50 results on '"Pellegatta, Serena"'
Search Results
2. ERBB3 overexpression due to miR-205 inactivation confers sensitivity to FGF, metabolic activation, and liability to ERBB3 targeting in glioblastoma
- Author
-
De Bacco, Francesca, Orzan, Francesca, Erriquez, Jessica, Casanova, Elena, Barault, Ludovic, Albano, Raffaella, D’Ambrosio, Antonio, Bigatto, Viola, Reato, Gigliola, Patanè, Monica, Pollo, Bianca, Kuesters, Geoffrey, Dell’Aglio, Carmine, Casorzo, Laura, Pellegatta, Serena, Finocchiaro, Gaetano, Comoglio, Paolo M., and Boccaccio, Carla
- Published
- 2021
- Full Text
- View/download PDF
3. High tumor mutational burden and T-cell activation are associated with long-term response to anti-PD1 therapy in Lynch syndrome recurrent glioblastoma patient
- Author
-
Anghileri, Elena, Di Ianni, Natalia, Paterra, Rosina, Langella, Tiziana, Zhao, Junfei, Eoli, Marica, Patanè, Monica, Pollo, Bianca, Cuccarini, Valeria, Iavarone, Antonio, Rabadan, Raul, Finocchiaro, Gaetano, and Pellegatta, Serena
- Published
- 2021
- Full Text
- View/download PDF
4. B7-H3-redirected chimeric antigen receptor T cells target glioblastoma and neurospheres
- Author
-
Nehama, Dean, Di Ianni, Natalia, Musio, Silvia, Du, Hongwei, Patané, Monica, Pollo, Bianca, Finocchiaro, Gaetano, Park, James J.H., Dunn, Denise E., Edwards, Drake S., Damrauer, Jeffrey S., Hudson, Hannah, Floyd, Scott R., Ferrone, Soldano, Savoldo, Barbara, Pellegatta, Serena, and Dotti, Gianpietro
- Published
- 2019
- Full Text
- View/download PDF
5. Next‐generation agents for fluorescence‐guided glioblastoma surgery.
- Author
-
Chirizzi, Cristina, Pellegatta, Serena, Gori, Alessandro, Falco, Jacopo, Rubiu, Emanuele, Acerbi, Francesco, and Bombelli, Francesca Baldelli
- Subjects
- *
GLIOBLASTOMA multiforme , *TISSUE differentiation , *SURGICAL excision , *BRAIN cancer , *RADIOTHERAPY safety ,TUMOR surgery - Abstract
Glioblastoma is a fast‐growing and aggressive form of brain cancer. Even with maximal treatment, patients show a low median survival and are often subjected to a high recurrence incidence. The currently available treatments require multimodal management, including maximal safe surgical resection, followed by radiation and chemotherapy. Because of the infiltrative glioblastoma nature, intraoperative differentiation of cancer tissue from normal brain parenchyma is very challenging, and this accounts for the low rate of complete tumor resection. For these reasons, clinicians have increasingly used various intraoperative adjuncts to improve surgical results, such as fluorescent agents. However, most of the existing fluorophores show several limitations such as poor selectivity, photostability, photosensitization and high costs. This could limit their application to successfully improve glioblastoma resection. In the present perspective, we highlight the possibility to develop next‐generation fluorescent tools able to more selectively label cancer cells during surgical resection. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Response assessment of GBM during immunotherapy by delayed contrast treatment response assessment maps.
- Author
-
Cuccarini, Valeria, Savoldi, Filippo, Mardor, Yael, Last, David, Pellegatta, Serena, Mazzi, Federica, Bruzzone, Maria Grazia, Anghileri, Elena, Pollo, Bianca, Maddaloni, Luisa, Russo, Camilla, Bocchi, Elisa, Pinzi, Valentina, Eoli, Marica, and Aquino, Domenico
- Subjects
TREATMENT delay (Medicine) ,KILLER cells ,BLOOD cell count ,IMMUNOTHERAPY ,RECEIVER operating characteristic curves ,GLIOBLASTOMA multiforme ,CONTRAST-enhanced magnetic resonance imaging - Abstract
Introduction: Assessing the treatment response of glioblastoma multiforme during immunotherapy (IT) is an open issue. Treatment response assessment maps (TRAMs) might help distinguish true tumor progression (TTP) and pseudoprogression (PsP) in this setting. Methods: We recruited 16 naïve glioblastoma patients enrolled in a phase II trial consisting of the Stupp protocol (a standardized treatment for glioblastoma involving combined radiotherapy and chemotherapy with temozolomide, followed by adjuvant temozolomide) plus IT with dendritic cells. Patients were followed up till progression or death; seven underwent a second surgery for suspected progression. Clinical, immunological, and MRI data were collected from all patients and histology in case of second surgery. Patients were classified as responders (progression-free survival, PFS > 12 months), and non-responders (PFS ≤ 12), HIGH-NK (natural killer cells, i.e., immunological responders), and LOW-NK (immunological non-responders) based on immune cell counts in peripheral blood. TRAMs differentiate contrast-enhancing lesions with different washout dynamics into hypothesized tumoral (conventionally blue-colored) vs. treatment-related (red-colored). Results: Using receiver operating characteristic (ROC) curves, a threshold of −0.066 in V
Blue /VCE (volume of the blue portion of tumoral area/volume of contrast enhancement) variation between values obtained in the MRI performed before PsP/TTP and at TTP/PSP allowed to discriminate TTP from PsP with a sensitivity of 71.4% and a specificity of 100%. Among HIGH-NK patients, at month 6 there was a significant reduction compared to baseline and month 2 in median “blue” volumes. Discussion: In conclusion, in our pilot study TRAMs support the discrimination between tumoral and treatment-related enhancing features in immunological responders vs. non-responders, the distinction between PsP and TTP, and might provide surrogate markers of immunological response. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
7. The Immunomodulatory Effects of Fluorescein-Mediated Sonodynamic Treatment Lead to Systemic and Intratumoral Depletion of Myeloid-Derived Suppressor Cells in a Preclinical Malignant Glioma Model.
