Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurooncol ; 167(1): 145-154, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457090

ABSTRACT

PURPOSE: Adult Diffuse midline glioma (DMG) is a very rare disease. DMGs are currently treated with radiotherapy and chemotherapy even if only a few retrospective studies assessed the impact on overall survival (OS) of these approaches. METHODS: We carried out an Italian multicentric retrospective study of adult patients with H3K27-altered DMG to assess the effective role of systemic therapy in the treatment landscape of this rare tumor type. RESULTS: We evaluated 49 patients from 6 Institutions. The median age was 37.3 years (range 20.1-68.3). Most patients received biopsy as primary approach (n = 30, 61.2%) and radiation therapy after surgery (n = 39, 79.6%). 25 (51.0%) of patients received concurrent chemotherapy and 26 (53.1%) patients received adjuvant temozolomide. In univariate analysis, concurrent chemotherapy did not result in OS improvement while adjuvant temozolomide was associated with longer OS (21.2 vs. 9.0 months, HR 0.14, 0.05-0.41, p < 0.001). Multivariate analysis confirmed the role of adjuvant chemotherapy (HR 0.1, 95%CI: 0.03-0.34, p = 0.003). In patients who progressed after radiation and/or chemotherapy the administration of a second-line systemic treatment had a significantly favorable impact on survival (8.0 vs. 3.2 months, HR 0.2, 95%CI 0.1-0.65, p = 0.004). CONCLUSION: In our series, adjuvant treatment after radiotherapy can be useful in improving OS of patients with H3K27-altered DMG. When feasible another systemic treatment after treatment progression could be proposed.


Subject(s)
Brain Neoplasms , Glioma , Adult , Humans , Young Adult , Middle Aged , Aged , Temozolomide/therapeutic use , Retrospective Studies , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Antineoplastic Agents, Alkylating/therapeutic use , Glioma/drug therapy , Glioma/pathology , Dacarbazine/therapeutic use , Chemotherapy, Adjuvant
2.
Anticancer Drugs ; 33(1): e28-e35, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34348358

ABSTRACT

To date, there are no standardized systemic treatment options for patients with metastatic pituitary carcinoma progressed to chemo and radiation therapy. Immune-checkpoint inhibitors (ICIs) have been successfully assessed in other solid malignancies and could be a concrete hope for these patients. We performed a critical review of the literature aimed to evaluate studies assessing ICIs in pituitary malignancies. We also conducted research about published translational data assessing immune-contexture in these malignancies. Some preliminary reports reported a successful administration of pembrolizumab or the combination between nivolumab and ipilimumab in patients with metastatic ACTH-secreting pituitary carcinomas. Translational data suggest that adenomas secreting growth hormone and ACTH have a suppressed immune-microenvironment, which could be more likely to benefit from ICIs. Immune-checkpoint inhibitors can be an effective treatment in patients with pituitary carcinoma and maybe also recurrent adenoma. Tumors secreting growth hormone and ACTH are more likely to benefit from ICIs due to a different immune-microenvironment.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/pathology , Adrenocorticotropic Hormone/biosynthesis , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Growth Hormone/biosynthesis , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Ipilimumab/therapeutic use , Neoplasm Metastasis , Nivolumab/therapeutic use , Tumor Microenvironment/drug effects
3.
J Neurooncol ; 159(2): 333-346, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35761160

ABSTRACT

PURPOSE: Artificial Intelligence (AI) involves several and different techniques able to elaborate a large amount of data responding to a specific planned outcome. There are several possible applications of this technology in neuro-oncology. METHODS: We reviewed, according to PRISMA guidelines, available studies adopting AI in different fields of neuro-oncology including neuro-radiology, pathology, surgery, radiation therapy, and systemic treatments. RESULTS: Neuro-radiology presented the major number of studies assessing AI. However, this technology is being successfully tested also in other operative settings including surgery and radiation therapy. In this context, AI shows to significantly reduce resources and costs maintaining an elevated qualitative standard. Pathological diagnosis and development of novel systemic treatments are other two fields in which AI showed promising preliminary data. CONCLUSION: It is likely that AI will be quickly included in some aspects of daily clinical practice. Possible applications of these techniques are impressive and cover all aspects of neuro-oncology.


Subject(s)
Neurology , Radiology , Artificial Intelligence , Humans , Machine Learning
4.
Oncologist ; 26(10): 865-878, 2021 10.
Article in English | MEDLINE | ID: mdl-34105205

ABSTRACT

Glioblastoma (GBM) is the most common primary tumor of the central nervous system. Arising from neuroepithelial glial cells, GBM is characterized by invasive behavior, extensive angiogenesis, and genetic heterogeneity that contributes to poor prognosis and treatment failure. Currently, there are several molecular biomarkers available to aid in diagnosis, prognosis, and predicting treatment outcomes; however, all require the biopsy of tumor tissue. Nevertheless, a tissue sample from a single location has its own limitations, including the risk related to the procedure and the difficulty of obtaining longitudinal samples to monitor treatment response and to fully capture the intratumoral heterogeneity of GBM. To date, there are no biomarkers in blood or cerebrospinal fluid for detection, follow-up, or prognostication of GBM. Liquid biopsy offers an attractive and minimally invasive solution to support different stages of GBM management, assess the molecular biology of the tumor, identify early recurrence and longitudinal genomic evolution, predict both prognosis and potential resistance to chemotherapy or radiotherapy, and allow patient selection for targeted therapies. The aim of this review is to describe the current knowledge regarding the application of liquid biopsy in glioblastoma, highlighting both benefits and obstacles to translation into clinical care. IMPLICATIONS FOR PRACTICE: To translate liquid biopsy into clinical practice, further prospective studies are required with larger cohorts to increase specificity and sensitivity. With the ever-growing interest in RNA nanotechnology, microRNAs may have a therapeutic role in brain tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , MicroRNAs , Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Glioblastoma/diagnosis , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Liquid Biopsy , Prognosis
5.
Oncologist ; 24(5): 664-670, 2019 05.
Article in English | MEDLINE | ID: mdl-30777895

ABSTRACT

BACKGROUND: Patients with low-grade gliomas (LGGs) with isocitrate dehydrogenase (IDH) mutation (mut) and 1p19q codeletion (codel) have a median overall survival of longer than 10 years. The aim of this study is to assess the role of postsurgical treatments. SUBJECTS, MATERIALS, AND METHODS: We evaluated patients with LGGs with IDH mut and 1p19q codel; IDH1/2 was performed by immunohistochemistry and quantitative polymerase chain reaction. In all wild-type cases, we performed next-generation sequencing. 1p19 codel analysis was performed by fluorescence in situ hybridization. RESULTS: Among the 679 patients, 93 with LGGs with IDH mutation and 1p19q codel were included. Median follow-up (FU) was 96.1 months. Eighty-four patients (90.3%) were high risk according to Radiation Therapy Oncology Group criteria. After surgery, 50 patients (53.7%) received only FU, 17 (18.3%) chemotherapy (CT), and 26 (30.1%) radiotherapy (RT) with (RT + CT, 8 patients, 8.6%) or without (RT, 18 patients, 19.4%) chemotherapy. Median progression-free survival (mPFS) was 46.3 months, 50.8 months, 103.6 months, and 120.2 months in patients with FU alone, with CT alone, with RT alone, or with RT + CT, respectively. Median PFS was significantly longer in patients who received postsurgical treatment (79.5 months, 95% confidence interval [CI]: 66.4-92.7) than patients who received FU (46.3 months, 95% CI: 36.0-56.5). Moreover, mPFS was longer in patients who received RT (alone or in combination with CT, n = 26, 113.8 months, 95% CI: 57.2-170.5) than those who did not (n = 67, 47.3 months, 95% CI: 36.4-58.2). In particular, temozolomide alone did not improve PFS with respect to FU. CONCLUSION: RT with or without chemotherapy, but not temozolomide alone, could extend PFS in IDH mut 1p19q codel LGGs. IMPLICATIONS FOR PRACTICE: Low-grade gliomas with high-risk features, defined according to Radiation Therapy Oncology Group criteria, receive radiotherapy and/or chemotherapy as postsurgical treatments. Radiotherapy, however, has serious long-term effects (cognitive impairment), which are to be taken into account in these young patients. Moreover, low-grade gliomas with isocitrate dehydrogenase mutation and 1p19q codeletion (oligodendrogliomas) have an extremely long survival and a better prognosis. This study suggests that postsurgical treatments prolong the time before tumor progression in patients with good prognosis as well as those with oligodendroglioma. Moreover, temozolomide alone might not be effective in prolonging progression-free survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Oligodendroglioma/therapy , Adult , Aged , Aged, 80 and over , Brain/pathology , Brain/surgery , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chemoradiotherapy, Adjuvant/methods , Chemoradiotherapy, Adjuvant/statistics & numerical data , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/statistics & numerical data , Chromosomes, Human, Pair 1/genetics , Female , Follow-Up Studies , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation , Neoplasm Grading , Oligodendroglioma/genetics , Oligodendroglioma/mortality , Oligodendroglioma/pathology , Progression-Free Survival , Temozolomide/therapeutic use
6.
Future Oncol ; 15(22): 2595-2601, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31339049

ABSTRACT

Aim: European Organization for Research and Treatment of Cancer (EORTC) and the Radiation Therapy Oncology Group (RTOG) criteria are used to choose treatment in low-grade gliomas. However, no data exist on their concordance. Methods: Low-grade glioma patients treated at our institution from 1998 to 2015 and assessable for both RTOG and EORTC criteria were included to analyze their concordance. Surgery extension, postsurgical treatments, molecular characteristics (IDH mutation, MGMT methylation and 1p/19q codeletion) were recorded. Results: We included 99 patients. The concordance was low (50.5%; K = 0.127; p = 0.021) but for two subgroups: EORTC high-risk patients were also RTOG high-risk patients (concordance: 97.5%) and RTOG low-risk patients were also EORTC low-risk patients (concordance: 90.9%). Conclusion: The concordance between RTOG and EORTC criteria is low. Thus, clinical trials adopting different risk criteria are not comparable.


Subject(s)
Glioma/epidemiology , Glioma/therapy , Prognosis , Adult , Aged , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Disease-Free Survival , Female , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Grading , Tumor Suppressor Proteins/genetics
7.
Future Oncol ; 14(11): 1063-1069, 2018 May.
Article in English | MEDLINE | ID: mdl-29741106

ABSTRACT

AIM: To identify patients with recurrent glioblastoma after temozolomide (TMZ) concurrent with and adjuvant to radiotherapy who could benefit from TMZ rechallenge at the time of disease progression. METHODS: We retrospectively evaluated 106 glioblastoma patients who had nonprogressive disease at first magnetic resonance imaging after completion of TMZ concurrent with and adjuvant to radiotherapy, a treatment-free interval (TFI) of at least 8 weeks and received TMZ rechallenge or a nitrosourea at the time of progression. RESULTS: In patients with TFI ≥5 months, median survival was 17.7 and 11.6 months and median progression-free survival was 8.1 and 5.8 months in the TMZ and nitrosourea group, respectively. Longer TFI was associated with reduced risk for death (p = 0.002) and for disease progression (p = 0.005). CONCLUSION: TFI ≥5 months represents a predictor of retained TMZ sensitivity.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Disease-Free Survival , Female , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/pathology , Temozolomide , Treatment Outcome
8.
Future Oncol ; 14(16): 1559-1567, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29938525

