Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.119
Filter
1.
Rev Med Virol ; 34(5): e2584, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39304923

ABSTRACT

Neurotropic viruses have been implicated in altering the central nervous system microenvironment and promoting brain metastasis of breast cancer through complex interactions involving viral entry mechanisms, modulation of the blood-brain barrier, immune evasion, and alteration of the tumour microenvironment. This narrative review explores the molecular mechanisms by which neurotropic viruses such as Herpes Simplex Virus, Human Immunodeficiency Virus, Japanese Encephalitis Virus, and Rabies Virus facilitate brain metastasis, focusing on their ability to disrupt blood-brain barrier integrity, modulate immune responses, and create a permissive environment for metastatic cell survival and growth within the central nervous system. Current therapeutic implications and challenges in targeting neurotropic viruses to prevent or treat brain metastasis are discussed, highlighting the need for innovative strategies and multidisciplinary approaches in virology, oncology, and immunology.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Humans , Breast Neoplasms/pathology , Breast Neoplasms/virology , Breast Neoplasms/therapy , Brain Neoplasms/secondary , Brain Neoplasms/virology , Brain Neoplasms/therapy , Female , Blood-Brain Barrier/virology , Animals , Tumor Microenvironment , Rabies virus/physiology , Rabies virus/pathogenicity , Rabies virus/immunology , Simplexvirus/physiology
2.
Biomed Pharmacother ; 179: 117413, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260325

ABSTRACT

The epidermal growth factor receptor (EGFR) plays a pivotal role in tumor progression and is an essential therapeutic target for treating malignant gliomas. Small interfering RNA (siRNA) has the potential to selectively degrade EGFR mRNA, yet its clinical utilization is impeded by various challenges, such as inefficient targeting and limited escape from lysosomes. Our research introduces polyethylene glycol (PEG) and endoplasmic reticulum membrane-coated siEGFR nanoplexes (PEhCv/siEGFR NPs) as an innovative approach to brain glioma therapy by overcoming several obstacles: 1) Tumor-derived endoplasmic reticulum membrane modifications provide a homing effect, facilitating targeted accumulation and cellular uptake; 2) Endoplasmic reticulum membrane proteins mediate a non-degradable "endosome-Golgi-endoplasmic reticulum" transport pathway, circumventing lysosomal degradation. These nanoplexes demonstrated significantly enhanced siEGFR gene silencing in both in vitro and in vivo U87 glioma models. The findings of this study pave the way for the advanced design and effective application of nucleic acid-based therapeutic nanocarriers.


Subject(s)
Brain Neoplasms , Endoplasmic Reticulum , ErbB Receptors , Glioma , RNA, Small Interfering , Glioma/pathology , Glioma/therapy , Glioma/drug therapy , Glioma/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/metabolism , Humans , ErbB Receptors/metabolism , Cell Line, Tumor , Animals , Endoplasmic Reticulum/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , Mice, Nude , Polyethylene Glycols/chemistry , Nanoparticles/chemistry , Gene Silencing , Mice , Biological Transport , Biomimetics/methods , Mice, Inbred BALB C
3.
Clin Med (Lond) ; 24(5): 100240, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39233205

ABSTRACT

Primary brain tumours are rare but carry a significant morbidity and mortality burden. Malignant gliomas are the most common subtype and their incidence is increasing within our ageing population. The diagnosis and treatment of gliomas involves substantial interplay between multiple specialties, including general medical physicians, radiologists, pathologists, surgeons, oncologists and allied health professionals. At any point along this pathway, patients can present to acute medicine with complications of their cancer or anti-cancer therapy. Increasing the awareness of malignant gliomas among general physicians is paramount to delivering prompt radiological and histopathological diagnoses, facilitating access to earlier and individualised treatment options and allows for effective recognition and management of anticipated complications. This article discusses evidence-based real-world practice for malignant gliomas, encompassing patient presentation, diagnostic pathways, treatments and their complications, and prognosis to guide management outside of specialist centres.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/therapy , Glioma/diagnosis , Brain Neoplasms/therapy , Brain Neoplasms/diagnosis , Adult
4.
Theranostics ; 14(14): 5725-5742, 2024.
Article in English | MEDLINE | ID: mdl-39310093

ABSTRACT

Brain tumors pose formidable challenges in oncology due to the intricate biology and the scarcity of effective treatment modalities. The emergence of immunotherapy has opened new avenues for innovative therapeutic strategies. Chimeric antigen receptor, originally investigated in T cell-based therapy, has now expanded to encompass macrophages, presenting a compelling avenue for augmenting anti-tumor immune surveillance. This emerging frontier holds promise for advancing the repertoire of therapeutic options against brain tumors, offering potential breakthroughs in combating the formidable malignancies of the central nervous system. Tumor-associated macrophages constitute a substantial portion, ranging from 30% to 50%, of the tumor tissue and exhibit tumor-promoting phenotypes within the immune-compromised microenvironment. Constructing CAR-macrophages can effectively repolarize M2-type macrophages towards an M1-type phenotype, thereby eliciting potent anti-tumor effects. CAR-macrophages can recruit T cells to the brain tumor site, thereby orchestrating a remodeling of the immune niche to effectively inhibit tumor growth. In this review, we explore the potential limitations as well as strategies for optimizing CAR-M therapy, offering insights into the future direction of this innovative therapeutic approach.


Subject(s)
Brain Neoplasms , Immunotherapy, Adoptive , Macrophages , Receptors, Chimeric Antigen , Tumor Microenvironment , Humans , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Animals , Tumor Microenvironment/immunology , Immunotherapy, Adoptive/methods , Macrophages/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , T-Lymphocytes/immunology , Immunotherapy/methods
5.
Cells ; 13(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273050

ABSTRACT

Immunotherapy represents a transformative shift in cancer treatment. Among myriad immune-based approaches, chimeric antigen receptor (CAR) T-cell therapy has shown promising results in treating hematological malignancies. Despite aggressive treatment options, the prognosis for patients with malignant brain tumors remains poor. Research leveraging CAR T-cell therapy for brain tumors has surged in recent years. Pre-clinical models are crucial in evaluating the safety and efficacy of these therapies before they advance to clinical trials. However, current models recapitulate the human tumor environment to varying degrees. Novel in vitro and in vivo techniques offer the opportunity to validate CAR T-cell therapies but also have limitations. By evaluating the strengths and weaknesses of various pre-clinical glioma models, this review aims to provide a roadmap for the development and pre-clinical testing of CAR T-cell therapies for brain tumors.


Subject(s)
Brain Neoplasms , Glioma , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Glioma/therapy , Glioma/immunology , Glioma/pathology , Humans , Animals , Immunotherapy, Adoptive/methods , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Disease Models, Animal , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology
6.
Cells ; 13(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273062

ABSTRACT

Primary central nervous system tumors are the most frequent solid tumors in children, accounting for over 40% of all childhood brain tumor deaths, specifically high-grade gliomas. Compared with pediatric low-grade gliomas (pLGGs), pediatric high-grade gliomas (pHGGs) have an abysmal survival rate. The WHO CNS classification identifies four subtypes of pHGGs, including Grade 4 Diffuse midline glioma H3K27-altered, Grade 4 Diffuse hemispheric gliomas H3-G34-mutant, Grade 4 pediatric-type high-grade glioma H3-wildtype and IDH-wildtype, and infant-type hemispheric gliomas. In recent years, we have seen promising advancements in treatment strategies for pediatric high-grade gliomas, including immunotherapy, CAR-T cell therapy, and vaccine approaches, which are currently undergoing clinical trials. These therapies are underscored by the integration of molecular features that further stratify HGG subtypes. Herein, we will discuss the molecular features of pediatric high-grade gliomas and the evolving landscape for treating these challenging tumors.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/pathology , Glioma/therapy , Glioma/genetics , Child , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Brain Neoplasms/genetics , Neoplasm Grading , Immunotherapy/methods
7.
Eur J Pharm Biopharm ; 203: 114460, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218361

ABSTRACT

Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.


Subject(s)
Antineoplastic Agents , Blood-Brain Barrier , Brain Neoplasms , Drug Delivery Systems , Exosomes , Glioblastoma , Exosomes/metabolism , Glioblastoma/drug therapy , Glioblastoma/therapy , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Drug Delivery Systems/methods , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage
8.
Front Immunol ; 15: 1399613, 2024.
Article in English | MEDLINE | ID: mdl-39257576

ABSTRACT

Background: Cerebral radiation necrosis (RN), a severe complication of stereotactic radiotherapy (SRT), has been shown to significantly decrease patient survival time and quality of life. The purpose of this study was to analyze whether bevacizumab can prevent or reduce the occurrence of SRT-induced cerebral RN in non-small cell lung cancer (NSCLC) patients with brain metastases. Materials and methods: We retrospectively reviewed the clinical records of NSCLC patients with brain metastases from March 2013 to June 2023 who were treated with SRT. Patients were divided into two groups: those in the bevacizumab group received SRT with four cycles of bevacizumab, and patients in the control group received SRT only. Inverse probability of treatment weighting (IPTW) was performed based on a multinomial propensity score model to balance the baseline characteristics. The chi-square test was used. A Cox model was used to evaluate overall survival (OS). Results: A total of 80 patients were enrolled, namely, 28 patients in the bevacizumab group and 52 patients in the control group. The possibility of developing cerebral RN and/or symptomatic edema (RN/SE) was significantly decreased in patients treated with bevacizumab compared to those who did not receive bevacizumab before IPTW (p=0.036) and after IPTW (p=0.015) according to chi-square analysis. The IPTW-adjusted median OS was 47.7 months (95% CI 27.4-80.8) for patients in the bevacizumab group and 44.1 months (95% CI 36.7-68.0) (p=0.364) for patients in the control group. Conclusion: The application of bevacizumab concurrent with SRT may prevent or reduce the occurrence of cerebral RN in NSCLC patients with brain metastases.


Subject(s)
Bevacizumab , Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Necrosis , Radiation Injuries , Radiosurgery , Humans , Carcinoma, Non-Small-Cell Lung/therapy , Bevacizumab/therapeutic use , Bevacizumab/administration & dosage , Male , Female , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Middle Aged , Radiosurgery/adverse effects , Aged , Retrospective Studies , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Antineoplastic Agents, Immunological/therapeutic use
9.
Nat Genet ; 56(9): 1903-1913, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39223316

ABSTRACT

Inhibiting epigenetic modulators can transcriptionally reactivate transposable elements (TEs). These TE transcripts often generate unique peptides that can serve as immunogenic antigens for immunotherapy. Here, we ask whether TEs activated by epigenetic therapy could appreciably increase the antigen repertoire in glioblastoma, an aggressive brain cancer with low mutation and neoantigen burden. We treated patient-derived primary glioblastoma stem cell lines, an astrocyte cell line and primary fibroblast cell lines with epigenetic drugs, and identified treatment-induced, TE-derived transcripts that are preferentially expressed in cancer cells. We verified that these transcripts could produce human leukocyte antigen class I-presented antigens using liquid chromatography with tandem mass spectrometry pulldown experiments. Importantly, many TEs were also transcribed, even in proliferating nontumor cell lines, after epigenetic therapy, which suggests that targeted strategies like CRISPR-mediated activation could minimize potential side effects of activating unwanted genomic regions. The results highlight both the need for caution and the promise of future translational efforts in harnessing treatment-induced TE-derived antigens for targeted immunotherapy.


Subject(s)
Antigens, Neoplasm , Brain Neoplasms , DNA Transposable Elements , Epigenesis, Genetic , Glioblastoma , Transcription, Genetic , Glioblastoma/genetics , Glioblastoma/therapy , Glioblastoma/immunology , Humans , DNA Transposable Elements/genetics , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Gene Expression Regulation, Neoplastic , Immunotherapy/methods
10.
Neurosurg Rev ; 47(1): 541, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39231832

ABSTRACT

Despite unprecedented survival in patients with glioblastoma (GB), the aggressive primary brain cancer remains largely incurable and its mechanisms of treatment resistance have gained particular attention. The cytokine interleukin 6 (IL-6) and its receptor weave through the hallmarks of malignant gliomas and may represent a key vulnerability to GB. Known for activating the STAT3 pathway in autocrine fashion, IL-6 is amplified in GB and has been recognized as a negative biomarker for GB prognosis, rendering it a putative target of novel GB therapies. While it has been recognized as a biologically active component of GB for three decades only with concurrent advances in understanding of complementary immunotherapy has the concept of targeting IL-6 for a human clinical trial gained scientific footing.


Subject(s)
Brain Neoplasms , Glioblastoma , Interleukin-6 , Glioblastoma/therapy , Humans , Interleukin-6/metabolism , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Drug Resistance, Neoplasm , Receptors, Interleukin-6 , STAT3 Transcription Factor/metabolism , Immunotherapy/methods
11.
Cancer Biomark ; 41(1): 1-24, 2024.
Article in English | MEDLINE | ID: mdl-39240627

ABSTRACT

Glioma is the most common primary tumor of the central nervous system (CNS). Glioblastoma (GBM) is incurable with current treatment strategies. Additionally, the treatment of recurrent GBM (rGBM) is often referred to as terminal treatment, necessitating hospice-level care and management. The presence of the blood-brain barrier (BBB) gives GBM a more challenging or "cold" tumor microenvironment (TME) than that of other cancers and gloma stem cells (GSCs) play an important role in the TME remodeling, occurrence, development and recurrence of giloma. In this review, our primary focus will be on discussing the following topics: niche-associated GSCs and macrophages, new theories regarding GSC and TME involving pyroptosis and ferroptosis in GBM, metabolic adaptations of GSCs, the influence of the cold environment in GBM on immunotherapy, potential strategies to transform the cold GBM TME into a hot one, and the advancement of GBM immunotherapy and GBM models.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplastic Stem Cells , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Immunotherapy/methods , Animals , Blood-Brain Barrier/metabolism
12.
J Neuroinflammation ; 21(1): 226, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285276

ABSTRACT

Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates. Tumor-associated neutrophils (TANs) are essential components of the tumor microenvironment (TME) in glioma and play a crucial role in glioma cell proliferation, invasion and proneural-mesenchymal transition. Besides the interactions between TANs and tumor cells, the multi-dimensional crosstalk between TANs and other components within TME have been reported to participate in glioma progression. More importantly, several therapies targeting TANs have been developed and relevant preclinical and clinical studies have been conducted in cancer therapy. In this review, we introduce the origin of TANs and the functions of TANs in malignant behaviors of glioma, highlighting the microenvironmental regulation of TANs. Moreover, we focus on summarizing the TANs-targeted methods in cancer therapy, aiming to provide insights into the mechanisms and therapeutic opportunities of TANs in the malignant glioma microenvironment.


Subject(s)
Brain Neoplasms , Glioma , Neutrophils , Tumor Microenvironment , Humans , Tumor Microenvironment/physiology , Tumor Microenvironment/immunology , Glioma/pathology , Glioma/therapy , Glioma/immunology , Neutrophils/pathology , Neutrophils/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Animals
13.
Cancer Res Commun ; 4(9): 2514-2524, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39240078

ABSTRACT

Cancer immunotherapy using immune checkpoint inhibitors and its combination with other anticancer therapies has emerged as a new standard of care because of the encouraging therapeutic effects in various solid cancers. Nonetheless, glioblastoma and pancreatic cancer remain resistant to immunotherapy and represent intractable cancers with the poorest prognosis. We investigated the therapeutic effects of next-generation chimeric antigen receptor (CAR) T cells producing IL7 and chemokine (C-C motif) ligand 19 (CCL19; referred to as 7 × 19 CAR-T) in these intractable cancers. Cytotoxic activities and therapeutic effects of 7 × 19 CAR-T were evaluated in vitro and in vivo, in a model using EGFR variant III (EGFRvIII)-positive glioblastoma and anti-EGFRvIII CAR-T generated from healthy donor peripheral blood mononuclear cells (PBMC), or a model using HER2-positive pancreatic cancer organoids and anti-HER2 CAR-T generated from the same patient's PBMC. Anti-EGFRvIII 7 × 19 CAR-T exhibited cytotoxic activity specific to EGFRvIII-positive tumor, induced complete rejection of glioblastoma with massive T-cell infiltration and tumor cell death in the tumor tissues, and consequently prolonged mouse survival. Anti-HER2 7 × 19 CAR-T demonstrated a potent cytotoxic activity against autologous HER2-positive pancreatic cancer organoids and induced complete rejection of autologous tumor along with prolonged mouse survival. Our results suggest that 7 × 19 CAR-T could become a therapeutic option for glioblastoma and pancreatic cancer. To the best of our knowledge, this is the first study to demonstrate the therapeutic efficacy of next-generation CAR-T in an autologous model using patient-derived tumor organoids and CAR-T generated from the same patient's PBMC, in which unwanted allogeneic immune responses are fully excluded. SIGNIFICANCE: Despite the clinical development of CAR T-cell therapy, its efficacy in solid cancers has yet to be established. This study explored the therapeutic potential and immunologic mechanisms of IL7/CCL19-producing CAR-T therapy in preclinical solid cancer models of glioblastoma and pancreatic cancer. We found that IL7/CCL19-producing CAR-T cells generated from the patient's PBMC showed potent therapeutic effects against the solid cancer model established by inoculating organoids from the autologous tumor tissue.


Subject(s)
Glioblastoma , Immunotherapy, Adoptive , Interleukin-7 , Pancreatic Neoplasms , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Glioblastoma/therapy , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Mice , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Interleukin-7/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , ErbB Receptors/immunology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Female
14.
Neurosurg Rev ; 47(1): 673, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39316196

ABSTRACT

The treatment of brain tumors is significantly hindered by the Blood-Brain Barrier (BBB), a selective barrier that restricts the passage of therapeutic agents to the brain. Recent advancements in BBB-targeting therapies offer promising strategies to overcome this challenge, providing new avenues for the effective treatment of brain cancer. This article reviews innovative approaches, including Convection-Enhanced Delivery (CED) and RNA-based therapeutics, which enhance drug delivery directly to tumor sites, bypassing the BBB and reducing systemic toxicity. Additionally, the use of theranostic nanoparticles and CRISPR-Cas9 gene editing presents novel opportunities for real-time monitoring and precision-targeted therapy, respectively. Techniques such as magnetic nanoparticles, intranasal drug administration, and focused ultrasound with microbubbles are also being refined to improve drug penetration across the BBB. Furthermore, peptide-based delivery systems and small molecules designed to mimic endogenous transport pathways are accelerating the discovery of more effective therapies. The exploration of combination therapies that synergize BBB-penetrant drugs with conventional chemotherapeutic agents or immunotherapies holds the potential to enhance treatment efficacy and patient outcomes. Continued research and interdisciplinary collaboration are essential to develop predictive models, personalized treatment strategies, and alternative delivery methods that ensure the long-term safety and effectiveness of these novel therapies. Advancements in BBB-targeting therapeutics are poised to transform the landscape of brain cancer treatment, offering renewed hope for improved survival rates and quality of life for patients.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Drug Delivery Systems , Precision Medicine , Humans , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Precision Medicine/methods , Drug Delivery Systems/methods , Treatment Outcome , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage
15.
BMC Cancer ; 24(1): 1146, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272048

ABSTRACT

BACKGROUND: The Multidisciplinary Tumor Board (MTB) is a collaborative platform involving specialists in oncology, surgery, radiology, pathology, and radiotherapy, and aims to optimize diagnostics and treatments. Despite MTB's widespread benefits, limited literature addresses its application in pediatric neuro-oncology. After a literature revision on pediatric neuro-oncology MTB, our study describes our institute's pediatric neuro-oncology MTB, focuses on evaluating its impact and the neuroradiologist's role in patient-centric approaches, considering recent genetic insights into pediatric brain tumors. MATERIALS AND METHODS: Literature Review concerning pediatric neuro-oncology MTB from January 2002 to June 2024. CLINICAL DATA: retrospective study of all patient files presented in the pediatric neuro-oncology MTB (pnMTB) between 2019 and 2022. Statistical analysis was mainly carried out by directly comparing the absolute or relative values of the respective parameters examined; qualitative variables compared mainly with the chi-square test, quantitative variables mainly with the t-test. RESULTS: Literature Review: 7 papers encompass a multidisciplinary approach for the pediatric CNS tumors. CLINICAL DATA: A total of 236 discussions were analyzed representing 107 patients. Median age was 14,3 years (range: 6 months - 17 years). The requests for case evaluations primarily came from the pediatric oncologists (83%) and neurosurgeons (14.8%), and they were mainly addressed to the neuroradiologists (70.3%). Proposals during pnMTB mainly involved imaging follow-up (47.8%) and management with chemotherapy (34.7%). Changes in patient treatment (CPT) occurred in 115 cases, and pediatric neuroradiologist intervention contributed to 72.4% of these changes. CONCLUSION: Thanks to their multidisciplinarity, high number of cases discussed, and usual respect for their proposals, the pnMTB has made it possible to improve the coordination among specialties involved in patient management, to apply the recent protocols, and to exchange knowledge among teams managing pediatric CNS tumors.


Subject(s)
Central Nervous System Neoplasms , Humans , Child , Adolescent , Retrospective Studies , Child, Preschool , Female , Male , Infant , Central Nervous System Neoplasms/therapy , Patient Care Team , Medical Oncology/methods , Brain Neoplasms/therapy
16.
Magn Reson Imaging Clin N Am ; 32(4): 673-679, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39322356

ABSTRACT

Malignant gliomas (MGs) are the most common primary brain tumors in adults. Despite recent advances in understanding the biology and potential therapeutic vulnerabilities of MGs, treatment options remain limited as the delivery of drugs is often impeded by the blood-brain barrier (BBB), and safe, complete surgical resection may not always be possible, especially for deep-seated tumors. In this review, the authors highlight emerging applications for MR imaging-guided focused ultrasound (MRgFUS) as a noninvasive treatment modality for MGs. Specifically, the authors discuss MRgFUS's potential role in direct tumor cell killing, opening the BBB, and modulating antitumor immunity.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/diagnostic imaging , Glioma/therapy , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Magnetic Resonance Imaging, Interventional/methods , Magnetic Resonance Imaging/methods , High-Intensity Focused Ultrasound Ablation/methods , Brain/diagnostic imaging
17.
Front Immunol ; 15: 1447879, 2024.
Article in English | MEDLINE | ID: mdl-39324140

ABSTRACT

Background: This study aimed to identify prognostic factors for survival and develop a prognostic nomogram to predict the survival probability of elderly patients with lower-grade gliomas (LGGs). Methods: Elderly patients with histologically confirmed LGG were recruited from the Surveillance, Epidemiology, and End Results (SEER) database. These individuals were randomly allocated to the training and validation cohorts at a 2:1 ratio. First, Kaplan-Meier survival analysis and subgroup analysis were performed. Second, variable screening of all 13 variables and a comparison of predictive models based on full Cox regression and LASSO-Cox regression analyses were performed, and the key variables in the optimal model were selected to construct prognostic nomograms for OS and CSS. Finally, a risk stratification system and a web-based dynamic nomogram were constructed. Results: A total of 2307 elderly patients included 1220 males and 1087 females, with a median age of 72 years and a mean age of 73.30 ± 6.22 years. Among them, 520 patients (22.5%) had Grade 2 gliomas, and 1787 (77.5%) had Grade 3 gliomas. Multivariate Cox regression analysis revealed four independent prognostic factors (age, WHO grade, surgery, and chemotherapy) that were used to construct the full Cox model. In addition, LASSO-Cox regression analysis revealed five prognostic factors (age, WHO grade, surgery, radiotherapy, and chemotherapy), and a LASSO model was constructed. A comparison of the two models revealed that the LASSO model with five variables had better predictive performance than the full Cox model with four variables. Ultimately, five key variables based on LASSO-Cox regression were utilized to develop prognostic nomograms for predicting the 1-, 2-, and 5-year OS and CSS rates. The nomograms exhibited relatively good predictive ability and clinical utility. Moreover, the risk stratification system based on the nomograms effectively divided patients into low-risk and high-risk subgroups. Conclusion: Variable screening based on LASSO-Cox regression was used to determine the optimal prediction model in this study. Prognostic nomograms could serve as practical tools for predicting survival probabilities, categorizing these patients into different mortality risk subgroups, and developing personalized decision-making strategies for elderly patients with LGGs. Moreover, the web-based dynamic nomogram could facilitate its use in the clinic.


Subject(s)
Brain Neoplasms , Glioma , Neoplasm Grading , Nomograms , Proportional Hazards Models , Humans , Female , Male , Aged , Glioma/mortality , Glioma/therapy , Glioma/diagnosis , Prognosis , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Brain Neoplasms/diagnosis , SEER Program , Cohort Studies , Aged, 80 and over , Kaplan-Meier Estimate
18.
Chin Clin Oncol ; 13(Suppl 1): AB011, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39295329

ABSTRACT

BACKGROUND: World Health Organization (WHO) grade 4 astrocytoma is a high-grade brain tumour in adults. Tumour treating fields (TTF) has been shown to improve overall survival (OS). Few studies have explored quality-of-life (QoL) in these patients. This study aims to assess the QoL of TTF patients and OS. METHODS: This was a prospective multicenter study of adult patients diagnosed with WHO grade 4 astrocytoma from 2018 to 2023 receiving TTF for >1 month after completing standard therapy. A propensity-score matched comparison with a 1:2 ratio with historical control was performed for OS analysis. The patients completed European Organisation for Research and Treatment of Cancer (EORTC) QLQ-30/BN20 questionnaires before TTF and at 3-month interval. Primary outcomes included OS, and secondary outcomes included QoL and TTF-associated adverse effects at 3 months. RESULTS: A total of 141 patients were reviewed, with TTF patients (n=47, 33%) and propensity-score matched controls (n=94). The mean duration of TTF use was 10±8 months. The mean age of the TTF group was 54±13 years, and for the control group 52±13 years. Sixty percent (n=28) were male, similar to the control group with 71% (n=67) (P=0.16). Seventy-two percent of TTF patients had preoperative Karnofsky Performance Scale (KPS) score ≥80, while controls had 70% (P=0.79). Five (11%) TTF patients and 8 (9%) controls were IDH1 mutant (P=0.70). Twenty (43%) TTF patients and 42 (45%) controls were O6-methylguanine-DNA methyltransferase promoter (pMGMT) methylated (P=0.81). Twenty-one (45%) of TTF patients and 55 (59%) of controls had gross total resection (P=0.72). After adjusting for independent predictors for OS, the median OS of the TTF group was 22.4 months [interquartile range (IQR): 18.6-26.5 months], significantly longer than the control group (17.2 months; IQR: 12.1-22.3 months) (log-rank test: P=0.01). Forty-seven TTF patients and 40 control patients completed EORTC questionnaires. There was no difference for EORTC functional and symptom scores between the TTF and control group [P=0.45, analysis of variance (ANOVA)] at 3 months. Thirty-two (67%) of TTF patients reported associated RTOG grade I scalp dermatitis. CONCLUSIONS: TTF for WHO grade 4 astrocytoma patients is an independent predictor for OS. QoL between the groups was similar, and overall QoL over time for TTF patients was not affected. TTF is a novel and effective outpatient treatment with minimal adverse effects.


Subject(s)
Astrocytoma , Propensity Score , Quality of Life , Humans , Male , Astrocytoma/therapy , Astrocytoma/mortality , Female , Prospective Studies , Middle Aged , Adult , Neoplasm Grading , Brain Neoplasms/therapy , Aged , World Health Organization
19.
Chin Clin Oncol ; 13(Suppl 1): AB010, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39295328