- Author
-
Pellegatta, Serena, Corradino, Nicoletta, Zingarelli, Manuela, Porto, Edoardo, Gionso, Matteo, Berlendis, Arianna, Durando, Gianni, Maffezzini, Martina, Musio, Silvia, Aquino, Domenico, DiMeco, Francesco, and Prada, Francesco
- Subjects
- *
GLIOMA treatment , *CELL metabolism , *BIOLOGICAL models , *IN vitro studies , *ULTRASONIC imaging , *IN vivo studies , *ANIMAL experimentation , *MACROPHAGES , *GLIOMAS , *MYELOID-derived suppressor cells , *TREATMENT effectiveness , *GENE expression , *SURVIVAL rate , *COMPARATIVE studies , *RESEARCH funding , *ULTRASONIC therapy , *FLUORESCENT dyes , *T cells , *IMMUNOTHERAPY , *MICE , *EVALUATION - Abstract
Simple Summary: Sonodynamic therapy (SDT) is emerging as a promising innovative technique for treating malignant gliomas in a noninvasive fashion. The use of fluorescein, instead of 5-aminolevulinic acid, could extend the application of sonodynamic therapy to intracranial tumors other than gliomas. Our aim was to investigate the feasibility of sonodynamic therapy with fluorescein in an intracranial mouse model of malignant glioma, its efficacy, and its effects on the immune microenvironment. Fluorescein-mediated sonodynamic therapy (FL-SDT) is an extremely promising approach for glioma treatment, resulting from the combination of low-intensity focused ultrasound (FUS) with a sonosensitizer. In the present study, we evaluated the efficacy and immunomodulation of SDT with fluorescein as the sonosensitizer in immunocompetent GL261 glioma mice for the first time. In vitro studies demonstrated that the exposure of GL261 cells to FL-SDT induced immunogenic cell death and relevant upregulation of MHC class I, CD80 and CD86 expression. In vivo studies were then performed to treat GL261 glioma-bearing mice with FL-SDT, fluorescein alone, or FUS alone. Perturbation of the glioma-associated macrophage subset within the immune microenvironment was induced by all the treatments. Notably, a relevant depletion of myeloid-derived suppressor cells (MDSCs) and concomitant robust infiltration of CD8+ T cells were observed in the SDT-FL-treated mice, resulting in a significant radiological delay in glioma progression and a consequent improvement in survival. Tumor control and improved survival were also observed in mice treated with FL alone (median survival 41.5 days, p > 0.0001 compared to untreated mice), reflecting considerable modulation of the immune microenvironment. Interestingly, a high circulating lymphocyte-to-monocyte ratio and a very low proportion of MDSCs were predictive of better survival in FL- and FL-SDT-treated mice than in untreated and FUS-treated mice, in which elevated monocyte and MDSC frequencies correlated with worse survival. The immunostimulatory potential of FL-SDT treatment and the profound modulation of most immunosuppressive components within the microenvironment encouraged the exploration of the combination of FL-SDT with immunotherapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. ER–mitochondria contacts control surface glycan expression and sensitivity to killer lymphocytes in glioma stem‐like cells
- Author
-
Bassoy, Esen Yonca, Kasahara, Atsuko, Chiusolo, Valentina, Jacquemin, Guillaume, Boydell, Emma, Zamorano, Sebastian, Riccadonna, Cristina, Pellegatta, Serena, Hulo, Nicolas, Dutoit, Valérie, Derouazi, Madiha, Dietrich, Pierre Yves, Walker, Paul R, and Martinvalet, Denis
- Published
- 2017
- Full Text
- View/download PDF
9. Constitutive and TNFα-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: Implications for CAR-T cell therapy
- Author
-
Pellegatta, Serena, Savoldo, Barbara, Di Ianni, Natalia, Corbetta, Cristina, Chen, Yuhui, Patané, Monica, Sun, Chuang, Pollo, Bianca, Ferrone, Soldano, DiMeco, Francesco, Finocchiaro, Gaetano, and Dotti, Gianpietro
- Published
- 2018
- Full Text
- View/download PDF
10. Resetting cancer stem cell regulatory nodes upon MYC inhibition
- Author
-
Galardi, Silvia, Savino, Mauro, Scagnoli, Fiorella, Pellegatta, Serena, Pisati, Federica, Zambelli, Federico, Illi, Barbara, Annibali, Daniela, Beji, Sara, Orecchini, Elisa, Alberelli, Maria Adele, Apicella, Clara, Fontanella, Rosaria Anna, Michienzi, Alessandro, Finocchiaro, Gaetano, Farace, Maria Giulia, Pavesi, Giulio, Ciafrè, Silvia Anna, and Nasi, Sergio
- Published
- 2016
- Full Text
- View/download PDF
11. MET inhibition overcomes radiation resistance of glioblastoma stem‐like cells
- Author
-
De Bacco, Francesca, D'Ambrosio, Antonio, Casanova, Elena, Orzan, Francesca, Neggia, Roberta, Albano, Raffaella, Verginelli, Federica, Cominelli, Manuela, Poliani, Pietro L, Luraghi, Paolo, Reato, Gigliola, Pellegatta, Serena, Finocchiaro, Gaetano, Perera, Timothy, Garibaldi, Elisabetta, Gabriele, Pietro, Comoglio, Paolo M, and Boccaccio, Carla
- Published
- 2016
- Full Text
- View/download PDF
12. Transforming Fusions of FGFR and TACC Genes in Human Glioblastoma
- Author
-
Singh, Devendra, Chan, Joseph Minhow, Zoppoli, Pietro, Niola, Francesco, Sullivan, Ryan, Castano, Angelica, Liu, Eric Minwei, Reichel, Jonathan, Porrati, Paola, Pellegatta, Serena, Qiu, Kunlong, Gao, Zhibo, Ceccarelli, Michele, Riccardi, Riccardo, Brat, Daniel J., Guha, Abhijit, Aldape, Ken, Golfinos, John G., Zagzag, David, Mikkelsen, Tom, Finocchiaro, Gaetano, Lasorella, Anna, Rabadan, Raul, and Iavarone, Antonio
- Published
- 2012
- Full Text
- View/download PDF
13. EGFR Amplified and Overexpressing Glioblastomas and Association With Better Response to Adjuvant Metronomic Temozolomide
- Author
-
Cominelli, Manuela, Grisanti, Salvatore, Mazzoleni, Stefania, Branca, Caterina, Buttolo, Luciano, Furlan, Daniela, Liserre, Barbara, Bonetti, Maria Fausta, Medicina, Daniela, Pellegrini, Vilma, Buglione, Michela, Liserre, Roberto, Pellegatta, Serena, Finocchiaro, Gaetano, Dalerba, Piero, Facchetti, Fabio, Pizzi, Marina, Galli, Rossella, and Poliani, Pietro Luigi
- Published
- 2015
- Full Text
- View/download PDF
14. Reelin affects chain-migration and differentiation of neural precursor cells
- Author
-
Massalini, Simone, Pellegatta, Serena, Pisati, Federica, Finocchiaro, Gaetano, Farace, Maria Giulia, and Ciafrè, Silvia Anna
- Published
- 2009
- Full Text
- View/download PDF
15. Glioblastoma stem cells express non‐canonical proteins and exclusive mesenchymal‐like or non‐mesenchymal‐like protein signatures.
- Author
-
Babačić, Haris, Galardi, Silvia, Umer, Husen M., Hellström, Mats, Uhrbom, Lene, Maturi, Nagaprathyusha, Cardinali, Deborah, Pellegatta, Serena, Michienzi, Alessandro, Trevisi, Gianluca, Mangiola, Annunziato, Lehtiö, Janne, Ciafrè, Silvia Anna, and Pernemalm, Maria
- Abstract
Glioblastoma (GBM) cancer stem cells (GSCs) contribute to GBM's origin, recurrence, and resistance to treatment. However, the understanding of how mRNA expression patterns of GBM subtypes are reflected at global proteome level in GSCs is limited. To characterize protein expression in GSCs, we performed in‐depth proteogenomic analysis of patient‐derived GSCs by RNA‐sequencing and mass‐spectrometry. We quantified > 10 000 proteins in two independent GSC panels and propose a GSC‐associated proteomic signature characterizing two distinct phenotypic conditions; one defined by proteins upregulated in proneural and classical GSCs (GPC‐like), and another by proteins upregulated in mesenchymal GSCs (GM‐like). The GM‐like protein set in GBM tissue was associated with necrosis, recurrence, and worse overall survival. Through proteogenomics, we discovered 252 non‐canonical peptides in the GSCs, i.e., protein sequences that are variant or derive from genome regions previously considered non‐protein‐coding, including variants of the heterogeneous ribonucleoproteins implicated in RNA splicing. In summary, GSCs express two protein sets that have an inverse association with clinical outcomes in GBM. The discovery of non‐canonical protein sequences questions existing gene models and pinpoints new protein targets for research in GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. A critical role for regulatory T cells in driving cytokine profiles of Th17 cells and their modulation of glioma microenvironment
- Author
-
Cantini, Gabriele, Pisati, Federica, Mastropietro, Alfonso, Frattini, Véronique, Iwakura, Yoichiro, Finocchiaro, Gaetano, and Pellegatta, Serena
- Published
- 2011
- Full Text
- View/download PDF
17. The potential of stem cells for the treatment of brain tumors and globoid cell leukodystrophy
- Author
-
Tunici, Patrizia, Pellegatta, Serena, and Finocchiaro, Gaetano
- Published
- 2003
- Full Text
- View/download PDF
18. The therapeutic potential of neural stem/progenitor cells in murine globoid cell leukodystrophy is conditioned by macrophage/microglia activation
- Author
-
Pellegatta, Serena, Tunici, Patrizia, Poliani, Pietro Luigi, Dolcetta, Diego, Cajola, Laura, Colombelli, Cristina, Ciusani, Emilio, Di Donato, Stefano, and Finocchiaro, Gaetano
