Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Curr Opin Neurol ; 36(6): 557-563, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37865836

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to discuss the molecular pathways governing the development of seizures in glioma patients. RECENT FINDINGS: The intrinsic epileptogenicity of the neuronal component of glioneuronal and neuronal tumors is the most relevant factor for seizure development. The two major molecular alterations behind epileptogenicity are the rat sarcoma virus (RAS)/mitogen-activated protein kinase / extracellular signal-regulated kinase (MAPK/ERK) and phosphatidylinositol-3-kinase / protein kinase B / mammalian target of rapamycin (P13K/AKT/mTOR) pathways. The BRAFv600E mutation has been shown in experimental models to contribute to epileptogenicity, and its inhibition is effective in controlling both seizures and tumor growth. Regarding circumscribed astrocytic gliomas, either BRAFv600E mutation or mTOR hyperactivation represent targets of treatment. The mechanisms of epileptogenicity of diffuse lower-grade gliomas are different: in addition to enhanced glutamatergic mechanisms, the isocitrate dehydrogenase (IDH) 1/2 mutations and their product D2-hydroxyglutarate (D2HG), which is structurally similar to glutamate, exerts excitatory effects on neurons also dependent on the presence of astrocytes. In preclinical models IDH1/2 inhibitors seem to impact both tumor growth and seizures. Conversely, the molecular factors behind the epileptogenicity of glioblastoma are unknown. SUMMARY: This review summarizes the current state of molecular knowledge on epileptogenicity in gliomas and highlights the relationships between epileptogenicity and tumor growth.


Subject(s)
Brain Neoplasms , Epilepsy , Glioma , Humans , Brain Neoplasms/complications , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Isocitrate Dehydrogenase/genetics , Glioma/complications , Glioma/genetics , Glioma/metabolism , Epilepsy/genetics , Seizures/complications , Mutation , Risk Factors , TOR Serine-Threonine Kinases/genetics
2.
J Neurooncol ; 163(2): 301-311, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37231231

ABSTRACT

BACKGROUND: Serum albumin has been demonstrated as prognostic parameter in non-Hodgkin lymphoma (NHL). Primary central nervous system lymphoma (PCNSL) is a rare extranodal NHL with highly aggressive behavior. In this study, we aimed at creating a novel prognostic model for PCNSL based on serum albumin levels. METHODS: We compared several commonly used laboratory nutritional parameters for predicting the survival of PCNSL patients using overall survival (OS) for outcome analysis and receiver operating characteristic curve analysis to determine the optimal cut-off values. Parameters associated with OS were evaluated by univariate and multivariate analyses. Independent prognostic parameters for OS were selected for risk stratification, including albumin ≤ 4.1 g/dL, ECOG PS > 1, and LLR > 166.8, which were associated with shorter OS; albumin > 4.1 g/dL, ECOG PS 0-1 and LLR ≤ 166.8, which were associated with longer OS, and five-fold cross-validation was used for evaluating predictive accuracy of identified prognostic model. RESULTS: By univariate analysis, age, ECOG PS, MSKCC score, Lactate dehydrogenase-to-lymphocyte ratio (LLR), total protein, albumin, hemoglobin, and albumin to globulin ratio (AGR) resulted statistically associated with the OS of PCNSL. By multivariate analysis, albumin ≤ 4.1 g/dL, ECOG PS > 1, and LLR > 166.8 were confirmed to be significant predictors of inferior OS. We explored several PCNSL prognostic models based on albumin, ECOG PS and LLR with 1 point assigned to each parameter. Eventually, a novel and effective PCNSL prognostic model based on albumin and ECOG PS successfully classified patients into three risk groups with 5-year survival rates of 47.5%, 36.9%, and 11.9%, respectively. CONCLUSIONS: The novel two-factor prognostic model based on albumin and ECOG PS we propose represents a simple but significant prognostic tool for assessing newly diagnosed patients with PCNSL.


Subject(s)
Lymphoma, Non-Hodgkin , Serum Albumin , Humans , Prognosis , Serum Albumin/metabolism , Lymphoma, Non-Hodgkin/diagnosis , Lymphoma, Non-Hodgkin/therapy , Lymphocytes , Retrospective Studies
3.
Curr Oncol Rep ; 25(7): 777-785, 2023 07.
Article in English | MEDLINE | ID: mdl-37071295

ABSTRACT

PURPOSE OF REVIEW: To summarize the mechanisms of tumor angiogenesis and resistance to antiangiogenic therapy, and the influence on tumor microenvironment. RECENT FINDINGS: Several clinical trials have investigated the activity of anti-VEGF monoclonal antibodies and tyrosine kinase inhibitors in glioblastoma, shedding the light on their limitations in terms of disease control and survival. We have outlined the mechanisms of resistance to antiangiogenic therapy, including vessel co-option, hypoxic signaling in response to vessel destruction, modulation of glioma stem cells, and trafficking of tumor-associated macrophages in tumor microenvironment. Moreover, novel generation of antiangiogenic compounds for glioblastoma, including small interfering RNAs and nanoparticles, as a delivery vehicle, could enhance selectivity and reduce side effects of treatments. There is still a rationale for the use of antiangiogenic therapy, but a better understanding of vascular co-option, vascular mimicry, and dynamic relationships between immunosuppressive microenvironment and blood vessel destruction is crucial to develop next-generation antiangiogenic compounds.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Bevacizumab/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/pathology , Vascular Endothelial Growth Factor A , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Tumor Microenvironment
4.
Int J Mol Sci ; 24(13)2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37445846

ABSTRACT

Research regarding the mechanisms of brain damage following radiation treatments for brain tumors has increased over the years, thus providing a deeper insight into the pathobiological mechanisms and suggesting new approaches to minimize this damage. This review has discussed the different factors that are known to influence the risk of damage to the brain (mainly cognitive disturbances) from radiation. These include patient and tumor characteristics, the use of whole-brain radiotherapy versus particle therapy (protons, carbon ions), and stereotactic radiotherapy in various modalities. Additionally, biological mechanisms behind neuroprotection have been elucidated.


Subject(s)
Brain Neoplasms , Radiosurgery , Humans , Cranial Irradiation , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Brain , Radiosurgery/adverse effects , Combined Modality Therapy
5.
J Neurooncol ; 160(2): 389-402, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36309895

ABSTRACT

PURPOSE: In the phase 2 REGOMA trial, regorafenib improved overall survival, as compared with lomustine, in glioblastoma (GBM) patients at first progression after chemoradiation. Recently, some real-life trials showed similar impact on survival but a higher rate of adverse events than in REGOMA, thus raising concerns over tolerability. The aim of this study was to assess the efficacy and tolerability of a lower intensity regorafenib regimen. PATIENTS AND METHODS: Regorafenib daily dose was gradually increased from 80 to 160 mg across the first 2 cycles. Progression-free survival (PFS) and overall survival (OS) were defined as time from regorafenib initiation and disease progression or death. RESULTS: Sixty-six GBM patients were included. Median age was 60.0 years. Median PFS and OS following regorafenib were 2.7 and 7.1 months, respectively. Best RANO response to regorafenib were partial response (PR) in 10 (15.1%), stable disease in 17 (25.8%), and progressive disease in 39 (59.1%) patients. Forty-six (69.7%) patients presented adverse events of any grade, and 21 (31.8%) grade 3-4 toxicity. In a multivariable analysis, higher age and absence of MGMTp methylation were significantly associated with poorer disease control after regorafenib. CONCLUSIONS: Our study is the largest observational real-life study on the use of regorafenib. Our lower intensity regimen proved as effective as the standard 160 mg daily schedule (mPFS and mOS being 2.7 vs 2.0 months and 7.1 vs 7.4 months in our study vs REGOMA, respectively). Moreover, we observed a higher rate of PRs as compared with REGOMA (15.0% vs 3.0%).


Subject(s)
Glioblastoma , Humans , Middle Aged , Glioblastoma/drug therapy , Drug Tapering , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/chemically induced , Phenylurea Compounds/adverse effects
6.
Curr Oncol Rep ; 24(8): 985-993, 2022 08.
Article in English | MEDLINE | ID: mdl-35384591

ABSTRACT

PURPOSE OF REVIEW: To review state of art and relevant advances in the molecular genetics and management of ependymomas of children and adults. RECENT FINDINGS: Ependymomas may occur either in the brain or in the spinal cord. Compared with intracranial ependymomas, spinal ependymomas are less frequent and exhibit a better prognosis. The new WHO classification of CNS tumors of 2021 has subdivided ependymomas into different histomolecular subgroups with different outcome. The majority of studies have shown a major impact of extent of resection; thus, a complete resection must be performed, whenever possible, at first surgery or at reoperation. Conformal radiotherapy is recommended for grade 3 or incompletely resected grade II tumors. Proton therapy is increasingly employed especially in children to reduce the risk of neurocognitive and endocrine sequelae. Craniospinal irradiation is reserved for metastatic disease. Chemotherapy is not useful as primary treatment and is commonly employed as salvage treatment for patients failing surgery and radiotherapy. Standard treatments are still the mainstay of treatment: the discovery of new druggable pathways will hopefully increase the therapeutic armamentarium in the near future.