ABSTRACT

BACKGROUND: Anaplastic astrocytoma [AA; World Health Organization (WHO) grade III] is a diffusely infiltrative astrocytic brain tumor with anaplasia and represents 3.3% of primary brain tumors. Overall, 5-year median survival can range from 22% to 50%, depending on various prognostic features, including the patient's age, tumor location and genetics, resection, etc. Given the higher grade and increased likelihood of transformation to WHO-grade IV tumors (glioblastomas), these tumors are generally treated aggressively upfront. Headache and seizures are the most common symptoms, occurring in about 50% of the cases. Other symptoms, including memory loss, motor weakness, language deficit, and cognitive and personality changes, occur in 20% of cases. Standard treatment involves surgical resection, radiotherapy, and chemotherapy, but treatment options are greatly limited for progression and recurrence. This paper highlights the case of a 48-year-old male who presents with chronic progressive cephalgia and a new-onset seizure. We review the diagnostic and therapeutic challenges associated with the treatment of AA. CASE DESCRIPTION: We describe a patient who presented with chronic progressive cephalgia, gradual right-sided weakness, an asymmetrical face, slurred speech, and a new-onset focal-to-bilateral seizure. A cranial magnetic resonance imaging revealed a mass in the left frontoparietal region, causing herniation of the cerebri to the right. The patient had a maximal tumor resection, and the histopathology showed tissue sections containing tumors that were infiltrative in the stroma, forming a diffuse pattern consisting of proliferation of oval, round, polygonal, spindle, pleomorphic oval nucleated cells, hyperchromatic, some nucleoli appearing prominent, and cytoplasmaeosinophilic. There were areas of stromal necrosis and mitosis [3/10 high power field (HPF)]. The pathology result was reported with AA. The patient underwent concomitant chemoradiation and followed oral chemotherapy with temozolomid. Subsequent imaging revealed a significant decrease in the tumor's size and a resolution of the compression of the brain parenchyma underneath. The Response Assessment in Neuro-Oncology (RANO) evaluation showed partial responses with good clinical improvement. CONCLUSIONS: The case presented an AA that was responsive to radiotherapy and temozolomid chemotherapy. Despite being rare, knowledge of this malignant tumor type and a multidisciplinary approach to case management are essential to optimizing treatment results.


Subject(s)
Astrocytoma , Humans , Male , Astrocytoma/therapy , Astrocytoma/complications , Astrocytoma/pathology , Middle Aged , Brain Neoplasms/therapy , Brain Neoplasms/complications , Brain Neoplasms/pathology , World Health Organization , Neoplasm Grading
20.
Chin Clin Oncol ; 13(Suppl 1): AB023, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39295341

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is one of the worst prognostic cancers and one of the reasons is that GBM patients rapidly deteriorated and half of them had a Karnofsky performance status (KPS) with 70 or less at the initial presentation. We discuss the significance of early diagnosis and treatment to the clinical outcomes of GBM patients. METHODS: Data of IDH-wildtype GBM patients treated at our institution between 2010 and 2019 were retrospectively reviewed. Patients were classified into early or late diagnosis groups with a threshold of 14 days from initial symptoms. Also, patients were divided into early, intermediate, and late surgery groups with thresholds of 21 and 35 days. Patients were also divided into two groups: neurosurgeon and non-neurosurgeon groups, based on the primary doctor whom patients sought for an initial evaluation. Representative symptoms and patient prognosis were examined. RESULTS: Of 153 patients, 72 and 81 were classified into the early and late diagnosis groups. The proportion of patients with preoperative KPS scores ≥90 was 48.6% and 29.6% in the early and late diagnosis groups (P=0.01). Median overall survival was significantly longer in the early surgery group (n=43) than in the late surgery group (n=86) (28.4 vs. 18.7 months, P=0.006). The neurosurgeon group exhibited a significantly shorter duration from the first hospital visit to the first surgery than the non-neurosurgeon group (18 vs. 29 days, P<0.001). Furthermore, the overall survival of the neurosurgeon group was significantly more prolonged than that of the non-neurosurgeon group (22.9 vs. 14.0 months, P=0.04). CONCLUSIONS: Early diagnosis and surgery within 3 weeks from the initial symptoms are associated with improved patient survival. Early GBM treatment will be beneficial for patients with GBM. A short duration from the first hospital visit to the first surgery is essential in enhancing GBM patient prognosis.


Subject(s)
Glioblastoma , Humans , Glioblastoma/mortality , Glioblastoma/therapy , Female , Male , Middle Aged , Retrospective Studies , Aged , Early Diagnosis , Adult , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Prognosis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL