8 results on '"Sheila Spada"'
Search Results
2. Combination of chemotherapy and PD-1 blockade induces T cell responses to tumor non-mutated neoantigens
- Author
-
Gian Paolo Spinelli, Rosalba Caccavale, Carmela Martire, Ilenia Cammarata, Paolo Visca, Sheila Spada, Felice Giangaspero, Paola Nisticò, Silvia Piconese, Silverio Tomao, Chiara Focaccetti, Fabiana Letizia Cecere, Gabriella Girelli, Julio Rodrigo Giròn Berrìos, Flavia Longo, Federica Buzzacchino, Carmine Mancone, Marino Paroli, Giovanna Peruzzi, Vincenzo Barnaba, Marta Buccilli, Mariangela Panetta, Alessio Grimaldi, Nicoletta D'Alessandris, and Francesco Facciolo
- Subjects
0301 basic medicine ,Male ,Cancer ,immunology ,chemotherapy ,Lung Neoplasms ,medicine.medical_treatment ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Medicine (miscellaneous) ,Settore MED/04 ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,lcsh:QH301-705.5 ,Antigen Presentation ,Immunity, Cellular ,Immune cell death ,Middle Aged ,Combined Modality Therapy ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Pd 1 blockade ,Female ,Immunotherapy ,General Agricultural and Biological Sciences ,medicine.drug ,T cell ,Biology ,complex mixtures ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immunoediting ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Humans ,Aged ,Cisplatin ,Chemotherapy ,In vitro ,030104 developmental biology ,lcsh:Biology (General) ,Apoptosis ,Case-Control Studies ,Cancer research ,Drug Screening Assays, Antitumor ,CD8 - Abstract
Here, we developed an unbiased, functional target-discovery platform to identify immunogenic proteins from primary non-small cell lung cancer (NSCLC) cells that had been induced to apoptosis by cisplatin (CDDP) treatment in vitro, as compared with their live counterparts. Among the multitude of proteins identified, some of them were represented as fragmented proteins in apoptotic tumor cells, and acted as non-mutated neoantigens (NM-neoAgs). Indeed, only the fragmented proteins elicited effective multi-specific CD4+ and CD8+ T cell responses, upon a chemotherapy protocol including CDDP. Importantly, these responses further increased upon anti-PD-1 therapy, and correlated with patients’ survival and decreased PD-1 expression. Cross-presentation assays showed that NM-neoAgs were unveiled in apoptotic tumor cells as the result of caspase-dependent proteolytic activity of cellular proteins. Our study demonstrates that apoptotic tumor cells generate a repertoire of immunogenic NM-neoAgs that could be potentially used for developing effective T cell-based immunotherapy across multiple cancer patients., Grimaldi and Cammarata et al. develop a proteomics-based, target discovery platform to identify immunogenic proteins specific to apoptotic tumor cells. This study highlights the importance of protein modifications in apoptotic tumor cells as a mechanism of generating immunogenic neoantigens that can be targeted for T cell-based immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
3. The actin modulator <scp>hMENA</scp> regulates <scp>GAS</scp> 6‐ <scp>AXL</scp> axis and pro‐tumor cancer/stromal cell cooperation
- Author
-
Francesco Facciolo, Rita T. Lawlor, Sheila Spada, Francesca Di Modugno, Gian Luca Grazi, Anna Di Carlo, Roberta Melchionna, Emily I. Chen, Aldo Scarpa, Barbara Antoniani, Daniel D'Andrea, Isabella Sperduti, Anna Maria Mileo, Michele Milella, Paolo Visca, Enzo Gallo, Mariangela Panetta, Lorenzo Piemonti, Paola Nisticò, Melchionna, Roberta, Spada, Sheila, Di Modugno, Francesca, D'Andrea, Daniel, Di Carlo, Anna, Panetta, Mariangela, Mileo, Anna Maria, Sperduti, Isabella, Antoniani, Barbara, Gallo, Enzo, Lawlor, Rita T, Piemonti, Lorenzo, Visca, Paolo, Milella, Michele, Grazi, Gian Luca, Facciolo, Francesco, Chen, Emily, Scarpa, Aldo, and Nisticò, Paola
- Subjects
Proteomics ,Lung Neoplasms ,actin cytoskeleton ,Stromal cell ,Biology ,AXL ,GAS6 ,cancer-associated fibroblasts ,lung cancer ,Biochemistry ,Article ,NO ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Tandem Mass Spectrometry ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Genetics ,Humans ,LS4_6 ,Epithelial–mesenchymal transition ,Molecular Biology ,Cancer ,030304 developmental biology ,0303 health sciences ,cancer‐associated fibroblasts ,Microfilament Proteins ,Articles ,Actin cytoskeleton ,Actins ,Pancreatic Neoplasms ,Cancer cell ,Cancer research ,Tumor promotion ,Stromal Cells ,Cell Adhesion, Polarity & Cytoskeleton ,Signal transduction ,030217 neurology & neurosurgery ,Chromatography, Liquid - Abstract
The dynamic interplay between cancer cells and cancer‐associated fibroblasts (CAFs) is regulated by multiple signaling pathways, which can lead to cancer progression and therapy resistance. We have previously demonstrated that hMENA, a member of the actin regulatory protein of Ena/VASP family, and its tissue‐specific isoforms influence a number of intracellular signaling pathways related to cancer progression. Here, we report a novel function of hMENA/hMENAΔv6 isoforms in tumor‐promoting CAFs and in the modulation of pro‐tumoral cancer cell/CAF crosstalk via GAS6/AXL axis regulation. LC‐MS/MS proteomic analysis reveals that CAFs that overexpress hMENAΔv6 secrete the AXL ligand GAS6, favoring the invasiveness of AXL‐expressing pancreatic ductal adenocarcinoma (PDAC) and non‐small cell lung cancer (NSCLC) cells. Reciprocally, hMENA/hMENAΔv6 regulates AXL expression in tumor cells, thus sustaining GAS6‐AXL axis, reported as crucial in EMT, immune evasion, and drug resistance. Clinically, we found that a high hMENA/GAS6/AXL gene expression signature is associated with a poor prognosis in PDAC and NSCLC. We propose that hMENA contributes to cancer progression through paracrine tumor–stroma crosstalk, with far‐reaching prognostic and therapeutic implications for NSCLC and PDAC., This study reveals that inhibition of hMENA/hMENADv6 expression reduces pro‐tumor CAF‐cancer cell crosstalk and inhibits cancer cell invasiveness.
- Published
- 2020
- Full Text
- View/download PDF
4. hMENA isoforms impact NSCLC patient outcome through fibronectin/β1 integrin axis
- Author
-
Paola Nisticò, Anna Di Benedetto, Irene Terrenato, Anna Di Carlo, Sheila Spada, Mina J. Bissell, Francesca Di Modugno, Barbara Antoniani, Isabella Sperduti, Martin A. Schwartz, Belinda Palermo, Francesco Gandolfi, Francesco Facciolo, Pierluigi Iapicca, Marcella Mottolese, Emily I. Chen, Angela Santoni, and Paolo Visca
- Subjects
0301 basic medicine ,Gene isoform ,Cancer Research ,Lung Neoplasms ,Stromal cell ,Clinical Sciences ,Oncology and Carcinogenesis ,Matrix metalloproteinase ,Article ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Serum response factor ,Tumor Microenvironment ,Genetics ,2.1 Biological and endogenous factors ,Humans ,Protein Isoforms ,Oncology & Carcinogenesis ,Aetiology ,Non-Small-Cell Lung ,Lung ,Molecular Biology ,Transcription factor ,Cancer ,Neoplastic ,biology ,Integrin beta1 ,Carcinoma ,Lung Cancer ,Microfilament Proteins ,Extracellular Matrix ,Fibronectins ,3. Good health ,Gene Expression Regulation, Neoplastic ,Fibronectin ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Signal Transduction - Abstract
We demonstrated previously that the splicing of the actin regulator, hMENA, generates two alternatively expressed isoforms, hMENA11a and hMENAΔv6, which have opposite functions in cell invasiveness. Their mechanisms of action have remained unclear. Here we report two major findings: (i) hMENA regulates β1 integrin expression. This was shown by depleting total hMENA, which led to loss of nuclear expression of serum response factor (SRF)-coactivator myocardin-related transcription factor 1 (MRTF-A), leading to an increase in the G-actin/F-actin ratio crucial for MRTF-A localization. This in turn inhibited SRF activity and the expression of its target gene β1 integrin. (ii) hMENA11a reduces and hMENAΔv6 increases β1 integrin activation and signaling. Moreover, exogenous expression of hMENA11a in hMENAΔv6-positive cancer cells dramatically reduces secretion of extracellular matrix (ECM) components, including β1 integrin ligands and metalloproteinases. On the other hand, overexpression of the pro-invasive hMENAΔv6 increases fibronectin production. In primary tumors high hMENA11a correlates with low stromal fibronectin and a favorable clinical outcome of early node-negative non-small-cell lung cancer patients. These data provide new insights into the roles of hMENA11a and hMENAΔv6 in the druggable β1 integrin-ECM signaling axis and allow stratification of patient risk, guiding their clinical management.
- Published
- 2018
- Full Text
- View/download PDF
5. Exosomes shuttle TREX1-sensitive IFN-stimulatory dsDNA from irradiated cancer cells to DCs
- Author
-
Nils Rudqvist, Sheila Spada, Yasmeen Sarfraz, Karsten A. Pilones, Claire Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Julie M. Diamond, Jessica R. Chapman, and Beatrix Ueberheide
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Immunology ,Mammary Neoplasms, Animal ,CD8-Positive T-Lymphocytes ,Exosomes ,Cancer Vaccines ,Article ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Downregulation and upregulation ,medicine ,Tumor Cells, Cultured ,Animals ,Mice, Inbred BALB C ,Chemistry ,Immunogenicity ,Cancer ,Immunotherapy ,DNA, Neoplasm ,Dendritic Cells ,medicine.disease ,Phosphoproteins ,Immune checkpoint ,Microvesicles ,Mice, Inbred C57BL ,030104 developmental biology ,Exodeoxyribonucleases ,Cancer cell ,Interferon Type I ,Cancer research ,Female ,CD8 ,Spleen - Abstract
Radiotherapy (RT) used at immunogenic doses leads to accumulation of cytosolic double-stranded DNA (dsDNA) in cancer cells, which activates type I IFN (IFN-I) via the cGAS/STING pathway. Cancer cell–derived IFN-I is required to recruit BATF3-dependent dendritic cells (DC) to poorly immunogenic tumors and trigger antitumor T-cell responses in combination with immune checkpoint blockade. We have previously demonstrated that the exonuclease TREX1 regulates radiation immunogenicity by degrading cytosolic dsDNA. Tumor-derived DNA can also activate cGAS/STING-mediated production of IFN-I by DCs infiltrating immunogenic tumors. However, how DNA from cancer cells is transferred to the cytoplasm of DCs remains unclear. Here, we showed that tumor-derived exosomes (TEX) produced by irradiated mouse breast cancer cells (RT-TEX) transfer dsDNA to DCs and stimulate DC upregulation of costimulatory molecules and STING-dependent activation of IFN-I. In vivo, RT-TEX elicited tumor-specific CD8+ T-cell responses and protected mice from tumor development significantly better than TEX from untreated cancer cells in a prophylactic vaccination experiment. We demonstrated that the IFN-stimulatory dsDNA cargo of RT-TEX is regulated by TREX1 expression in the parent cells. Overall, these results identify RT-TEX as a mechanism whereby IFN-stimulatory dsDNA is transferred from irradiated cancer cells to DCs. We have previously shown that the expression of TREX1 is dependent on the RT dose size. Thus, these data have important implications for the use of RT with immunotherapy. Cancer Immunol Res; 6(8); 910–20. ©2018 AACR.
- Published
- 2018
6. Abstract 5224: hMENA isoforms impact NSCLC patient outcome through fibronectin/β1 integrin axis
- Author
-
Irene Terrenato, Francesco Gandolfi, Paolo Visca, Anna Di Carlo, Pierluigi Iapicca, Francesca Di Modugno, Francesco Facciolo, Sheila Spada, Isabella Sperduti, Barbara Antoniani, Angela Santoni, Marcella Mottolese, Paola Nisticò, Anna Di Benedetto, Martin A. Schwartz, Belinda Palermo, Mina J. Bissell, and Emily Chen
- Subjects
Gene isoform ,Cancer Research ,biology ,Integrin ,Cancer ,medicine.disease ,Fibronectin ,Oncology ,Tumor progression ,Cancer cell ,Serum response factor ,biology.protein ,Cancer research ,medicine ,Paxillin - Abstract
The splicing of the actin regulator hMENA generates different isoforms and we have demonstrated that the two alternatively expressed isoforms, hMENA11a and hMENAΔv6, have opposite functions in cell invasiveness. This general mechanism is of great clinical relevance in early NSCLC patients, where the pattern of hMENA isoform expression is a powerful prognostic factor. However the mechanism of action of the two isoforms have remained unclear. Herein, we evaluated whether hMENA and its isoforms influence β1 integrin expression and signaling considering the role of this integrin in cancer cell invasiveness and tumor progression. We performed hMENA silencing by siRNA and shRNA, to evaluate by QRT-PCR and biochemical approaches the expression of β1 integrin; by immunofluorescence the MRTF1 localization, by in vivo assay G-Actin/F-Actin ratio and by luciferase reporter assay the SRF activity. β1 integrin activation and signaling was evaluated by flow cytometry using an antibody specific for the β1 active conformation and by biochemical analysis of the phosphorylation of FAK, SRC and Paxillin. The secretoma of hMENA11a transfected cancer cell lines was analyzed by LC-MS/MS. Immunohistochemical analysis was performed using pan-hMENA, hMENA11a, and fibronectin antibodies in primary cancer tissues from node negative NSCLC patients. The Chi-Square or Fisher Exact tests were used to estimate associations among categorical variables and disease-free survival was calculated by the Kaplan-Meier product limit method. We show that the depletion of all hMENA isoforms inhibits the Serum Response Factor (SRF) activity, and the expression of its target gene β1 Integrin, by affecting G-Actin/F-Actin ratio, critical for the nuclear localization of the SRF co-factor myocardin related transcription factor 1 (MRTF1). Furthermore, we provide new insights into the mechanisms involved in the opposite functions of hMENA11a and hMENAΔv6 in cell invasiveness and we identify a new role of these isoforms in the β1 integrin-ECM signalling axis. Indeed, hMENAΔv6-drives cancer cell invasion by increasing β1 integrin activation and signalling, which is reduced by the anti-invasive hMENA11a isoform. Moreover, exogenous expression of hMENA11a in hMENAΔv6 positive cancer cells dramatically reduces secretion of extracellular matrix (ECM) components, including β1 integrin ligands and metalloproteinases. On the other hand overexpression of the pro-invasive hMENAΔv6 increases fibronectin production. In primary tumors high hMENA11a correlates with low stromal fibronectin and favorable clinical outcome of early node-negative non-small cell lung cancer patients. This newly discovered signature, which pays attention to the alternative splicing of hMENA and ECM components such as fibronectin in the stroma, might help fill in the gap in the still controversial clinical management of early node-negative NSCLC patients. Citation Format: Francesca Di Modugno, Sheila Spada, Belinda Palermo, Paolo Visca, Pierluigi Iapicca, Anna Di Carlo, Barbara Antoniani, Isabella Sperduti, Anna Di Benedetto, Irene Terrenato, Marcella Mottolese, Francesco Gandolfi, Francesco Facciolo, Emily Chen, Martin A. Schwartz, Angela Santoni, Mina J. Bissell, Paola Nisticò. hMENA isoforms impact NSCLC patient outcome through fibronectin/β1 integrin axis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5224.
- Published
- 2018
- Full Text
- View/download PDF
7. Abstract 4316: hMENA11a contributes to HER3-mediated resistance to PI3K inhibitors in HER2 overexpressing breast cancer cells
- Author
-
Paola Nisticò, Ruggero De Maria, Francesca Di Modugno, Paola Trono, Silvia Matteoni, Sheila Spada, Mariangela Panetta, Rita Circo, Silvia Soddu, Belinda Palermo, and Roberta Melchionna
- Subjects
MAPK/ERK pathway ,Cancer Research ,medicine.medical_specialty ,Cell growth ,business.industry ,Cancer ,medicine.disease ,Endocrinology ,Breast cancer ,Oncology ,Internal medicine ,Cancer cell ,Cancer research ,Medicine ,skin and connective tissue diseases ,business ,Protein kinase B ,Transcription factor ,PI3K/AKT/mTOR pathway - Abstract
Human Mena (hMENA), an actin regulatory protein of the ENA/VASP family, cooperates with ErbB receptor family signaling in breast cancer. It is overexpressed in high-risk preneoplastic lesions and in primary breast tumors where it correlates with HER2 overexpression and an activated status of AKT and MAPK. The concomitant overexpression of hMENA and HER2 identifies breast cancer patients with a worse prognosis. hMENA is expressed along with alternatively expressed isoforms, hMENA11a and hMENAΔv6 with opposite functions. By Reverse Phase Protein Assay, we identified a novel role for the epithelial associated hMENA11a isoform in sustaining HER3 activation and pro-survival pathways in HER2 overexpressing breast luminal cancer cells. Since HER3 activation is crucial in mechanisms of cell resistance to PI3K inhibitors, we explored whether hMENA11a is involved in these resistance mechanisms. The specific hMENA11a depletion switched off the HER3-related pathway activated by PI3K inhibitors and impaired the nuclear accumulation of HER3 transcription factor FOXO3a induced by PI3K inhibitors. On the other hand, PI3K inhibitors activated hMENA11a phosphorylation and affected its localization. At the functional level, we found that hMENA11a sustains cell proliferation and survival in response to PI3K inhibitor treatment whereas hMENA11a silencing increases molecules involved in cancer cell apoptosis. As shown in three-dimensional cultured breast cancer cells hMENA11a contributes to cancer cells resistance to PI3K inhibition since the depletion of hMENA11a drastically reduced cell viability upon treatment with PI3K inhibitor BEZ235. Altogether, these results indicate that hMENA11a in HER2 overexpressing breast cancer cells sustains HER3/AKT axis activation and contributes to HER3-mediated resistance mechanisms to PI3K inhibitors. Thus, hMENA11a expression can be proposed as a marker of HER3 activation and of resistance to PI3K inhibition therapies, to select patients who can benefit from these combined targeted treatments. hMENA11a activity may represent a new target for anti-proliferative therapies in breast cancer. Citation Format: Paola Trono, Francesca Di Modugno, Rita Circo, Sheila Spada, Roberta Melchionna, Belinda Palermo, Mariangela Panetta, Silvia Matteoni, Silvia Soddu, Ruggero De Maria, Paola Nisticò. hMENA11a contributes to HER3-mediated resistance to PI3K inhibitors in HER2 overexpressing breast cancer cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4316. doi:10.1158/1538-7445.AM2015-4316
- Published
- 2015
- Full Text
- View/download PDF
8. Abstract 1035: hMENA splicing program and TGF-β1-mediated EMT in pancreatic cancer
- Author
-
Maria Grazia Diodoro, Novella Gualtieri, Sheila Spada, Paola Nisticò, Pierluigi Iapicca, Francesca Di Modugno, Roberta Melchionna, Giuliana Falasca, Gian Luca Grazi, Paola Trono, and Mina J. Bissell
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,biology ,Cancer ,Vimentin ,medicine.disease ,Actin cytoskeleton ,Paracrine signalling ,Oncology ,Pancreatic tumor ,Pancreatic cancer ,medicine ,Cancer research ,biology.protein ,Epithelial–mesenchymal transition - Abstract
Background Epithelial to mesenchymal transition (EMT) is an early event in pancreatic cancer and has been involved in cancer invasiveness. An intense stromal reaction, peculiar to the pancreatic tumor microenvironment, includes cancer-associated fibroblasts(CAFs), abundant cells in the tumor stroma, recently linked to the induction of EMT. On the other hand, the EMT process requires a dynamic remodeling of the actin cytoskeleton, and the splicing program of hMENA, a regulator of actin, has been associated with the EMT process. We have described two alternatively expressed isoforms, hMENA11a and hMENAΔv6, with opposite functions in invasiveness in breast cancer (1). hMENA expression was detected in human pancreatic ductal adenocarcinoma samples (PDAC) (2), but no data are available on the alternative isoform expression in this neoplasia. The aim of this study is to investigate the role of hMENA splicing in TGF-β -mediated EMT in pancreatic cancer, the mechanisms involved in hMENA induction in PDAC and the role of CAFs in this process. Methods hMENA isoform expression was evaluated in PDAC tissues by immunohistochemistry using isoform-specific antibodies. Human PDAC cell lines, untreated or TGF-β treated, were characterized for the expression of hMENA isoforms and markers of EMT by qRT-PCR and WB analysis. The effects of both hMENAΔv6 knockdown or overexpression were also evaluated. Pancreatic cancer associated-fibroblasts were isolated from primary PDAC tissues. To study the role of fibroblast-cancer cell interactions on hMENA expression, a noncontact coculture system was used. Results IHC analysis of PDAC tissues revealed that epithelial hMENA11a is rarely expressed in primary pancreatic tumors that express a high level of hMENA and hMENAΔv6 isoforms. In a panel of pancreatic cancer cell lines, hMENA11a expression correlates with an epithelial phenotype, wherea hMENAΔv6 expression is associated with a mesechymal phenotype. TGF-β treatment specifically upregulated the invasive hMENAΔv6 isoform expression. Knockdown of endogenous hMENA/hMENAΔv6 isoform reduced cell invasiveness, reverted cells to an epithelial -“like” phenotype with an increased E-cadherin expression and impaired the TGF-β-mediated vimentin up-regulation. Conversely, overexpression of hMENAΔv6 increased the expression of the mesenchymal marker vimentin. Freshly explanted CAFs expressed the “mesenchymal” hMENAΔv6, and not hMENA11a and produced paracrine factors involved in the induction of hMENA isoforms in tumor cells. Conclusions These data provide new and critical insights into the role of hMENA splicing in TGF-β mediated EMT and identify the hMENA splicing program as a promising pathway for the development of new diagnostics and therapeutics in PDAC. (1) Di Modugno F. et al PNAS 2012 (2) Pino S. et al Clin Cancer Res 2008 Citation Format: Roberta Melchionna, Pierluigi Iapicca, Francesca Di Modugno, Paola Trono, Novella Gualtieri, Maria Grazia Diodoro, Sheila Spada, Giuliana Falasca, Gian Luca Grazi, Mina J Bissell, Paola Nisticò. hMENA splicing program and TGF-β1-mediated EMT in pancreatic cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1035. doi:10.1158/1538-7445.AM2014-1035
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.