Subject(s)
Brain Neoplasms , Ependymoma , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Child , Ependymoma/diagnosis , Ependymoma/genetics , Ependymoma/therapy , Humans , Prognosis , Salvage Therapy
7.
Curr Treat Options Oncol ; 22(12): 110, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34693454

ABSTRACT

OPINION STATEMENT: Systemic therapy for brain metastases (BM) is quickly moving from conventional cytotoxic chemotherapy toward targeted therapies, that allow a disruption of driver molecular pathways. The discovery of actionable driver mutations has led to the development of an impressive number of tyrosine kinase inhibitors (TKIs), that target the epidermal growth factor receptor (EGFR) mutations, anaplastic-lymphoma-kinase (ALK) rearrangements, and other rare molecular alterations in patients bearing metastatic non-small cell lung cancer (NSCLC) in the brain, with remarkable results in terms of intracranial disease control and overall survival. Moreover, these drugs may delay the use of local therapies, such as stereotactic radiosurgery (SRS) or whole-brain radiotherapy (WBRT). New drugs with higher molecular specificity and ability to cross the CNS barriers (BBB, BTB and blood-CSF) are being developed. Two major issues are related to targeted therapies. First, the emergence of a resistance is a common event, and a deeper understanding of molecular pathways that are involved is critical for the successful development of effective new targeted agents. Second, an early detection of tumor progression is of utmost importance to avoid the prolongation of an ineffective therapy while changing to another drug. In order to monitor over time the treatment to targeted therapies, liquid biopsy, that allows the detection in biofluids of either circulating tumor cells (CTCs) or circulating tumor DNA (ctDNA) or exosomes, is increasingly employed in clinical trials: with respect to BM the monitoring of both blood and CSF is necessary. Also, radiomics is being developed to predict the mutational status of the BM on MRI.For patients without druggable mutations or who do not respond to targeted agents, immunotherapy with checkpoint inhibitors is increasingly employed, alone or in combination with radiotherapy. Pseudoprogression after immunotherapy alone maybe a challenge for several months after the start of treatment, and the same is true for radionecrosis after the combination of immunotherapy and SRS. In this regard, the value of advanced MRI techniques and PET imaging for a better distinction of pseudoprogression/radionecrosis and true tumor progression is promising, but needs validation in large prospective datasets. Last, a new frontier in the near future will be chemoprevention (primary and secondary), but we need to identify among solid tumors those subgroups of patients with a higher risk of relapsing into the brain and novel drugs, active on either neoplastic or normal cells of the microenvironment, that are cooperating in the invasion of brain tissue.


Subject(s)
Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Anaplastic Lymphoma Kinase/genetics , Blood-Brain Barrier/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Chemoradiotherapy , Circulating Tumor DNA/blood , Circulating Tumor DNA/cerebrospinal fluid , Computational Biology , Gene Rearrangement , Genes, erbB-1/genetics , Humans , Lung Neoplasms/pathology , Magnetic Resonance Imaging , Molecular Targeted Therapy , Neoplastic Cells, Circulating/metabolism
8.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884457

ABSTRACT

The presence of barriers, such as the blood-brain barrier (BBB) and brain-tumor barrier (BTB), limits the penetration of antineoplastic drugs into the brain, resulting in poor response to treatments. Many techniques have been developed to overcome the presence of these barriers, including direct injections of substances by intranasal or intrathecal routes, chemical modification of drugs or constituents of BBB, inhibition of efflux pumps, physical disruption of BBB by radiofrequency electromagnetic radiation (EMP), laser-induced thermal therapy (LITT), focused ultrasounds (FUS) combined with microbubbles and convection enhanced delivery (CED). However, most of these strategies have been tested only in preclinical models or in phase 1-2 trials, and none of them have been approved for treatment of brain tumors yet. Concerning the treatment of brain metastases, many molecules have been developed in the last years with a better penetration across BBB (new generation tyrosine kinase inhibitors like osimertinib for non-small-cell lung carcinoma and neratinib/tucatinib for breast cancer), resulting in better progression-free survival and overall survival compared to older molecules. Promising studies concerning neural stem cells, CAR-T (chimeric antigen receptors) strategies and immunotherapy with checkpoint inhibitors are ongoing.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms/therapy , Blood-Brain Barrier/drug effects , Brain Neoplasms/secondary , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Neural Stem Cells/transplantation , Receptors, Chimeric Antigen/metabolism
9.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360713

ABSTRACT

Rare central nervous system (CNS) tumours represent a unique challenge. Given the difficulty of conducting dedicated clinical trials, there is a lack of therapies for these tumours supported by high quality evidence, and knowledge regarding the impact of standard treatments (i.e., surgery, radiotherapy or chemotherapy) is commonly based on retrospective studies. Recently, new molecular techniques have led to the discovery of actionable molecular alterations. The aim of this article is to review recent progress in the molecular understanding of and therapeutic options for rare brain tumours, both in children and adults. We will discuss options such as targeting the mechanistic target of rapamycin (mTOR) pathway in subependymal giant cells astrocytomas (SEGAs) of tuberous sclerosis and BRAF V600E mutation in rare glial (pleomorphic xanthoastrocytomas) or glioneuronal (gangliogliomas) tumours, which are a model of how specific molecular treatments can also favourably impact neurological symptoms (such as seizures) and quality of life. Moreover, we will discuss initial experiences in targeting new molecular alterations in gliomas, such as isocitrate dehydrogenase (IDH) mutations and neurotrophic tyrosine receptor kinase (NTRK) fusions, and in medulloblastomas such as the sonic hedgehog (SHH) pathway.


Subject(s)
Brain Neoplasms , Molecular Targeted Therapy , Mutation , Neoplasm Proteins , Rare Diseases , Signal Transduction , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Rare Diseases/genetics , Rare Diseases/metabolism , Rare Diseases/therapy
10.
BMC Cancer ; 20(1): 410, 2020 May 12.
Article in English | MEDLINE | ID: mdl-32398144

ABSTRACT

BACKGROUND: Neurosurgical resection represents an important treatment option in the modern, multimodal therapy approach of brain metastases (BM). Guidelines for perioperative imaging exist for primary brain tumors to guide postsurgical treatment. Optimal perioperative imaging of BM patients is so far a matter of debate as no structured guidelines exist. METHODS: A comprehensive questionnaire about perioperative imaging was designed by the European Association of Neuro-Oncology (EANO) Youngsters Committee. The survey was distributed to physicians via the EANO network to perform a descriptive overview on the current habits and their variability on perioperative imaging. Chi square test was used for dichotomous variables. RESULTS: One hundred twenty physicians worldwide responded to the survey. MRI was the preferred preoperative imaging method (93.3%). Overall 106/120 (88.3%) physicians performed postsurgical imaging routinely including MRI alone (62/120 [51.7%]), postoperative CT (29/120 [24.2%]) and MRI + CT (15/120 [12.5%]). No correlation of postsurgical MRI utilization in academic vs. non-academic hospitals (58/89 [65.2%] vs. 19/31 [61.3%], p = 0.698) was found. Early postoperative MRI within ≤72 h after resection is obtained by 60.8% of the participants. The most frequent reason for postsurgical imaging was to evaluate the extent of tumor resection (73/120 [60.8%]). In case of residual tumor, 32/120 (26.7%) participants indicated to adjust radiotherapy, 34/120 (28.3%) to consider re-surgery to achieve complete resection and 8/120 (6.7%) to evaluate both. CONCLUSIONS: MRI was the preferred imaging method in the preoperative setting. In the postoperative course, imaging modalities and timing showed high variability. International guidelines for perioperative imaging with special focus on postoperative MRI to assess residual tumor are warranted to optimize standardized management and adjuvant treatment decisions for BM patients.


Subject(s)
Brain Neoplasms/pathology , Neoplasm, Residual/pathology , Neuroimaging/methods , Neurosurgical Procedures/methods , Perioperative Care , Brain Neoplasms/surgery , Europe , Humans , Neoplasm, Residual/surgery , Prognosis , Surveys and Questionnaires
11.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198331

ABSTRACT

The management of breast cancer (BC) has rapidly evolved in the last 20 years. The improvement of systemic therapy allows a remarkable control of extracranial disease. However, brain (BM) and leptomeningeal metastases (LM) are frequent complications of advanced BC and represent a challenging issue for clinicians. Some prognostic scales designed for metastatic BC have been employed to select fit patients for adequate therapy and enrollment in clinical trials. Different systemic drugs, such as targeted therapies with either monoclonal antibodies or small tyrosine kinase molecules, or modified chemotherapeutic agents are under investigation. Major aims are to improve the penetration of active drugs through the blood-brain barrier (BBB) or brain-tumor barrier (BTB), and establish the best sequence and timing of radiotherapy and systemic therapy to avoid neurocognitive impairment. Moreover, pharmacologic prevention is a new concept driven by the efficacy of targeted agents on macrometastases from specific molecular subgroups. This review aims to provide an overview of the clinical and molecular factors involved in the selection of patients for local and/or systemic therapy, as well as the results of clinical trials on advanced BC. Moreover, insight on promising therapeutic options and potential directions of future therapeutic targets against BBB and microenvironment are discussed.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/therapy , Estrogen Receptor alpha/metabolism , Meningeal Neoplasms/secondary , Receptor, ErbB-2/metabolism , Triple Negative Breast Neoplasms/therapy , Animals , Antineoplastic Agents/pharmacology , Blood-Brain Barrier , Brain Neoplasms/therapy , Breast Neoplasms/pathology , Clinical Trials as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Meningeal Neoplasms/therapy , Mice , Neoplasm Recurrence, Local , Prognosis , Triple Negative Breast Neoplasms/pathology
12.
Int J Mol Sci ; 21(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979374

ABSTRACT

The neurotrophic tropomyosin receptor kinase (NTRK) genes (NTRK1, NTRK2, and NTRK3) code for three transmembrane high-affinity tyrosine-kinase receptors for nerve growth factors (TRK-A, TRK-B, and TRK-C) which are mainly involved in nervous system development. Loss of function alterations in these genes can lead to nervous system development problems; conversely, activating alterations harbor oncogenic potential, promoting cell proliferation/survival and tumorigenesis. Chromosomal rearrangements are the most clinically relevant alterations of pathological NTRK activation, leading to constitutionally active chimeric receptors. NTRK fusions have been detected with extremely variable frequencies in many pediatric and adult cancer types, including central nervous system (CNS) tumors. These alterations can be detected by different laboratory assays (e.g., immunohistochemistry, FISH, sequencing), but each of these approaches has specific advantages and limitations which must be taken into account for an appropriate use in diagnostics or research. Moreover, therapeutic targeting of this molecular marker recently showed extreme efficacy. Considering the overall lack of effective treatments for brain neoplasms, it is expected that detection of NTRK fusions will soon become a mainstay in the diagnostic assessment of CNS tumors, and thus in-depth knowledge regarding this topic is warranted.


Subject(s)
Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Gene Fusion , Membrane Glycoproteins/genetics , Protein Kinase Inhibitors/therapeutic use , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Animals , Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/drug therapy , Humans , Membrane Glycoproteins/metabolism , Precision Medicine , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Signal Transduction/genetics
13.
Curr Opin Oncol ; 31(6): 493-500, 2019 11.
Article in English | MEDLINE | ID: mdl-31414987

ABSTRACT

PURPOSE OF REVIEW: The current article reviews the state of art of prevention strategies for brain metastases from solid tumors and touches both old pivotal studies and new directions of personalized molecular approaches. RECENT FINDINGS: Prophylactic cranial irradiation (PCI) has a definite role in the prevention of relapse into the brain for patients with small cell lung cancer (SCLC) responding to chemotherapy and radiotherapy as it prolongs overall survival (OS). However, the risk of late cognitive deficit following whole brain radiotherapy (WBRT) in this patient population is still not well known. Conversely, PCI significantly reduces the incidence of brain metastases and prolongs the disease-free interval in patients with non-SCLC (NSCLC), but does not improve OS thus far. Pharmacologic prevention is a new concept driven by the efficacy of targeted agents on macrometastases from specific molecular subgroups. SUMMARY: The future challenges for prevention of brain metastases are represented by the identification of subgroups of patients at higher risk of relapse into the brain coupled with either new WBRT strategies to better preserve cognition or effective molecular agents to target micrometastases.


