Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Clin Neurol Neurosurg ; 210: 106953, 2021 11.
Article in English | MEDLINE | ID: mdl-34607197

ABSTRACT

OBJECTIVE: In patients with brain lesion, awareness of cognitive deficits is an important aspect of disease awareness. Glioblastoma (GBM) and anaplastic astrocytoma (AA) can cause cognitive deficits, but, to date, awareness of these deficits has not been documented. This study aimed to test cognitive awareness in these patients after the end of treatment. METHODS: Fifty patients with GBM or AA were assessed using the Multiple Ability Self-Report Questionnaire (MASQ), State-Trait Anxiety Inventory (STAI), Self Rating Depression Scale (SRDS), and memory, attention, mental speed, abstract reasoning, and flexibility neuropsychological tests. Cognitive awareness was calculated as the concordance between the composite score of neuropsychological performance (PEC) and the total MASQ score. The controls were 48 healthy subjects. Analysis of variance and regression analysis compared subject groups and explored variables predicting perceived abilities. RESULTS: Patients with GBM or AA showed similar attention, memory, and executive deficits compared with controls. Cognitive awareness was fair/full in 64% of patients. In the entire patients group, the worst MASQ scores were associated with neuropsychological deficits, anxiety, depression, and glioma location in the right hemisphere . In patients with fair/full awareness, MASQ scores were related to affective status and neuropsychological performance, whereas, in those with scarce/no awareness, they were related only to affective status. CONCLUSIONS: After treatment, many patients with GBM or AA are aware of their cognitive deficits. Anxiety, depression, and right hemisphere tumour exacerbate the perceived difficulties. This neurocognitive approach expands the behavioural phenotypes of high-grade gliomas and may have therapeutic implications over the course of the disease.


Subject(s)
Awareness/physiology , Brain Neoplasms/psychology , Cognition/physiology , Glioma/psychology , Adult , Anxiety/psychology , Attention/physiology , Depression/psychology , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Quality of Life/psychology , Young Adult
2.
J Neurol ; 261(5): 894-904, 2014 May.
Article in English | MEDLINE | ID: mdl-24590402

ABSTRACT

This study analyzed the subjective facets of quality of life (QoL) and their relation to the type of brain tumor (BT) and phase of disease. Two hundred and ninety-one patients with pinealoblastoma, medulloblastoma, low-grade glioma, anaplastic astrocytoma, or glioblastoma were evaluated. With respect to 110 healthy controls, patients in the phases of radiotherapy/chemotherapy, stable disease, or tumor recurrence were significantly more anxious and depressed compared with patients in the early postoperative period. All patients were impaired in mental flexibility and memory, with preservation of abstract reasoning. The Functional Living Index-Cancer (FLIC), previously validated in cancer and BT patients, yielded six subjective factors (disease perception, affective well-being, role and leisure, personal base, nausea, sharing). None of the FLIC factors were predicted by tumor type, which only related to the physical and cognitive performances and mood scores. Affective well-being, role and leisure, and sharing were predicted by the phase of disease. Personal base, including self-perception and confidence, was independent on tumor progression and treatment. To conclude, QoL encompasses different subjective aspects, which vary in relation to the phase of disease and clinical burden. However, some person-related facets appear independent on tumor progression and treatment, indicating individual resources. Knowing this may guide tailored interventions supporting QoL.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/psychology , Cost of Illness , Disease Progression , Quality of Life/psychology , Adult , Anxiety/diagnosis , Anxiety/psychology , Cross-Sectional Studies , Depression/diagnosis , Depression/psychology , Female , Humans , Male , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/psychology
3.
J Neurooncol ; 94(1): 57-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19212704

ABSTRACT

BACKGROUND: The prognosis of patients with glioblastoma is very poor with a mean survival of 10-12 months. Currently available treatment options are multimodal, which include surgery, radiotherapy, and chemotherapy. However, these have been shown to improve survival only marginally in glioblastoma multiforme (GBM) patients. Methylated methylguanine methyltransferase (MGMT) promoter is correlated with improved progression-free and overall survival in patients treated with alkylating agents. Strategies to overcome MGMT-mediated chemoresistance are being actively investigated. METHODS: A retrospective analysis on 160 adult patients (> or =16 years) treated for histologically confirmed GBM between 2003 and 2005 at our Institution was performed. All patients were treated with conventional fractionated radiotherapy and a combined chemotherapy treatment with Cisplatin (CDDP) (100 mg/sqm on day 1) and carmustine (BCNU) (160 mg/sqm on day 2); the treatment was repeated every 6 weeks for five cycles. Toxicity, progression free survival and overall survival were assessed. RESULTS: The median number of chemotherapy cycles delivered to each patient was 5 (range 3-6), with no patients discontinuing treatment because of refusal or toxicity. Dose reduction was required in 16 patients (10%), and all patients completed radiotherapy, whereas 5 patients discontinued chemotherapy before completing all planned cycles for disease progression. The primary toxicities were: neutropenia (grade 3-4: 23%), thrombocytopenia (grade 3-4: 18.5%), and nausea and vomiting (13%). Median progression-free survival times and 1-year progression free survival were 7.6 months (95% CI 6.6-8.5) and 17.3%, respectively. OS was 15.6 months (95% CI 14.1-17.1). CONCLUSIONS: Our results for PFS and overall survival are comparable with those obtained with temozolomide, but the toxicity occurring in our series was more frequent and persistent. The toxicity, and mainly the modalities of administration associated with cisplatin and BCNU combination, argues against future use in the treatment of patients with GBM.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Carmustine/therapeutic use , Cisplatin/therapeutic use , Glioblastoma/drug therapy , Adult , Aged , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Combined Modality Therapy , Dose-Response Relationship, Drug , Female , Glioblastoma/mortality , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Male , Middle Aged , Radiotherapy, Adjuvant , Retrospective Studies , Survival Analysis , Treatment Outcome , Young Adult
4.
J Neurooncol ; 88(1): 105-13, 2008 May.
Article in English | MEDLINE | ID: mdl-18283418

ABSTRACT

In this study, the records of 276 adult patients with recurrent glioblastoma (GBM) treated at recurrence at our institution between 2004 and 2006 were reviewed for progression-free survival (PFS), overall survival (OS), and toxicity. At recurrence, all patients underwent systemic treatment with temozolomide (200 mg/sqm on days 1-5 every 28 days) until tumor progression. Patients, whose tumor was judged resectable without risk of adjunctive neurological deficit, underwent a second surgery with or without positioning of a Rickam/Ommaya reservoir. The reservoir was used for locoregional chemotherapy with mitoxantrone. Two hundred seventy-six rGBL patients (pts) were divided into three subgroups: A 161 pts treated only with temozolomide, B 50 pts re-operated-on +temozolomide, and C 65 pts re-operated on + temozolomide + locoregional CHT. For group A, the 6 month PFS and 6 month survival (ST) were 39.3 and 43%, respectively, with a median survival time (mST) of 5 months (range 4-6) and 25% of pts alive at 9 months. For group B, the 6 month PFS and 6 month survivors were 64 and 74.1%, respectively, with a mST of 8 months (range 6-10) and 25% of pts alive at 12 months. For group C, the 6 month PFS and 6 month survivors were 70.7 and 87.7%, respectively, with a mST of 11 months (range 9-13) and 25% of pts alive at 18 months (A vs. B vs. C, log-rank P < 0.001) (B vs. C, P = 0.041) (A vs. B P = 0.009). Cox proportional hazard model was used to obtain Hazard Ratio (HR) for type of treatment corrected by age and time (in months) between diagnosis and first recurrence: second tumor debulking was statistically effective for survival, reducing by 36% the risk of death (HR = 0.64; 0.46-0.89), but the most significant favorable prognostic factor for survival was the local delivery of mitoxantrone which reduced the risk of death to 50% (HR = 0.50; 0.38-0.68).


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Mitoxantrone/administration & dosage , Mitoxantrone/therapeutic use , Adult , Aged , Aging/physiology , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/pathology , Combined Modality Therapy , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Disease Progression , Female , Glioblastoma/pathology , Humans , Magnetic Resonance Spectroscopy , Male , Middle Aged , Mitoxantrone/adverse effects , Neoplasm Recurrence, Local , Neurosurgical Procedures , Positron-Emission Tomography , Retrospective Studies , Survival , Temozolomide , Treatment Outcome
5.
J Neurooncol ; 87(2): 143-51, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17576523

