127 results on '"Pellegatta, Serena"'
Search Results
102. Immunotherapy for glioma: getting closer to the clinical arena?
- Author
-
Finocchiaro, Gaetano and Pellegatta, Serena
- Published
- 2011
- Full Text
- View/download PDF
103. 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
104. 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
105. 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
106. ADVANCED MRI ASSESSMENT DURING DENDRITIC CELL IMMUNOTHERAPY
- Author
-
Eoli, Marica, Cuccarini, Valeria, Domenico Aquino, Anghileri, Elena, Pellegatta, Serena, Finocchiaro, Gaetano, and Bruzzone, Maria Grazia
107. GLIOBLASTOMA STEM-LIKE CELLS ARE MORE FREQUENTLY GENERATED BY TUMORS WITH UNMETHYLATED MGMT
- Author
-
Pellegatta, Serena, Orzan, Francesca, Elena Anghileri, Guzzetti, Sara, Porrati, Paola, Eoli, Marcia, and Finocchiaro, Gaetano
108. NEUROSPHERES FROM GLIOBLASTOMA MULTIFORME EXPRESS HIGH LEVELS OF EZH2 AND ARISE MORE FREQUENTLY FROM TUMORS WITH LOW/ABSENT MGMT METHYLATION
- Author
-
Orzan, Francesca, Pellegatta, Serena, Guzzetti, Sara, Villa, Giulia, Menghi, Francesca, Poliani, Luigi, Pisati, Federica, Caldera, Valentina, Stucchi, Elena, Bettolini, Rossana, Porrati, Paola, Eoli, Marica, Farinotti, Mariangela, Marras, Carlo E., Tunici, Patrizia, maria grazia bruzzone, Schiffer, Davide, and Finocchiaro, Gaetano
109. HIGH LEVELS OF IL-8 AND IL-13 AT BASELINE AND ABSENCE OF EARLY DECREASE OF VEGF IN PLASMA OF PATIENTS WITH RECURRENT GLIOBLASTOMA NOT RESPONDING TO BEVACIZUMAB
- Author
-
Eoli, Marica, Rabascio, Cristin, Cuppini, Lucia, Elena Anghileri, Pellegatta, Serena, Calleri, Angelica, Mancuso, Patrizia, Porrati, Paola, Bertolini, Francesco, and Finocchiaro, Gaetano
110. TUMOR DEBULKING AND CD56+CELL RESPONSES ARE ASSOCIATED WITH PROLONGED SURVIVAL IN RELAPSING GLIOBLASTOMAS TREATED BY TUMOR-LOADED DENDRITIC CELLS
- Author
-
Pellegatta, Serena, Eoli, Marica, Antozzi, Carlo, Frigerio, Simona, Bruzzone, Maria Grazia, Cuppini, Lucia, Nava, Sara, Anghileri, Elena, Cantini, Gabriele, Prodi, Elena, Ciusani, Emilio, Paolo Ferroli, Saini, Marco, Broggi, Giovanni, Mantegazza, Renato, Parati, Eugenio A., and Finocchiaro, Gaetano
111. RELEVANCE OF CD109+CIRCULATING ENDOTHELIAL CELLS IN RECURRENT HIGH GRADE GLIOMAS TREATED WITH BEVACIZUMAB AND IRINOTECAN
- Author
-
Eoli, Marica, Calleri, Angelica, Cuppini, Lucia, Anghileri, Elena, Pellegatta, Serena, Prodi, Elena, maria grazia bruzzone, Bertolini, Francesco, and Finocchiaro, Gaetano
112. Survival gain in glioblastoma patients treated with dendritic cell immunotherapy is associated with increased NK but not CD8+T cell activation in the presence of adjuvant temozolomide
- Author
-
Pellegatta, Serena, Eoli, Marica, Cuccarini, Valeria, Anghileri, Elena, Pollo, Bianca, Pessina, Sara, Frigerio, Simona, Servida, Maura, Cuppini, Lucia, Antozzi, Carlo, Cuzzubbo, Stefania, Corbetta, Cristina, Paterra, Rosina, Acerbi, Francesco, Ferroli, Paolo, DiMeco, Francesco, Fariselli, Laura, Parati, Eugenio A., Bruzzone, Maria Grazia, and Finocchiaro, Gaetano
- Abstract
ABSTRACTIn a two-stage phase II study, 24 patients with first diagnosis of glioblastoma (GBM) were treated with dendritic cell (DC) immunotherapy associated to standard radiochemotherapy with temozolomide (TMZ) followed by adjuvant TMZ.Three intradermal injections of mature DC loaded with autologous GBM lysate were administered before adjuvant TMZ, while 4 injections were performed during adjuvant TMZ. According to a two-stage Simon design, to proceed to the second stage progression-free survival (PFS) 12 months after surgery was expected in at least 8 cases enrolled in the first stage. Evidence of immune response and interaction with chemotherapy were investigated. After a median follow up of 17.4 months, 9 patients reached PFS12. In these patients (responders, 37.5%), DC vaccination induced a significant, persistent activation of NK cells, whose increased response was significantly associated with prolonged survival. CD8+T cells underwent rapid expansion and priming but, after the first administration of adjuvant TMZ, failed to generate a memory status. Resistance to TMZ was associated with robust expression of the multidrug resistance protein ABCC3 in NK but not CD8+T cells. The negative effect of TMZ on the formation of T cell-associated antitumor memory deserves consideration in future clinical trials including immunotherapy.
- Published
- 2018
- Full Text
- View/download PDF
113. 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
114. 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
115. DNA Damage Repair in Glioblastoma: A Novel Approach to Combat Drug Resistance.
- Author
-
Gaiaschi L, Casali C, Stabile A, D'Amico S, Ravera M, Gabano E, Galluzzo A, Favaron C, Gola F, De Luca F, Pellegatta S, and Bottone MG
- Abstract
Due to the lack of effective therapeutic approach, glioblastoma (GBM) remains one of the most malignant brain tumour. By in vitro investigations on primary GBM stem cells, we highlighted one of the underlying mechanisms of drug resistance to alkylating agents, the DNA damage responses. Here, flow cytometric analysis and viability and repopulation assays were used to assess the long-term cytotoxic effect induced by the administration of a fourth-generation platinum prodrug, the (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato) platinum(IV) named Pt(IV)Ac-POA, in comparison to the most widely used Cisplatin. The immunofluorescence studies revealed changing pathways involved in the DNA damage response mechanisms in response to the two chemotherapies, suggesting in particular the role of Poly (ADP-Ribose) polymerases in the onset of resistance to Cisplatin-induced cytotoxicity. Thus, this research provides a proof of concept for how the use of a prodrug which allows the co-administration of Cisplatin and an Histone DeACetylase inhibitors, could suppress DNA repair mechanisms, suggesting a novel effective approach in GBM treatment., (© 2025 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.)
- Published
- 2025
- Full Text
- View/download PDF
116. Response assessment of GBM during immunotherapy by delayed contrast treatment response assessment maps.
- Author
-
Cuccarini V, Savoldi F, Mardor Y, Last D, Pellegatta S, Mazzi F, Bruzzone MG, Anghileri E, Pollo B, Maddaloni L, Russo C, Bocchi E, Pinzi V, Eoli M, and Aquino D
- 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., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Cuccarini, Savoldi, Mardor, Last, Pellegatta, Mazzi, Bruzzone, Anghileri, Pollo, Maddaloni, Russo, Bocchi, Pinzi, Eoli and Aquino.)- Published
- 2024
- Full Text
- View/download PDF
117. 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 S, Corradino N, Zingarelli M, Porto E, Gionso M, Berlendis A, Durando G, Maffezzini M, Musio S, Aquino D, DiMeco F, and Prada F
- Abstract
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.
- Published
- 2024
- Full Text
- View/download PDF
118. Next-generation agents for fluorescence-guided glioblastoma surgery.
- Author
-
Chirizzi C, Pellegatta S, Gori A, Falco J, Rubiu E, Acerbi F, and Bombelli FB
- 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., Competing Interests: Dr. Francesco Acerbi received honoraria from Carl Zeiss Meditec for lectures in International Meetings. Other authors declare no conflict of interest., (© 2023 The Authors. Bioengineering & Translational Medicine published by Wiley Periodicals LLC on behalf of American Institute of Chemical Engineers.)
- Published
- 2023
- Full Text
- View/download PDF
119. Coexisting cancer stem cells with heterogeneous gene amplifications, transcriptional profiles, and malignancy are isolated from single glioblastomas.
- Author
-
De Bacco F, Orzan F, Crisafulli G, Prelli M, Isella C, Casanova E, Albano R, Reato G, Erriquez J, D'Ambrosio A, Panero M, Dall'Aglio C, Casorzo L, Cominelli M, Pagani F, Melcarne A, Zeppa P, Altieri R, Morra I, Cassoni P, Garbossa D, Cassisa A, Bartolini A, Pellegatta S, Comoglio PM, Finocchiaro G, Poliani PL, and Boccaccio C
- Subjects
- Humans, Gene Amplification, Neoplastic Stem Cells metabolism, Carcinogenesis pathology, Cell Line, Tumor, Glioblastoma pathology, Brain Neoplasms metabolism
- Abstract
Glioblastoma (GBM) is known as an intractable, highly heterogeneous tumor encompassing multiple subclones, each supported by a distinct glioblastoma stem cell (GSC). The contribution of GSC genetic and transcriptional heterogeneity to tumor subclonal properties is debated. In this study, we describe the systematic derivation, propagation, and characterization of multiple distinct GSCs from single, treatment-naive GBMs (GSC families). The tumorigenic potential of each GSC better correlates with its transcriptional profile than its genetic make-up, with classical GSCs being inherently more aggressive and mesenchymal more dependent on exogenous growth factors across multiple GBMs. These GSCs can segregate and recapitulate different histopathological aspects of the same GBM, as shown in a paradigmatic tumor with two histopathologically distinct components, including a conventional GBM and a more aggressive primitive neuronal component. This study provides a resource for investigating how GSCs with distinct genetic and/or phenotypic features contribute to individual GBM heterogeneity and malignant escalation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
120. ERBB3 overexpression due to miR-205 inactivation confers sensitivity to FGF, metabolic activation, and liability to ERBB3 targeting in glioblastoma.
- Author
-
De Bacco F, Orzan F, Erriquez J, Casanova E, Barault L, Albano R, D'Ambrosio A, Bigatto V, Reato G, Patanè M, Pollo B, Kuesters G, Dell'Aglio C, Casorzo L, Pellegatta S, Finocchiaro G, Comoglio PM, and Boccaccio C
- Subjects
- Antibodies metabolism, Apoptosis, Cell Line, Tumor, Fibroblast Growth Factor 2, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Humans, MicroRNAs genetics, Oligodendroglia metabolism, Phosphatidylinositol 3-Kinases metabolism, Prognosis, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptor, ErbB-3 antagonists & inhibitors, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction, Spheroids, Cellular pathology, TOR Serine-Threonine Kinases metabolism, Glioblastoma genetics, MicroRNAs metabolism, Receptor, ErbB-3 metabolism
- Abstract
In glioblastoma (GBM), the most frequent and lethal brain tumor, therapies suppressing recurrently altered signaling pathways failed to extend survival. However, in patient subsets, specific genetic lesions can confer sensitivity to targeted agents. By exploiting an integrated model based on patient-derived stem-like cells, faithfully recapitulating the original GBMs in vitro and in vivo, here, we identify a human GBM subset (∼9% of all GBMs) characterized by ERBB3 overexpression and nuclear accumulation. ERBB3 overexpression is driven by inheritable promoter methylation or post-transcriptional silencing of the oncosuppressor miR-205 and sustains the malignant phenotype. Overexpressed ERBB3 behaves as a specific signaling platform for fibroblast growth factor receptor (FGFR), driving PI3K/AKT/mTOR pathway hyperactivation, and overall metabolic upregulation. As a result, ERBB3 inhibition by specific antibodies is lethal for GBM stem-like cells and xenotransplants. These findings highlight a subset of patients eligible for ERBB3-targeted therapy., Competing Interests: Declaration of interests G.K. is a shareholder of Merrimack stock. All the other authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
121. 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 E, Di Ianni N, Paterra R, Langella T, Zhao J, Eoli M, Patanè M, Pollo B, Cuccarini V, Iavarone A, Rabadan R, Finocchiaro G, and Pellegatta S
- Subjects
- Adult, Biopsy, Colorectal Neoplasms, Hereditary Nonpolyposis diagnosis, Colorectal Neoplasms, Hereditary Nonpolyposis drug therapy, Combined Modality Therapy, Female, Glioblastoma diagnostic imaging, Glioblastoma therapy, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Immunohistochemistry, Magnetic Resonance Imaging, Molecular Targeted Therapy, Neoplasm Recurrence, Local, Neuroimaging, Programmed Cell Death 1 Receptor antagonists & inhibitors, Retreatment, T-Lymphocytes drug effects, Treatment Outcome, Exome Sequencing, Biomarkers, Tumor, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Glioblastoma pathology, Mutation, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Background: Glioblastomas (GBMs) in patients harboring somatic or germinal mutations of mismatch-repair (MMR) genes exhibit a hypermutable phenotype. Here, we describe a GBM patient with increased tumor mutational burden and germline MMR mutations, treated using anti-PD1 therapy., Methods: A woman with newly diagnosed GBM (nGBM) was treated by surgery, radiotherapy, and temozolomide. The tumor recurred after 13 months leading to a second surgery and treatment with nivolumab. Whole-exome sequencing was performed on the nGBM, recurrent GBM (rGBM), and blood. Immune infiltration was investigated by immunohistochemistry and the immune response in the blood during treatment was analyzed by flow cytometry., Results: High density of infiltrating CD163 + cells was found in both GBM specimens. Large numbers of CD3 + and CD8 + T cells were homogeneously distributed in the nGBM. The infiltration of CD4 + T cells and a different CD8 + T cell density were observed in the rGBM. Both GBM shared 12,431 somatic mutations, with 113 substitutions specific to the nGBM and 1,683 specific to the rGBM. Germline variants included pathogenic mutation in the MSH2 (R359S) gene, suggesting the diagnosis of Lynch syndrome. Systemic immunophenotyping revealed the generation of CD8 + T memory cells and persistent activation of CD4 + T cells. The patient is still receiving nivolumab 68 months after the second surgery., Conclusions: Our observations indicate that the hypermutator phenotype associated with germinal mutations of MMR genes and abundant T-cell infiltration contributes to a durable clinical benefit sustained by a persistent and robust immune response during anti-PD1 therapy.
- Published
- 2021
- Full Text
- View/download PDF
122. PGE 2 Is Crucial for the Generation of FAST Whole- Tumor-Antigens Loaded Dendritic Cells Suitable for Immunotherapy in Glioblastoma.
- Author
-
Nava S, Lisini D, Frigerio S, Pogliani S, Pellegatta S, Gatti L, Finocchiaro G, Bersano A, and Parati EA
- 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.- Published
- 2020
- Full Text
- View/download PDF
123. B7-H3-redirected chimeric antigen receptor T cells target glioblastoma and neurospheres.
- Author
-
Nehama D, Di Ianni N, Musio S, Du H, Patané M, Pollo B, Finocchiaro G, Park JJH, Dunn DE, Edwards DS, Damrauer JS, Hudson H, Floyd SR, Ferrone S, Savoldo B, Pellegatta S, and Dotti G
- Subjects
- Animals, Antigens, Neoplasm immunology, B7 Antigens genetics, Biomarkers, Brain Neoplasms immunology, Brain Neoplasms mortality, Brain Neoplasms therapy, Cell Line, Tumor, Disease Models, Animal, Glioblastoma immunology, Glioblastoma mortality, Glioblastoma therapy, Humans, Immunophenotyping, Immunotherapy, Adoptive, Mice, Neoplastic Stem Cells immunology, Neoplastic Stem Cells metabolism, T-Lymphocytes immunology, Xenograft Model Antitumor Assays, B7 Antigens metabolism, Brain Neoplasms metabolism, Glioblastoma metabolism, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism, T-Lymphocytes metabolism
- Abstract
Background: The dismal survival of glioblastoma (GBM) patients urgently calls for the development of new treatments. Chimeric antigen receptor T (CAR-T) cells are an attractive strategy, but preclinical and clinical studies in GBM have shown that heterogeneous expression of the antigens targeted so far causes tumor escape, highlighting the need for the identification of new targets. We explored if B7-H3 is a valuable target for CAR-T cells in GBM., Methods: We compared mRNA expression of antigens in GBM using TCGA data, and validated B7-H3 expression by immunohistochemistry. We then tested the antitumor activity of B7-H3-redirected CAR-T cells against GBM cell lines and patient-derived GBM neurospheres in vitro and in xenograft murine models., Findings: B7-H3 mRNA and protein are overexpressed in GBM relative to normal brain in all GBM subtypes. Of the 46 specimens analyzed by immunohistochemistry, 76% showed high B7-H3 expression, 22% had detectable, but low B7-H3 expression and 2% were negative, as was normal brain. All 20 patient-derived neurospheres showed ubiquitous B7-H3 expression. B7-H3-redirected CAR-T cells effectively targeted GBM cell lines and neurospheres in vitro and in vivo. No significant differences were found between CD28 and 4-1BB co-stimulation, although CD28-co-stimulated CAR-T cells released more inflammatory cytokines., Interpretation: We demonstrated that B7-H3 is highly expressed in GBM specimens and neurospheres that contain putative cancer stem cells, and that B7-H3-redirected CAR-T cells can effectively control tumor growth. Therefore, B7-H3 represents a promising target in GBM. FUND: Alex's Lemonade Stand Foundation; Il Fondo di Gio Onlus; National Cancer Institute; Burroughs Wellcome Fund., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
124. Survival gain in glioblastoma patients treated with dendritic cell immunotherapy is associated with increased NK but not CD8 + T cell activation in the presence of adjuvant temozolomide.
- Author
-
Pellegatta S, Eoli M, Cuccarini V, Anghileri E, Pollo B, Pessina S, Frigerio S, Servida M, Cuppini L, Antozzi C, Cuzzubbo S, Corbetta C, Paterra R, Acerbi F, Ferroli P, DiMeco F, Fariselli L, Parati EA, Bruzzone MG, and Finocchiaro G
- Abstract
In a two-stage phase II study, 24 patients with first diagnosis of glioblastoma (GBM) were treated with dendritic cell (DC) immunotherapy associated to standard radiochemotherapy with temozolomide (TMZ) followed by adjuvant TMZ. Three intradermal injections of mature DC loaded with autologous GBM lysate were administered before adjuvant TMZ, while 4 injections were performed during adjuvant TMZ. According to a two-stage Simon design, to proceed to the second stage progression-free survival (PFS) 12 months after surgery was expected in at least 8 cases enrolled in the first stage. Evidence of immune response and interaction with chemotherapy were investigated. After a median follow up of 17.4 months, 9 patients reached PFS12. In these patients (responders, 37.5%), DC vaccination induced a significant, persistent activation of NK cells, whose increased response was significantly associated with prolonged survival. CD8
+ T cells underwent rapid expansion and priming but, after the first administration of adjuvant TMZ, failed to generate a memory status. Resistance to TMZ was associated with robust expression of the multidrug resistance protein ABCC3 in NK but not CD8+ T cells. The negative effect of TMZ on the formation of T cell-associated antitumor memory deserves consideration in future clinical trials including immunotherapy.- Published
- 2018
- Full Text
- View/download PDF
125. Hypermutations in gliomas: a potential immunotherapy target.
- Author
-
Finocchiaro G, Langella T, Corbetta C, and Pellegatta S
- Subjects
- Animals, Dacarbazine therapeutic use, Glioma drug therapy, Humans, Mutation, Temozolomide, Antineoplastic Agents, Alkylating therapeutic use, Dacarbazine analogs & derivatives, Glioma genetics, Immunotherapy methods
- Abstract
Checkpoint inhibitors, like ipilimumab, nivolumab, and pembrolizumab, have provided a breakthrough in cancer immunotherapy, such as in the treatment of melanoma and colorectal and lung cancer. The close relationship between the number of mutations (mutational load) and the response to checkpoint immunotherapy has been convincingly demonstrated in these cancers. Hypermutations in tumors are caused by environmental factors, like UV radiations or cigarette smoking, or by germinal mutations affecting genes of the Mismatch Repair (MMR) machinery, as in the Lynch syndrome. In the context of a high mutational load, a number of neoantigens become visible to the immune system, creating the basis for effective T cell responses. In low- and high-grade gliomas, the most frequent brain tumors, germinal MMR defects are rare; however, hypermutations associated with mutations or decreased expression of MMR genes are rather frequent, occurring in 20-60% of the tumors at recurrence after alkylating chemotherapy with temozolomide. Ongoing clinical trials and genomic investigations will clarify if temozolomide-induced hypermutations, which usually occur in the presence of methylation of the methylguanine methyltransferase gene (MGMT), will be effectively targeted by immunotherapy with checkpoint inhibitors or dendritic cell immunotherapy, thus improving the survival expectations for patients affected by these tumors.
- Published
- 2017
126. Novel mechanisms and approaches in immunotherapy for brain tumors.
- Author
-
Finocchiaro G and Pellegatta S
- Subjects
- Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Antigens, Neoplasm therapeutic use, Brain Neoplasms genetics, Brain Neoplasms pathology, Dendritic Cells immunology, Dendritic Cells pathology, ErbB Receptors genetics, ErbB Receptors immunology, Glioblastoma genetics, Glioblastoma pathology, Humans, T-Lymphocytes immunology, T-Lymphocytes pathology, Brain Neoplasms immunology, Brain Neoplasms therapy, Glioblastoma immunology, Glioblastoma therapy
- Abstract
Converging data indicate that the immune system is able to recognize cancer epitopes as non-self and mount an immune reaction that may erase, or temporarily block, tumor growth. The immune pressure supports the amplification of immune resistant tumor clones, creating an immune suppressive environment that leads to the formation of a clinically relevant tumor. These general observations also apply to brain tumors and specifically to gliomas. Cancer immunotherapy strategies are aimed at reverting such immune suppression. Two approaches are already used in the clinics. The first one, peptide immunotherapy, has been oriented to the most aggressive glioma, glioblastoma (GBM) where, in the context of EGFR (epidermal growth factor receptor) amplification, a large deletion arises and creates a novel, cancer-specific antigen, EGFRvIII. The second one is dendritic cell immunotherapy. Dendritic cells are potent antigen presenting cells that can be pulsed with autologous tumor lysate or peptide pp65 from cytomegalovirus (CMV) that is present in GBM but not in normal brain. Antigen presentation by dendritic cells is bolstered by preconditioning their injection site with the tetanus/diphtheria toxoid. The third approach is adoptive cell therapy (ACT) in which tumor-specific T cells can be amplified ex vivo and subsequently re-injected to the patient to lyse cells expressing tumor antigens, increasing survival durably in a fraction of melanoma patients. ACT may also be based on T cell transduction of tumor specific receptors or chimeric antigen receptors (CARs). CARs are powerful tools for immunotherapy but off-target toxicity may be an issue as they do not request MHC presentation for activation. Upcoming clinical trial results will clarify the most effective direction for cancer immunotherapy in gliomas and other cancers with poor prognosis.
- Published
- 2015
127. Dendritic cell vaccines for cancer stem cells.
- Author
-
Pellegatta S and Finocchiaro G
- Subjects
- Animals, Biological Assay, Cell Adhesion, Cell Line, Cell Proliferation, Coculture Techniques, Cytotoxicity, Immunologic, Flow Cytometry, Humans, Mice, Spleen cytology, Spleen immunology, Vaccination, Cancer Vaccines immunology, Cell Culture Techniques methods, Dendritic Cells immunology, Neoplastic Stem Cells immunology, Neoplastic Stem Cells pathology
- Abstract
Accumulating evidence suggests that only a fraction of neoplastic cells, defined as cancer stem cells (CSC), are responsible for tumor perpetuation. Recent data suggest that neurospheres (NS) from glioblastoma multiforme (GBM) are enriched in CSC. The characterization of this subpopulation of brain tumor cells with a potent tumorigenic activity supports the cancer stem cell hypothesis in solid tumors and may imply that cancer cells are differentially targeted by treatments, including dendritic cell (DC) immunotherapy. To test therapeutic strategies, a good model mimicking the characteristics of GBM-NS and GBM-AC (Adherent Cells) was necessary. One of the most frequently used murine brain tumor models is the GL261 glioma cell line. To see whether GL261 cells could mimic the growth of human GBM-CSC we let them grow in EGF/bFGF without serum. After 5 days neurospheres were visible in the culture medium and were proliferating continuously. The characterization in vivo and in vitro demonstrates that GL261-NS satisfy criteria used to identify CSC and are more immunogenic than AC. DC loaded with GL261-NS lysates protect mice against tumors from both GL261-NS and GL261-AC. Our results suggest that only DC vaccination against neurospheres can restrain the growth of a highly infiltrating and aggressive model of glioma and may have implications for the design of novel, more effective immunotherapy trials for malignant glioma and possibly other malignancies.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.