Subject(s)
Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Cranial Irradiation/methods , Brain Neoplasms/radiotherapy , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Clinical Trials, Phase III as Topic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Randomized Controlled Trials as Topic
14.
J Neurooncol ; 145(1): 115-123, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31556015

ABSTRACT

INTRODUCTION: The optimal management of high risk WHO grade II gliomas after surgery is debated including the role of initial temozolomide to delay radiotherapy and risk of cognitive defects. METHODS: A post-hoc analysis of a phase II multicenter study on high risk WHO grade II gliomas, receiving initial temozolomide alone, has re-evaluated the long-term results within the molecular subgroups of WHO 2016. The primary endpoint of the study was response according to RANO, being seizure response, PFS and OS secondary endpoints. RESULTS: Response rate among oligodendrogliomas IDH-mutant and 1p/19q codeleted (76%) was significantly higher than that among diffuse astrocytomas either mutant (55%) or wild-type (36%). A reduction of seizure frequency > 50% was observed in 87% of patients and a seizure freedom in 72%. The probability of seizure reduction > 50% was significantly associated with the presence of an IDH mutation. Median PFS, PFS at 5 and 10 years, median OS and OS at 5 and 10 years were significantly longer in oligodendrogliomas IDH-mutant and 1p/19q codeleted. Sixty-seven percent of patients with oligodendroglioma IDH mutant and 1p/19q codeleted did not recur with a median follow up of 9.3 years, while 59% did not receive radiotherapy at recurrence with a median follow up of 8.2 years. CONCLUSIONS: The beneficial effects of initial temozolomide prevail in oligodendrogliomas IDH-mutant and 1p/19q codeleted: thus, these tumors, when incompletely resected or progressive after surgery alone, or with intractable seizures, should receive temozolomide as initial treatment with salvage radiotherapy and/o reoperation and/or second-line chemotherapy at recurrence.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Temozolomide/therapeutic use , Adult , Aged , Brain Neoplasms/classification , Brain Neoplasms/pathology , Female , Follow-Up Studies , Glioma/classification , Glioma/pathology , Humans , Male , Middle Aged , Neoplasm Grading , Survival Rate , World Health Organization
16.
Surg Technol Int ; 35: 432-440, 2019 11 10.
Article in English | MEDLINE | ID: mdl-31373379

ABSTRACT

BACKGROUND: A relationship between the extent of resection (EOR) and survival has been demonstrated in patients with glioblastomas (GBMs). However, despite gross total resection (GTR) of the enhancing nodule (EN), GBMs usually relapse, generally near the surgical cavity. OBJECTIVE: The aim of this study was to determine the prognostic role of FLAIR resection of GBMs by analyzing pre- and post-operative MRIs to estimate the EOR of EN, FLAIR-hyperintense regions and total tumor volume (TTV). METHODS: Radiologic and clinical outcomes were analyzed retrospectively. Pre- and post-operative EN volume, pre- and postoperative FLAIR volume (POFV), and pre- and postoperative TTV were analyzed. EOR was then calculated for each component. Time-dependent ROC curves and cut-off values for pre- and post-operative volumes and EOR were calculated. A Kaplan-Meier analysis with the log-rank test and Cox regression analysis were then used to analyze progression-free survival (PFS) and overall survival (OS). RESULTS: We did not find any correlation between EOR of FLAIR-altered regions and patient survival. On the other hand, there were statistically significant relationships between the prognosis and both a preoperative EN volume less than 31.35 cm3 (p=0.032) and a postoperative EN volume less than 0.57 cm3 (p=0.015). Moreover, an EOR of EN greater than 96% was significantly associated with the prognosis (p=0.0051 for OS and p=0.022 for PFS). CONCLUSION: Our retrospective, multi-center study suggests that survival in patients with GBM is not affected by the extent of resection of FLAIR-hyperintense areas.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/surgery , Glioblastoma/surgery , Humans , Magnetic Resonance Imaging , Neoplasm Recurrence, Local , Neurosurgical Procedures , Retrospective Studies , Treatment Outcome
17.
J Neurooncol ; 136(1): 105-114, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29030718

ABSTRACT

To report the efficacy and tolerability of lacosamide as an add-on treatment in patients with gliomas and uncontrolled seizures despite conventional antiepileptic drugs (AEDs). We conducted an observational study on 71 patients to describe patterns of response to lacosamide and the association between clinico-pathological factors and seizure control. We observed at 3, 6 and 9 months a seizure reduction ≥ 50% in 74.6, 76 and 86.2% of patients and a seizure freedom in 42.2, 43 and 50%, respectively. The median number of seizures in the 3 months before treatment was 13, and decreased to 3 between baseline and 6 months, and to 0.5 between 6 and 9 months. The best seizure response was observed at 3 months (62%). Sixty per cent of patients displayed the maximum seizure control with doses of lacosamide of 100-250 mg/day, while 21% needed doses up to 400 mg/day. Seizure reduction ≥ 50% and seizure freedom were higher in patients who received lacosamide as first add-on compared to those who received a later adjunctive therapy. A reduction ≥ 50% of seizures was observed in a proportion of patients with progressive disease on MRI. Age > 45 years (OR 0.11, 95% CI 0.02-0.63, p = 0.013) was a significant predictor of seizure freedom at 9 months on multivariate analysis. The study suggests that lacosamide, when added to any baseline AEDs, is effective in obtaining a high seizure reduction and seizure freedom regardless of the tumor activity and response to antineoplastic therapies.


Subject(s)
Anticonvulsants/therapeutic use , Brain Neoplasms/complications , Glioma/complications , Lacosamide/therapeutic use , Seizures/drug therapy , Adult , Antineoplastic Combined Chemotherapy Protocols , Female , Humans , Male , Middle Aged , Seizures/complications , Treatment Outcome
18.
Q J Nucl Med Mol Imaging ; 62(3): 225-238, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29696949

ABSTRACT

High-grade gliomas (HGGs) are the most common primary tumors of the central nervous system, which include anaplastic gliomas (grade III) and glioblastomas (GBM, grade IV). Surgery is the mainstay of treatment in HGGs in order to achieve a histological and molecular characterization, as well as relieve neurological symptoms and improve seizure control. Combinations of some molecular factors, such as IDH 1-2 mutations, 1p/19q codeletion and MGMT methylation status, allow to classify different subtypes of gliomas and identify patients with different outcome. The SOC in HGGs consists in a combination of radiotherapy and chemotherapy with alkylating agents. Despite this therapeutic approach, tumor recurrence occurs in HGGs, and new surgical debulking, reirradiation or second-line chemotherapy are needed. Considering the poor results in terms of survival, several clinical trials have explored the efficacy and tolerability of antiangiogenic agents, targeted therapies against epidermal growth factor receptor (EGFR) and different immunotherapeutic approaches in recurrent and newly-diagnosed GBM, including immune checkpoint inhibitors (ICIs), and cell- or peptide-based vaccination with unsatisfactory results in term of disease control. In this review we describe the major updates in molecular biology of HGGs according to 2016 WHO Classification, the current management in newly-diagnosed and recurrent GBM and grade III gliomas, and the results of the most relevant clinical trials on targeted agents and immunotherapy.


Subject(s)
Glioma/pathology , Glioma/therapy , Glioma/drug therapy , Glioma/immunology , Humans , Immunotherapy , Molecular Targeted Therapy , Neoplasm Grading , Recurrence , Reference Standards
19.
Future Oncol ; 14(17): 1765-1774, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29956562

ABSTRACT

Breast cancer (BC) is a heterogeneous disease. Different subtypes of BC exhibit a peculiar natural history, metastatic potential and outcome. Stereotactic radiosurgery is the most used treatment for brain metastases (BM), while surgery is reserved for large and symptomatic lesions. Whole-brain radiotherapy is employed in multiple BM not amendable to radiosurgery or surgery, and it is not employed any more following local treatments of a limited number of BM. A critical issue is the distinction from pseudoprogression or radionecrosis, and tumor regrowth. Considering the increase of long-term survivors after combined or novel treatments for BM, cognitive dysfunctions following whole-brain radiotherapy represent an issue of utmost importance. Neuroprotective drugs and innovative radiotherapy techniques are being investigated to reduce this risk of cognitive sequelae. Leptomeningeal disease represents a devastating complication, either alone or in association to BM, thus targeted therapies are employed in HER2-positive BC brain and leptomeningeal metastases.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Cranial Irradiation , Neuroprotective Agents/therapeutic use , Radiosurgery , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Female , Humans , Prognosis , Quality of Life
SELECTION OF CITATIONS
SEARCH DETAIL