at postmortem analysis is discussed. In autopsies the most observed pattern is diffuse alveolar damage with alveolar-epithelial type-II cell hyperplasia, hyaline membranes, and frequent thromboembolic disease. However, these observations cannot explain some clinical, radiological and physiopathological features observed in SARS-CoV-2 interstitial pneumonia, including the occurrence of vascular enlargement on CT and preserved lung compliance in subjects even presenting with or developing respiratory failure. Histological investigation on early-phase pneumonia on perioperative samples and lung biopsies revealed peculiar morphological and morpho-phenotypical changes including hyper-expression of phosphorylated STAT3 and immune checkpoint molecules (PD-L1 and IDO) in alveolar-epithelial and endothelial cells. These features might explain in part these discrepancies.
- Published
- 2021
- Full Text
- View/download PDF
15. Phenotypical Characterization and Isolation of Tumor-Derived Mouse Myeloid-Derived Suppressor Cells
- Author
-
Stefano Ugel, Chiara Musiu, Roza Maria Barouni, Vincenzo Bronte, and Stefania Canè
- Subjects
0301 basic medicine ,Myeloid-derived suppressor cells (MDSCs) ,Myeloid ,Cell ,Population ,Biology ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,law ,medicine ,Myeloid-derived suppressor cells (MDSCs), Tumor digestion, Immunosuppression, ELISPOT, Cell sorting ,education ,education.field_of_study ,ELISPOT ,Cell sorting ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,Tumor digestion ,030220 oncology & carcinogenesis ,Cancer research ,Myeloid-derived Suppressor Cell ,Suppressor ,Immunosuppression - Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population composed of mature and immature cells of myeloid origin that play a major role in tumor progression by inhibiting the antitumor immune responses mediated by T cells. In this chapter, we describe protocols for isolation, phenotypical and functional evaluation of MDSCs isolated from mouse tumors, with the aim at unifying and standardizing protocols set up by different laboratories.
- Published
- 2020
- Full Text
- View/download PDF
16. Deciphering the state of immune silence in fatal COVID-19 patients
- Author
-
Federica Facciotti, Francesco De Sanctis, Alessandra Fiore, Cristina Anselmi, Evelina Tacconelli, Rosalinda Trovato, Davide Gibellini, Pasquale De Nardo, Stefano Ugel, Pierre Bost, Simone Caligola, Leonardo Gottin, Roza Maria Barouni, Ido Amit, Enrico Polati, Alessia Lamolinara, Katia Donadello, Monica Castellucci, David Eyal, Annarita Mazzariol, Stefania Canè, Benno Schwikowski, Manuela Iezzi, Vincenzo Bronte, Biologie systémique - Systems Biology, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Weizmann Institute of Science [Rehovot, Israël], University and Hospital Trust of Verona, Università degli studi di Verona = University of Verona (UNIVR), University of G. D Annunzio of Chieti-Pescara [Chieti, Italy], European Institute of Oncology IRCCS [Milan, Italy] (EIO), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), University of Verona (UNIVR), Bost, P, De Sanctis, F, Cane, S, Ugel, S, Donadello, K, Castellucci, M, Eyal, D, Fiore, A, Anselmi, C, Barouni, R, Trovato, R, Caligola, S, Lamolinara, A, Iezzi, M, Facciotti, F, Mazzariol, A, Gibellini, D, De Nardo, P, Tacconelli, E, Gottin, L, Polati, E, Schwikowski, B, Amit, I, and Bronte, V
- Subjects
Male ,0301 basic medicine ,Myeloid ,Neutrophils ,T-Lymphocytes ,General Physics and Astronomy ,Disease ,CD8-Positive T-Lymphocytes ,Monocyte ,Monocytes ,0302 clinical medicine ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Medicine ,Myeloid Cells ,CD4-positive T cells ,Myeloid Cell ,Aged, 80 and over ,0303 health sciences ,Multidisciplinary ,Neutrophil ,Middle Aged ,3. Good health ,Cytokine release syndrome ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Female ,Case-Control Studie ,Viral load ,Human ,Coronavirus disease 2019 (COVID-19) ,Science ,Article ,General Biochemistry, Genetics and Molecular Biology ,Sepsis ,03 medical and health sciences ,Immune system ,Humans ,In patient ,Cytokine ,Aged ,030304 developmental biology ,Hemophagocytic lymphohistiocytosis ,Lung ,SARS-CoV-2 ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Case-control study ,COVID-19 ,CD8-Positive T-Lymphocyte ,General Chemistry ,medicine.disease ,030104 developmental biology ,T-Lymphocyte ,Viral infection ,Case-Control Studies ,Immunology ,business ,Ex vivo ,030217 neurology & neurosurgery - Abstract
Since the beginning of the SARS-CoV-2 pandemic, COVID-19 appeared as a unique disease with unconventional tissue and systemic immune features. Here we show a COVID-19 immune signature associated with severity by integrating single-cell RNA-seq analysis from blood samples and broncho-alveolar lavage fluids with clinical, immunological and functional ex vivo data. This signature is characterized by lung accumulation of naïve lymphoid cells associated with a systemic expansion and activation of myeloid cells. Myeloid-driven immune suppression is a hallmark of COVID-19 evolution, highlighting arginase-1 expression with immune regulatory features of monocytes. Monocyte-dependent and neutrophil-dependent immune suppression loss is associated with fatal clinical outcome in severe patients. Additionally, our analysis shows a lung CXCR6+ effector memory T cell subset is associated with better prognosis in patients with severe COVID-19. In summary, COVID-19-induced myeloid dysregulation and lymphoid impairment establish a condition of ‘immune silence’ in patients with critical COVID-19., Integrated studies of matched tissue sites and cell types in COVID-19 patients are important to define the immune mechanisms of pathology. Here, the authors describe an immune signature in fatal COVID-19 patients harmonizing single-cell RNA sequencing of blood and matched BAL cells with deep clinical, immunological and functional data.
- Published
- 2020
- Full Text
- View/download PDF
17. Acute Lung injury evolution in Covid-19
- Author
-
Giulio Rossi, Antonella Arcadu, Vanni Agnoletti, Luca Donati, Federica Pedica, Alessandra Dubini, Claudio Doglioni, Marco Chilosi, Elisabetta Fabbri, Vittorio Sambri, Silvia Puglisi, Claudia Ravaglia, Venerino Poletti, Carmela Grosso, Sara Piciucchi, Stefano Maitan, Vincenzo Bronte, Stefano Ugel, Giovanni Pizzolo, Simona di Cesare, Lorenza Pecciarini, Athol U. Wells, Emanuele Russo, Antonio Vizzuso, Giovanni Poletti, Franco Stella, and Emiliano Gamberini
- Subjects
Pathology ,medicine.medical_specialty ,Lung ,business.industry ,Microangiopathy ,Lung biopsy ,Hyperplasia ,Lung injury ,medicine.disease ,Pathogenesis ,medicine.anatomical_structure ,medicine ,Cytokine storm ,business ,Diffuse alveolar damage - Abstract
BackgroundPathogenesis of Coronavirus disease 2019 (Covid-19) is poorly understood. Most histologic studies come from post-mortem analysis, with existing data indicating that histologic features of acute respiratory distress syndrome are typically present in fatal cases. However, this observation may be misleading, due to confounding factors in pre-terminal disease, including injury resulting from prolonged mechanical ventilation. Ante-mortem lung biopsy may provide major pathogenetic insights, potentially providing a basis for novel treatment approaches.AimThis comparative, multicenter, prospective, observational study was planned to identify ante-mortem histological profile and immunohistochemical features of lung tissue in patients with Covid-19 in early and late phases of the disease, including markers of inflammatory cells and major pathways involved in the cytokine storm triggering.MethodsEnrolled patients underwent lung biopsy, according to the study protocol approved by local Ethical Committee, either within 15 days of the first symptoms appearing (early phase) or after >15 days (more advanced disease). Key exclusion criteria were excessive or uncorrectable bleeding risk and cardiovascular disease with heart failure. Lung samples were obtained by conventional transbronchial biopsy, trans-bronchial lung cryobiopsy or surgical lung biopsy.Results23 patients were enrolled: 12 patients underwent lung biopsy within 15 days and 11 patients more than 15 days after the onset of symptoms. Early biopsies were characterized by spots of patchy acute lung injury (ALI) with alveolar type II cells hyperplasia and significant vascular abnormalities (disordered angiogenesis with alveolar capillary hyperplasia, luminal enlargement and thickened walls of pulmonary venules, perivascular CD4-T-cell infiltration), with no hyaline membranes. In the later stages, the alveolar architecture appeared disrupted, with areas of organizing ALI, venular congestion and capillary thromboembolic microangiopathy. Striking phenotypic features were demonstrated in hyperplastic pneumocytes and endothelial cells, including the expression of phospho-STAT3 and molecules involved in immunoinhibitory signals (PD-L1 and IDO-1). Alveolar macrophages exhibited macrophage-related markers (CD68, CD11c, CD14) together with unusual markers, such as DC-Lamp/CD208, CD206, CD123/IL3AR.ConclusionA morphologically distinct “Covid pattern” was identified in the earlier stages of the disease, with prominent epithelial and endothelial cell abnormalities, that may be potentially reversible, differing strikingly from findings in classical diffuse alveolar damage. These observations may have major therapeutic implications, justifying studies of early interventions aimed at mitigating inflammatory organ injury.
- Published
- 2020
- Full Text
- View/download PDF
18. Aptamers against mouse and human tumor-infiltrating myeloid cells as reagents for targeted chemotherapy
- Author
-
Jimmy Caroli, Serena Zilio, Paolo Serafini, Tan A. Ince, Silvio Bicciato, Vincenzo Bronte, Adriana C. De La Fuente, Dimitri Van Simaeys, Donald T. Weed, and Emilia Maria Cristina Mazza
- Subjects
Aptamer ,Antineoplastic Agents ,Article ,Mice ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Medicine ,Doxorubicin ,Fibrosarcoma ,Tumor microenvironment ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Myeloid-Derived Suppressor Cells ,General Medicine ,Aptamers, Nucleotide ,medicine.disease ,Head and neck squamous-cell carcinoma ,Blood chemistry ,Head and Neck Neoplasms ,Cancer research ,Indicators and Reagents ,business ,Ex vivo ,Systematic evolution of ligands by exponential enrichment ,medicine.drug - Abstract
Local delivery of anticancer agents has the potential to maximize treatment efficacy and minimize the acute and long-term systemic toxicities. Here we used unsupervised systematic evolution of ligands by exponential enrichment (SELEX) to identify four RNA aptamers that specifically recognized mouse and human myeloid cells infiltrating tumors but not their peripheral or circulating counterparts in multiple mouse models and from patients with head and neck squamous cell carcinoma (HNSCC). The use of these aptamers conjugated to doxorubicin enhanced the accumulation and bystander release of the chemotherapeutic drug in both primary and metastatic tumor sites in breast and fibrosarcoma mouse models. In the 4T1 mammary carcinoma model, these doxorubicin-conjugated aptamers outperformed Doxil, the first clinically approved highly optimized nanoparticle for targeted chemotherapy, by promoting tumor regression with no detected toxicity measured as weight loss and by blood chemistry after just 3 administrations. These RNA aptamers recognized tumor infiltrating myeloid cells in a variety of mouse tumors in vivo and from human HNSCC ex vivo. This suggests their use for the detection of myeloid-derived suppressor cells (MDSCs) in human and for a targeted delivery of chemotherapy to the tumor microenvironment in multiple malignancies.
- Published
- 2020
- Full Text
- View/download PDF
19. Emerging Trends in COVID-19 Treatment: Learning from Inflammatory Conditions Associated with Cellular Therapies
- Author
-
Patrick J. Hanley, Patricia R. M. Rocco, Rachele Ciccocioppo, Jaap Jan Boelens, Catherine M. Bollard, Vincenzo Bronte, Bruce L. Levine, Daniel J. Weiss, and Maria Cancio
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Hemophagocytic ,Passive ,0302 clinical medicine ,Adrenal Cortex Hormones ,Immune Reconstitution Inflammatory Syndrome ,Global health ,Immunology and Allergy ,Killer Cells ,Genetics(clinical) ,Viral ,Hyperimmune response ,Genetics (clinical) ,Lymphohistiocytosis ,Plasmapheresis ,Cellular Therapy ,Killer Cells, Natural ,Cytokine release syndrome ,STAT Transcription Factors ,Oncology ,030220 oncology & carcinogenesis ,Natural ,Immunotherapy ,Coronavirus Infections ,Cytokine Release Syndrome ,CRS ,HLH ,Secondary Hemophagocytic Lymphohistiocytosis ,medicine.medical_specialty ,Immunology ,Pneumonia, Viral ,Context (language use) ,Lymphohistiocytosis, Hemophagocytic ,Article ,03 medical and health sciences ,Immune reconstitution inflammatory syndrome ,medicine ,Humans ,Intensive care medicine ,Cytokine release ,Pandemics ,Transplantation ,business.industry ,Interleukin-6 ,SARS-CoV-2 ,Immunization, Passive ,COVID-19 ,IRIS ,Cell Biology ,Pneumonia ,medicine.disease ,COVID-19 Drug Treatment ,Clinical trial ,030104 developmental biology ,Immunization ,business ,Interleukin-1 - Abstract
Coronavirus disease 2019 (SARS-CoV2) is an active global health threat for which treatments are desperately being sought. Even though most people infected experience mild to moderate respiratory symptoms and recover with supportive care, certain vulnerable hosts develop severe clinical deterioration. While several drugs are currently being investigated in clinical trials, there are currently no approved treatments or vaccines for COVID-19 and hence there is an unmet need to explore additional therapeutic options. At least three inflammatory disorders or syndromes associated with immune dysfunction have been described in the context of cellular therapy. Specifically, Cytokine Release Syndrome (CRS), Immune Reconstitution Inflammatory Syndrome (IRIS), and Secondary Hemophagocytic Lymphohistiocytosis (sHLH) all have clinical and laboratory characteristics in common with COVID19 and associated therapies that could be worth testing in the context of clinical trials. Here we discuss these diseases, their management, and potential applications of these treatment in the context of COVID-19. We also discuss current cellular therapies that are being evaluated for the treatment of COVID-19 and/or its associated symptoms.
- Published
- 2020
20. mTOR and STAT3 Pathway Hyper-Activation is Associated with evated Interleukin-6 Levels in Patients with Shwachman-Diamond Syndrome: Further Evidence of Lymphoid Lineage Impairment
- Author
-
Elena Marinelli Busilacchi, Claudio Sorio, Alice Giacomazzi, Tiziana Cestari, Valentino Bezzerri, Monica Borgatti, Seth J. Corey, Marco Cipolli, Giovanna D'Amico, Vincenzo Bronte, Antonella Poloni, Antonio Vella, Elisabetta D'Aversa, Marisole Allegri, Martina Api, Benedetta Fabrizzi, Giulia Breveglieri, Antonio Benedetti, Chiara Bovo, and Gloria Bedini
- Subjects
0301 basic medicine ,lymphocytes ,Cancer Research ,Myeloid ,Bone Marrow Failure Syndromes ,Lymphocytes ,MTOR ,STAT3 ,lcsh:RC254-282 ,Article ,stat3 ,bone marrow failure syndromes ,NO ,03 medical and health sciences ,0302 clinical medicine ,mtor ,medicine ,Interleukin 6 ,mTOR ,PI3K/AKT/mTOR pathway ,Shwachman–Diamond syndrome ,biology ,business.industry ,Lymphoblast ,Mesenchymal stem cell ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,n/a ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Bone marrow ,business ,human activities - Abstract
Keywords: STAT3, mTOR, Bone Marrow Failure Syndromes, lymphocytes
- Published
- 2020
21. Macrophages Instruct Aberrant Glycosylation in Colon Cancer by Chemokine and Cytokine Signals
- Author
-
Vincenzo Bronte
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Glycosylation ,Colorectal cancer ,medicine.medical_treatment ,Immunology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,medicine ,CCL17 ,Humans ,biology ,Macrophages ,Mucin-1 ,medicine.disease ,Colitis ,Ulcerative colitis ,digestive system diseases ,030104 developmental biology ,Cytokine ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Colonic Neoplasms ,biology.protein ,Cancer research ,Cytokines ,Colitis, Ulcerative ,Chemokines - Abstract
Patients with ulcerative colitis (UC) have an increased risk of developing colitis-associated colon cancer (CACC). Changes in glycosylation of the oncoprotein MUC1 commonly occur in chronic inflammation, including UC, and this abnormally glycosylated MUC1 promotes cancer development and progression. It is not known what causes changes in glycosylation of MUC1. Gene expression profiling of myeloid cells in inflamed and malignant colon tissues showed increased expression levels of inflammatory macrophage-associated cytokines compared to normal tissues. We analyzed the involvement of macrophage-associated cytokines in the induction of aberrant MUC1 glycoforms. A co-culture system was used to examine the effects of M1 and M2 macrophages on glycosylation-related enzymes in colon cancer cells. M2-like macrophages induced the expression of the glycosyltransferase ST6GALNAC1, an enzyme that adds sialic acid to O-linked GalNAc residues, promoting the formation of tumor-associated sialyl-Tn (sTn) O-glycans. Immunostaining of UC and CACC tissue samples confirmed the elevated number of M2-like macrophages as well as high expression of ST6GALNAC1 and the altered MUC1-sTn glycoform on colon cells. Cytokine arrays and blocking antibody experiments indicated that the macrophage-dependent ST6GALNAC1 activation was mediated by IL-13 and CCL17. We demonstrated that IL-13 promoted phosphorylation of STAT6 to activate transcription of ST6GALNAC1. A computational model of signaling pathways was assembled and used to test IL-13 inhibition as a possible therapy. Our findings revealed a novel cellular cross-talk between colon cells and macrophages within the inflamed and malignant colon that contributes to the pathogenesis of UC and CACC.
- Published
- 2020
22. Platelets promote thromboinflammation in SARS-CoV-2 pneumonia
- Author
-
Giancarlo Mansueto, Francesco Dima, Manuela Iezzi, Stefania Canè, Simone De Nitto, Cristiano Fava, Marco Benati, Evelina Tacconelli, Giovanni Poli, Vincenzo Bronte, Fulvia Mazzaferri, Pietro Minuz, Giuseppe Lippi, Francesco Taus, Elena Carrara, Varvara Petrova, Simone Romano, Roza Maria Barouni, Gian Luca Salvagno, and Andrea Dalbeni
- Subjects
Male ,0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,blood platelets ,Pneumonia, Viral ,Inflammation ,030204 cardiovascular system & hematology ,Betacoronavirus ,03 medical and health sciences ,0302 clinical medicine ,Cytokines metabolism ,Thromboembolism ,Humans ,Medicine ,Platelet ,Pandemics ,Pulmonary thrombosis ,thrombosis ,Aged ,Aged, 80 and over ,SARS-CoV-2 ,business.industry ,COVID-19 ,Middle Aged ,Flow Cytometry ,Prognosis ,medicine.disease ,Thrombosis ,Immunity, Innate ,interferons ,Pneumonia ,030104 developmental biology ,inflammation ,Immunology ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Cytokines ,Female ,medicine.symptom ,Coronavirus Infections ,Cardiology and Cardiovascular Medicine ,business ,monocytes ,Megakaryocytes ,Clinical and Population Studies - Abstract
Supplemental Digital Content is available in the text., Objective: Pulmonary thrombosis is observed in severe acute respiratory syndrome coronavirus 2 pneumonia. Aim was to investigate whether subpopulations of platelets were programmed to procoagulant and inflammatory activities in coronavirus disease 2019 (COVID-19) patients with pneumonia, without comorbidities predisposing to thromboembolism. Approach and Results: Overall, 37 patients and 28 healthy subjects were studied. Platelet-leukocyte aggregates, platelet-derived microvesicles, the expression of P-selectin, and active fibrinogen receptor on platelets were quantified by flow cytometry. The profile of 45 cytokines, chemokines, and growth factors released by platelets was defined by immunoassay. The contribution of platelets to coagulation factor activity was selectively measured. Numerous platelet-monocyte (mean±SE, 67.9±4.9%, n=17 versus 19.4±3.0%, n=22; P
- Published
- 2020
23. Detection and functional evaluation of arginase-1 isolated from human PMNs and murine MDSC
- Author
-
Vincenzo Bronte and Stefania Canè
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Functional evaluation ,Tumor microenvironment ,Arginase ,medicine.medical_treatment ,030303 biophysics ,Immunosuppression ,Immunotherapy ,Biology ,Arginase inhibitors ,Human neutrophils ,Murine myeloid-derived suppressor cells ,law.invention ,03 medical and health sciences ,Immune system ,Enzyme ,chemistry ,law ,medicine ,Cancer research ,Suppressor - Abstract
Immunotherapy has emerged as a potent alternative for cancer treatment, unfortunately, the clinical benefit remains limited to few patients and immunotherapy resistance due to immunosuppressive tumor microenvironment represents the major reason of such a failure. Arginase-1 is one of the enzymes contributing to the establishment of such immunosuppression. Among the human immune cells, polymorphonuclear cells (PMNs) represent the major source of arginase-1, while myeloid-derived suppressor cells (MDSCs) are the main arginase-1 producing cells in mice. Due to arginase-1 potential impact in dampening the immune response, there is a growing interest in assaying arginase-1 levels and functions. Thus, in this chapter we propose how to evaluate the expression and activity of arginase in human peripheral blood-derived PMNs and in MDSCs isolated from tumor-bearing mice.
- Published
- 2020
24. Wnt–β-catenin as an epigenetic switcher in colonic Treg cells
- Author
-
Vincenzo Bronte and Stefania Canè
- Subjects
0301 basic medicine ,B catenin ,business.industry ,Colorectal cancer ,Immunology ,Wnt signaling pathway ,Tregs ,medicine.disease ,Treg cell ,Inflammatory bowel disease ,digestive system diseases ,Proinflammatory cytokine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,RAR-related orphan receptor gamma ,Catenin ,medicine ,Cancer research ,Immunology and Allergy ,Epigenetics ,B catenin, Tregs, epigenetic ,business ,epigenetic ,030215 immunology - Abstract
In the colonic environment, sustained Wnt–β-catenin activation in regulatory T cells promotes epigenetic rewiring toward proinflammatory RORγt+ Treg cells, whose expansion parallels the disease progression from inflammatory bowel disease (IBD) to manifest colorectal cancer (CRC).
- Published
- 2021
- Full Text
- View/download PDF
25. The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO
- Author
-
Ursula Grohmann, Stefano Ugel, Vincenzo Bronte, and Giada Mondanelli
- Subjects
EXPRESSION ,0301 basic medicine ,medicine.medical_treatment ,INHIBITION ,Arginase 1 (ARG1) and indoleamine 2 ,Biology ,INDOLEAMINE 2,3-DIOXYGENASE ,SUPPRESSOR-CELLS ,T-CELLS ,TUMOR MICROENVIRONMENT ,TRYPTOPHAN CATABOLISM ,ARGINASE ACTIVITY ,ARGININE ,TOLERANCE ,03 medical and health sciences ,3-dioxygenase 1 (IDO1) ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Transforming Growth Factor beta ,Immunity ,Neoplasms ,Drug Discovery ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Indoleamine 2,3-dioxygenase ,INDOLEAMINE 2 ,Pharmacology ,chemistry.chemical_classification ,Tumor microenvironment ,Arginase ,3-DIOXYGENASE ,Tryptophan ,Peripheral tolerance ,Dendritic Cells ,Arginase 1 (ARG1) and indoleamine 2,3-dioxygenase 1 (IDO1) ,biochemical phenomena, metabolism, and nutrition ,3. Good health ,Cell biology ,Amino acid ,030104 developmental biology ,Enzyme ,chemistry ,030220 oncology & carcinogenesis ,Immunology - Abstract
Some enzymes degrading amino acids have evolved in mammals to dampen immune responses and maintain peripheral tolerance. The enzymes metabolizing l-arginine and l-tryptophan are particularly powerful, contributing to restrain immunity towards fetal tissues and establish neonatal tolerance. Solid tumors can hijack these formidable pathways to construct a microenvironment highly unfavorable to anti-tumor T lymphocytes able to recognize them, one of mechanisms for their immune evasion. In this review, we analyze emerging concepts in the cross-talk between cells expressing these enzymes, their immune regulatory functions and pharmacological approaches that can target them to enhance cancer immunotherapy.
- Published
- 2017
- Full Text
- View/download PDF
26. Identifying baseline immune-related biomarkers to predict clinical outcome of immunotherapy
- Author
-
Vincenzo Bronte, Vaios Karanikas, Marcus O. Butler, Lisa H. Butterfield, Brent A. Hanks, Jérôme Galon, Lance D. Miller, Isabelle Tanneau, Jon M. Wigginton, Samir N. Khleif, Dolores J. Schendel, John M. Kirkwood, Sacha Gnjatic, Mary L. Disis, Leif Håkansson, and Laura Rosa Brunet
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Immunology ,Cancer immunotherapy ,Review ,Bioinformatics ,Outcome (game theory) ,lcsh:RC254-282 ,03 medical and health sciences ,Immune checkpoint inhibitors ,0302 clinical medicine ,Immune system ,Immunity ,Antigens, Neoplasm ,Neoplasms ,medicine ,Biomarkers, Tumor ,Immunology and Allergy ,Humans ,Baseline (configuration management) ,Baseline immunity ,Biomarkers ,Tumor microenvironment ,Pharmacology ,B-Lymphocytes ,business.industry ,Myeloid-Derived Suppressor Cells ,Cancer ,Immunotherapy ,medicine.disease ,Prognosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Molecular Medicine ,business - Abstract
As cancer strikes, individuals vary not only in terms of factors that contribute to its occurrence and development, but as importantly, in their capacity to respond to treatment. While exciting new therapeutic options that mobilize the immune system against cancer have led to breakthroughs for a variety of malignancies, success is limited to a subset of patients. Pre-existing immunological features of both the host and the tumor may contribute to how patients will eventually fare with immunotherapy. A broad understanding of baseline immunity, both in the periphery and in the tumor microenvironment, is needed in order to fully realize the potential of cancer immunotherapy. Such interrogation of the tumor, blood, and host immune parameters prior to treatment is expected to identify biomarkers predictive of clinical outcome as well as to elucidate why some patients fail to respond to immunotherapy. To approach these opportunities for progress, the Society for Immunotherapy of Cancer (SITC) reconvened the Immune Biomarkers Task Force. Comprised of an international multidisciplinary panel of experts, Working Group 4 sought to make recommendations that focus on the complexity of the tumor microenvironment, with its diversity of immune genes, proteins, cells, and pathways naturally present at baseline and in circulation, and novel tools to aid in such broad analyses.
- Published
- 2017
- Full Text
- View/download PDF
27. GCN2 drives macrophage and MDSC function and immunosuppression in the tumor microenvironment
- Author
-
Marie Jo Halaby, Vincenzo Bronte, Andreas Kloetgen, Trevor J. Pugh, David G. Brooks, Kebria Hezaveh, M. Teresa Ciudad, Tracy L. McGaha, Daniel D. De Carvalho, Mengdi Guo, David H. Munn, Sara Lamorte, Aristotelis Tsirigos, Bethany L. MacLeod, Marcus O. Butler, Stefano Ugel, Tiago Medina, Ankur Chakravarthy, and Pamela S. Ohashi
- Subjects
0301 basic medicine ,Small interfering RNA ,Myeloid ,T cell ,Immunology ,Macrophage polarization ,CD8-Positive T-Lymphocytes ,Protein Serine-Threonine Kinases ,Biology ,Article ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology, tumor Immunology ,Melanoma ,Transcription factor ,Cells, Cultured ,Tumor microenvironment ,tumor Immunology ,Macrophages ,Myeloid-Derived Suppressor Cells ,General Medicine ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research - Abstract
General control nonderepressible 2 (GCN2) is an environmental sensor controlling transcription and translation in response to nutrient availability. Although GCN2 is a putative therapeutic target for immuno-oncology, its role in shaping the immune response to tumors is poorly understood. Here, we used mass cytometry, transcriptomics, and transcription factor-binding analysis to determine the functional impact of GCN2 on the myeloid phenotype and immune responses in melanoma. We found that myeloid-lineage deletion of GCN2 drives a shift in the phenotype of tumor-associated macrophages and myeloid-derived suppressor cells (MDSCs) that promotes antitumor immunity. Time-of-flight mass cytometry (CyTOF) and single-cell RNA sequencing showed that this was due to changes in the immune microenvironment with increased proinflammatory activation of macrophages and MDSCs and interferon-γ expression in intratumoral CD8+ T cells. Mechanistically, GCN2 altered myeloid function by promoting increased translation of the transcription factor CREB-2/ATF4, which was required for maturation and polarization of macrophages and MDSCs in both mice and humans, whereas targeting Atf4 by small interfering RNA knockdown reduced tumor growth. Last, analysis of patients with cutaneous melanoma showed that GCN2-dependent transcriptional signatures correlated with macrophage polarization, T cell infiltrates, and overall survival. Thus, these data reveal a previously unknown dependence of tumors on myeloid GCN2 signals for protection from immune attack.
- Published
- 2019
- Full Text
- View/download PDF
28. Sustained Type I interferon signaling as a mechanism of resistance to PD-1 blockade
- Author
-
Takahiro Yamazaki, Paule Opolon, Connie P.M. Duong, Pierre Olivier Gaudreau, Gabriele Madonna, Vancheswaran Gopalakrishnan, Antje Sucker, Miles C. Andrews, Dirk Schadendorf, Laurence Zitvogel, Meriem Messaoudene, Aurélien Marabelle, Christophe Klein, Guido Kroemer, Ilaria Marigo, Sonia Becharef, Gautier Stoll, Maria Paula Roberti, Vincenzo Bronte, Shin Foong Ngiow, David Enot, Stefano Ugel, Marie Vétizou, Jennifer A. Wargo, Jean-Charles Soria, Armelle Prévost-Blondel, Mark J. Smyth, Loic Verlingue, Romain Daillère, Paolo A. Ascierto, Gladys Ferrere, Nicolas Jacquelot, Alexander M.M. Eggermont, Immunologie des tumeurs et immunothérapie (UMR 1015), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Dynamique de l'information génétique : bases fondamentales et cancer (DIG CANCER), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS), UMR 996 - DHU Hépatinov, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Gustave Roussy (IGR), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Service de Bioinformatique (CURIE-BIOINFO), Institut Curie [Paris], Veneto Institute of Oncology, IOV - IRCCS, Innovation en Immonomonitoring et Immunothérapie (P3I), Centre de Lutte contre le Cancer (CLCC) de Lyon-Centre Léon Bérard [Lyon], Institut Cochin (IC UM3 (UMR 8104 / U1016)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Plateforme histo-cyto-pathologies (PHCP), Analyse moléculaire, modélisation et imagerie de la maladie cancéreuse (AMMICa), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Istituto Nazionale Tumori IRCCS, Istituto Nazionale Tumori Fondazione Pascale [Naples, Italy], German Cancer Consortium, Department of Dermatology [Essen, Germany], University Hospital [Essen, Germany]-West German Cancer Center [Essen, Germany]-University Duisburg-Essen [Germany], Oncologie thoracique, Département de médecine oncologique [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Apoptose, cancer et immunité (Equipe labellisée Ligue contre le cancer - CRC - Inserm U1138), Institut Gustave Roussy (IGR)-Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École pratique des hautes études (EPHE), The University of Texas M.D. Anderson Cancer Center [Houston], Centre d'Investigation Clinique en Biotherapie des cancers (CIC 1428 , CBT 507 ), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Curie-Centre National de la Recherche Scientifique (CNRS), École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Institut Curie, Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)
- Subjects
medicine.medical_treatment ,T-Lymphocytes ,[SDV]Life Sciences [q-bio] ,Programmed Cell Death 1 Receptor ,Medizin ,Drug Resistance ,Nitric Oxide Synthase Type II ,Drug resistance ,Kaplan-Meier Estimate ,Inbred C57BL ,T-Lymphocytes, Regulatory ,B7-H1 Antigen ,Mice ,0302 clinical medicine ,Interferon ,Neoplasms ,Monoclonal ,Animals ,Antibodies, Monoclonal ,Cell Line, Tumor ,Dendritic Cells ,Drug Resistance, Neoplasm ,Humans ,Interferon Type I ,Melanoma ,Mice, Inbred C57BL ,Signal Transduction ,ComputingMilieux_MISCELLANEOUS ,0303 health sciences ,Tumor ,biology ,Nitric oxide synthase 2 ,Regulatory ,3. Good health ,Tumour immunology ,medicine.drug ,Cancer microenvironment ,Immunology ,Article ,Antibodies ,Cell Line ,03 medical and health sciences ,medicine ,Antigen-presenting cell ,Molecular Biology ,030304 developmental biology ,Cell Biology ,Immunotherapy ,medicine.disease ,Blockade ,Cell culture ,Cancer research ,biology.protein ,Neoplasm ,030217 neurology & neurosurgery - Abstract
PD-1 blockade represents a major therapeutic avenue in anticancer immunotherapy. Delineating mechanisms of secondary resistance to this strategy is increasingly important. Here, we identified the deleterious role of signaling via the type I interferon (IFN) receptor in tumor and antigen presenting cells, that induced the expression of nitric oxide synthase 2 (NOS2), associated with intratumor accumulation of regulatory T cells (Treg) and myeloid cells and acquired resistance to anti-PD-1 monoclonal antibody (mAb). Sustained IFNβ transcription was observed in resistant tumors, in turn inducing PD-L1 and NOS2 expression in both tumor and dendritic cells (DC). Whereas PD-L1 was not involved in secondary resistance to anti-PD-1 mAb, pharmacological or genetic inhibition of NOS2 maintained long-term control of tumors by PD-1 blockade, through reduction of Treg and DC activation. Resistance to immunotherapies, including anti-PD-1 mAb in melanoma patients, was also correlated with the induction of a type I IFN signature. Hence, the role of type I IFN in response to PD-1 blockade should be revisited as sustained type I IFN signaling may contribute to resistance to therapy. CA extern
- Published
- 2019
- Full Text
- View/download PDF
29. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3
- Author
-
Salvatore Paiella, Rosalinda Trovato, Rita T. Lawlor, Rosalba Giugno, Roberto Salvia, Carmine Carbone, Geny Piro, Francesca Hofer, Ornella Poffe, Samantha Solito, Stefania Canè, Vincenzo Bronte, Luciano Cascione, Susanna Mandruzzato, Silvia Sartoris, Francesco De Sanctis, Alessandra Fiore, Laura Pinton, Cristina Anselmi, Stefano Ugel, Aldo Scarpa, Claudio Bassi, Sara Sartori, and Vincenzo Corbo
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Myeloid ,Pancreatic ductal adenocarcinoma (PDAC) ,medicine.medical_treatment ,Lipopolysaccharide Receptors ,Cell Separation ,0302 clinical medicine ,Tumor Microenvironment ,Immunology and Allergy ,Medicine ,Prospective Studies ,Cells, Cultured ,Innate immunity ,Aged, 80 and over ,Tumor-associated immunosuppression ,Immunosuppression ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Flow Cytometry ,Prognosis ,Tumor progression ,medicine.anatomical_structure ,Oncology ,Myeloid-derived suppressor cells (MDSC) ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Research Article ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,STAT3 Transcription Factor ,CD14 ,Primary Cell Culture ,Immunology ,lcsh:RC254-282 ,03 medical and health sciences ,Immune system ,Humans ,Pancreas ,Survival rate ,Aged ,Pharmacology ,Arginase ,business.industry ,Gene Expression Profiling ,Myeloid-Derived Suppressor Cells ,Monocyte ,Gene signature ,Survival Analysis ,Pancreatic Neoplasms ,030104 developmental biology ,Cancer research ,Myeloid-derived Suppressor Cell ,Tumor Escape ,business - Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly devastating disease with an overall 5-year survival rate of less than 8%. New evidence indicates that PDAC cells release pro-inflammatory metabolites that induce a marked alteration of normal hematopoiesis, favoring the expansion and accumulation of myeloid-derived suppressor cells (MDSCs). We report here that PDAC patients show increased levels of both circulating and tumor-infiltrating MDSC-like cells. Methods The frequency of MDSC subsets in the peripheral blood was determined by flow cytometry in three independent cohorts of PDAC patients (total analyzed patients, n = 117). Frequency of circulating MDSCs was correlated with overall survival of PDAC patients. We also analyzed the frequency of tumor-infiltrating MDSC and the immune landscape in fresh biopsies. Purified myeloid cell subsets were tested in vitro for their T-cell suppressive capacity. Results Correlation with clinical data revealed that MDSC frequency was significantly associated with a shorter patients’ overall survival and metastatic disease. However, the immunosuppressive activity of purified MDSCs was detectable only in some patients and mainly limited to the monocytic subset. A transcriptome analysis of the immunosuppressive M-MDSCs highlighted a distinct gene signature in which STAT3 was crucial for monocyte re-programming. Suppressive M-MDSCs can be characterized as circulating STAT3/arginase1-expressing CD14+ cells. Conclusion MDSC analysis aids in defining the immune landscape of PDAC patients for a more appropriate diagnosis, stratification and treatment.
- Published
- 2019
- Full Text
- View/download PDF
30. MAP Kinase inhibition reshapes tumor microenvironment of mouse pancreatic cancer by depleting anti-inflammatory macrophages
- Author
-
Pietro Delfino, Christian Neander, Dea Filippini, Sabrina L. D’Agosto, Caterina Vicentini, Elena Fiorini, Francesca Lupo, Claudia Fiorini, Borislav Rusev, Gael D. Temgue Tane, Francesco De Sanctis, Michele Simbolo, Stefano Ugel, Vincenzo Bronte, Matteo Fassan, Rita T. Lawlor, Federico Boschi, Stefano Barbi, Aldo Scarpa, Phyllis F. Cheung, Jens T. Siveke, and Vincenzo Corbo
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Trametinib ,Tumor microenvironment ,medicine.diagnostic_test ,biology ,Chemistry ,education ,medicine.disease ,3. Good health ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,In vivo ,030220 oncology & carcinogenesis ,Mitogen-activated protein kinase ,Pancreatic cancer ,medicine ,biology.protein ,Cancer research - Abstract
The RAF/MEK/ERK (MAP Kinase) pathway is the index oncogenic signaling towards which many compounds have been developed and tested for the treatment of KRAS-driven cancers, including pancreatic ductal adenocarcinoma (PDA). Here, we explored the immunological changes induced by targeted MEK1/2 inhibition (MEKi) using trametinib in preclinical mouse models of PDA. We evaluated the dynamic changes in the immune contexture of mouse PDA upon MEKi using a multidimensional approach (mRNA analyses, flow cytometry, and immunophenotyping). Effect of MEKi on the viability and metabolism of macrophages was investigated in vitro. We showed that transcriptional signatures of MAP Kinase activation are enriched in aggressive human PDA subtype (squamous/basal-like/quasimesenchymal), while short term MEKi treatment in mouse PDA induced subtype switching. Integrative mRNA expression and immunophenotypic analyses showed that MEKi reshapes the immune landscape of PDA by depleting rather than reprogramming macrophages, while augmenting infiltration by neutrophils. Depletion of macrophages is observed early in the course of in vivo treatment and is at least partially due to their higher sensitivity to MEKi. Tumor-associated macrophages were consistently reported to interfere with gemcitabine uptake by PDA cells. Here, our in vivo studies show a superior antitumor activity upon combination of MEKi and gemcitabine using a sequential rather than simultaneous dosing protocol. Our results show that MEK inhibition induces a dramatic remodeling of the tumor microenvironment of mouse PDA through depletion of macrophages, which substantially improves the antitumor activity of gemcitabine.
- Published
- 2019
- Full Text
- View/download PDF
31. Immunoevolution of mouse pancreatic organoid isografts from preinvasive to metastatic disease
- Author
-
Francesca Lupo, Borislav Rusev, Vincenzo Bronte, Francesco De Sanctis, Sabrina D' Agosto, Giampaolo Tortora, Pietro Delfino, Dea Filippini, Michele Simbolo, Aldo Scarpa, Stefano Ugel, Geny Piro, Vincenzo Corbo, Michele Milella, Lisa Veghini, Carmine Carbone, and Cinzia Cantù
- Subjects
0301 basic medicine ,Male ,T-CELL EXCLUSION ,medicine.medical_treatment ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,NT5E ,Mice ,0302 clinical medicine ,TUMOR ,Tumor Microenvironment ,lcsh:Science ,Cancer ,education.field_of_study ,DUCTAL ADENOCARCINOMA ,7 C7 ,CANCER ,SUPPRESSOR ,INHIBITION ,IMMUNOTHERAPY ,MAINTENANCE ,PROGENITORS ,Multidisciplinary ,Isografts ,Neoplasm Proteins ,Female ,Carcinoma, Pancreatic Ductal ,Stromal cell ,Population ,Mice, Transgenic ,Biology ,Article ,03 medical and health sciences ,health services administration ,medicine ,Organoid ,Animals ,Humans ,Progenitor cell ,education ,Cancer models ,Pancreas ,Macrophages ,lcsh:R ,Immunotherapy ,Pancreatic Neoplasms ,030104 developmental biology ,Cancer cell ,Cancer research ,lcsh:Q ,030217 neurology & neurosurgery ,CD8 - Abstract
Pancreatic ductal adenocarcinoma (PDA) has a highly immunosuppressive microenvironment, which is contributed by the complex interaction between cancer cells and a heterogeneous population of stromal cells. Therefore, facile and trackable models are needed for integrative and dynamic interrogation of cancer-stroma interaction. Here, we tracked the immunoevolution of PDA in a genetically-defined transplantable model of mouse pancreatic tumour organoids that recapitulates the progression of the disease from early preinvasive lesions to metastatic carcinomas. We demonstrated that organoid-derived isografts (ODI) can be used as a biological source of biomarkers (NT5E, TGFB1, FN1, and ITGA5) of aggressive molecular subtypes of human PDA. In ODI, infiltration from leukocytes is an early event during progression of the disease as observed for autochthonous models. Neoplastic progression was associated to accumulation of Maf+ macrophages, which inversely correlated with CD8+ T cells infiltration. Consistently, levels of MAF were enriched in human PDA subtypes characterized by abundance of macrophage-related transcripts and indicated poor patients’ survival. Density of MAF+ macrophages was higher in human PDA tissues compared to preinvasive lesions. Our results suggest that ODIs represent a suitable system for genotypic-immunophenotypic studies and support the hypothesis of MAF+ macrophages as a prominent immunosuppressive population in PDA.
- Published
- 2019
32. Co-delivery of RNAi and Chemokine by Polyarginine Nanocapsules Enables the Modulation of Myeloid-Derived Suppressor Cells
- Author
-
Vincenzo Bronte, Ben J. Boyd, Ilaria Marigo, Marcos Garcia-Fuentes, María J. Alonso, Maria Stella Sasso, Adriana M. Ledo, Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas, and Universidade de Santiago de Compostela. Departamento de Farmacoloxía, Farmacia e Tecnoloxía Farmacéutica
- Subjects
Chemokine ,Cells ,medicine.medical_treatment ,Pharmaceutical Science ,MDSCs ,02 engineering and technology ,Inbred C57BL ,Small Interfering ,Nanocapsules ,Cell Line ,03 medical and health sciences ,Mice ,Cancer immunotherapy ,RNA interference ,Cell Line, Tumor ,C/EBPβ ,CCL2 ,RNAi ,medicine ,Animals ,Humans ,Immunologic Factors ,RNA, Small Interfering ,Cells, Cultured ,Chemokine CCL2 ,030304 developmental biology ,Mice, Inbred C57BL ,Myeloid-Derived Suppressor Cells ,Peptides ,RAW 264.7 Cells ,RNA Interference ,0303 health sciences ,Tumor ,Cultured ,biology ,Chemistry ,021001 nanoscience & nanotechnology ,Drug vehicle ,Cell biology ,Monocyte differentiation ,Drug delivery ,biology.protein ,Myeloid-derived Suppressor Cell ,RNA ,0210 nano-technology - Abstract
Myeloid-Derived Suppressor Cells (MDSCs), immunosuppressive cells that promote tumor growth, represent an attractive target in cancer immunotherapy. However, the clinical success of this strategy is limited by the lack of efficient drug delivery vehicles targeting this cell compartment. The objective of this work was to develop a delivery carrier, multilayer polymer nanocapsules, with the capacity to co-encapsulate two types of immunomodulatory drugs, a chemokine and an RNAi sequence, aimed at reverting MDSC-mediated immunosuppression. The chemokine CCL2, intended to attract monocyte-macrophage MDSCs, was encapsulated within the L2 inverse micellar aqueous domains of the lipid core of these nanocapsules. On the other hand, two different RNAi sequences that modulate the CCAAT/enhancer-binding protein beta (C/EBPβ) pathway, shC/EBPβ and miR 142-3p, were successfully associated to their polymer shell. These RNAi sequences were covered by subsequent layers of polyarginine and hyaluronic acid, thereby creating multi-layered assemblies that protected them and facilitated their targeted delivery. The in vitro studies performed in primary MDSCs cultures showed the capacity of miR 142-3p-loaded nanocapsules to reduce the highly immunosuppressive monocyte- macrophage subset. Additionally, the encapsulation of CCL2 within the nanocapsules induced a potent monocyte-macrophage chemoattraction that could be used to direct the therapy to these cell subsets. Finally, in vitro and in vivo studies showed the capacity of shC/EBPβ-loaded nanocapsules to downregulate C/EBPβ levels in MDSCs and to reduce monocyte differentiation into tumor-associated macrophages in an MCA-203 fibrosarcoma mice model. In conclusion, the multilayer polymer nanocapsules described here are efficient vehicles for the co-delivery of proteins and RNA, and are potential candidates as nanomedicines for the modulation of MDSCs. This work was supported by LYMPHOTARG (ERA-NET EuroNanoMed ProgramISCIII, ref PS09/02670) and NICHE projects (ERA-NET EuroNanoMed II framework by ISCIII through CIBER- BBN), which received funding from the European Union's Seventh Framework Programme. The SAXS measurements were completed on the SAXS/WAXS beamLine at the Australian Synchrotron. A. M. L. was supported by a FPU fellowship from the Spanish Ministry of Education SI
- Published
- 2019
33. Danger-associated extracellular ATP counters MDSC therapeutic efficacy in acute GVHD
- Author
-
Jonathan S. Serody, Jenny P.-Y. Ting, Christopher J. Farady, Vincenzo Bronte, Michael Loschi, Brent H. Koehn, Takao Iwawaki, Asim Saha, Govindarajan Thangavelu, Mark E. Cooper, Cameron McDonald-Hyman, Jamie Panthera, Lie Ma, Keli L. Hippen, Walker Krepps, Bruce R. Blazar, Josh Dysthe, Jeffrey S. Miller, Stephen C. Jameson, Robert Zeiser, Peter J. Murray, William J. Murphy, David H. Munn, Michael Zaiken, and Geoffrey R. Hill
- Subjects
Inflammasomes ,Receptor expression ,Immunology ,Graft vs Host Disease ,Inflammation ,Biochemistry ,Mice ,Adenosine Triphosphate ,In vivo ,medicine ,Extracellular ,Animals ,Receptor ,Mice, Knockout ,Apyrase ,Chemistry ,Myeloid-Derived Suppressor Cells ,Inflammasome ,Cell Biology ,Hematology ,medicine.disease ,Graft-versus-host disease ,Cancer research ,Female ,medicine.symptom ,medicine.drug - Abstract
Myeloid-derived suppressor cells (MDSCs) can subdue inflammation. In mice with acute graft-versus-host disease (GVHD), donor MDSC infusion enhances survival that is only partial and transient because of MDSC inflammasome activation early posttransfer, resulting in differentiation and loss of suppressor function. Here we demonstrate that conditioning regimen-induced adenosine triphosphate (ATP) release is a primary driver of MDSC dysfunction through ATP receptor (P2x7R) engagement and NLR pyrin family domain 3 (NLRP3) inflammasome activation. P2x7R or NLRP3 knockout (KO) donor MDSCs provided significantly higher survival than wild-type (WT) MDSCs. Although in vivo pharmacologic targeting of NLRP3 or P2x7R promoted recipient survival, indicating in vivo biologic effects, no synergistic survival advantage was seen when combined with MDSCs. Because activated inflammasomes release mature interleukin-1�� (IL-1��), we expected that IL-1�� KO donor MDSCs would be superior in subverting GVHD, but such MDSCs proved inferior relative to WT. IL-1�� release and IL-1 receptor expression was required for optimal MDSC function, and exogenous IL-1�� added to suppression assays that included MDSCs increased suppressor potency. These data indicate that prolonged systemic NLRP3 inflammasome inhibition and decreased IL-1�� could diminish survival in GVHD. However, loss of inflammasome activation and IL-1�� release restricted to MDSCs rather than systemic inhibition allowed non-MDSC IL-1�� signaling, improving survival. Extracellular ATP catalysis with peritransplant apyrase administered into the peritoneum, the ATP release site, synergized with WT MDSCs, as did regulatory T-cell infusion, which we showed reduced but did not eliminate MDSC inflammasome activation, as assessed with a novel inflammasome reporter strain. These findings will inform future clinical using MDSCs to decrease alloresponses in inflammatory environments.
- Published
- 2019
- Full Text
- View/download PDF
34. Characterization of Myeloid-derived Suppressor Cells in a Patient With Lung Adenocarcinoma Undergoing Durvalumab Treatment: A Case Report
- Author
-
Emilio Bria, Vincenzo Bronte, Giacomo Bertelli, Stefano Ugel, Rosalinda Trovato, Sara Pilotto, and Michele Milella
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,Durvalumab ,Lung Neoplasms ,medicine.medical_treatment ,MDSC ,Adenocarcinoma of Lung ,NSCLC ,B7-H1 Antigen ,Tumor-induced immunosuppression ,Immunomodulation ,Antineoplastic Agents, Immunological ,Immunotherapy ,Predictive biomarkers ,Medicine ,Humans ,Lung cancer ,Neoplasm Staging ,Settore MED/06 - ONCOLOGIA MEDICA ,Lung ,biology ,business.industry ,Myeloid-Derived Suppressor Cells ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Prognosis ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Myeloid-derived Suppressor Cell ,Adenocarcinoma ,Neoplasm staging ,Female ,Antibody ,business - Published
- 2019
35. Close to the Bone: Tissue-Specific Checkpoint Immunotherapy Evasion
- Author
-
Vincenzo Bronte
- Subjects
medicine.medical_treatment ,Cell ,Immunology ,Biology ,Bone tissue ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Immunology, Immunotheray ,medicine ,Tumor Microenvironment ,Humans ,Immunotheray ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,Evasion (ethics) ,Immune checkpoint ,medicine.anatomical_structure ,Cancer research ,bacteria ,030217 neurology & neurosurgery - Abstract
Primary or secondary resistance to immune checkpoint therapy is often analyzed in primary tumors rather than metastases. In this issue of Cell, Jiao et al. show that the site of metastatic diffusion matters and bones empower a particularly hostile environment.
- Published
- 2019
36. The endless saga of monocyte diversity
- Author
-
Stefania Canè, Francesco De Sanctis, Silvia Sartoris, Ilaria Marigo, Vincenzo Bronte, Rosalinda Trovato, and Stefano Ugel
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,medicine.medical_treatment ,Immunology ,Cell Plasticity ,Review ,Biology ,Monocyte continuum ,Monocytes ,metastatic niche ,monocyte continuum ,monocytes heterogeneity ,primary tumor ,targeting of monocytes ,03 medical and health sciences ,Primary tumor ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Metastatic niche ,Monocytes heterogeneity ,Targeting of monocytes ,Animals ,CCAAT-Enhancer-Binding Protein-alpha ,Humans ,Immunotherapy ,Myeloid-Derived Suppressor Cells ,Neoplasms ,medicine ,Immunology and Allergy ,Epigenetics ,Tumor microenvironment ,Monocyte ,medicine.disease ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,lcsh:RC581-607 ,CD8 ,030215 immunology - Abstract
Cancer immunotherapy relies on either restoring or activating the function of adaptive immune cells, mainly CD8+ T lymphocytes. Despite impressive clinical success, cancer immunotherapy remains ineffective in many patients due to the establishment of tumor resistance, largely dependent on the nature of tumor microenvironment. There are several cellular and molecular mechanisms at play, and the goal is to identify those that are clinically significant. Among the hematopoietic-derived cells, monocytes are endowed with high plasticity, responsible for their pro- and anti-tumoral function. Indeed, monocytes are involved in several cancer-associated processes such as immune-tolerance, metastatic spread, neoangiogenesis, and chemotherapy resistance; on the other hand, by presenting cancer-associated antigens, they can also promote and sustain anti-tumoral T cell response. Recently, by high throughput technologies, new findings have revealed previously underappreciated, profound transcriptional, epigenetic, and metabolic differences among monocyte subsets, which complement and expand our knowledge on the monocyte ontogeny, recruitment during steady state, and emergency hematopoiesis, as seen in cancer. The subdivision into discrete monocytes subsets, both in mice and humans, appears an oversimplification, whereas continuum subsets development is best for depicting the real condition. In this review, we examine the evidences sustaining the existence of a monocyte heterogeneity along with functional activities, at the primary tumor and at the metastatic niche. In particular, we describe how tumor-derived soluble factors and cell-cell contact reprogram monocyte function. Finally, we point out the role of monocytes in preparing and shaping the metastatic niche and describe relevant targetable molecules altering monocyte activities. We think that exploiting monocyte complexity can help identifying key pathways important for the treatment of cancer and several conditions where these cells are involved.
- Published
- 2019
37. How to Reprogram Myeloma-Associated Macrophages: Target IKZF1
- Author
-
Francesco De Sanctis and Vincenzo Bronte
- Subjects
0301 basic medicine ,Cancer Research ,Macrophages ,Immunology ,Macrophage polarization ,Macrophage Activation ,Biology ,Molecular pathway ,medicine.disease ,Ikaros Transcription Factor ,03 medical and health sciences ,Phenotype ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Interferon Regulatory Factors ,Cancer cell ,Cancer research ,medicine ,Humans ,Multiple Myeloma ,IRF5 ,Multiple myeloma - Abstract
Cancer cells hijack tumor-associated macrophages to sustain their outgrowth. In this issue, Mougiakakos and colleagues identify the IKZF1–IRF4/IRF5 axis as a key molecular pathway regulating macrophage polarization in multiple myeloma. These results suggest targeting IKZF1 could provide a new strategy to reprogram myeloma-associated macrophages toward a tumoricidal and immune-activating phenotype. See article by Mougiakakos et al., p. 265
- Published
- 2021
- Full Text
- View/download PDF
38. Prostate-specific membrane antigen (PSMA) assembles a macromolecular complex regulating growth and survival of prostate cancer cells 'in vitro' and correlating with progression 'in vivo'
- Author
-
Giulio Fracasso, Vincenzo Bronte, Silvia Grasso, Dunia Ramarli, Tiziana Cestari, Marco Colombatti, Matteo Brunelli, Maria Elisa Perico, Hassan Y. Naim, Enrico Moiso, Guido Martignoni, and Enrico Munari
- Subjects
0301 basic medicine ,Male ,Cell Survival ,MAP Kinase Signaling System ,Cell Growth Processes ,Filamin ,urologic and male genital diseases ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cell Line, Tumor ,LNCaP ,BCAR1 ,PSMA ,castration-resistant prostate adenocarcinoma ,p130CAS ,phospho-EGFR receptor ,Medicine ,Humans ,Phosphorylation ,business.industry ,Cell growth ,TOR Serine-Threonine Kinases ,Prostatic Neoplasms ,Prostate-Specific Antigen ,medicine.disease ,ErbB Receptors ,Oncogene Protein v-akt ,Prostate-specific antigen ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Disease Progression ,Kallikreins ,bcl-Associated Death Protein ,business ,Research Paper - Abstract
The expression of Prostate Specific-Membrane Antigen (PSMA) increases in high-grade prostate carcinoma envisaging a role in growth and progression. We show here that clustering PSMA at LNCaP or PC3-PSMA cell membrane activates AKT and MAPK pathways thus promoting proliferation and survival. PSMA activity was dependent on the assembly of a macromolecular complex including filamin A, beta1 integrin, p130CAS, c-Src and EGFR. Within this complex beta1 integrin became activated thereby inducing a c-Src-dependent EGFR phosphorylation at Y1086 and Y1173 EGF-independent residues. Silencing or blocking experiments with drugs demonstrated that all the complex components were required for full PSMA-dependent promotion of cell growth and/or survival in 3D culture, but that p130CAS and EGFR exerted a major role. All PSMA complex components were found assembled in multiple samples of two high-grade prostate carcinomas and associated with EGFR phosphorylation at Y1086. The expression of p130CAS and pEGFRY1086 was thus analysed by tissue micro array in 16 castration-resistant prostate carcinomas selected from 309 carcinomas and stratified from GS 3+4 to GS 5+5. Patients with Gleason Score ≤5 resulted negative whereas those with GS≥5 expressed p130CAS and pEGFRY1086 in 75% and 60% of the cases, respectively. Collectively, our results demonstrate for the first time that PSMA recruits a functionally active complex which is present in high-grade patients. In addition, two components of this complex, p130CAS and the novel pEGFRY1086, correlate with progression in castration-resistant patients and could be therefore useful in therapeutic or surveillance strategies of these patients.
- Published
- 2016
39. Immune suppressive mechanisms in the tumor microenvironment
- Author
-
Vincenzo Bronte and David H. Munn
- Subjects
0301 basic medicine ,Stromal cell ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Biology ,Immunotherapy, Adoptive ,Article ,natural immunosuppressive and tolerogenic mechanisms, tumor-specific T cells ,Cell therapy ,03 medical and health sciences ,Immune system ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,tumor-specific T cells ,natural immunosuppressive and tolerogenic mechanisms ,Tumor microenvironment ,Cancer ,Immunotherapy ,medicine.disease ,Blockade ,030104 developmental biology ,medicine.anatomical_structure - Abstract
Effective immunotherapy, whether by checkpoint blockade or adoptive cell therapy, is limited in most patients by a key barrier: the immunosuppressive tumor microenvironment. Suppression of tumor-specific T cells is orchestrated by the activity of a variety of stromal myeloid and lymphoid cells. These often display inducible suppressive mechanisms that are triggered by the same anti-tumor inflammatory response that the immunotherapy intends to create. Therefore, a more comprehensive understanding of how the immunosuppressive milieu develops and persists is critical in order to harness the full power of immunotherapy of cancer.
- Published
- 2016
- Full Text
- View/download PDF
40. Feasibility of Telomerase-Specific Adoptive T-cell Therapy for B-cell Chronic Lymphocytic Leukemia and Solid Malignancies
- Author
-
Chiara Cavallini, Sara Bobisse, Manuela Iezzi, Federico Boschi, Sara Sandri, Silvia Sartoris, Andrea Sbarbati, Vincenzo Bronte, Kelly Moxley, Giovanna Ferrarini, Maria Teresa Scupoli, Rudi W. Hendriks, Stefano Ugel, Francesco De Sanctis, Alessia Lamolinara, Michael I. Nishimura, Giulio Fracasso, and Pulmonary Medicine
- Subjects
0301 basic medicine ,HLA-A2–restricted T-cell receptor ,Cancer Research ,Adoptive cell transfer ,Telomerase ,mice ,T cell ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Biology ,telomerase ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,medicine ,Cytotoxic T cell ,Animals ,Humans ,B cell ,Immunotherapy ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Mice, Inbred C57BL ,Disease Models, Animal ,in vivo ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Humanized mouse ,Cancer research ,adoptive immunotherapy ,high-avidity T-cell receptor ,Feasibility Studies ,T-Lymphocytes, Cytotoxic - Abstract
Telomerase (TERT) is overexpressed in 80% to 90% of primary tumors and contributes to sustaining the transformed phenotype. The identification of several TERT epitopes in tumor cells has elevated the status of TERT as a potential universal target for selective and broad adoptive immunotherapy. TERT-specific cytotoxic T lymphocytes (CTL) have been detected in the peripheral blood of B-cell chronic lymphocytic leukemia (B-CLL) patients, but display low functional avidity, which limits their clinical utility in adoptive cell transfer approaches. To overcome this key obstacle hindering effective immunotherapy, we isolated an HLA-A2–restricted T-cell receptor (TCR) with high avidity for human TERT from vaccinated HLA-A*0201 transgenic mice. Using several relevant humanized mouse models, we demonstrate that TCR-transduced T cells were able to control human B-CLL progression in vivo and limited tumor growth in several human, solid transplantable cancers. TERT-based adoptive immunotherapy selectively eliminated tumor cells, failed to trigger a self–MHC-restricted fratricide of T cells, and was associated with toxicity against mature granulocytes, but not toward human hematopoietic progenitors in humanized immune reconstituted mice. These data support the feasibility of TERT-based adoptive immunotherapy in clinical oncology, highlighting, for the first time, the possibility of utilizing a high-avidity TCR specific for human TERT. Cancer Res; 76(9); 2540–51. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
41. Methods to Measure MDSC Immune Suppressive Activity In Vitro and In Vivo
- Author
-
Samantha Solito, Stefano Ugel, Francesco De Sanctis, Ilaria Marigo, Laura Pinton, Susanna Mandruzzato, and Vincenzo Bronte
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Myeloid ,immunomagnetic sorting ,T cell ,MDSC ,Immunology ,Biology ,Inbred C57BL ,Lymphocyte Activation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,In vivo ,medicine ,Cytotoxic T cell ,Animals ,Humans ,adoptive cell transfer ,Inbred BALB C ,Mice, Inbred BALB C ,chromium release test ,immunosuppression ,Myeloid-Derived Suppressor Cells ,General Medicine ,Flow Cytometry ,In vitro ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,flow cytometry ,thymidine incorporation ,Immunosuppressive Agents ,Myeloid-derived Suppressor Cell ,Cancer research ,030215 immunology - Abstract
This unit presents methods to assess the immunosuppressive properties of immunoregulatory cells of myeloid origin, such as myeloid-derived suppressor cells (MDSCs), both in vitro and in vivo in mice, as well as in biological samples from cancer patients. These methods could be adapted to test the impact of different suppressive populations on T cell activation, proliferation, and cytotoxic activity; moreover, they could be useful to assess the influence exerted by genetic modifications, chemical inhibitors, and drugs on immune suppressive pathways © 2018 by John Wiley & Sons, Inc.
- Published
- 2018
42. Peripheral blood immunophenotyping in a large cohort of patients with Shwachman-Diamond syndrome
- Author
-
Seth J. Corey, Elena Nicolis, Antonio Vella, Riccardo Ortolani, Marco Cipolli, Valentino Bezzerri, Vincenzo Bronte, Gianfranco Di Gennaro, Benedetta Fabrizzi, and Simone Cesaro
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Myeloid ,Adolescent ,Anemia ,Neutropenia ,Article ,Immunophenotyping ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,neutropenia ,Humans ,Lipomatosis ,Child ,Bone Marrow Diseases ,Shwachman–Diamond syndrome ,B-Lymphocytes ,Shwachman-Diamond syndrome ,bone marrow failure ,double-negative T cells ,immunophenotype ,business.industry ,Bone marrow failure ,Infant ,Hematology ,medicine.disease ,Prognosis ,Shwachman-Diamond Syndrome ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Leukocytes, Mononuclear ,Exocrine Pancreatic Insufficiency ,Female ,Bone marrow ,business ,CD8 ,030215 immunology ,Follow-Up Studies - Abstract
Shwachman-Diamond syndrome (SDS) is one of the more common inherited bone marrow failure syndromes, characterized by neutropenia, occasional thrombocytopenia, and anemia. Bone marrow evaluation reveals an increased number of monocytes and mature B cells along with decreased granulocytes. However, little is known about the subpopulations of peripheral blood cells, and few previous publications have been based on a small number of patients. Here, we report a comprehensive immunophenotypic analysis from a cohort of 37 SDS patients who display impairment mostly in the myeloid compartment with a deficiency also in the number of B cells and CD4/CD8 double-negative T cells.
- Published
- 2018
43. The expanding constellation of immune checkpoints: a DNAMic control by CD155
- Author
-
Vincenzo Bronte
- Subjects
0301 basic medicine ,T-Lymphocytes ,Disease ,03 medical and health sciences ,Immune system ,Downregulation and upregulation ,Killer Cells ,Medicine ,Humans ,Neoplasm Invasiveness ,CD155 ,Tumor Escape ,Up-Regulation ,Killer Cells, Natural ,biology ,business.industry ,Cancer ,General Medicine ,T lymphocyte ,medicine.disease ,030104 developmental biology ,Cancer cell ,Natural ,biology.protein ,Cancer research ,Commentary ,business ,Research Article - Abstract
Critical immune-suppressive pathways beyond programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) require greater attention. Nectins and nectin-like molecules might be promising targets for immunotherapy, since they play critical roles in cell proliferation and migration and exert immunomodulatory functions in pathophysiological conditions. Here, we show CD155 expression in both malignant cells and tumor-infiltrating myeloid cells in humans and mice. Cd155–/– mice displayed reduced tumor growth and metastasis via DNAM-1 upregulation and enhanced effector function of CD8+ T and NK cells, respectively. CD155-deleted tumor cells also displayed slower tumor growth and reduced metastases, demonstrating the importance of a tumor-intrinsic role of CD155. CD155 absence on host and tumor cells exerted an even greater inhibition of tumor growth and metastasis. Blockade of PD-1 or both PD-1 and CTLA4 was more effective in settings in which CD155 was limiting, suggesting the clinical potential of cotargeting PD-L1 and CD155 function.
- Published
- 2018
44. The mesenchymal and myeloid regulation of immunity: Power is nothing without control
- Author
-
Vincenzo Bronte
- Subjects
0301 basic medicine ,Cell signaling ,Immunity, Cellular ,Myeloid ,Cellular differentiation ,Immunology ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Communication ,Biology ,Immunomodulation ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Immunity ,Myeloid cells ,Cancer research ,medicine ,Immunology and Allergy ,Animals ,Humans ,Myeloid Cells - Published
- 2018
45. Induction of immunosuppressive functions and NF-κB by FLIP in monocytes
- Author
-
Alessia Lamolinara, Davide Melisi, Peter J. Murray, Aldo Scarpa, Manuela Iezzi, Giulio Fracasso, Samantha Solito, Carmine Carbone, Fulvia Vascotto, Silvio Bicciato, Bruce R. Blazar, Francesco De Sanctis, Alessandra Fiore, Geny Piro, Ugur Sahin, Giada Mondanelli, Sara Sandri, Vincenzo Bronte, Matteo Fassan, Keli L. Hippen, Stefano Ugel, Rosalinda Trovato, Silvia Sartoris, Ursula Grohmann, Rita T. Lawlor, and Susanna Mandruzzato
- Subjects
0301 basic medicine ,medicine.medical_treatment ,CASP8 and FADD-Like Apoptosis Regulating Protein ,General Physics and Astronomy ,Apoptosis ,Cells, Cultured ,Humans ,Immunosuppression ,Lentivirus ,Lentivirus Infections ,Monocytes ,Myeloid Cells ,NF-kappa B ,Chemistry (all) ,Biochemistry, Genetics and Molecular Biology (all) ,Physics and Astronomy (all) ,Cells ,Cultured ,Macrophage differentiation ,Expression ,Death ,Chemotherapy ,Inhibition ,Cancer ,Activation ,Resistance ,ACTIVATION ,chemistry.chemical_compound ,lcsh:Science ,Multidisciplinary ,DEATH ,CHEMOTHERAPY ,CANCER ,3. Good health ,APOPTOSIS ,medicine.anatomical_structure ,EXPRESSION ,Science ,Necroptosis ,T cell ,INHIBITION ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,MACROPHAGE DIFFERENTIATION ,CELLS ,RESISTANCE ,Immunosuppression Therapy ,NF-κB ,General Chemistry ,Immunotherapy ,030104 developmental biology ,chemistry ,Tumor progression ,Flip ,Cancer research ,lcsh:Q - Abstract
Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells., Signaling and transcriptional regulation of MDSC activity remains largely undefined. Here the authors show that monocytic MDSC immunosuppression is triggered by c-FLIP and requires NFκB, implicate this axis in cancer prognosis and response to therapy, and employ ectopic FLIP to treat immunopathology.
- Published
- 2018
46. Deciphering Macrophage and Monocyte Code to Stratify Human Breast Cancer Patients
- Author
-
Vincenzo Bronte
- Subjects
0301 basic medicine ,Cancer Research ,Breast Neoplasms ,CCL8 ,Article ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,stomatognathic system ,Humans ,Macrophage ,Medicine ,skin and connective tissue diseases ,business.industry ,Macrophages ,Monocyte ,Cancer ,Cell Biology ,Prognosis ,medicine.disease ,Biomarkers ,Female ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,business ,Human breast ,Patient stratification - Abstract
The roles of tumor-associated macrophages (TAMs) and circulating monocytes in human cancer are poorly understood. Here, we show that monocyte subpopulation distribution and transcriptomes are significantly altered by the presence of endometrial and breast cancer. Furthermore, TAMs from endometrial and breast cancers are transcriptionally distinct from monocytes and their respective tissue-resident macrophages. We identified a breast TAM signature that is highly enriched in aggressive breast cancer subtypes and associated with shorter disease-specific survival. We also identified an auto-regulatory loop between TAMs and cancer cells driven by tumor necrosis factor alpha involving SIGLEC1 and CCL8, which is self-reinforcing through the production of CSF1. Together these data provide direct evidence that monocyte and macrophage transcriptional landscapes are perturbed by cancer, reflecting patient outcomes.
- Published
- 2019
- Full Text
- View/download PDF
47. GVHD-associated, inflammasome-mediated loss of function in adoptively transferred myeloid-derived suppressor cells
- Author
-
Jessica M. Haverkamp, Robert Zeiser, Bruce R. Blazar, Vincenzo Bronte, Jenny P.-Y. Ting, Valarie McCullar, William J. Murphy, David H. Munn, Brent H. Koehn, Peter J. Murray, Jakub Tolar, Jeffrey S. Miller, Dmitry I. Gabrilovich, Petya Apostolova, Jonathan S. Serody, and Willie June Brickey
- Subjects
Adoptive cell transfer ,Inflammasomes ,Interleukin-1beta ,Immunology ,Population ,Graft vs Host Disease ,Bone Marrow Cells ,Biology ,graft-versus-host disease (GvHD) ,Biochemistry ,Mice ,Immune system ,inflammatory responses ,immune system diseases ,medicine ,Animals ,Humans ,Myeloid Cells ,education ,Cells, Cultured ,Transplantation ,education.field_of_study ,CD11 Antigens ,Cell Differentiation ,Inflammasome ,Cell Biology ,Hematology ,Myeloid-derived suppressor cells (MDSC), inflammatory responses, graft-versus-host disease (GvHD) ,medicine.disease ,Adoptive Transfer ,Haematopoiesis ,surgical procedures, operative ,Graft-versus-host disease ,medicine.anatomical_structure ,Myeloid-derived suppressor cells (MDSC) ,Myeloid-derived Suppressor Cell ,Bone marrow ,medicine.drug - Abstract
Myeloid-derived suppressor cells (MDSCs) are a naturally occurring immune regulatory population associated with inhibition of ongoing inflammatory responses. In vitro generation of MDSCs from bone marrow has been shown to enhance survival in an acute model of lethal graft-versus-host disease (GVHD). However, donor MDSC infusion only partially ameliorates GVHD lethality. In order to improve the potential therapeutic benefit and ultimately survival outcomes, we set out to investigate the fate of MDSCs after transfer in the setting of acute GVHD (aGVHD). MDSCs transferred to lethally irradiated recipients of allogeneic donor hematopoietic grafts are exposed to an intense inflammatory environment associated with aGVHD, which we now show directly undermines their suppressive capacity. Under a conditioning regimen and GVHD inflammatory settings, MDSCs rapidly lose suppressor function and their potential to inhibit GVHD lethality, which is associated with their induced conversion toward a mature inflammasome-activated state. We find even brief in vitro exposure to inflammasome-activating mediators negates the suppressive potential of cultured murine and human-derived MDSCs. Consistent with a role for the inflammasome, donor MDSCs deficient in the adaptor ASC (apoptosis-associated speck-like protein containing a CARD), which assembles inflammasome complexes, conferred improved survival of mice developing GVHD compared with wild-type donor MDSCs. These data suggest the use of MDSCs as a therapeutic approach for preventing GVHD and other systemic inflammatory conditions will be more effective when combined with approaches limiting in vivo MDSC inflammasome activation, empowering MDSCs to maintain their suppressive potential.
- Published
- 2015
- Full Text
- View/download PDF
48. Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells
- Author
-
Vincenzo Bronte and Ainhoa Arina
- Subjects
autochthonous tumorigenesis ,Adoptive cell transfer ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunology ,Cell Communication ,Major histocompatibility complex ,Immunotherapy, Adoptive ,Immunomodulation ,Interleukin 21 ,Neoplasms ,Tumor Microenvironment ,adoptive T cell therapy (ATT) ,myeloid-derived suppressor cells (MDSC) ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Cytotoxic T cell ,Myeloid Cells ,Tumor microenvironment ,biology ,business.industry ,Immunotherapy ,Adoptive Transfer ,Treatment Outcome ,medicine.anatomical_structure ,Myeloid-derived Suppressor Cell ,biology.protein ,business - Abstract
Novel models of autochthonous tumorigenesis and adoptive T cell therapy (ATT) are providing new clues regarding the pro-tumorigenic and immunosuppressive effects of myeloid-derived suppressor cells (MDSC), and their interaction with T cells. New findings are shifting the perception of the main level at which MDSC act, from direct cell-to-cell suppression to others, such as limiting T cell infiltration. Adoptively transferred, high-avidity T cells recognizing peptides with high-affinity for MHC-I eliminated large tumors. However, low-avidity T cells or low-affinity peptides resulted in failure to eradicate tumors. Manipulation of intratumoral myeloid cells improved the outcome of otherwise unsuccessful ATT. Therefore, therapeutic intervention directed at the tumor stroma might be required when using suboptimal T cells for ATT.
- Published
- 2015
- Full Text
- View/download PDF
49. Interfering with CCL5/CCR5 at the Tumor-Stroma Interface
- Author
-
Emilio Bria and Vincenzo Bronte
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Receptors, CCR5 ,Colorectal cancer ,Macrophage polarization ,Soft Tissue Neoplasms ,CCL5 ,03 medical and health sciences ,0302 clinical medicine ,Receptors ,Humans ,Medicine ,Receptor ,Chemokine CCL5 ,biology ,business.industry ,Cancer ,Cell Biology ,Colorectal Neoplasms ,medicine.disease ,Blockade ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Cancer cell ,biology.protein ,Cancer research ,business ,CCR5 - Abstract
In this issue of Cancer Cell, Halama et al. (2016) further advance chemokine interference as a therapeutic option for cancer by demonstrating the effect of CCR5 blockade in reshaping macrophage polarization toward an anti-tumor functional state in patient-derived tumor models and liver metastases of colorectal cancer patients.
- Published
- 2016
- Full Text
- View/download PDF
50. Bone marrow mesenchymal stromal cells induce nitric oxide synthase-dependent differentiation of CD11b+ cells that expedite hematopoietic recovery
- Author
-
Ilaria Marigo, Francesco Dazzi, Luigi Dolcetti, Marta Serafini, Chun Yin Wang, Vincenzo Bronte, Antonio Galleu, Cristina Trento, Alice Pievani, Trento, C, Marigo, I, Pievani, A, Galleu, A, Dolcetti, L, Wang, C, Serafini, M, Bronte, V, and Dazzi, F
- Subjects
0301 basic medicine ,Stromal cell ,Knockout ,medicine.medical_treatment ,CD34 ,Clinical uses of mesenchymal stem cells ,Nitric Oxide Synthase Type II ,Bone Marrow Cells ,Hematopoietic stem cell transplantation ,Biology ,Inbred C57BL ,Mice ,03 medical and health sciences ,Animals ,CD11b Antigen ,Cell Self Renewal ,Hematopoietic Stem Cell Transplantation ,Homeostasis ,Humans ,Mesenchymal Stem Cells ,Mice, Inbred C57BL ,Mice, Knockout ,Myeloid Cells ,Cell Differentiation ,Hematopoiesis ,Homeostasi ,medicine ,Myeloid Cell ,Animal ,Bone Marrow Microenvironment ,Mesenchymal stem cell ,Hematopoietic stem cell ,Articles ,Hematology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Mesenchymal Stem Cell ,Immunology ,Cancer research ,Bone Marrow Cell ,Bone marrow ,Human - Abstract
Bone marrow microenvironment is fundamental for hematopoietic homeostasis. Numerous efforts have been made to reproduce or manipulate its activity to facilitate engraftment after hematopoietic stem cell transplantation but clinical results remain unconvincing. This probably reflects the complexity of the hematopoietic niche. Recent data have demonstrated the fundamental role of stromal and myeloid cells in regulating hematopoietic stem cell self-renewal and mobilization in the bone marrow. In this study we unveil a novel interaction by which bone marrow mesenchymal stromal cells induce the rapid differentiation of CD11b+ myeloid cells from bone marrow progenitors. Such an activity requires the expression of nitric oxide synthase-2. Importantly, the administration of these mesenchymal stromal cell-educated CD11b+ cells accelerates hematopoietic reconstitution in bone marrow transplant recipients. We conclude that the liaison between mesenchymal stromal cells and myeloid cells is fundamental in hematopoietic homeostasis and suggests that it can be harnessed in clinical transplantation.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.