- Published
- 2006
- Full Text
- View/download PDF
19. Brain cancer immunoediting: novel examples provided by immunotherapy of malignant gliomas
- Author
-
Pellegatta, Serena, Cuppini, Lucia, and Finocchiaro, Gaetano
- Published
- 2011
- Full Text
- View/download PDF
20. MR-Spectroscopy and Survival in Mice with High Grade Glioma Undergoing Unrestricted Ketogenic Diet.
- Author
-
Ciusani, Emilio, Vasco, Chiara, Rizzo, Ambra, Girgenti, Vita, Padelli, Francesco, Pellegatta, Serena, Fariselli, Laura, Bruzzone, Maria Grazia, and Salmaggi, Andrea
- Subjects
KETOGENIC diet ,CELL culture ,ANIMAL experimentation ,GLIOMAS ,NUCLEAR magnetic resonance spectroscopy ,MITOCHONDRIA ,SURVIVAL analysis (Biometry) ,RESEARCH funding ,MICE - Abstract
Glioblastoma multiforme (GBM) is considered the most malignant form of primary brain tumor. Despite multimodal treatment, prognosis remains poor. Ketogenic diet (KD) has been suggested for the treatment of GBM. In this study, the syngenic, orthotopic GL261 mouse glioma model was used to evaluate the effects of KD on the metabolic responses of the tumor using 7T magnetic resonance imaging/spectroscopy. GL261 cells were injected into the caudate nucleus of mice. Following implantation, animals were fed with standard chow or underwent a KD. 18 days after initiating the diet, mice fed with KD displayed significantly higher plasmatic levels of ketone bodies and survived longer than those fed with the standard diet. Decreased concentrations of gamma-aminobutyric acid, N-Acetyl-Aspartate and N-acetylaspartylglutamate were found in tumor tissue after 9 days into the KD, while a huge increase in beta-hydroxybutyrate (bHB) was detected in tumor tissue as compared to normal brain. The accumulation of bHB in the tumor tissue in mice undergoing the KD, may suggest either elevated uptake/release of bHB by tumor cells, or the inability of tumor cells in this context to use it for mitochondrial metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
21. Revisiting the Immunological Aspects of Temozolomide Considering the Genetic Landscape and the Immune Microenvironment Composition of Glioblastoma.
- Author
-
Di Ianni, Natalia, Maffezzini, Martina, Eoli, Marica, and Pellegatta, Serena
- Subjects
THERAPEUTICS ,TEMOZOLOMIDE ,ALKYLATING agents ,GLIOBLASTOMA multiforme ,IMMUNOTHERAPY ,SPEECH apraxia - Abstract
The microenvironment (ME) plays a critical role in causing glioblastoma (GBM) to be a moving and incurable target. The main features governing the interaction between cancer cells and the ME include dependency, promotion, and in rare cases, even competition. In the original Stupp protocol, the alkylating agent temozolomide (TMZ) is the first-line chemotherapy drug to treat GBM, and it is broadly used together or after radiotherapy. Some studies have described TMZ as an adjuvant to other therapeutic approaches including immunotherapy because of its ability to induce an immunogenic death of cancer cells. TMZ also exerts immunomodulatory effects on the tumor and immune ME. These findings support the coexistence of two circuits, i.e., one that subverts local immunosuppressive mechanisms and another that exerts a harmful influence on the peripheral immune response. A bias toward the latter can drive the failure of treatments based on the combination of chemotherapy and immunotherapy approaches. In this review, we will reanalyze how intrinsic and acquired resistance to TMZ impacts the immunomodulatory effects previously described by way of inducing a functional alteration of local immune cells and promoting immunosuppression and how different components of the immune ME, with particular attention to tumor-associated macrophages and microglia, can cause TMZ resistance to circumvent potential local immunogenic mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
22. Altered function of the glutamate–aspartate transporter GLAST, a potential therapeutic target in glioblastoma.
- Author
-
Corbetta, Cristina, Di Ianni, Natalia, Bruzzone, Maria Grazia, Patanè, Monica, Pollo, Bianca, Cantini, Gabriele, Cominelli, Manuela, Zucca, Ileana, Pisati, Federica, Poliani, Pietro Luigi, Finocchiaro, Gaetano, and Pellegatta, Serena
- Abstract
In glioma patients, high levels of glutamate can cause brain edema and seizures. GLAST, a glutamate–aspartate transporter expressed by astrocytes with a role in glutamate uptake, is highly expressed on the plasma membrane of glioblastoma (GBM) cells, and its expression significantly correlates with shortened patient survival. Here, it was demonstrated that inhibition of GLAST expression limited the progression and invasion of GBM xenografts. Magnetic resonance spectroscopy was used to measure glutamate in GLAST‐expressing gliomas showing that these tumors exhibit increased glutamate concentration compared to GLAST‐depleted glioma. Despite their GLAST expression, GBM stem‐like cells (GSCs) released rather than taking up glutamate due to their lack of Na+/K+‐ATPase. Overexpression of Na+/K+‐ATPase in these cells restored glutamate uptake and induced apoptosis. The therapeutic relevance of targeting GLAST in gliomas was assessed using the inhibitor UCPH‐101. In glioma‐bearing mice, a single intratumoral injection of UCPH‐101 significantly increased survival by decreasing GLAST expression and inducing apoptosis. Thus, GLAST has a novel role in GBM that appears to have crucial relevance in glutamate trafficking and may thus be a new therapeutic target. What's new? Increased expression of the cystine/glutamate antiporter xCT is associated with glutamate‐promoted glioblastoma (GBM) invasion into surrounding brain tissue. In this study, GLAST, an astrocyte transporter physiologically devoted to glutamate uptake, was also found to serve a role in GBM aggressiveness. GLAST was highly expressed in GBM specimens and, in association with Na+/K+/ATPase downregulation, exhibited impaired glutamate uptake, resulting in elevated extracellular glutamate levels and GMB cell protection against apoptosis. GLAST inhibition, by contrast, decreased extracellular glutamate, increased GBM cell apoptosis, and prolonged survival in glioma‐bearing mice. The findings provide preliminary background for translational research of GLAST inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
23. Genetic Evolution of Glioblastoma Stem-Like Cells From Primary to Recurrent Tumor.
- Author
-
Orzan, Francesca, De Bacco, Francesca, Crisafulli, Giovanni, Pellegatta, Serena, Mussolin, Benedetta, Siravegna, Giulia, D'Ambrosio, Antonio, Comoglio, Paolo M., Finocchiaro, Gaetano, and Boccaccio, Carla
- Abstract
Glioblastoma (GBM) is a lethal tumor that displays remarkable genetic heterogeneity. It is also known that GBM contains a cell hierarchy driven by GBM stem-like cells (GSCs), responsible for tumor generation, therapeutic resistance, and relapse. An important and still open issue is whether phylogenetically related GSCs can be found in matched primary and recurrent GBMs, and reflect tumor genetic evolution under therapeutic pressure. To address this, we analyzed the mutational profile of GSCs isolated from either human primary GBMs (primary GSCs) or their matched tumors recurring after surgery and chemoradiotherapy (recurrent GSCs). We found that recurrent GSCs can accumulate temozolomide-related mutations over primary GSCs, following both linear and branched patterns. In the latter case, primary and recurrent GSCs share a common set of lesions, but also harbor distinctive mutations indicating that primary and recurrent GSCs derive from a putative common ancestor GSC by divergent genetic evolution. Interestingly, TP53 mutations distinctive of recurrent GSCs were detectable at low frequency in the corresponding primary tumors and likely marked pre-existent subclones that evolved under therapeutic pressure and expanded in the relapsing tumor. Consistently, recurrent GSCs displayed in vitro greater therapeutic resistance than primary GSCs. Overall, these data indicate that (a) phylogenetically related GSCs are found in matched primary and recurrent GBMs and (b) recurrent GSCs likely pre-exist in the untreated primary tumor and are both mutagenized and positively selected by chemoradiotherapy. Stem Cells 2017;35:2218-2228 [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
24. Decitabine Treatment of Glioma-Initiating Cells Enhances Immune Recognition and Killing.
- Author
-
Riccadonna, Cristina, Yacoub Maroun, Céline, Vuillefroy de Silly, Romain, Boehler, Margaux, Calvo Tardón, Marta, Jueliger, Simone, Taverna, Pietro, Barba, Leticia, Marinari, Eliana, Pellegatta, Serena, Bassoy, Esen Yonca, Martinvalet, Denis, Dietrich, Pierre-Yves, and Walker, Paul R.
- Subjects
DECITABINE ,GLIOMA treatment ,IMMUNE recognition ,BRAIN tumors ,FAS proteins ,PROTEIN expression ,PROGNOSIS - Abstract
Malignant gliomas are aggressive brain tumours with very poor prognosis. The majority of glioma cells are differentiated (glioma-differentiated cells: GDCs), whereas the smaller population (glioma-initiating cells, GICs) is undifferentiated and resistant to conventional therapies. Therefore, to better target this pool of heterogeneous cells, a combination of diverse therapeutic approaches is envisaged. Here we investigated whether the immunosensitising properties of the hypomethylating agent decitabine can be extended to GICs. Using the murine GL261 cell line, we demonstrate that decitabine augments the expression of the death receptor FAS both on GDCs and GICs. Interestingly, it had a higher impact on GICs and correlated with an enhanced sensitivity to FASL-mediated cell death. Moreover, the expression of other critical molecules involved in cognate recognition by cytotoxic T lymphocytes, MHCI and ICAM-1, was upregulated by decitabine treatment. Consequently, T-cell mediated killing of both GDCs and GICs was enhanced, as was T cell proliferation after reactivation. Overall, although GICs are described to resist classical therapies, our study shows that hypomethylating agents have the potential to enhance glioma cell recognition and subsequent destruction by immune cells, regardless of their differentiation status. These results support the development of combinatorial treatment modalities including epigenetic modulation together with immunotherapy in order to treat heterogenous malignancies such as glioblastoma. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
25. Immunotherapy with dendritic cells loaded with glioblastoma stem cells: from preclinical to clinical studies.
- Author
-
Finocchiaro, Gaetano and Pellegatta, Serena
- Subjects
- *
CANCER immunotherapy , *CANCER stem cells , *GLIOBLASTOMA multiforme treatment , *DENDRITIC cells , *ANTINEOPLASTIC agents , *CANCER vaccines - Abstract
Different approaches have been explored to raise effective antitumor responses against glioblastoma (GBM), the deadliest of primary brain tumors. In many clinical studies, cancer vaccines have been based on dendritic cells (DCs) loaded with peptides, representing one or more specific tumor antigens or whole lysates as a source of multiple antigens. Randomized clinical trials using DCs are ongoing, and results of efficacy are not yet available. Such strategies are feasible and safe; however, immune-suppressive microenvironment, absence of appropriate specific epitopes to target, and cancer immunoediting can limit their efficacy. The aim of this review is to describe how the definition of novel and more specific targets may increase considerably the possibility of successful DC immunotherapy. By proposing to target glioblastoma stem-like cells (GSCs), the immune response will be pointed to eradicating factors and pathways highly relevant to GBM biology. Preclinical observations on efficacy, and preliminary results of immunotherapy trials, encourage exploring the clinical efficacy of DC immunotherapy in GBM patients using high-purity, GSC-loaded DC vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
26. Safe and Reproducible Preparation of Functional Dendritic Cells for Immunotherapy in Glioblastoma Patients.
- Author
-
NAVA, SARA, LISINI, DANIELA, POGLIANI, SIMONA, DOSSENA, MARTA, BERSANO, ANNA, PELLEGATTA, SERENA, PARATI, EUGENIO, FINOCCHIARO, GAETANO, and FRIGERIO, SIMONA
- Published
- 2015
- Full Text
- View/download PDF
27. Effective immuno-targeting of the IDH1 mutation R132H in a murine model of intracranial glioma.
- Author
-
Pellegatta, Serena, Valletta, Lorella, Corbetta, Cristina, Patanè, Monica, Zucca, Ileana, Sirtori, Federico Riccardi, Bruzzone, Maria Grazia, Fogliatto, Gianpaolo, Isacchi, Antonella, Pollo, Bianca, and Finocchiaro, Gaetano
- Subjects
- *
ISOCITRATE dehydrogenase , *IMMUNOTHERAPY , *MASS spectrometry - Abstract
The R132H mutation of cytosolic isocitrate dehydrogenase (IDH1) is present in the majority of low grade gliomas. Immunotherapy in these tumors has an interesting, still unexploited, therapeutic potential, as they are less immunosuppressive than glioblastomas. Using site-directed mutagenesis we introduced the R132H mutation into the murine glioma cell line GL261, creating mIDH1-GL261. Presence of the mutation was confirmed by immunoblotting and production of the oncometabolite 2-hydroxyglutarate (2HG), demonstrated by mass spectrometry (LC-MS/MS) performed on cell supernatant. In vitro mIDH1-GL261 had different morphology but similar growth rate than parental GL261 (p-GL261). After intracranial injection, MRI suggested that the initial growth rate was slower in mIDH1-GL261 than p-GL261 gliomas but overall survival was similar. mIDH1-GL261 gliomas showed evidence of R132H expression and of intratumoral 2HG production (evaluated by MRS and LC-MS/MS). Immunizations were performed nine days after intracranial implantation of mIDH1- or p-GL261 cells by three subcutaneous injections of five different peptides encompassing the IDH1 mutation site, all emulsified with Montanide ISA-51, in association with GM-CSF. Control mice were injected with four ovalbumin peptides or vehicle. Mice with mIDH1-GL261 but not p-GL261 gliomas treated with mIDH1 peptides survived longer than controls; 25% of them were cured. Immunized mice showed higher amounts of peripheral CD8+ T cells, higher production of IFN-γ, and evidence of anti-mIDH1 antibodies. Immunizations led to intratumoral up-regulation of IFN-γ, granzyme-b and perforin-1 and down-regulation of TGF-β2 and IL-10. These results support the translational potential of immunotherapeutic targeting of gliomas carrying IDH1 mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
28. Brain tumor immunotherapy: what have we learned so far?
- Author
-
Eyrich, Matthias, Pellegatta, Serena, and Van Gool, Stefaan Willy
- Subjects
BRAIN tumors ,IMMUNOTHERAPY ,GLIOMAS ,RADIOTHERAPY ,CANCER chemotherapy ,GLIOBLASTOMA multiforme - Abstract
High grade glioma is a rare brain cancer, incurable in spite of modern neurosurgery, radiotherapy, and chemotherapy. Novel approaches are in research, and immunotherapy emerges as a promising strategy. Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, we developed an immunotherapy schedule and we categorized patients into clinically defined risk profiles. We learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, we developed a mode of care within academic centers to organize cell-based therapies for experimental clinical trials in a large number of patients. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
29. Perspectives for immunotherapy in glioblastoma treatment.
- Author
-
Finocchiaro, Gaetano and Pellegatta, Serena
- Published
- 2014
- Full Text
- View/download PDF
30. The integrated landscape of driver genomic alterations in glioblastoma.
- Author
-
Frattini, Veronique, Trifonov, Vladimir, Chan, Joseph Minhow, Castano, Angelica, Lia, Marie, Abate, Francesco, Keir, Stephen T, Ji, Alan X, Zoppoli, Pietro, Niola, Francesco, Danussi, Carla, Dolgalev, Igor, Porrati, Paola, Pellegatta, Serena, Heguy, Adriana, Gupta, Gaurav, Pisapia, David J, Canoll, Peter, Bruce, Jeffrey N, and McLendon, Roger E
- Subjects
GENOMICS ,GLIOBLASTOMA multiforme ,SOMATIC mutation ,LIGASES ,STEM cells ,MITOGENS - Abstract
Glioblastoma is one of the most challenging forms of cancer to treat. Here we describe a computational platform that integrates the analysis of copy number variations and somatic mutations and unravels the landscape of in-frame gene fusions in glioblastoma. We found mutations with loss of heterozygosity in LZTR1, encoding an adaptor of CUL3-containing E3 ligase complexes. Mutations and deletions disrupt LZTR1 function, which restrains the self renewal and growth of glioma spheres that retain stem cell features. Loss-of-function mutations in CTNND2 target a neural-specific gene and are associated with the transformation of glioma cells along the very aggressive mesenchymal phenotype. We also report recurrent translocations that fuse the coding sequence of EGFR to several partners, with EGFR-SEPT14 being the most frequent functional gene fusion in human glioblastoma. EGFR-SEPT14 fusions activate STAT3 signaling and confer mitogen independence and sensitivity to EGFR inhibition. These results provide insights into the pathogenesis of glioblastoma and highlight new targets for therapeutic intervention. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
31. The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates.
- Author
-
Pellegatta, Serena, Eoli, Marica, Frigerio, Simona, Antozzi, Carlo, Bruzzone, Maria Grazia, Cantini, Gabriele, Nava, Sara, Anghileri, Elena, Cuppini, Lucia, Cuccarini, Valeria, Ciusani, Emilio, Dossena, Marta, Pollo, Bianca, Mantegazza, Renato, Parati, Eugenio A., and Finocchiaro, Gaetano
- Subjects
- *
KILLER cells , *DENDRITIC cells , *TUMORS , *DISEASE relapse , *IMMUNOTHERAPY , *TRANSCRIPTION factors - Abstract
Recurrent glioblastomas (GBs) are highly aggressive tumors associated with a 6-8 mo survival rate. In this study, we evaluated the possible benefits of an immunotherapeutic strategy based on mature dendritic cells (DCs) loaded with autologous tumor-cell lysates in 15 patients affected by recurrent GB. The median progression-free survival (PFS) of this patient cohort was 4.4 mo, and the median overall survival (OS) was 8.0 mo. Patients with small tumors at the time of the first vaccination (< 20 cm3; n = 8) had significantly longer PFS and OS than the other patients (6.0 vs. 3.0 mo, p = 0.01; and 16.5 vs. 7.0 mo, p = 0.003, respectively). CD8+ T cells, CD56+ natural killer (NK) cells and other immune parameters, such as the levels of transforming growth factor β, vascular endothelial growth factor, interleukin-12 and interferon γ (IFNγ), were measured in the peripheral blood and serum of patients before and after immunization, which enabled us to obtain a vaccination/baseline ratio (V/B ratio). An increased V/B ratio for NK cells, but not CD8+ T cells, was significantly associated with prolonged PFS and OS. Patients exhibiting NK-cell responses were characterized by high levels of circulating IFNγ and E4BP4, an NK-cell transcription factor. Furthermore, the NK cell V/B ratio was inversely correlated with the TGFβ2 and VEGF V/B ratios. These results suggest that tumor-loaded DCs may increase the survival rate of patients with recurrent GB after effective tumor debulking, and emphasize the role of the NK-cell response in this therapeutic setting. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
32. Frequency of NFKBIA deletions is low in glioblastomas and skewed in glioblastoma neurospheres.
- Author
-
Patanè, Monica, Porrati, Paola, Bottega, Elisa, Morosini, Sara, Cantini, Gabriele, Girgenti, Vita, Rizzo, Ambra, Eoli, Marica, Pollo, Bianca, Sciacca, Francesca L., Pellegatta, Serena, and Finocchiaro, Gaetano
- Subjects
TRANSCRIPTION factors ,QUANTITATIVE research ,POLYMERASE chain reaction ,EPIDERMAL growth factor receptors ,X chromosome ,GENE amplification - Abstract
The NF-kB family of transcription factors is up-regulated in inflammation and different cancers. Recent data described heterozygous deletions of the NF-kB Inhibitor alpha gene (NFKBIA) in about 20 % of glioblastomas (GBM): deletions were mutually exclusive with epidermal growth factor receptor (EGFR) amplification, a frequent event in GBM. We reassessed the status of NFKBIA and EGFR in 69 primary GBMs and in corresponding neurospheres (NS). NFKBIA deletion was investigated by the copy number variation assay (CNV); EGFR amplification by CNV ratio with HGF; expression of EGFR and EGFRvIII by quantitative PCR or ReverseTranscriptase PCR. Heterozygous deletions of NFKBIA were present in 3 of 69 primary GBMs and, surprisingly, in 30 of 69 NS. EGFR amplification was detected in 36 GBMs: in corresponding NS, amplification was lost in 13 cases and reduced in 23 (10 vs 47 folds in NS vs primary tumors; p < 0.001). The CNV assay was validated investigating HPRT1 on chromosome X in females and males. Results of array-CGH performed on 3 primary GBMs and 1 NS line were compatible with the CNV assay. NS cells with NFKBIA deletion had increased nuclear activity of p65 (RelA) and increased expression of the NF-kB target IL-6. In absence of EGF in the medium, EGFR amplification was more conserved and NFKBIA deletion less frequent. Our data point to a low frequency of NFKBIA in GBM and suggest that EGF in the culture medium of NS may affect frequency not only of EGFR amplifications but also of NFKBIA deletions. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
33. A Radial Glia Gene Marker, Fatty Acid Binding Protein 7 (FABP7), Is Involved in Proliferation and Invasion of Glioblastoma Cells.
- Author
-
De Rosa, Antonella, Pellegatta, Serena, Rossi, Marco, Tunici, Patrizia, Magnoni, Letizia, Speranza, Maria Carmela, Malusa, Federico, Miragliotta, Vincenzo, Mori, Elisa, Finocchiaro, Gaetano, and Bakker, Annette
- Subjects
- *
GLIOBLASTOMA multiforme , *GENETIC markers , *CANCER cells , *STEM cells , *DNA microarrays , *CELL proliferation - Abstract
Glioblastoma multiforme (GBM) is among the most deadly cancers. A number of studies suggest that a fraction of tumor cells with stem cell features (Glioma Stem-like Cells, GSC) might be responsible for GBM recurrence and aggressiveness. GSC similarly to normal neural stem cells, can form neurospheres (NS) in vitro, and seem to mirror the genetic features of the original tumor better than glioma cells growing adherently in the presence of serum. Using cDNA microarray analysis we identified a number of relevant genes for glioma biology that are differentially expressed in adherent cells and neurospheres derived from the same tumor. Fatty acid-binding protein 7 (FABP7) was identified as one of the most highly expressed genes in NS compared to their adherent counterpart. We found that down-regulation of FABP7 expression in NS by small interfering RNAs significantly reduced cell proliferation and migration. We also evaluated the potential involvement of FABP7 in response to radiotherapy, as this treatment may cause increased tumor infiltration. Migration of irradiated NS was associated to increased expression of FABP7. In agreement with this, in vivo reduced tumorigenicity of GBM cells with down-regulated expression of FABP7 was associated to decreased expression of the migration marker doublecortin. Notably, we observed that PPAR antagonists affect FABP7 expression and decrease the migration capability of NS after irradiation. As a whole, the data emphasize the role of FABP7 expression in GBM migration and provide translational hints on the timing of treatment with anti-FABP7 agents like PPAR antagonists during GBM evolution. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
34. An Optimized Method for Manufacturing a Clinical Scale Dendritic Cell-Based Vaccine for the Treatment of Glioblastoma.
- Author
-
Nava, Sara, Dossena, Marta, Pogliani, Simona, Pellegatta, Serena, Antozzi, Carlo, Baggi, Fulvio, Gellera, Cinzia, Pollo, Bianca, Parati, Eugenio A., Finocchiaro, Gaetano, and Frigerio, Simona
- Subjects
DENDRITIC cells ,CANCER patients ,IMMUNOLOGY ,LEUKOCYTES ,CANCER cells - Abstract
Immune-based treatments represent a promising new class of therapy designed to boost the immune system to specifically eradicate malignant cells. Immunotherapy may generate specific anti-tumor immune responses, and dendritic cells (DC), professional antigen-presenting cells, are widely used in experimental cancer immunotherapy. Several reports describe methods for the generation of mature, antigen-pulsed DC for clinical use. Improved quality and standardization are desirable to obtain GMP-compliant protocols. In this study we describe the generation of DC from 31 Glioblastoma (GB) patients starting from their monocytes isolated by immunomagnetic CD14 selection using the CliniMACS® device. Upon differentiation of CD14+ with IL-4 and GM-CSF, DC were induced to maturation with TNF-α, PGE2, IL-1β, and IL-6. Whole tumor lysate was obtained, for the first time, in a closed system using the semi-automated dissociator GentleMACSH. The yield of proteins improved by 130% compared to the manual dissociation method. Interestingly the Mean Fluorescence Intensity for CD83 increased significantly in DC pulsed with ''new method'' lysate compared to DC pulsed with ''classical method'' lysate. Our results indicate that immunomagnetic isolation of CD14
+ monocytes using the CliniMACS® device and their pulsing with whole tumor lysate proteins is a suitable method for clinical-scale generation of high quality, functional DC under GMP-grade conditions. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
35. Rai is a New Regulator of Neural Progenitor Migration and Glioblastoma Invasion.
- Author
-
Ortensi, Barbara, Osti, Daniela, Pellegatta, Serena, Pisati, Federica, Brescia, Paola, Fornasari, Lorenzo, Levi, Daniel, Gaetani, Paolo, Colombo, Piergiuseppe, Ferri, Anna, Nicolis, Silvia, Finocchiaro, Gaetano, and Pelicci, Giuliana
- Subjects
GLIOBLASTOMA multiforme ,CANCER cells ,NEURAL stem cells ,PROGENITOR cells ,CELL migration ,ADAPTOR proteins - Abstract
The invasive nature of glioblastoma (GBM) is one important reason for treatment failure. GBM stem/progenitor cells retain the migratory ability of normal neural stem/progenitor cells and infiltrate the brain parenchyma. Here, we identify Rai (ShcC/N-Shc), a member of the family of Shc-like adaptor proteins, as a new regulator of migration of normal and cancer stem/progenitor cells. Rai is expressed in neurogenic areas of the brain and its knockdown impairs progenitor migration to the olfactory bulb. Its expression is retained in GBM stem/progenitor cells where it exerts the same promigratory activity. Rai silencing in cancer stem/progenitor cells isolated from different patients causes significant decrease in cell migration and invasion, both in vitro and in vivo, providing survival benefit. Rai depletion is associated with alteration of multiple-signaling pathways, yet it always leads to reduced expression of proinvasive genes. S TEM C ELLS 2012;30:817-832 [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
36. Immunotherapy for glioma: getting closer to the clinical arena?
- Author
-
Finocchiaro, Gaetano and Pellegatta, Serena
- Published
- 2011
- Full Text
- View/download PDF
37. DNA Microarray Analysis Identifies CKS2 and LEPR as Potential Markers of Meningioma Recurrence.
- Author
-
Menghi, Francesca, Orzan, Francesca N., Eoli, Marica, Farinotti, Mariangela, Maderna, Emanuela, Pisati, Federica, Bianchessi, Donatella, Valletta, Lorella, Lodrini, Sandro, Galli, Giuseppe, Anghileri, Elena, Pellegatta, Serena, Pollo, Bianca, and Finocchiaro, Gaetano
- Subjects
BIOMARKERS ,DNA ,GENE expression ,HISTOLOGY ,IMMUNOHISTOCHEMISTRY ,LONGITUDINAL method ,MENINGIOMA ,POLYMERASE chain reaction ,T-test (Statistics) ,DISEASE relapse ,BIOCHIPS - Abstract
Meningiomas are the most frequent intracranial tumors. Surgery can be curative, but recurrences are possible. We performed gene expression analyses and loss of heterozygosity (LOH) studies looking for new markers predicting the recurrence risk. We analyzed expression profiles of 23 meningiomas (10 grade I, 10grade II, and 3 grade III) and validated the data using quantitative polymerase chain reaction (qPCR). We performed LOH analysis on 40 meningiomas, investigating chromosomal regions on 1p, 9p, 10q, 14q, and22q. We found 233 and 268 probe sets to be significantly down- and up regulated, respectively, in grade II or III meningiomas. Genes down regulated in high-grade meningiomas were overrepresented on chromosomes 1, 6, 9, 10, and 14. Based on functional enrichment analysis, we selected LIM domain and actin binding 1 (LIMA1), tissue inhibitor of metalloproteinases 3 (TIMP3), cyclin-dependent kinases regulatory subunit 2 (CKS2), leptin receptor (LEPR), and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) for validation using qPCR and confirmed their differential expression in the two groups of tumors. We calculated ΔCt values of CKS2 and LEPR and found that their differential expression (C-L index) was significantly higher in grade I than in grade II or III meningiomas (p < .0001). Interestingly, the C-L index of nine grade I meningiomas from patients who relapsed in <5 years was significantly lower than in grade I meningiomas from patients who did not relapse. These findings indicate that the C-L index may be relevant to define the progression risk in meningioma patients, helping guide their clinical management. A prospective analysis on a larger number of cases is warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
38. Intra-tumoral dendritic cells increase efficacy of peripheral vaccination by modulation of glioma microenvironment.
- Author
-
Pellegatta, Serena, Poliani, Pietro Luigi, Stucchi, Elena, Corno, Daniela, Colombo, Chiara Agnese, Orzan, Francesca, Ravanini, Maria, and Finocchiaro, Gaetano
- Published
- 2010
- Full Text
- View/download PDF
39. Deciphering the Labyrinthine System of the Immune Microenvironment in Recurrent Glioblastoma: Recent Original Advances and Lessons from Clinical Immunotherapeutic Approaches.
- Author
-
Anghileri, Elena, Patanè, Monica, Di Ianni, Natalia, Sambruni, Irene, Maffezzini, Martina, Milani, Micaela, Maddaloni, Luisa, Pollo, Bianca, Eoli, Marica, and Pellegatta, Serena
- Subjects
GLIOMAS ,CELL physiology ,CANCER relapse ,LYMPHOCYTES ,TREATMENT effectiveness ,RISK assessment ,CANCER patients ,IMMUNOTHERAPY - Abstract
Simple Summary: Active communication between GBM cells and tumor-infiltrating immune components contributes to establishing an immunosuppressive environment where T cells are scarce and exhausted. This condition is particularly exacerbated upon recurrence, which is almost inevitable for GBM patients. Immunotherapeutic approaches, including checkpoint inhibitors, have demonstrated limited efficacy and failed to prolong survival or recurrent GBM patients. Nevertheless, many studies have shown that T cell priming is possible in the GBM microenvironment, and that T cell exhaustion or dysfunction can be reprogrammed. We will revisit data from the literature and report original results obtained from recurrent GBM patients treated with dendritic cells to demonstrate the role of the microenvironment in predicting immunotherapy response and influencing decisions for personalized therapies. The interpretation of the presence and function of immune infiltration in glioblastoma (GBM) is still debated. Over the years, GBM has been considered a cold tumor that is less infiltrated by effector cells and characterized by a high proportion of immunosuppressive innate immune cells, including GBM-associated microglia/macrophages (GAMs). In this context, the failure of checkpoint inhibitors, particularly in recurrent GBM (rGBM), caused us to look beyond the clinical results and consider the point of view of immune cells. The tumor microenvironment in rGBM can be particularly hostile, even when exposed to standard immunomodulatory therapies, and tumor-infiltrating lymphocytes (TILs), when present, are either dysfunctional or terminally exhausted. However, after checkpoint blockade therapy, it was possible to observe specific recruitment of adaptive immune cells and an efficient systemic immune response. In this review article, we attempt to address current knowledge regarding the tumor and immune microenvironment in rGBM. Furthermore, immunosuppression induced by GAMs and TIL dysfunction was revisited to account for genetic defects that can determine resistance to therapies and manipulate the immune microenvironment upon recurrence. Accordingly, we reevaluated the microenvironment of some of our rGBM patients treated with dendritic cell immunotherapy, with the goal of identifying predictive immune indicators of better treatment response. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
40. NG2/CSPG4 in glioblastoma: about flexibility.
- Author
-
Finocchiaro, Gaetano and Pellegatta, Serena
- Published
- 2019
- Full Text
- View/download PDF
41. Altered Metabolism in Glioblastoma: Myeloid-Derived Suppressor Cell (MDSC) Fitness and Tumor-Infiltrating Lymphocyte (TIL) Dysfunction.
- Author
-
Di Ianni, Natalia, Musio, Silvia, Pellegatta, Serena, and Hau, Peter
- Subjects
MYELOID-derived suppressor cells ,LIPID metabolism ,LYMPHOCYTES ,METABOLISM ,GLIOBLASTOMA multiforme ,BRAIN tumors - Abstract
The metabolism of glioblastoma (GBM), the most aggressive and lethal primary brain tumor, is flexible and adaptable to different adverse conditions, such as nutrient deprivation. Beyond glycolysis, altered lipid metabolism is implicated in GBM progression. Indeed, metabolic subtypes were recently identified based on divergent glucose and lipid metabolism. GBM is also characterized by an immunosuppressive microenvironment in which myeloid-derived suppressor cells (MDSCs) are a powerful ally of tumor cells. Increasing evidence supports the interconnection between GBM and MDSC metabolic pathways. GBM cells exert a crucial contribution to MDSC recruitment and maturation within the tumor microenvironment, where the needs of tumor-infiltrating lymphocytes (TILs) with antitumor function are completely neglected. In this review, we will discuss the unique or alternative source of energy exploited by GBM and MDSCs, exploring how deprivation of specific nutrients and accumulation of toxic byproducts can induce T-cell dysfunction. Understanding the metabolic programs of these cell components and how they impact fitness or dysfunction will be useful to improve treatment modalities, including immunotherapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
42. Sonodynamic Therapy for the Treatment of Intracranial Gliomas.
- Author
-
D'Ammando, Antonio, Raspagliesi, Luca, Gionso, Matteo, Franzini, Andrea, Porto, Edoardo, Di Meco, Francesco, Durando, Giovanni, Pellegatta, Serena, Prada, Francesco, and Acerbi, Francesco
- Subjects
CEREBELLAR tumors ,GLIOMAS ,PROGRESSION-free survival ,BRAIN tumors ,ULTRASONIC imaging - Abstract
High-grade gliomas are the most common and aggressive malignant primary brain tumors. Current therapeutic schemes include a combination of surgical resection, radiotherapy and chemotherapy; even if major advances have been achieved in Progression Free Survival and Overall Survival for patients harboring high-grade gliomas, prognosis still remains poor; hence, new therapeutic options for malignant gliomas are currently researched. Sonodynamic Therapy (SDT) has proven to be a promising treatment combining the effects of low-intensity ultrasound waves with various sound-sensitive compounds, whose activation leads to increased immunogenicity of tumor cells, increased apoptotic rates and decreased angiogenetic potential. In addition, this therapeutic technique only exerts its cytotoxic effects on tumor cells, while both ultrasound waves and sensitizing compound are non-toxic per se. This review summarizes the present knowledge regarding mechanisms of action of SDT and currently available sonosensitizers and focuses on the preclinical and clinical studies that have investigated its efficacy on malignant gliomas. To date, preclinical studies implying various sonosensitizers and different treatment protocols all seem to confirm the anti-tumoral properties of SDT, while first clinical trials will soon start recruiting patients. Accordingly, it is crucial to conduct further investigations regarding the clinical applications of SDT as a therapeutic option in the management of intracranial gliomas. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
43. PGE2 Is Crucial for the Generation of FAST Whole- Tumor-Antigens Loaded Dendritic Cells Suitable for Immunotherapy in Glioblastoma.
- Author
-
Nava, Sara, Lisini, Daniela, Frigerio, Simona, Pogliani, Simona, Pellegatta, Serena, Gatti, Laura, Finocchiaro, Gaetano, Bersano, Anna, and Parati, Eugenio Agostino
- Subjects
DENDRITIC cells ,GLIOBLASTOMA multiforme ,IMMUNOTHERAPY ,CURRENT good manufacturing practices ,CYTOTOXIC T cells ,IMMUNE response ,MONOCYTES ,T cells - Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells, strongly inducers of T cell-mediated immune responses and, as such, broadly used as vaccine adjuvant in experimental clinical settings. DC are widely generated from human monocytes following in vitro protocols which require 5–7 days of differentiation with GM-CSF and IL-4 followed by 2–3 days of activation/maturation. In attempts to shorten the vaccine's production, Fast-DC protocols have been developed. Here we reported a Fast-DC method in compliance with good manufacturing practices for the production of autologous mature dendritic cells loaded with antigens derived from whole tumor lysate, suitable for the immunotherapy in glioblastoma patients. The feasibility of generating Fast-DC pulsed with whole tumor lysate was assessed using a series of small-scale cultures performed in parallel with clinical grade large scale standard method preparations. Our results demonstrate that this Fast protocol is effective only in the presence of PGE
2 in the maturation cocktail to guarantee that Fast-DC cells exhibit a mature phenotype and fulfill all requirements for in vivo use in immunotherapy approaches. Fast-DC generated following this protocol were equally potent to standard DC in inducing Ag-specific T cell proliferation in vitro. Generation of Fast-DC not only reduces labor, cost, and time required for in vitro clinical grade DC development, but can also minimizes inter-preparations variability and the risk of contamination. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
44. ABCC3 Expressed by CD56dim CD16+ NK Cells Predicts Response in Glioblastoma Patients Treated with Combined Chemotherapy and Dendritic Cell Immunotherapy.
- Author
-
Pellegatta, Serena, Di Ianni, Natalia, Pessina, Sara, Paterra, Rosina, Anghileri, Elena, Eoli, Marica, and Finocchiaro, Gaetano
- Subjects
- *
KILLER cells , *GLIOBLASTOMA multiforme , *DENDRITIC cells , *MULTIDRUG resistance , *SINGLE nucleotide polymorphisms , *IMMUNOTHERAPY - Abstract
Recently, we found that temozolomide (TMZ) can upregulate the expression of the multidrug-resistance protein ABCC3 in NK cells from both glioma-bearing mice and glioblastoma patients treated with dendritic cell immunotherapy combined with TMZ, allowing NK cells to escape apoptosis and favoring their role as antitumor effector cells. Here, we demonstrate that CD56dim NK cells expressing CD16+ are predominant in patients surviving more than 12 months after surgery without disease progression. CD56dim CD16+ NK cells co-expressed high levels of ABCC3 and IFN-γ. Notably, not only basal but also TMZ-induced ABCC3 expression was related to a strong, long-term NK cell response and a better prognosis of patients. The identification of the single nucleotide polymorphism (SNP) rs35467079 with the deletion of a cytosine (−897DelC) in the promoter region of the ABCC3 gene resulted associated with a better patient outcome. ABCC3 expression in patients carrying DelC compared to patients with reference haplotype was higher and modulated by TMZ. The transcription factor NRF2, involved in ABCC3 induction, was phosphorylated in CD56dim CD16+ NK cells expressing ABCC3 under TMZ treatment. Thus, ABCC3 protein and the SNP −897DelC can play a predictive role in patients affected by GBM, and possibly other cancers, treated with dendritic cell immunotherapy combined with chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
45. Advanced MRI Assessment during Dendritic Cell Immunotherapy Added to Standard Treatment against Glioblastoma.
- Author
-
Cuccarini, Valeria, Aquino, Domenico, Gioppo, Andrea, Anghileri, Elena, Pellegatta, Serena, Schettino, Carla, Mazzi, Federica, Finocchiaro, Gaetano, Bruzzone, Maria Grazia, and Eoli, Marica
- Subjects
GLIOBLASTOMA multiforme ,DENDRITIC cells ,KILLER cells ,BLOOD volume ,IMMUNOTHERAPY - Abstract
Evaluating changes induced by immunotherapies (IT) on conventional magnetic resonance imaging (MRI) is difficult because those treatments may produce inflammatory responses. To explore the potential contribution of advanced MRI to distinguish pseudoprogression (PsP) and true tumor progression (TTP), and to identify patients obtaining therapeutic benefit from IT, we examined aMRI findings in newly diagnosed glioblastoma treated with dendritic cell IT added to standard treatment. We analyzed longitudinal MRIs obtained in 22 patients enrolled in the EUDRACT N° 2008-005035-15 trial. According to RANO criteria, we observed 18 TTP and 8 PsP. Comparing MRI performed at the time of TTP/PsP with the previous exam performed two months before, a difference in cerebral blood volume ΔrCBV
max ≥ 0.47 distinguished TTP from PsP with a sensitivity of 67% and specificity of 75% (p = 0.004). A decrease in minimal apparent diffusion coefficient rADCmin (1.15 vs. 1.01, p = 0.003) was observed after four vaccinations only in patients with a persistent increase of natural killer cells (response effectors during IT) in peripheral blood. Basal rADCmin > 1 was independent predictor of longer progression free (16.1 vs. 9 months, p = 0.0001) and overall survival (32.8 vs. 17.5 months, p = 0.0005). In conclusion, rADC predicted response to immunotherapy and survival; Apparent Diffusion Coefficient (ADC) and Cerebral Blood Volume (CBV) modifications over time help differentiating PsP from TTP at onset. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
46. The multidrug-resistance transporter Abcc3 protects NK cells from chemotherapy in a murine model of malignant glioma.
