Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Quintino Giorgio D’Alessandris is active.

Publication


Featured researches published by Quintino Giorgio D’Alessandris.


American Journal of Clinical Pathology | 2012

Prognostic Relevance of c-Myc and BMI1 Expression in Patients With Glioblastoma

Tonia Cenci; Maurizio Martini; Nicola Montano; Quintino Giorgio D’Alessandris; Maria Laura Falchetti; Daniela Annibali; Mauro Savino; Federico Bianchi; Francesco Pierconti; Sergio Nasi; Roberto Pallini; Luigi Maria Larocca

Although the c-Myc oncogene is frequently deregulated in human cancer, its involvement in the pathogenesis of glioblastoma is not clear. We conducted immunohistochemical analysis of the expression of c-Myc, polycomb ring finger oncogene (BMI1), and acetylation of the lysine 9 (H3K9Ac) of histone 3 in 48 patients with glioblastoma who underwent surgery followed by radiotherapy and temozolomide treatment. The expression of c-Myc, BMI1, and H3K9ac was correlated with clinical characteristics and outcome. We found that overexpression of c-Myc was significantly associated with that of BMI1 (P = .009), and that patients who harbored glioblastomas overexpressing c-Myc and BMI1 showed significantly longer overall survival (P < .0001 and P = .0009, respectively). Our results provide the first evidence of the prognostic value of c-Myc and associated genes in patients with glioblastoma. The favorable effect of c-Myc and BMI1 expression on survival is likely mediated by the sensitization of cancer cells to radiotherapy and temozolomide through the activation of apoptotic pathways.


Scientific Reports | 2016

Metabolic / Proteomic Signature Defines Two Glioblastoma Subtypes With Different Clinical Outcome

Giovanna Marziali; Michele Signore; Mariachiara Buccarelli; Sveva Grande; Alessandra Palma; Mauro Biffoni; Antonella Rosi; Quintino Giorgio D’Alessandris; Maurizio Martini; Luigi Maria Larocca; R. De Maria; Roberto Pallini; Lucia Ricci-Vitiani

Glioblastoma (GBM) is one of the deadliest human cancers. Because of the extremely unfavorable prognosis of GBM, it is important to develop more effective diagnostic and therapeutic strategies based on biologically and clinically relevant subclassification systems. Analyzing a collection of seventeen patient-derived glioblastoma stem-like cells (GSCs) by gene expression profiling, NMR spectroscopy and signal transduction pathway activation, we identified two GSC clusters, one characterized by a pro-neural-like phenotype and the other showing a mesenchymal-like phenotype. Evaluating the levels of proteins differentially expressed by the two GSC clusters in the TCGA GBM sample collection, we found that SRC activation is associated with a GBM subgroup showing better prognosis whereas activation of RPS6, an effector of mTOR pathway, identifies a subgroup with a worse prognosis. The two clusters are also differentiated by NMR spectroscopy profiles suggesting a potential prognostic stratification based on metabolic evaluation. Our data show that the metabolic/proteomic profile of GSCs is informative of the genomic/proteomic GBM landscape, which differs among tumor subtypes and is associated with clinical outcome.


World Neurosurgery | 2015

Olfactory groove meningioma: report of 99 cases surgically treated at the Catholic University School of Medicine, Rome.

Roberto Pallini; Eduardo Fernandez; Liverana Lauretti; Francesco Doglietto; Quintino Giorgio D’Alessandris; Nicola Montano; Gabriele Capo; Mario Meglio; Giulio Maira

OBJECTIVE We reviewed our series of olfactory groove meningiomas (OGMs) with the aim to relate the surgical approach with outcome and to define clinical and pathologic predictors of prognosis. METHODS Ninety-nine patients who underwent 113 craniotomies at our Institution between 1984 and 2010 were entered this study. The relationship between surgical approach (bifrontal, fronto-orbito-basal, and pterional) and either tumor diameter, extent of tumor resection, complication rate, need of reoperation, and Karnofsky Performance Status (KPS) was analyzed. The impact of age (≤ 70 vs. > 70 years), sex, tumor diameter (< 6 vs. ≥ 6 cm), pre- and postoperative KPS (< 80 vs. ≥ 80), Simpson grade (I-II vs. III-IV), and World Health Organization (WHO) histologic grade (I vs. II-III) on survival was assessed. Kaplan-Meier survival curves were plotted and differences in survival between groups of patients were compared. A multivariate analysis adjusted for age, pre- and postoperative KPS, Simpson grade, tumor diameter, and WHO histologic grade also was performed. RESULTS The fronto-orbito-basal approach (n = 22) allowed a significantly greater percentage of Simpson I-II removals than the bifrontal (n = 70) and pterional approach (n = 21) (P = 0.0354 and P = 0.0485, respectively). The risk of life-threatening complications trended to be lower in patients operated upon either via the fronto-orbito-basal and via the pterional approach than in those treated via the bifrontal approach. Retraction-related brain swelling did not occur in any case after the fronto-orbito-basal approach (P = 0.0384); however, this approach was associated with a greater rate of cerebrospinal fluid leak (P = 0.0011). Among prognostic factors, age ≤ 70 years (P = 0.0044), tumor diameter <6 cm (P = 0.0455), pre- and postoperative KPS ≥ 80 (both P < 0.0001), Simpson grade I-II (P = 0.0096), and WHO histologic grade I (P = 0.0112) were significantly associated with longer overall survival. Age (P = 0.0393) and WHO histologic grade (P = 0.0418) emerged as independent prognostic factors for overall survival on multivariate analysis. CONCLUSION In the largest series of OGMs published to date, the bifrontal approach was associated with a greater risk of life-threatening complications compared with the lateral pterional and fronto-orbito-basal approaches. The fronto-orbito-basal approach provided greater chances of total tumor removal than the bifrontal and pterional approaches. Two independent factors for overall survival of patients with OGM were identified, namely age and WHO grade.


Stem Cell Research & Therapy | 2017

Human mesenchymal stromal cells inhibit tumor growth in orthotopic glioblastoma xenografts

Simone Pacioni; Quintino Giorgio D’Alessandris; Stefano Giannetti; Liliana Morgante; Valentina Coccè; Arianna Bonomi; Mariachiara Buccarelli; Luisa Pascucci; Giulio Alessandri; Augusto Pessina; Lucia Ricci-Vitiani; Maria Laura Falchetti; Roberto Pallini

BackgroundMesenchymal stem/stromal cells (MSCs) represent an attractive tool for cell-based cancer therapy mainly because of their ability to migrate to tumors and to release bioactive molecules. However, the impact of MSCs on tumor growth has not been fully established. We previously demonstrated that murine MSCs show a strong tropism towards glioblastoma (GBM) brain xenografts and that these cells are able to uptake and release the chemotherapeutic drug paclitaxel (PTX), maintaining their tropism towards the tumor. Here, we address the therapy-relevant issue of using MSCs from human donors (hMSCs) for local or systemic administration in orthotopic GBM models, including xenografts of patient-derived glioma stem cells (GSCs).MethodsU87MG or GSC1 cells expressing the green fluorescent protein (GFP) were grafted onto the striatum of immunosuppressed rats. Adipose hMSCs (Ad-hMSCs), fluorescently labeled with the mCherry protein, were inoculated adjacent to or into the tumor. In rats bearing U87MG xenografts, systemic injections of Ad-hMSCs or bone marrow (BM)-hMSCs were done via the femoral vein or carotid artery. In each experiment, either PTX-loaded or unloaded hMSCs were used. To characterize the effects of hMSCs on tumor growth, we analyzed survival, tumor volume, tumor cell proliferation, and microvascular density.ResultsOverall, the AD-hMSCs showed remarkable tropism towards the tumor. Intracerebral injection of Ad-hMSCs significantly improved the survival of rats with U87MG xenografts. This effect was associated with a reduction in tumor growth, tumor cell proliferation, and microvascular density. In GSC1 xenografts, intratumoral injection of Ad-hMSCs depleted the tumor cell population and induced migration of resident microglial cells. Overall, PTX loading did not significantly enhance the antitumor potential of hMSCs. Systemically injected Ad- and BM-hMSCs homed to tumor xenografts. The efficiency of hMSC homing ranged between 0.02 and 0.5% of the injected cells, depending both on the route of cell injection and on the source from which the hMSCs were derived. Importantly, systemically injected PTX-loaded hMSCs that homed to the xenograft induced cytotoxic damage to the surrounding tumor cells.ConclusionshMSCs have a therapeutic potential in GBM brain xenografts which is also expressed against the GSC population. In this context, PTX loading of hMSCs seems to play a minor role.


PLOS ONE | 2015

A BMP7 Variant Inhibits Tumor Angiogenesis In Vitro and In Vivo through Direct Modulation of Endothelial Cell Biology

Courtney M. Tate; Jacquelyn Mc Entire; Roberto Pallini; Eliza Vakana; Lisa Wyss; Wayne Blosser; Lucia Ricci-Vitiani; Quintino Giorgio D’Alessandris; Liliana Morgante; Stefano Giannetti; Luigi Maria Larocca; Matilde Todaro; Antonina Benfante; Maria Luisa Colorito; Giorgio Stassi; Ruggero De Maria; Scott W. Rowlinson; Louis Stancato

Bone morphogenetic proteins (BMPs), members of the TGF-β superfamily, have numerous biological activities including control of growth, differentiation, and vascular development. Using an in vitro co-culture endothelial cord formation assay, we investigated the role of a BMP7 variant (BMP7v) in VEGF, bFGF, and tumor-driven angiogenesis. BMP7v treatment led to disruption of neo-endothelial cord formation and regression of existing VEGF and bFGF cords in vitro. Using a series of tumor cell models capable of driving angiogenesis in vitro, BMP7v treatment completely blocked cord formation. Pre-treatment of endothelial cells with BMP7v significantly reduced their cord forming ability, indicating a direct effect on endothelial cell function. BMP7v activated the canonical SMAD signaling pathway in endothelial cells but targeted gene knockdown using shRNA directed against SMAD4 suggests this pathway is not required to mediate the anti-angiogenic effect. In contrast to SMAD activation, BMP7v selectively decreased ERK and AKT activation, significantly decreased endothelial cell migration and down-regulated expression of critical RTKs involved in VEGF and FGF angiogenic signaling, VEGFR2 and FGFR1 respectively. Importantly, in an in vivo angiogenic plug assay that serves as a measurement of angiogenesis, BMP7v significantly decreased hemoglobin content indicating inhibition of neoangiogenesis. In addition, BMP7v significantly decreased angiogenesis in glioblastoma stem-like cell (GSLC) Matrigel plugs and significantly impaired in vivo growth of a GSLC xenograft with a concomitant reduction in microvessel density. These data support BMP7v as a potent anti-angiogenic molecule that is effective in the context of tumor angiogenesis.


Acta Neurochirurgica | 2015

Ultrasound evaluation in traumatic peripheral nerve lesions: from diagnosis to surgical planning and follow-up

Liverana Lauretti; Quintino Giorgio D’Alessandris; Giuseppe Granata; Luca Padua; Romeo Roselli; Rina Di Bonaventura; Eduardo Fernandez

Surgical treatment of traumatic nerve lesions is a matter of debate, mostly about the timing and technical aspects of the procedure. In deciding about and planning the operation, it is often necessary to repeat the electrophysiological and neuroradiological studies several times. Here we present our experience with ultrasonography taken before and after surgery: this simple and handy tool allowed clear visualization of the preoperative anatomy, thorough preparation and fast carrying out of surgery, and accurate postoperative monitoring of the graft’s vitality at follow-up. Though this is a limited series, the importance of ultrasonographic evaluation in traumatic peripheral nerve lesions appears remarkable.


Operative Neurosurgery | 2009

Calcified extra-axial cavernoma involving lower cranial nerves: technical case report.

Alessio Albanese; Carmelo Lucio Sturiale; Quintino Giorgio D’Alessandris; Gennaro Capone; Giulio Maira

OBJECTIVE Extra-axial cavernomas involving cranial nerves (CNs) are uncommon vascular malformations and may cause neurological deficits. We report what is, to our knowledge, a unique case of a calcified extra-axial cerebellopontine angle (CPA) cavernoma involving the lower CNs. CLINICAL PRESENTATION A 48-year-old man was admitted to our department with a 5-month history of gait instability and loss in tone of voice. A clinical examination documented gait disturbances and hoarseness but was otherwise unremarkable. Neuroradiological studies revealed a calcified mass in the lower third of the CPA cistern that was angiographically occult. It was associated with 3 additional lesions with a radiological appearance suggestive of multiple cavernomas. INTERVENTION The patient underwent a retrosigmoid approach, and the calcified mass, tightly adherent to the lower CNs, was gently removed. The histopathological examination was consistent with a cavernoma. The postoperative course was characterized by a further lowering in the patients tone of voice. At the 3-month follow-up examination, the patient showed significant improvement. CONCLUSION CPA cavernomas are an extremely rare entity. Symptoms are generally related to CN compression, and subarachnoid hemorrhage is a very rare occurrence. The clinical and radiological appearance may mimic that of other CPA tumors (meningiomas, schwannomas). In spite of the benign nature and the very low risk of hemorrhage, we believe, with support from the literature, that surgical treatment is mandatory to prevent significant neurological deficits owing to the chronic CN compression.


Oncotarget | 2017

Type 5 phosphodiesterase regulates glioblastoma multiforme aggressiveness and clinical outcome

Valeriana Cesarini; Maurizio Martini; Lucia Ricci Vitiani; Giovanni Luca Gravina; Silvia Di Agostino; Grazia Graziani; Quintino Giorgio D’Alessandris; Roberto Pallini; Luigi Maria Larocca; Pellegrino Rossi; Emmanuele A. Jannini; Susanna Dolci

Expression of type 5 phosphodiesterase (PDE5), a cGMP-specific hydrolytic enzyme, is frequently altered in human cancer, but its specific role in tumorigenesis remains controversial. Herein, by analyzing a cohort of 69 patients affected by glioblastoma multiforme (GBM) who underwent chemo- and radiotherapy after surgical resection of the tumor, we found that PDE5 was strongly expressed in cancer cells in about 50% of the patients. Retrospective analysis indicated that high PDE5 expression in GBM cells significantly correlated with longer overall survival of patients. Furthermore, silencing of endogenous PDE5 by short hairpin lentiviral transduction (sh-PDE5) in the T98G GBM cell line induced activation of an invasive phenotype. Similarly, pharmacological inhibition of PDE5 activity strongly enhanced cell motility and invasiveness in T98G cells. This invasive phenotype was accompanied by increased secretion of metallo-proteinase 2 (MMP-2) and activation of protein kinase G (PKG). Moreover, PDE5 silencing markedly enhanced DNA damage repair and cell survival following irradiation. The enhanced radio-resistance of sh-PDE5 GBM cells was mediated by an increase of poly(ADP-ribosyl)ation (PARylation) of cellular proteins and could be counteracted by poly(ADP-ribose) polymerase (PARP) inhibitors. Conversely, PDE5 overexpression in PDE5-negative U87G cells significantly reduced MMP-2 secretion, inhibited their invasive potential and interfered with DNA damage repair and cell survival following irradiation. These studies identify PDE5 as a favorable prognostic marker for GBM, which negatively affects cell invasiveness and survival to ionizing radiation. Moreover, our work highlights the therapeutic potential of targeting PKG and/or PARP activity in this currently incurable subset of brain cancers.


Cell Death and Disease | 2018

Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis

Mariachiara Buccarelli; Matteo Marconi; Simone Pacioni; Ivana De Pascalis; Quintino Giorgio D’Alessandris; Maurizio Martini; Barbara Ascione; Walter Malorni; Luigi Maria Larocca; Roberto Pallini; Lucia Ricci-Vitiani; Paola Matarrese

The role of autophagy in cancer onset and progression appears still controversial. On one hand, autophagy allows cancer cell to survive in unfavorable environmental conditions, on the other hand, once internal energy resources are exhausted, it leads to cell death. In addition, autophagy interpheres with cell cycle progression, de facto exerting a cytostatic activity. Hence, it represents an important target for anticancer therapy. For example, temozolomide (TMZ), of use for glioblastoma (GBM) treatment, appears as capable of inducing autophagy partially inhibiting cancer cell proliferation. However, GBM, a very aggressive brain tumor with poor prognosis even after surgery and radio-chemotherapy, invariably recurs and leads to patient death. Since cancer stem cells have been hypothesized to play a role in refractory/relapsing cancers, in the present work we investigated if autophagy could represent a constitutive cytoprotection mechanism for glioblastoma stem-like cells (GSCs) and if the modulation of autophagic process could affect GBM growth and survival. Thus, in the present study we first evaluated the relevance of autophagy in GBM tumor specimens, then its occurrence in GSCs and, finally, if modulation of autophagy could influence GSC response to TMZ. Our results suggested that, in vitro, the impairing autophagic process with quinacrine, a compound able to cross the blood-brain barrier, increased GSC susceptibility to TMZ. Death of GSCs was apparently due to the iron dependent form of programmed cell death characterized by the accumulation of lipid peroxides called ferroptosis. These results underscore the relevance of the modulation of autophagy in the GSC survival and death and suggest that triggering of ferroptosis in GSCs could represent a novel and important target for the management of glioblastoma.


Translational cancer research | 2016

Emerging role for USP1 in glioblastoma stem cell maintenance and radioresistance: a potential target for glioblastoma therapy

Quintino Giorgio D’Alessandris; Lucia Ricci-Vitiani

Glioblastoma (GBM) is the most common primary brain malignancy with limited treatment options. The standard of care for GBM, including maximal safe resection, radiotherapy, and concomitant and adjuvant temozolomide is currently recommended for all patients (1) but only a small proportion of patients survive 2 years or longer and, eventually, virtually every tumor recurs (2). GBM is characterized by a high degree of intra-tumoral cellular heterogeneity. Recently, genome-wide expression profiling in the context of The Cancer Genome Atlas (TCGA) project has led to the identification of initially three, and then four main subtypes of GBM (3,4), namely proneural (P), mesenchymal (M), neural (N) and proliferative (or classical, C), thus revealing the existence of a profound degree of inter-tumoral cellular heterogeneity.

Collaboration


Dive into the Quintino Giorgio D’Alessandris's collaboration.

Top Co-Authors

Avatar

Roberto Pallini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Luigi Maria Larocca

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Maurizio Martini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Lucia Ricci-Vitiani

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Liverana Lauretti

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Nicola Montano

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Eduardo Fernandez

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Ivana De Pascalis

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Liliana Morgante

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Simone Pacioni

Catholic University of the Sacred Heart

View shared research outputs
Researchain Logo
Decentralizing Knowledge