39 research outputs found

    Tumor derived Microvesicles enhance cross-processing ability of clinical grade Dendritic Cells

    Get PDF
    Tumor cells release extracellular microvesicles (MVs) in the microenvironment to deliver biological signals to neighbouring cells as well as to cells in distant tissues. Tumor-derived MVs appear to play contradictory role promoting both immunosuppression and tumor growth and both evoking tumor specific immune response. Recent evidences indicate that tumor-derived MVs can positively impact Dendritic Cells (DCs) immunogenicity by reprogramming DC antigen processing machinery and intracellular signaling pathways, thus promoting anti-tumor response. DCs are considered pivot cells of the immune system due to their exclusive ability to coordinate the innate and acquired immune responses, cross-present exogenous antigens and prime naĂŻve T cells. DCs are required for the induction and maintenance of long-lasting anti-tumor immunity and their exploitation has been extensively investigated for the design of anti-tumor vaccines. However, the clinical grade culture conditions that are required to generate DCs for therapeutic use can strongly affect their functions. Here, we investigated the immunomodulatory impact of MVs carrying the MUC1 tumor glycoantigen (MVsMUC1) as immunogen formulation on clinical grade DCs grown in X-VIVO 15 (X-DCs). Results indicated that X-DCs displayed reduced performance of the antigen processing machinery in term of diminished phagocytosis and acidification of the phagosomal compartment suggesting an altered immunogenicity of clinical grade DCs. Pulsing DCs with MVsMUC1 restored phagosomal alkalinization, triggering ROS increase. This was not observed when a soluble MUC1 protein was employed (rMUC1). Concurrently, MVsMUC1 internalization by X-DCs allowed MUC1 cross-processing. Most importantly, MVsMUC1 pulsed DCs activated IFNÎł response mediated by MUC1 specific CD8+ T cells. These results strongly support the employment of tumor-derived MVs as immunogen platforms for the implementation of DC-based vaccine

    Triple peptide vaccination as consolidation treatment in women affected by ovarian and breast cancer: clinical and immunological data of a phase I/II clinical trial

    Get PDF
    Vaccination with priming and expansion of tumour reacting T cells is an important therapeutic option to be used in combination with novel checkpoint inhibitors to increase the specificity of the T cell infiltrate and the efficacy of the treatment. In this phase I/II study, 14 high-risk disease-free ovarian (OC) and breast cancer (BC) patients after completion of standard therapies were vaccinated with MUC1, ErbB2 and carcinoembryonic antigen (CEA) HLA-A2+-restricted peptides and Montanide. Patients were subjected to 6 doses of vaccine every two weeks and a recall dose after 3 months. ECOG grade 2 toxicity was observed at the injection site. Eight out of 14 patients showed specific CD8+ T cells to at least one antigen. None of 4 patients vaccinated for compassionate use showed a CD8 activation. An OC patient who suffered from a lymph nodal recurrence, showed specific anti-ErbB2 CD8+ T cells in the bulky aortic lymph nodes suggesting homingof the activated T cells. Results confirm that peptide vaccination strategy is feasible, safe and well tolerated. In particular OC patients appear to show a higher response rate compared to BC patients. Vaccination generates a long-lasting immune response, which is strongly enhanced by recall administrations. The clinical outcome of patients enrolled in the trial appears favourable, having registered no deceased patients with a minimum follow-up of 8 years. These promising data, in line with the results of similar studies, the high compliance of patients observed and the favourable toxicity profile, support future trials of peptide vaccination in clinically disease-free patients who have completed standard treatments

    Tumor-derived microvesicles modulate antigen cross-processing via reactive oxygen species-mediated alkalinization of phagosomal compartment in dendritic cells

    Get PDF
    Dendritic cells (DCs) are the only antigen-presenting cells able to prime naĂŻve T cells and cross-prime antigen-specific CD8+ T cells. Their functionality is a requirement for the induction and maintenance of long-lasting cancer immunity. Albeit intensively investigated, the in vivo mechanisms underlying efficient antigen cross-processing and presentation are not fully understood. Several pieces of evidence indicate that antigen transfer to DCs mediated by microvesicles (MVs) enhances antigen immunogenicity. This mechanism is also relevant for cross-presentation of those tumor-associated glycoproteins such as MUC1 that are blocked in HLA class II compartment when internalized by DCs as soluble molecules. Here, we present pieces of evidence that the internalization of tumor-derived MVs modulates antigen-processing machinery of DCs. Employing MVs derived from ovarian cancer ascites fluid and established tumor cell lines, we show that MV uptake modifies DC phagosomal microenvironment, triggering reactive oxygen species (ROS) accumulation and early alkalinization. Indeed, tumor MVs carry radical species and the MV uptake by DCs counteracts the chemically mediated acidification of the phagosomal compartment. Further pieces of evidence suggest that efficacious antigen cross-priming of the MUC1 antigen carried by the tumor MVs results from the early signaling induced by MV internalization and the function of the antigen-processing machinery of DCs. These results strongly support the hypothesis that tumor-derived MVs impact antigen immunogenicity by tuning the antigen-processing machinery of DCs, besides being carrier of tumor antigens. Furthermore, these findings have important implications for the exploitation of MVs as antigenic cell-free immunogen for DC-based therapeutic strategies

    Tumor-Derived Microvesicles Enhance Cross-Processing Ability of Clinical Grade Dendritic Cells

    Get PDF
    Tumor cells release extracellular microvesicles (MVs) in the microenvironment to deliver biological signals to neighboring cells as well as to cells in distant tissues. Tumor-derived MVs appear to play contradictory role promoting both immunosuppression and tumor growth and both evoking tumor specific immune response. Recent evidences indicate that tumor-derived MVs can positively impact Dendritic Cells (DCs) immunogenicity by reprogramming DC antigen processing machinery and intracellular signaling pathways, thus promoting anti-tumor response. DCs are considered pivot cells of the immune system due to their exclusive ability to coordinate the innate and acquired immune responses, cross-present exogenous antigens, and prime naĂŻve T cells. DCs are required for the induction and maintenance of long-lasting anti-tumor immunity and their exploitation has been extensively investigated for the design of anti-tumor vaccines. However, the clinical grade culture conditions that are required to generate DCs for therapeutic use can strongly affect their functions. Here, we investigated the immunomodulatory impact of MVs carrying the MUC1 tumor glycoantigen (MVsMUC1) as immunogen formulation on clinical grade DCs grown in X-VIVO 15 (X-DCs). Results indicated that X-DCs displayed reduced performance of the antigen processing machinery in term of diminished phagocytosis and acidification of the phagosomal compartment suggesting an altered immunogenicity of clinical grade DCs. Pulsing DCs with MVsMUC1 restored phagosomal alkalinization, triggering ROS increase. This was not observed when a soluble MUC1 protein was employed (rMUC1). Concurrently, MVsMUC1 internalization by X-DCs allowed MUC1 cross-processing. Most importantly, MVsMUC1 pulsed DCs activated IFNÎł response mediated by MUC1 specific CD8+ T cells. These results strongly support the employment of tumor-derived MVs as immunogen platforms for the implementation of DC-based vaccines

    Seasonal modulation of the C-type lectin MGL on human DCs

    No full text
    The C-type lectin MGL is a pathogen recognition receptor, expressed by dendritic cells (DCs) and macrophages (Ms), able to bind GalNAc (Tn)- carrying structures. This receptor also recogni- zed Tn-TAAs that were internalized, processed and presented by DCs to T cells and it acted as an adjuvant on DCs, highlighting its possible ap- plication in anti-cancer vaccination. In this work, we found that this receptor present a seasonal modulation: its expression is higher in winter rather than in summer. The percentage of MGL+ donors displayed a negative trend that dropped to 33% during the summer and increased up to 100% in winter. This modulation could be also ascribed to the circa-annual variation of gluco- corticoids, in fact MGL is up-regulated in pres- ence of dexamethasone in vitro. The seasonal variation of this receptor could be an important point in the field of tumor vaccination strategies.The C-type lectin MGL is a pathogen recognition receptor, expressed by dendritic cells (DCs) and macrophages (Ms), able to bind GalNAc (Tn)- carrying structures. This receptor also recogni- zed Tn-TAAs that were internalized, processed and presented by DCs to T cells and it acted as an adjuvant on DCs, highlighting its possible ap- plication in anti-cancer vaccination. In this work, we found that this receptor present a seasonal modulation: its expression is higher in winter rather than in summer. The percentage of MGL+ donors displayed a negative trend that dropped to 33% during the summer and increased up to 100% in winter. This modulation could be also ascribed to the circa-annual variation of gluco- corticoids, in fact MGL is up-regulated in pres- ence of dexamethasone in vitro. The seasonal variation of this receptor could be an important point in the field of tumor vaccination strategies

    ANTIMICROBIAL LIPASE ANTIBODIES THEIR NUCLEOTIDE AND AMINOACID SEQUENCES AND USES THEREOF”.

    No full text
    Polypeptides and antibodies able to recognize microbial lipases and to enhance the hydrolyitic activity of these enzymes are disclosed. The nucleotide and aminoacid sequences of a preferred embodiment of these antibodies are also provided as seq IDN 1 - 4 in the sequence listing. In the present application the industrial uses of these antibodies are also disclose

    Immune response to the polymorphic epithelial mucin (PEM)

    No full text
    Several tumor associated antigens have been shown to be able to induce an previous termimmunenext termprevious termresponsenext term. The identification of such antigens and the effector mechanisms involved is a first step for the development of useful cancer vaccines. PEM is a glycoprotein localized on the luminal surface ofmost simple epithelia. In cancer cells it is overexpressed and undergoes a process of aberrant glycosylation. Cryptic epitopes within the core protein of the extracellular domain are therefore exposed and could be a target for an previous termimmunenext termprevious termresponsenext term. An MHC unrestricted T cell previous termresponsenext term mediated by CD8 cells was described and explained by the particular structure of the molecule made up of tandem repeats (TR). Lymphocytes from tumor draining lymph nodes from patients with gynecological malignancies were utilized to study both the humoral and cellular previous termimmuneprevious termresponses of these patients. The human antibodies produced by these patients were directed against different epitopes within the TR sequence of PEM and were able to recognize the tumor associated glycoforms of the molecule. T cell clones were isolated that were able to proliferate in the presence of specific peptides corresponding to the TR of PEM presented by autologous B cells. The functional analysis of these clones revealed a Th phenotype. The possible contribution of T helper cells in generating and maintaining anti-tumor immunity opens new possibilities for effective immunological approaches in cancer therapy
    corecore