ABSTRACT

The purpose of this study was to evaluate safety and efficacy of Procarbazine (PCB) and fotemustine (FTM) combination in the treatment of pre-temozolomide treated, recurrent GBM patients. The primary end-point was progression free survival at 6 months (PFS-6). Secondary end-points were overall survival, response rates (CR + PR) and toxicity. About 54 patients (41 men and 13 women) aged 26-68 years (median age, 53.5 years) with recurrent GBM were treated. PCB was administered as an oral dosage of 450 mg on days 1-2 and a total dose of 300 mg on day 3. FTM was administered on day 3, 3 h after the last PCB intake at a dose of 110 mg/mq/BSA. The treatment was repeated every 5 weeks. Treatment was continued for a maximum of six cycles or until disease progression. After two cycles of chemotherapy: 6 patients (11.2%) experienced a neuroradiographic partial response (PR), 29 patients (53.7%) had stable disease (SD), and 19 patients (35.1%) had progressive disease (PD). For the whole group of patients, the median PFS was 19.3 weeks (95% CI, 14.1-24.4 weeks), and PFS-6 was 26.7% (95% CI, 10.6-42.8%). Overall MST from the beginning of PCB + FTM chemotherapy was 28.7 weeks (95% CI, 24.8-32.7 weeks). At 6 and 12 months, 64.4% (95% CI, 51.5-77.3%) and 23.6% (95% CI, 10.1-37.1%) of patients were alive. The median survival time calculated from the first diagnosis was 20.8 months (95% CI, 16.7-24.8). We concluded that the PCB + FTM combination as done in the current trial for patients with recurrent GBM after treatment with TMZ showed some benefit with regards to increased survival and that a Phase III trial is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy/methods , Adult , Aged , Brain Neoplasms/mortality , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Disease-Free Survival , Female , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/adverse effects , Procarbazine/administration & dosage , Procarbazine/adverse effects , Temozolomide
6.
Neuro Oncol ; 10(1): 79-87, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17993634

ABSTRACT

Reliable data on large cohorts of patients with glioblastoma are needed because such studies differ importantly from trials that have a strong bias toward the recruitment of younger patients with a higher performance status. We analyzed the outcome of 676 patients with histologically confirmed newly diagnosed glioblastoma who were treated consecutively at a single institution over a 7-year period (1997-2003) with follow-up to April 30, 2006. Survival probabilities were 57% at 1 year, 16% at 2 years, and 7% at 3 years. Progression-free survival was 15% at 1 year. Prolongation of survival was significantly associated with surgery in patients with a good performance status, whatever the patient's age, with an adjusted hazard ratio of 0.55 (p < 0.001) or a 45% relative decrease in the risk of death. Radiotherapy and chemotherapy improved survival, with adjusted hazard ratios of 0.61 (p = 0.001) and 0.89 (p = 0.04), respectively, regardless of age, performance status, or residual tumor volume. Recurrence occurred in 99% of patients throughout the follow-up. Reoperation was performed in one-fourth of these patients but was not effective, whether performed within 9 months (hazard ratio, 0.86; p = 0.256) or after 9 months (hazard ratio, 0.98; p = 0.860) of initial surgery, whereas second-line chemotherapy with procarbazine, lomustine, and vincristine (PCV) or with temozolomide improved survival (hazard ratio, 0.77; p = 0.008). Surgery followed by radiotherapy and chemotherapy should be considered in all patients with glioblastoma, and these treatments should not be withheld because of increasing age alone. The benefit of second surgery at recurrence is uncertain, and new trials are needed to assess its effectiveness. Chemotherapy with PCV or temozolomide seems to be a reasonable option at tumor recurrence.


Subject(s)
Brain Neoplasms/mortality , Brain Neoplasms/therapy , Glioblastoma/mortality , Glioblastoma/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Brain Neoplasms/pathology , Combined Modality Therapy , Disease-Free Survival , Female , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/therapy , Neurosurgical Procedures , Prognosis , Radiotherapy , Treatment Outcome
9.
Clin Cancer Res ; 13(9): 2606-13, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473190

ABSTRACT

PURPOSE: Recent data suggest that methylation of the DNA repair gene O(6)-methylguanine DNA methyltransferase (MGMT), by increasing the chemosensitivity of glioblastoma multiforme, is significantly associated with improved prognosis. Results in contradiction with these findings, however, are present in the literature and the clinical and genetic context framing MGMT methylation is poorly characterized. EXPERIMENTAL DESIGN: To address these issues, we have investigated the MGMT methylation status, clinical and magnetic resonance imaging characteristics, and relevant genetic features (loss of heterozygosity on 17p and 19q, EGFR amplification, and p53 mutations) in a retrospective study on 86 patients affected by glioblastoma multiforme: 72 patients had a clinical history indicating de novo insurgence of the tumor and the remaining 14 were secondary glioblastoma multiforme. RESULTS: MGMT methylation was detected by methylation-specific PCR in 41 of 86 cases (47.7%; Meth+). Progression-free survival and overall survival were significantly longer in Meth+ than in Meth- patients [10 versus 7 months (P=0.003, log-rank test) and 18 versus 14 months (P=0.0003, log-rank test), respectively]. Mixed-nodular enhancement at magnetic resonance imaging was significantly more frequent in Meth+ and secondary glioblastoma multiforme and ring enhancement in Meth- and primary glioblastoma multiforme (P<0.005). MGMT methylation was more present in secondary glioblastoma multiforme (P=0.006) and associated with loss of heterozygosity on 17p and/or 19q (P=0.005). CONCLUSIONS: These observations suggest that MGMT methylation is part of a genetic signature of glioblastomas that developed from lower-grade gliomas.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Glioblastoma/genetics , Loss of Heterozygosity , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adult , Aged , Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 19/genetics , DNA, Neoplasm/metabolism , ErbB Receptors/genetics , Female , Gene Amplification , Glioblastoma/diagnosis , Glioblastoma/secondary , Glioma/genetics , Glioma/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Prognosis , Survival , Tumor Suppressor Protein p53/genetics
10.
Oncol Res ; 16(10): 453-63, 2007.
Article in English | MEDLINE | ID: mdl-18196869

ABSTRACT

Valproic acid (VPA) has been recently investigated for its anticancer properties in different tumors, including malignant gliomas. The aim of the present work was to evaluate the effects of VPA, alone or in combination with other chemotherapeutic drugs, on in vitro growth of human glioma cell lines. A172, U373, U138, U87, and SW1783 were treated with VPA alone or in combination with mitoxantrone, etoposide, or 1,3-bis(2-chloroethyl)-l-nitrosourea (BCNU). The effects of treatments on cell growth were assessed with crystal violet staining and analyzed using the combination index (CI). The percentage of apoptotic cells and the DNA content for cell cycle phases detection were also investigated by flow cytometry. Despite a certain variability, glioma cell lines were rather resistant to the drugs tested. Addition of VPA decreased the IC50 of the chemotherapeutic agents in all cell lines tested. This effect was more evident with BCNU. The synergic effect of the association of VPA and BCNU was related to an increased block of cell cycle with accumulation in S-G2/M phases of cell cycle rather than an increased programmed cell death. In our experimental model, VPA showed anticancer properties per se on human glioma cell lines and our data support the hypothesis that, if used in association with conventional chemotherapy, it might improve the effects of single chemotherapeutic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Astrocytoma/drug therapy , Carmustine/pharmacology , Glioblastoma/drug therapy , Valproic Acid/pharmacology , Acetylation/drug effects , Apoptosis/drug effects , Astrocytoma/metabolism , Astrocytoma/pathology , Carmustine/administration & dosage , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Synergism , Etoposide/administration & dosage , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mitoxantrone/administration & dosage , Valproic Acid/administration & dosage
11.
Glia ; 54(8): 850-60, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16981197

ABSTRACT

We investigated in vitro the properties of selected populations of cancer stem-like cells defined as tumorospheres that were obtained from human glioblastoma. We also assessed their potential and capability of differentiating into mature cells of the central nervous system. In vivo, their tumorigenicity was confirmed after transplantation into the brain of non-obese diabetic/severe combined immunodeficient (NOD-SCID) mice. The angiogenic potential of tumorospheres and glioblastoma-derived cells grown as adherent cells was revealed by evaluating the release of angiogenic factors such as vascular endothelial growth factor and CXCL12 by ELISA, as well as by rat aortic ring assay. The proliferative response of tumorospheres in the presence of CXCL12 was observed for the first time. Multidrug resistance-associated proteins 1 and 3 as well as other molecules conferring multidrug resistance were higher when compared with primary adherent cells derived from the same tumor. Finally, we obtained cells from the tumor developing after grafting that clearly expressed the putative neural stem cell marker CD133 as shown by FACS analysis and also nestin and CXCR4. The cells' positivity for glial fibrillary acidic protein was very low. Moreover these cells preserved their angiogenic potential. We conclude that human glioblastoma could contain tumor cell subsets with angiogenic and chemoresistance properties and that this chemoresistance potential is highly preserved by immature cells whereas the angiogenic potential is, to a higher extent, a property of mature cells. A better understanding of the features of these cell subsets may favor the development of more specifically targeted therapies.


Subject(s)
Brain Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Glioblastoma/metabolism , Neovascularization, Pathologic/physiopathology , Spheroids, Cellular/metabolism , Stem Cells/metabolism , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Aged , Antigens, CD/metabolism , Blood Vessels/growth & development , Blood Vessels/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Cell Differentiation/physiology , Cell Lineage/physiology , Chemokine CXCL12 , Chemokines, CXC/metabolism , Drug Resistance, Multiple/physiology , Female , Glioblastoma/blood supply , Glioblastoma/drug therapy , Glycoproteins/metabolism , Humans , Intermediate Filament Proteins/metabolism , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Nestin , Peptides/metabolism , Receptors, CXCR4/metabolism , Spheroids, Cellular/drug effects , Stem Cells/drug effects , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
12.
Neurosurgery ; 59(2): E433-4; discussion E433-4, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16883158

ABSTRACT

OBJECTIVE: To investigate the safety and feasibility of a novel form of treatment offered by the direct intraoperative application of a Surgifoam-mitoxantrone mix into a glioblastoma multiforme postresection cavity. A technique for the placement of an intracavity catheter connected with a subcutaneous reservoir for further locoregional mitoxantrone administration is also described. METHODS: Between January and December 2004, 22 consecutive recurrent glioblastoma multiforme patients (14 men, 8 women; age, 56-72 yr; average, 64 yr; median, 65 yr) were enrolled in this study. All patients underwent image-assisted gross total resection of the pathological tissue. A Surgifoam-mitoxantrone mix (1 g Surgifoam powder, 3 ml physiological solution, and 12 mg mitoxantrone in 6 ml) was used to fill the surgical cavity. A ventricular catheter, connected to a Rickham subcutaneous reservoir, was then positioned in the surgical cavity for future mitoxantrone administration. RESULTS: Toxic effects caused by mitoxantrone administration were not observed in any patients during the first postoperative month. On postoperative Days 1, 7, and 30, computed tomographic scans excluded surgical complications. In three patients, residual tumor was disclosed. CONCLUSION: A mix of Surgifoam and mitoxantrone could be safely applied intraoperatively into the post-glioblastoma multiforme resection cavity without any observable side effects. This technique may benefit both the surgeon and the patient by taking advantage of the drug's hemostatic and cytostatic properties.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/surgery , Fibrin Foam/administration & dosage , Glioblastoma/drug therapy , Glioblastoma/surgery , Mitoxantrone/administration & dosage , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain Edema/etiology , Brain Edema/physiopathology , Brain Edema/therapy , Brain Neoplasms/diagnosis , Catheters, Indwelling/standards , Catheters, Indwelling/trends , Epilepsy/etiology , Epilepsy/physiopathology , Epilepsy/therapy , Feasibility Studies , Female , Fibrin Foam/pharmacokinetics , Glioblastoma/diagnosis , Humans , Infusion Pumps, Implantable/standards , Infusion Pumps, Implantable/trends , Magnetic Resonance Imaging , Male , Middle Aged , Mitoxantrone/pharmacokinetics , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Neoplasm Recurrence, Local/surgery , Postoperative Complications/etiology , Postoperative Complications/physiopathology , Postoperative Complications/therapy , Radiotherapy , Steroids/adverse effects , Tomography, X-Ray Computed , Treatment Outcome
13.
Cancer Biol Ther ; 5(7): 827-32, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16760646

ABSTRACT

Both clinical and biological features have been reported as prognostic factors in low-grade gliomas. Among these, histotype, tumor size, enhancement, age and genetic pattern. Microvessel density (MVD) has been correlated to clinical outcome in astrocytomas, but its impact in oligodendrogliomas and mixed tumors is not sure. The pro-angiogenic chemokine stromal cell-derived factor (SDF-1/CXCL12) and its receptor CXC chemokine receptor 4 (CXCR4) have been described in low-grade gliomas, with a correlation between CXCL12 expression and shorter time to progression (TTP). The intermediate filament Nestin is expressed in proliferating vessels. Platelet-derived growth factor B (PDGF-B) and its receptor PDGFR-beta are also involved in angiogenesis and malignant progression in gliomas.


Subject(s)
Astrocytoma/blood supply , Astrocytoma/diagnosis , Brain Neoplasms/blood supply , Brain Neoplasms/diagnosis , Chemokines, CXC/analysis , Neovascularization, Pathologic/diagnosis , Oligodendroglioma/blood supply , Oligodendroglioma/diagnosis , Adolescent , Adult , Aged , Astrocytoma/pathology , Brain Neoplasms/pathology , Capillaries/pathology , Chemokine CXCL12 , Female , Humans , Intermediate Filament Proteins/analysis , Male , Middle Aged , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/analysis , Nestin , Oligodendroglioma/pathology , Prognosis , Proto-Oncogene Proteins c-sis/analysis , Receptor, Platelet-Derived Growth Factor beta/analysis , Receptors, CXCR4/analysis
14.
Int J Cancer ; 119(1): 84-90, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16432842

ABSTRACT

Oligoastrocytomas (OAs) are WHO grade II or III tumors composed of a mixture of 2 neoplastic cell types morphologically resembling the cells in oligodendrogliomas and diffuse astrocytomas. Investigations on the genetic profile of OAs may yield important information for their classification and help for their clinical management. We have studied, in 94 OAs (46 WHO grade II and 48 WHO grade III), the patterns of loss of heterozygosity (LOH) of 4 genomic regions: 1p, 19q, 17p and 10q. Results were as follows: LOH 1p was present in 46% of the tumors; LOH 19q in 45%; LOH 17p in 22%; LOH 10q in 16%. LOH 1p and 19q were associated in 32%, other LOH associations were rare (<3%). Patients had a median follow-up of 30 months. Patients without LOH on 1p had shorter progression free survival than patients with LOH on 1p: 30 vs. 132 months, p < 0.0001. MRI indicated that tumors without LOH on 1p were often temporal (p < 0.02), and showed signal inhomogeneity on T1 and T2 images (p < 0.02) and contrast enhancement (p < 0.04). Thus, LOH on 1p identifies two subgroups of OAs. OAs without LOH on 1p behave like WHO grade II or III diffuse astrocytomas: they have shorter survival, MRI characteristics implying malignancy and genetic alterations associated with tumor progression. OAs with LOH on 1p, on the other hand, behave like WHO grade II or III oligodendrogliomas with 1p loss: they are associated with longer survival and do not have MRI or genetic alterations associated with malignancy. These findings suggest that the definition of OAs or mixed gliomas could be reshaped in agreement with the genetic information.


Subject(s)
Astrocytoma/classification , Astrocytoma/genetics , Brain Neoplasms/classification , Brain Neoplasms/genetics , Loss of Heterozygosity , Adult , Analysis of Variance , Astrocytoma/pathology , Astrocytoma/surgery , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 19 , Disease Progression , Disease-Free Survival , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Survival Analysis
15.
Cancer Biol Ther ; 5(2): 204-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16357523

ABSTRACT

Malignant gliomas are associated with risk of thromboembolism, but the molecular link between tumor and peripheral pro-coagulant status has not been elucidated. Vascular Endothelial Growth Factor (VEGF), tissue-type Plasminogen Activator (tPA), Plasminogen Activator Inhibitor-1 (PAI-1) and lipoprotein (lp) (a) influence the pro-coagulant status. To assess whether the presence of the tumor influenced the peripheral levels of VEGF, tPA, PAI-1 and lp(a), we studied the expression and secretion of VEGF, tPA, PAI-1 and lp(a) in glioma specimens, in peripheral blood and in primary glioma-derived cultures. We also measured lp(a), VEGF, tPA and PAI-1 in the peripheral circulation of patients, before and after surgery for glioma. VEGF, tPA and PAI-1 were expressed in glioma specimens. Glioma cells were indeed a major source of tPA and PAI-1; these molecules were significantly more expressed in glioma than in patient's blood cells. Lp(a) was rarely expressed in glioma specimens and not expressed in blood cells. In glioma, VEGF, tPA and PAI-1 were localized mainly in tumor cells; tPA was localized also in the extracellular matrix and PAI-1 in tumor vascular lumen. Glioma cells were indeed able to produce and release VEGF, tPA and PAI-1. After surgery, peripheral levels of VEGF and PAI-1 were increased, while tPA and lp(a) were unchanged. The great amount of VEGF, tPA and PAI-1 produced by glioma could influence peripheral levels of these molecules. The partial resection of the tumor by surgery was not able to decrease plasma levels of these molecules.


Subject(s)
Brain Neoplasms/chemistry , Glioma/chemistry , Plasminogen Activator Inhibitor 1/analysis , Thrombosis/diagnosis , Tissue Plasminogen Activator/analysis , Vascular Endothelial Growth Factor A/analysis , Brain Neoplasms/complications , Brain Neoplasms/surgery , Female , Glioma/complications , Glioma/surgery , Humans , Male , Middle Aged , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/analysis , Thrombosis/etiology , Tissue Plasminogen Activator/blood , Tissue Plasminogen Activator/genetics , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics
16.
Oncol Res ; 16(5): 245-50, 2006.
Article in English | MEDLINE | ID: mdl-17294805

ABSTRACT

Heparins represent the first choice for prevention and treatment of venous thromboembolism. In particular, low molecular weight heparins (LMWHs) provide pharmacokinetic advantages compared to unfractionated heparin (UFH): longer half-life, better bioavailability, and lower binding to plasma proteins. In the last years results of preclinical and clinical studies have suggested that LMWH may be able to inhibit cell growth, cell invasion, and angiogenesis, which are key mechanisms involved in tumor progression, possibly influencing favorable clinical outcome in at least a proportion of cancer patients. In this work we investigated the effect of LMWH (enoxaparin) on cell growth and cell invasion in primary cell cultures obtained from high-grade glioma specimens: 5 anaplastic astrocytoma (AA) and 13 glioblastoma multiforme (GBM). Apoptosis and expression of the thrombin receptor PAR1 were also assessed. A significant decrease in tumor cell growth was observed after treatment with 10 U/ml (-21%; p = 0.001) and 100 U/ml (-26%; p < 0.001); tumor cells from AA (grade III; WHO) were more affected by LMWH treatment compared to cell lines from GBM (grade IV; WHO). The antiproliferative effect was more pronounced in cell cultures displaying higher expression of PAR1. Glioma cell cultures were able to invade a model of basement membrane (Matrigel matrix) in standard culture conditions, but migration was not modulated significantly by LMWH treatment at any of the concentrations tested (1, 10, 100 U/ml). In conclusion, our results confirm the antineoplastic effect of LMWH, suggesting a potential direct role on tumor cell growth in high grade gliomas.


Subject(s)
Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Enoxaparin/pharmacology , Glioma/drug therapy , Adolescent , Adult , Aged , Apoptosis/drug effects , Brain Neoplasms/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Humans , Male , Middle Aged , Molecular Weight , Neoplasm Invasiveness , Receptor, PAR-1/biosynthesis , Receptor, PAR-1/drug effects , Receptor, PAR-1/genetics , Sensitivity and Specificity , Structure-Activity Relationship , Tumor Cells, Cultured
17.
J Neurooncol ; 74(1): 87-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16078113

ABSTRACT

Mutations and instability of mitochondrial DNA (mtDNA) are frequent in tumors but their pathogenic relevance is not established. To assess their role in the clinical management of malignant gliomas we have studied the D loop of mtDNA in 42 such tumors. Alterations were found in 36% of the cases. The MRI and the clinical follow-up of these patients suggest that these mutations are not associated with increased aggressiveness. mtDNA could be amplified from post-surgical tumor cavities in patients undergoing a loco-regional treatment. These results imply that mtDNA mutations are unlikely to play a role in diagnostic or prognostic evaluations of gliomas: their detection, however, could be of use for the clinical follow-up of malignant gliomas.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , DNA, Mitochondrial/genetics , Glioma/genetics , Base Sequence , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Genomic Instability , Glioma/mortality , Glioma/pathology , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Genetic , Prognosis , Survival Analysis
18.
J Neurooncol ; 72(2): 125-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15925992

ABSTRACT

Twenty-six recurrent Glioblastoma (rGBM) patients sequentially treated at the National Neurological Institute 'C Besta' were enrolled for a second surgery in order to remove recurrent tumor and to place an Ommaya reservoire to allow local delivery of chemotherapy and local pre-targeted radio-immunotherapy (RIT). All patients had partial tumor resection and 75% of them had a residual tumor mass after exeresis larger than 2 cm. After surgery all patients were managed with a second line systemic chemotherapy (PCV). Moreover the protocol scheduled two cycles of local RIT (90 Yttrium 5- 25 mCi per cycle) with a 10 week interval. Locoregional mitoxantrone chemotherapy was locally delivered as a single dose of 4 mg every 20 days. Responses to treatment were assessed by monthly neurological examination and by MRI or contrast-enhanced CT scan performed every 2 months. For the whole group of patients the PFS after second surgery at 6 and 12 months was 61% and 22%, respectively and survival after recurrence at 6, 12 and 18 months was 80%, 53% and 42%, respectively. Neither major side effects occurred systemically nor related on the place of local injections. The percentage of long-term survivors was very high: 42% of patients were still alive at 18 months. We stress the concept that the combined treatments could be more effective if delivered into a smaller residual tumor mass and probably in an adjuvant setting, before tumour recurrence.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Mitoxantrone/administration & dosage , Radioimmunotherapy/methods , Yttrium Radioisotopes/administration & dosage , Adolescent , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Bone Marrow Diseases/chemically induced , Brain Neoplasms/surgery , Chemotherapy, Adjuvant/methods , Combined Modality Therapy , Disease-Free Survival , Drug Delivery Systems/methods , Glioblastoma/surgery , Humans , Infusions, Intralesional , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Recurrence, Local/surgery , Pilot Projects
19.
J Neurooncol ; 71(3): 271-6, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15735916

ABSTRACT

We report on our experience in the treatment of intracranial germinomas (18 pure germinomas and two germinomas with syncytiotrophoblastic giant cells) according to a strategy of radiotherapy doses and fields reduction after a neoadjuvant chemotherapy (Cisplatin-vinblastine and bleomycin combination). Radiation therapy was delivered after the completion of the third and last course of chemotherapy. For the solitary germinoma the target volume was the gross tumour volume. In the five multifocal germinoma patients the whole ventricle volume was irradiated. For the single disseminated germinoma patient we treated the whole central nervous system. The cumulative doses were 30 Gy for the pure germinomas. For the STGCs, a cumulative dose of 35 Gy was used. The median follow-up was 55 months (range 12-120). 18 patients were alive without recurrence of disease. In the two patients with STGCs the death took place 16 and 35 months after diagnosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Germinoma/drug therapy , Germinoma/radiotherapy , Adolescent , Adult , Antibiotics, Antineoplastic/administration & dosage , Bleomycin/administration & dosage , Chemotherapy, Adjuvant/methods , Cisplatin/administration & dosage , Combined Modality Therapy/methods , Female , Giant Cell Tumors/drug therapy , Giant Cell Tumors/therapy , Humans , Male , Middle Aged , Pineal Gland/pathology , Radiation Dosage , Survival Analysis , Treatment Outcome , Vinblastine/administration & dosage
20.
J Neurooncol ; 71(1): 19-25, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15719269

ABSTRACT

The survival of patients with malignant gliomas is still unsatisfactory despite multimodality treatment, therefore new therapeutic strategies are required. Tumor necrosis factor apoptosis related ligand (TRAIL/Apo2L), a member of the tumor necrosis factor superfamily, may induce apoptotic cell death in several tumors, but not in normal cells, upon binding with specific receptors. In the present study, the expression and function of TRAIL receptors (TRAIL-R1/DR4 and TRAIL-R2/DR5) has been investigated in five human glioma cell lines (U87, U138, U373, A172, SW1783) in ex vivo tumors and in primary cultures obtained from the tumors. Our data show that gliomas preferentially express TRAIL R2 and that treatment with topotecan, a topoisomerase I inhibitor, significantly up-regulates its expression as detected by flow cytometry and western blotting. Moreover, in most cases, treatment with topotecan resulted in an increased sensitivity to TRAIL-dependent apoptosis, although cyclohexymide had to be added to induce apoptosis. On glioma cell lines, the effects of irradiation on TRAIL receptors were also analysed. In our experimental conditions, irradiation with 2 Gy had a modest additive effect on TRAIL-dependent apoptosis and was not able to modulate TRAIL receptor expression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Glioma/metabolism , Membrane Glycoproteins/metabolism , Nervous System Neoplasms/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins , Cell Line, Tumor , Combined Modality Therapy , Glioma/therapy , Humans , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/radiation effects , Nervous System Neoplasms/therapy , Radiation, Ionizing , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/radiation effects , TNF-Related Apoptosis-Inducing Ligand , Topotecan/pharmacology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...