- Author
-
Pessina, Sara, Cantini, Gabriele, Kapetis, Dimos, Cazzato, Emanuela, Di Ianni, Natalia, Finocchiaro, Gaetano, and Pellegatta, Serena
- Subjects
MULTIDRUG resistance ,CARRIER proteins ,KILLER cells ,CANCER chemotherapy ,MURINE gammaherpesvirus diseases ,GLIOMAS ,ATP-binding cassette transporters - Abstract
Abcc3, a member of the ATP-binding cassette transporter superfamily, plays a role in multidrug resistance. Here, we found that Abcc3 is highly expressed in blood-derived NK cells but not in CD8+T cells. In GL261 glioma-bearing mice treated with the alkylating agent temozolomide (TMZ) for 5 d, an early increased frequency of NK cells was observed. We also found that Abcc3 is strongly upregulated and functionally active in NK cells from mice treated with TMZ compared to controls. We demonstrate that Abcc3 is critical for NK cell survival during TMZ administration; more importantly, Akt, involved in lymphocyte survival, is phosphorylated only in NK cells expressing Abcc3. The resistance of NK cells to chemotherapy was accompanied by increased migration and homing in the brain at early time points. Cytotoxicity, evaluated by IFNγ production and specific lytic activity against GL261 cells, increased peripherally in the later phases, after conclusion of TMZ treatment. Intra-tumor increase of the NK effector subset as well as in IFNγ, granzymes and perforin-1 expression, were found early and persisted over time, correlating with a profound modulation on glioma microenvironment induced by TMZ. Our findings reveal an important involvement of Abcc3 in NK cell resistance to chemotherapy and have important clinical implications for patients treated with chemo-immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
47. NK CELLS FROM GLIOMA-BEARING MICE TREATED WITH TEMOZOLOMIDE ARE ENRICHED FOR GENES RELATED TO MULTI-DRUG RESISTANCE AND CHEMOTAXIS.
- Author
-
Pellegatta, Serena, Pessina, Sara, Cantini, Gabriele, Kapetis, Dimos, Cazzato, Emanuela, and Finocchiaro, Gaetano
- Published
- 2014
- Full Text
- View/download PDF
48. Prognostic Value of CD109+ Circulating Endothelial Cells in Recurrent Glioblastomas Treated with Bevacizumab and Irinotecan.
- Author
-
Cuppini, Lucia, Calleri, Angelica, Bruzzone, Maria Grazia, Prodi, Elena, Anghileri, Elena, Pellegatta, Serena, Mancuso, Patrizia, Porrati, Paola, Di Stefano, Anna Luisa, Ceroni, Mauro, Bertolini, Francesco, Finocchiaro, Gaetano, and Eoli, Marica
- Subjects
CELL surface antigens ,ENDOTHELIAL cells ,GLIOBLASTOMA multiforme treatment ,IRINOTECAN ,BEVACIZUMAB ,PROGENITOR cells ,CANCER treatment ,VASCULAR endothelial growth factors ,THERAPEUTICS - Abstract
Background:Recent data suggest that circulating endothelial and progenitor cells (CECs and CEPs, respectively) may have predictive potential in cancer patients treated with bevacizumab, the antibody recognizing vascular endothelial growth factor (VEGF). Here we report on CECs and CEPs investigated in 68 patients affected by recurrent glioblastoma (rGBM) treated with bevacizumab and irinotecan and two Independent Datasets of rGBM patients respectively treated with bevacizumab alone (n=32, independent dataset A: IDA) and classical antiblastic chemotherapy (n=14, independent dataset B: IDB). Methods:rGBM patients with KPS ≥50 were treated until progression, as defined by MRI with RANO criteria. CECs expressing CD109, a marker of tumor endothelial cells, as well as other CEC and CEP subtypes, were investigated by six-color flow cytometry. Results:A baseline count of CD109+ CEC higher than 41.1/ml (1
st quartile) was associated with increased progression free survival (PFS; 20 versus 9 weeks, P=0.008) and overall survival (OS; 32 versus 23 weeks, P=0.03). Longer PFS (25 versus 8 weeks, P=0.02) and OS (27 versus 17 weeks, P=0.03) were also confirmed in IDA with CD109+ CECs higher than 41.1/ml but not in IDB. Patients treated with bevacizumab with or without irinotecan that were free from MRI progression after two months of treatment had significant decrease of CD109+ CECs: median PFS was 19 weeks; median OS 29 weeks. The presence of two non-contiguous lesions (distant disease) at baseline was an independent predictor of shorter PFS and OS (P<0.001). Conclusions:Data encourage further studies on the predictive potential of CD109+ CECs in GBM patients treated with bevacizumab. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
49. Sox2 Is Required to Maintain Cancer Stem Cells in a Mouse Model of High-Grade Oligodendroglioma.
- Author
-
Favaro, Rebecca, Appolloni, Irene, Pellegatta, Serena, Badiola Sanga, Alexandra, Pagella, Pierfrancesco, Gambini, Eleonora, Pisati, Federica, Ottolenghi, Sergio, Foti, Maria, Finocchiaro, Gaetano, Malatesta, Paolo, and Nicolis, Silvia K.
- Subjects
- *
TRANSCRIPTION factors , *CANCER cells , *STEM cells , *OLIGODENDROGLIOMAS , *CANCER research - Abstract
The stem cell-determining transcription factor Sox2 is required for the maintenance of normal neural stem cells. In this study, we investigated the requirement for Sox2 in neural cancer stem-like cells using a conditional genetic deletion mutant in a mouse model of platelet-derived growth factor-induced malignant oligodendroglioma. Transplanting wild-type oligodendroglioma cells into the brain generated lethal tumors, but mice transplanted with Sox2-deleted cells remained free of tumors. Loss of the tumor-initiating ability of Sox2-deleted cells was reversed by lentiviral-mediated expression of Sox2. In cell culture, Sox2-deleted tumor cells were highly sensitive to differentiation stimuli, displaying impaired proliferation, increased cell death, and aberrant differentiation. Gene expression analysis revealed an early transcriptional response to Sox2 loss. The observed requirement of oligodendroglioma stem cells for Sox2 suggested its relevance as a target for therapy. In support of this possibility, an immunotherapeutic approach based on immunization of mice with SOX2 peptides delayed tumor development and prolonged survival. Taken together, our results showed that Sox2 is essential for tumor initiation by mouse oligodendroglioma cells, and they illustrated a Sox2-directed strategy of immunotherapy to eradicate tumor-initiating cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
50. The MET Oncogene Is a Functional Marker of a Glioblastoma Stem Cell Subtype.
- Author
-
De Bacco, Francesca, Casanova, Elena, Medico, Enzo, Pellegatta, Serena, Orzan, Francesca, Albano, Raffaella, Luraghi, Paolo, Reato, Gigliola, D'Ambrosio, Antonio, Porrati, Paola, Patanè, Monica, Maderna, Emanuela, Pollo, Bianca, Comoglio, Paolo M., Finocchiaro, Gaetano, and Boccaccio, Carla
- Subjects
- *
CANCER treatment , *STEM cell treatment , *ONCOGENES , *GLIOBLASTOMA multiforme treatment , *CELL cycle , *GENOTYPE-environment interaction - Abstract
The existence of treatment-resistant cancer stem cells contributes to the aggressive phenotype of glioblastoma. However, the molecular alterations that drive stem cell proliferation in these tumors remain unknown. In this study, we found that expression of the MET oncogene was associated with neurospheres expressing the gene signature of mesenchymal and proneural subtypes of glioblastoma. Met expression was almost absent from neurospheres expressing the signature of the classical subtype and was mutually exclusive with amplification and expression of the EGF receptor (EGFR) gene. Met-positive and Met-negative neurospheres displayed distinct growth factor requirements, differentiated along divergent pathways, and generated tumors with distinctive features. The Methigh subpopulation within Met-pos neurospheres displayed clonogenic potential and long-term self-renewal ability in vitro and enhanced growth kinetics in vivo. In Methigh cells, the Met ligand HGF further sustained proliferation, clonogenicity, expression of self-renewal markers, migration, and invasion in vitro. Together, our findings suggest that Met is a functional marker of glioblastoma stem cells and a candidate target for identification and therapy of a subset of glioblastomas. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.