ABSTRACT

AIM: To evaluate relevance of clinical and molecular factors in adult low-grade gliomas (LGG) and to correlate with survival. METHODS: We reviewed records from adult LGG patients from 1991 to 2015 who received surgery and had sufficient tissue to molecular biomarkers characterization. RESULTS: 213 consecutive LGG patients were included: 17.4% were low-risk, according to Radiation Therapy Oncology Group (RTOG) risk assessment. IDH 1/2 mutation, 1p/19q co-deletion, MGMT methylation were found in 93, 50.8 and 65.3% of patients. Median follow-up was 98.3 months. In univariate analysis, overall survival was influenced by extent of resection (p = 0.011), IDH mutation (p < 0.001), 1p/19q co-deletion (p = 0.015) and MGMT methylation (p = 0.013). In multivariate analysis, RTOG clinical risk (p = 0.006), IDH mutation (p < 0.001) and 1p/19q co-deletion (p = 0.035) correlated with overall survival. RTOG clinical risk (p = 0.006), IDH mutation (p < 0.001) and 1p/19q co-deletion (p = 0.035) correlated with overall survival. CONCLUSION: Both clinical and molecular factors are essential to determine prognosis and treatment strategies.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/mortality , Glioma/genetics , Glioma/mortality , Adolescent , Adult , Aged , Brain Neoplasms/therapy , Chromosome Deletion , Chromosomes, Human, Pair 1 , Cohort Studies , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Glioma/therapy , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Multivariate Analysis , Mutation , Neurosurgical Procedures , Prognosis , Risk Factors , Tumor Suppressor Proteins/genetics
9.
Curr Neurol Neurosci Rep ; 16(2): 13, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26750128

ABSTRACT

Nitrosoureas represent one of the most active classes of agents in the treatment of high-grade gliomas and glioblastoma. In clinical practice, the most commonly used compounds are lomustine (either alone or in combination with procarbazine and vincristine), carmustine, and fotemustine. Given their toxicity profile and subsequent to the introduction of temozolomide in clinical practice, most of these agents were moved to the recurrent setting. This review focuses on the role of the nitrosoureas currently used in clinical practice for the treatment of malignant gliomas.


Subject(s)
Glioma , Dacarbazine/analogs & derivatives , Glioma/pathology , Humans , Lomustine , Nitrosourea Compounds , Organophosphorus Compounds , Procarbazine , Temozolomide , Vincristine
10.
Curr Treat Options Oncol ; 17(9): 49, 2016 09.
Article in English | MEDLINE | ID: mdl-27461038

ABSTRACT

OPINION STATEMENT: At the time of glioblastoma (GBM) recurrence, a sharp analysis of prognostic factors, disease characteristics, response to adjuvant treatment, and clinical conditions should be performed. A prognostic assessment could allow a careful selection between patients that could be proposed to intensified approaches or palliative setting. Participation in clinical trials aims to improve outcome, and should be encouraged due to dismal prognosis of GBM patients after recurrence. Reoperation should be proposed if the tumor is amenable to a complete resection and if prognostic factors suggest that patient could benefit from a second surgery. Second-line chemotherapy should be chosen based on MGMT status, time to disease recurrence, and toxicity profile. If enrollment into a clinical trial is not possible, a nitrosourea-based regimen is the preferred choice, carefully evaluating any previous temozolomide (TMZ)-related toxicity. In MGMT-methylated patients relapsing after TMZ completion, a rechallenge could be proposed. After second progression, the clinical advantage of subsequent lines of chemotherapy still needs to be clarified. However, based on performance status, patients' preference, and disease behavior, a third-line treatment could be considered. Available treatments include nitrosoureas, bevacizumab, or carboplatin plus etoposide. However, more effective therapeutic options are needed.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioblastoma/pathology , Glioblastoma/therapy , Brain Neoplasms/mortality , Combined Modality Therapy/methods , Glioblastoma/mortality , Humans , Randomized Controlled Trials as Topic , Recurrence , Retreatment , Treatment Outcome
11.
Future Oncol ; 12(8): 1039-44, 2016.
Article in English | MEDLINE | ID: mdl-26880307

ABSTRACT

BACKGROUND: The most appropriate management of recurrent glioblastoma is still controversial. In particular, the role of surgery at recurrence remains uncertain. PATIENTS & METHODS: From our Institutional data warehouse we analyzed 270 consecutive patients who received second surgery for recurrent glioblastoma, to assess survival after second surgery, and to evaluate prognostic factors. RESULTS: Complete resection was found in 128 (47.4%) and partial resection in 142 patients (52.6%). Median survival from second surgery was 11.4 months (95% CI: 10.0-12.7). Multivariate analysis showed that age (p = 0.001), MGMT methylation (p = 0.021) and extent of surgery (p < 0.001) are associated with better survival. CONCLUSION: A complete resection should be the goal for second resection and younger age and MGMT methylation status might be considered in the selection of patients.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/surgery , Glioblastoma/pathology , Glioblastoma/surgery , Adolescent , Adult , Aged , Biomarkers, Tumor , Brain Neoplasms/mortality , Combined Modality Therapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Disease Management , Female , Glioblastoma/mortality , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Retreatment , Tomography, X-Ray Computed , Treatment Outcome , Tumor Suppressor Proteins/genetics , Young Adult
12.
Oncologist ; 20(2): 166-75, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25568148

ABSTRACT

Bevacizumab, currently an option for treatment of different types of tumors including glioblastoma, has a peculiar toxicity profile related to its antiangiogenic effect. Because some bevacizumab-related adverse events can be life threatening, it is important to identify risk factors and to establish treatment protocols to minimize treatment-related morbidity and mortality. In glioblastoma patients, the risk of developing certain side effects, such as gastrointestinal perforation, venous thromboembolism, and intracranial hemorrhages, is slightly higher than in patients treated with bevacizumab for other tumor types. We performed a systematic review of the side effects of bevacizumab and their incidence, causal mechanisms, and available treatments. Finally, we identified risk factors and proposed preventive and therapeutic measures for these adverse events.


Subject(s)
Bevacizumab/adverse effects , Glioblastoma/drug therapy , Venous Thromboembolism/pathology , Angiogenesis Inhibitors , Bevacizumab/administration & dosage , Disease Management , Glioblastoma/pathology , Humans , Venous Thromboembolism/chemically induced
13.
J Neurooncol ; 121(2): 399-404, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25366365

ABSTRACT

The optimal end point for phase II studies for recurrent glioblastoma (GBM) is unclear and a matter of debate. Moreover, data about post-progression survival (PPS) after the first disease progression in GBM patients treated according to EORTC 26981/22981/NCIC CE.3 trial are limited. The aim of this study was to evaluate the PPS in GBM patients. The analysis was made with a database on 1,006 GBM patients followed prospectively between 06/2005 and 06/2010. Eligibility criteria for the study were: age ≥ 18 years; PS: 0-2; chemotherapy given at disease progression after RT/TMZ. 232 patients (mean age 52 years, range 18-77 years) were enrolled. The median PFS following second line chemotherapy (PFS2) was 2.5 months (95 % CI 2.1-2.9) and the rate of patients free of progression at 6 months (PFS2-6 mo), was 21.6 % (95 % CI 16.3-26.9 %). The median PPS was 8.6 months (95 % CI 7.4-9.8), PPS rates were: PPS-6: 66 % (95 % CI 60.3-72.9 %), PPS-9: 48.2 % (95 % CI 41.5-54.9 %) and PPS-12: 31.7 % (95 % CI 25.2-38.2 %). PPS in unselected patients treated with alkylating agents is about 8 months. PPS rates could be of interest as an end point in future studies in recurrent GBM.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Outcome Assessment, Health Care/methods , Adolescent , Adult , Aged , Brain Neoplasms/genetics , Combined Modality Therapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Databases, Factual , Disease Progression , Disease-Free Survival , Endpoint Determination , Female , Glioblastoma/genetics , Humans , Male , Middle Aged , Prospective Studies , Tumor Suppressor Proteins/genetics , Young Adult
14.
Future Oncol ; 10(8): 1417-25, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25052752

ABSTRACT

Angiogenesis is a key process for tumoral growth, which has become a main target for anticancer treatments. A wide number of agents targeting both VEGF and its receptor have recently become standard treatments for different tumor types. Unfortunately, most of the tumors become resistant to these agents after few months of treatment. Different mechanisms of resistance to antiangiogenic drugs have been proposed and investigated; some agents demonstrated to be able to restore sensitivity to antiangiogenic drugs by blocking pathways or molecules involved in the resistance in preclinical models. Biomarkers for the prediction of response or resistance to antiangiogenic agents are under evaluation.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Biomarkers , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Treatment Outcome , Vascular Endothelial Growth Factor A/antagonists & inhibitors
15.
Monaldi Arch Chest Dis ; 82(2): 68-74, 2014 Jun.
Article in Italian | MEDLINE | ID: mdl-25845089

ABSTRACT

Life expectancy in patients affected by cancer has recently increased because of early diagnosis and actual therapies. In recent years, Oncology and Cardiology developed a tight relationship because of common risk factors (i.e., obesity, smoking, alcool intake, etc...), and for preventing the prothrombotic status due to cancer and the potential cardiotoxicity of chemotherapy. Cardiotoxicity incidence is reported from 1% up to 70% in retrospective analyses of drug protocols, mainly representing by left ventricular dysfunction (both reversible or irreversible), but also by arrhythmias, hypertension, atrioventricular block, coronary spasm, and arterial or venous thromboembolism. The early detection of the chemoterapy induced cardiotoxicity is now mandatory and can be obtained through a proper patients selection for different treatments and a strict monitoring during the follow-up period. The role of biomarkers of early cardiac damage, mainly, troponin I and brain natriuretic peptide-BNP, has been recently challenged, and algorithms are currently available. In the present paper, we propose how to perform a cardiological evaluation of patients undergoing chemotherapy tailored by the known adverse effects of the drugs.


Subject(s)
Cardiotoxicity , Heart Diseases/diagnosis , Comorbidity , Heart/drug effects , Heart Diseases/chemically induced , Heart Diseases/epidemiology , Humans , Neoplasms/drug therapy , Neoplasms/epidemiology
16.
CNS Oncol ; 13(1): 2357532, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38873961

ABSTRACT

Aim: Glioneuronal and neuronal tumors are rare primary central nervous system malignancies with heterogeneous features. Due to the rarity of these malignancies diagnosis and treatment remains a clinical challenge. Methods: Here we performed a narrative review aimed to investigate the principal issues concerning the diagnosis, pathology, and clinical management of glioneuronal tumors. Results: Diagnostic criteria have been recently overturned thanks to a better characterization on a histological and molecular biology level. The study of genomic alterations occurring within these tumors has allowed us to identify potential therapeutic targets including BRAF, FGFR, and PDGFRA. Conclusion: Techniques allowing molecular sequencing DNA methylation assessment of the disease are essential diagnostic tools. Targeting agents should be included in the therapeutic armamentarium after loco-regional treatment failure.


[Box: see text].


Subject(s)
Brain Neoplasms , Humans , Young Adult , Brain Neoplasms/therapy , Brain Neoplasms/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Central Nervous System Neoplasms/therapy , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/drug therapy , Glioma/therapy , Glioma/genetics , Glioma/diagnosis , Glioma/pathology
17.
Pathol Res Pract ; 262: 155516, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39163733

ABSTRACT

BACKGROUND: Mutations of the TP53 oncosuppressor gene are frequent events in patients with malignant tumors including IDH-wildtype GBM (GBM IDH wt). However, the effective impact of TP53 mutations on prognosis has been poorly evaluated. METHODS: We performed a retrospective study investigating the impact of TP53 mutations on patients with GBM IDH wt. Only patients with PS=0-1, treated with temozolomide concurrent with and adjuvant to radiotherapy, and younger than 70 years assessed with NGS were included in the analysis. RESULTS: 97 GBM IDH wt have been selected. The median follow-up was 34.5 months (95 %CI, 30.6 - NA). Overall, 20 patients (19.4 %) presented a TP53 mutation. There were no significant differences in terms of TERT mutation (75 % vs 79.2 %) between TP53 mutated and TP53 wild-type (wt) patients. We detected 6 TP53 mutations not previously described within GBM IDH wt patients. The overall survival (OS) did not significantly differ between TP53 mutated and wt patients (HR 0.69, 95 %CI 0.37-1.27, p = 0.24). Considering only patients with an OS longer than 36 months (n = 10), the presence of a TP53 mutation was significantly associated with prolonged survival (45.6 months vs Not Reached, p = 0.037). CONCLUSION: The presence of a TP53 mutation does not appear to be correlated with overall survival in this patient cohort. While there is an association with survival for patients with an OS of 36 months or longer, the number of patients is low and there is no available evidence correlating TP53 mutations to long-term survivors.

18.
Front Oncol ; 13: 1206983, 2023.
Article in English | MEDLINE | ID: mdl-37397356

ABSTRACT

Chimeric antigen receptor (CAR-T) therapy has marked a paradigm shift in the treatment of hematological malignancies and represent a promising growing field also in solid tumors. Neurotoxicity is a well-recognized common complication of CAR-T therapy and is at the forefront of concerns for CAR-based immunotherapy widespread adoption, as it necessitates a cautious approach. The non-specific targeting of the CAR-T cells against normal tissues (on-target off-tumor toxicities) can be life-threatening; likewise, immune-mediate neurological symptoms related to CAR-T cell induced inflammation in central nervous system (CNS) must be precociously identified and recognized and possibly distinguished from non-specific symptoms deriving from the tumor itself. The mechanisms leading to ICANS (Immune effector Cell-Associated Neurotoxicity Syndrome) remain largely unknown, even if blood-brain barrier (BBB) impairment, increased levels of cytokines, as well as endothelial activation are supposed to be involved in neurotoxicity development. Glucocorticoids, anti-IL-6, anti-IL-1 agents and supportive care are frequently used to manage patients with neurotoxicity, but clear therapeutic indications, supported by high-quality evidence do not yet exist. Since CAR-T cells are under investigation in CNS tumors, including glioblastoma (GBM), understanding of the full neurotoxicity profile in brain tumors and expanding strategies aimed at limiting adverse events become imperative. Education of physicians for assessing individualized risk and providing optimal management of neurotoxicity is crucial to make CAR-T therapies safer and adoptable in clinical practice also in brain tumors.

19.
Cancers (Basel) ; 15(12)2023 Jun 20.
Article in English | MEDLINE | ID: mdl-37370860

ABSTRACT

The lack of significant improvement in the prognosis of patients with GB over the last decades highlights the need for innovative treatments aimed at fighting this malignancy and increasing survival outcomes. The results of the phase III clinical trial of DCVax-L (autologous tumor lysate-loaded dendritic cell vaccination), which has been shown to increase both median survival and long-term survival in newly diagnosed and relapsed glioblastoma, have been enthusiastically received by the scientific community. However, this study deserves some reflections regarding methodological issues related to the primary endpoint change, the long accrual period, and the suboptimal validity of the external control population used as the comparison arm.

20.
Expert Opin Drug Discov ; 18(3): 269-286, 2023 03.
Article in English | MEDLINE | ID: mdl-36718723

ABSTRACT

INTRODUCTION: Glioblastoma (GBM) is the most common primary brain tumor in adults. GBM treatment options have been the same for the past 30 years and have only modestly extended survival, despite aggressive multimodal treatments. The progressively better knowledge of GBM biology and a comprehensive analysis of its genomic profile have elucidated GBM heterogeneity, contributing to a more effective molecular classification and to the development of innovative targeted therapeutic approaches. AREAS COVERED: This article reports all the noteworthy innovations for immunotherapy and targeted therapy, providing insights into the current advances in trial designs, including combination therapies with immuno-oncology agents and target combinations. EXPERT OPINION: GBM molecular heterogeneity and brain anatomical characteristics critically restrain drug effectiveness. Nevertheless, stimulating insights for future research and drug development come from innovative treatment strategies for GBM, such as multi-specific 'off-the-shelf' CAR-T therapy, oncolytic viral therapy and autologous dendritic cell vaccination. Disappointing results from targeted therapies-clinical trials are mainly due to complex interferences between signaling pathways and biological processes leading to drug resistance: hence, it is imperative in the future to develop combinatorial approaches and multimodal therapies.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Virotherapy , Adult , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Immunotherapy/methods , Combined Modality Therapy , Oncolytic Virotherapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL