Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.584
Filtrar
1.
Med Oncol ; 41(11): 250, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39316248

RESUMO

For glioblastoma patients, the efficacy-targeted therapy is limited to date. Most of the molecular therapies previously studied are lacking efficacy in this population. More trials are needed to study the actual actionability of biomarkers in (recurrent) glioblastoma. This study aimed to assess the current clinical trial landscape to assess the role of molecular biomarkers in trials on recurrent glioblastoma treatment. The database ClinicalTrials.gov was used to identify not yet completed clinical trials on recurrent glioblastoma in adults. Recruiting studies were assessed to investigate the role of molecular criteria, which were retrieved as detailed as possible. Primary outcome was molecular criteria used as selection criteria for study participation. Next to this, details on moment and method of testing, and targets and drugs studied, were collected. In 76% (181/237) of the included studies, molecular criteria were not included in the study design. Of the remaining 56 studies, at least one specific genomic alteration as selection criterium for study participation was required in 33 (59%) studies. Alterations in EGFR, CDKN2A/B or C, CDK4/6, and RB were most frequently investigated, as were the corresponding drugs abemaciclib and ribociclib. Of the immunotherapies, CAR-T therapies were the most frequently studied therapies. Previously, genomics studies have revealed the presence of potentially actionable alterations in glioblastoma. Our study shows that the potential efficacy of targeted treatment is currently not translated into genome-driven trials in patients with recurrent glioblastoma. An intensification of genome-driven trials might help in providing evidence for (in)efficacy of targeted treatments.


Assuntos
Biomarcadores Tumorais , Neoplasias Encefálicas , Ensaios Clínicos como Assunto , Glioblastoma , Recidiva Local de Neoplasia , Humanos , Glioblastoma/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/terapia , Biomarcadores Tumorais/genética , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Terapia de Alvo Molecular/métodos
2.
Cancer Res Commun ; 4(9): 2514-2524, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39240078

RESUMO

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.


Assuntos
Glioblastoma , Imunoterapia Adotiva , Interleucina-7 , Neoplasias Pancreáticas , Receptores de Antígenos Quiméricos , Ensaios Antitumorais Modelo de Xenoenxerto , Glioblastoma/terapia , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Animais , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Camundongos , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Interleucina-7/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Receptores ErbB/imunologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Feminino
3.
Cancer Biomark ; 41(1): 1-24, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39240627

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Células-Tronco Neoplásicas , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/terapia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Imunoterapia/métodos , Animais , Barreira Hematoencefálica/metabolismo
4.
BMJ Open ; 14(9): e081689, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39260848

RESUMO

INTRODUCTION: There are no guidelines or prospective studies defining the optimal surgical treatment for glioblastomas in older patients (≥70 years), for those with a limited functioning performance at presentation (Karnofsky Performance Scale ≤70) or for those with tumours in certain locations (midline, multifocal). Therefore, the decision between resection and biopsy is varied, among neurosurgeons internationally and at times even within an institution. This study aims to compare the effects of maximal tumour resection versus tissue biopsy on survival, functional, neurological and quality of life outcomes in these patient subgroups. Furthermore, it evaluates which modality would maximise the potential to undergo adjuvant treatment. METHODS AND ANALYSIS: This study is an international, multicentre, prospective, two-arm cohort study of an observational nature. Consecutive patients with glioblastoma will be treated with resection or biopsy and matched with a 1:1 ratio. Primary endpoints are (1) overall survival and (2) proportion of patients that have received adjuvant treatment with chemotherapy and radiotherapy. Secondary endpoints are (1) proportion of patients with National Institute of Health Stroke Scale deterioration at 6 weeks, 3 months and 6 months after surgery; (2) progression-free survival (PFS); (3) quality of life at 6 weeks, 3 months and 6 months after surgery and (4) frequency and severity of serious adverse events. The total duration of the study is 5 years. Patient inclusion is 4 years; follow-up is 1 year. ETHICS AND DISSEMINATION: The study has been approved by the Medical Ethics Committee (METC Zuid-West Holland/Erasmus Medical Center; MEC-2020-0812). The results will be published in peer-reviewed academic journals and disseminated to patient organisations and media. TRIAL REGISTRATION NUMBER: NCT06146725.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Qualidade de Vida , Humanos , Glioblastoma/cirurgia , Glioblastoma/patologia , Glioblastoma/terapia , Estudos Prospectivos , Neoplasias Encefálicas/cirurgia , Neoplasias Encefálicas/patologia , Biópsia/métodos , Estudos Multicêntricos como Assunto , Procedimentos Neurocirúrgicos/métodos , Idoso , Feminino , Masculino
5.
Nat Genet ; 56(9): 1903-1913, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39223316

RESUMO

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.


Assuntos
Antígenos de Neoplasias , Neoplasias Encefálicas , Elementos de DNA Transponíveis , Epigênese Genética , Glioblastoma , Transcrição Gênica , Glioblastoma/genética , Glioblastoma/terapia , Glioblastoma/imunologia , Humanos , Elementos de DNA Transponíveis/genética , Linhagem Celular Tumoral , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Regulação Neoplásica da Expressão Gênica , Imunoterapia/métodos
6.
Neurosurg Rev ; 47(1): 541, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39231832

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Interleucina-6 , Glioblastoma/terapia , Humanos , Interleucina-6/metabolismo , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores de Interleucina-6 , Fator de Transcrição STAT3/metabolismo , Imunoterapia/métodos
7.
Chin Clin Oncol ; 13(Suppl 1): AB023, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39295341

RESUMO

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.


Assuntos
Glioblastoma , Humanos , Glioblastoma/mortalidade , Glioblastoma/terapia , Feminino , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Idoso , Diagnóstico Precoce , Adulto , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Prognóstico , Análise de Sobrevida
8.
Chin Clin Oncol ; 13(Suppl 1): AB047, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39295365

RESUMO

BACKGROUND: There remains controversy in the observed survival of gliomas worldwide, especially for glioblastoma (GBM). The 5-year survival rate ranged wildly, but comparable higher in several Asian countries, such as China showed almost 18% from CONCORD-3 data. Are there any special factors relating to long-term survivors (LTSs)? METHODS: We reviewed our single center real-world data for the last 20 years, of 536 GBM [World Health Organization (WHO) grade 4] patients revealed 5-year overall survival (OS) of 19.1%. We analyzed our GBM patients and searched for possible factors relating to LTSs. We collected tumor samples of 13 LTSs (OS >60 months) and 19 short-term survivors (OS <24 months), and performed whole exome sequencing and transcriptome sequencing. RESULTS: From treatment setting, besides surgical resection, post-operational adjutant treatment (radiotherapy plus chemotherapy) are the most important factor contributing to long-term survival. Whole exome sequencing analysis revealed a higher proportion of mutation signature 19 was associated with LTSs. Analysis of copy number variation (CNV) showed that the LTSs had higher copy number variants at the chromosomal level (P=0.049). At the arm level, the proportion of 19p amplification in the LTS was significantly higher than in the short-term survivors (P=0.001). And in The Cancer Genome Atlas (TCGA) GBM dataset, GBM patients with 19p amplification also had a better prognosis (log-rank P=0.04). Based on RNA sequencing (RNAseq) and differential expression analysis, the differentially expressed genes were enriched in hypoxia-related processes, apoptosis, and immune-related processes. CONCLUSIONS: From our single-institution data, the factors relating to GBM LTSs should be both clinical management and genomic alternation which could be potential novel targets be applied to future clinical practice.


Assuntos
Glioblastoma , Humanos , Glioblastoma/genética , Glioblastoma/terapia , Masculino , Feminino , Pessoa de Meia-Idade , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Adulto , Idoso , Sobreviventes de Câncer
9.
J Korean Med Sci ; 39(34): e244, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39228184

RESUMO

BACKGROUND: For treating recurrent glioblastoma, for which there is no established treatment, the antiangiogenic antibody, bevacizumab, is used alone or with irinotecan. This study was aimed at comparing the survival of patients with recurrent glioblastoma receiving bevacizumab monotherapy and those receiving bevacizumab plus irinotecan combination therapy (B+I) by using a nationwide population-based dataset. METHODS: Patients matching the International Classification of Diseases code C71.x were screened from the Health Insurance Review and Assessment Service database. From January 2008 to November 2021, patients who underwent surgery or biopsy and subsequent standard concurrent chemoradiation with temozolomide were included. Among them, those who received bevacizumab monotherapy or B+I were selected. Demographic characteristics, inpatient stay, prescription frequency, survival outcomes, and steroid prescription duration were compared between these two groups. RESULTS: Eight hundred and forty-six patients who underwent surgery or biopsy and received concurrent chemoradiotherapy with temozolomide were included. Of these, 450 and 396 received bevacizumab monotherapy and B+I, respectively. The corresponding median overall survival from the initial surgery was 22.60 months (95% confidence interval [CI], 20.50-24.21) and 20.44 months (95% CI, 18.55-22.60; P = 0.508, log-rank test). The B+I group had significantly more bevacizumab prescriptions (median 5 times; BEV group: median 3 times). Cox analysis, based on the postsurgery period, revealed that male sex (hazard ratio [HR], 1.28; P = 0.002), older age (HR, 1.01; P = 0.042), and undergoing biopsy instead of surgery (HR, 1.79; P < 0.0001) were significantly associated with decreased survival. Fewer radiotherapy cycles correlated with improved survival outcomes (HR, 0.63; P = 0.001). Cox analysis, conducted from the start of chemotherapy including bevacizumab, showed that male sex was the only variable significantly associated with decreased survival (HR, 1.18; P = 0.044). CONCLUSION: We found no significant difference in overall survival between the bevacizumab monotherapy and B+I groups. Considering the additional potential toxicity associated with irinotecan, bevacizumab monotherapy could be a suitable treatment option for treating recurrent glioblastoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Bevacizumab , Glioblastoma , Irinotecano , Recidiva Local de Neoplasia , Humanos , Bevacizumab/uso terapêutico , Bevacizumab/administração & dosagem , Irinotecano/uso terapêutico , Masculino , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Glioblastoma/terapia , Glioblastoma/patologia , Pessoa de Meia-Idade , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Idoso , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/mortalidade , Temozolomida/uso terapêutico , Estimativa de Kaplan-Meier , Estudos Retrospectivos , Quimiorradioterapia , Bases de Dados Factuais
10.
Neurosurg Focus ; 57(3): E7, 2024 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-39217635

RESUMO

OBJECTIVE: Sonodynamic therapy (SDT) is gaining attention as a promising new noninvasive brain tumor treatment that targets and selectively kills tumor cells, with limited side effects. This review examines the mechanisms of SDT and ongoing clinical trials looking at optimization of sonication parameters for potential treatment of glioblastoma (GBM) and diffuse intrinsic pontine glioma (DIPG). The results in the first patient with recurrent GBM treated at the Mayo Clinic are briefly discussed. METHODS: The authors of this literature review used electronic databases including PubMed, EMBASE, and OVID. Articles reporting relevant preclinical and clinical trials were identified by searching for text words/phrases and MeSH terms, including the following: "sonodynamic therapy," "SDT," "focused ultrasound," "5-ALA," "ALA," "brain tumors," "diffuse pontine glioma," "glioblastoma," and "high grade glioma." RESULTS: Preclinical and clinical trials investigating the specific use of SDT in brain tumors were reviewed. In preclinical models of high-grade glioma and GBM, SDT has shown evidence of targeted tumor cell death via the production of reactive oxygen species. Emerging clinical trial results within recurrent GBM and DIPG show evidence of successful treatment response, with minimal side effects experienced by recruited patients. So far, SDT has been shown to be a promising noninvasive cancer treatment that is well tolerated by patients. The authors present pilot data suggesting good radiological response of GBM to a single SDT treatment, with unpublished observation of a lack of off-target effects even after multiple (monthly) sonication outpatient treatments. The scope of the clinical trials of SDT is to investigate whether it can be the means by which the fatal diagnosis of GBM or DIPG is converted into that of a chronic, treatable disease. CONCLUSIONS: SDT is safe, repeatable, and better tolerated than both chemotherapy and radiotherapy. It has been shown to have an effect in human cancer therapy, but more clinical trials are needed to establish standardized protocols for sonosensitizer delivery, treatment parameters, and combination therapies. The most appropriate timing of treatment also remains to be determined-whether to prevent recurrence in the postoperative period, or as a salvage option in patients with recurrent GBM for which redo surgery is inappropriate. It is hoped that SDT will also be developed for a wider spectrum of clinical indications, such as metastases, meningioma, and low-grade glioma. Further clinical trials are in preparation.


Assuntos
Neoplasias Encefálicas , Humanos , Neoplasias Encefálicas/terapia , Terapia por Ultrassom/métodos , Glioblastoma/terapia , Neoplasias do Tronco Encefálico/terapia , Glioma Pontino Intrínseco Difuso/terapia
11.
Eur J Pharm Biopharm ; 203: 114460, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39218361

RESUMO

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.


Assuntos
Antineoplásicos , Barreira Hematoencefálica , Neoplasias Encefálicas , Sistemas de Liberação de Medicamentos , Exossomos , Glioblastoma , Exossomos/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/terapia , Humanos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Sistemas de Liberação de Medicamentos/métodos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem
12.
Radiol Artif Intell ; 6(5): e230489, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39166970

RESUMO

Purpose To develop and validate a deep learning (DL) method to detect and segment enhancing and nonenhancing cellular tumor on pre- and posttreatment MRI scans in patients with glioblastoma and to predict overall survival (OS) and progression-free survival (PFS). Materials and Methods This retrospective study included 1397 MRI scans in 1297 patients with glioblastoma, including an internal set of 243 MRI scans (January 2010 to June 2022) for model training and cross-validation and four external test cohorts. Cellular tumor maps were segmented by two radiologists on the basis of imaging, clinical history, and pathologic findings. Multimodal MRI data with perfusion and multishell diffusion imaging were inputted into a nnU-Net DL model to segment cellular tumor. Segmentation performance (Dice score) and performance in distinguishing recurrent tumor from posttreatment changes (area under the receiver operating characteristic curve [AUC]) were quantified. Model performance in predicting OS and PFS was assessed using Cox multivariable analysis. Results A cohort of 178 patients (mean age, 56 years ± 13 [SD]; 116 male, 62 female) with 243 MRI timepoints, as well as four external datasets with 55, 70, 610, and 419 MRI timepoints, respectively, were evaluated. The median Dice score was 0.79 (IQR, 0.53-0.89), and the AUC for detecting residual or recurrent tumor was 0.84 (95% CI: 0.79, 0.89). In the internal test set, estimated cellular tumor volume was significantly associated with OS (hazard ratio [HR] = 1.04 per milliliter; P < .001) and PFS (HR = 1.04 per milliliter; P < .001) after adjustment for age, sex, and gross total resection (GTR) status. In the external test sets, estimated cellular tumor volume was significantly associated with OS (HR = 1.01 per milliliter; P < .001) after adjustment for age, sex, and GTR status. Conclusion A DL model incorporating advanced imaging could accurately segment enhancing and nonenhancing cellular tumor, distinguish recurrent or residual tumor from posttreatment changes, and predict OS and PFS in patients with glioblastoma. Keywords: Segmentation, Glioblastoma, Multishell Diffusion MRI Supplemental material is available for this article. © RSNA, 2024.


Assuntos
Neoplasias Encefálicas , Aprendizado Profundo , Imagem de Difusão por Ressonância Magnética , Glioblastoma , Humanos , Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Glioblastoma/terapia , Glioblastoma/mortalidade , Masculino , Feminino , Pessoa de Meia-Idade , Estudos Retrospectivos , Imagem de Difusão por Ressonância Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/mortalidade , Adulto , Idoso , Interpretação de Imagem Assistida por Computador/métodos
13.
Cancer Cell ; 42(8): 1450-1466.e11, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39137729

RESUMO

Glioblastoma (GBM) is an aggressive brain cancer with limited therapeutic options. Natural killer (NK) cells are innate immune cells with strong anti-tumor activity and may offer a promising treatment strategy for GBM. We compared the anti-GBM activity of NK cells engineered to express interleukin (IL)-15 or IL-21. Using multiple in vivo models, IL-21 NK cells were superior to IL-15 NK cells both in terms of safety and long-term anti-tumor activity, with locoregionally administered IL-15 NK cells proving toxic and ineffective at tumor control. IL-21 NK cells displayed a unique chromatin accessibility signature, with CCAAT/enhancer-binding proteins (C/EBP), especially CEBPD, serving as key transcription factors regulating their enhanced function. Deletion of CEBPD resulted in loss of IL-21 NK cell potency while its overexpression increased NK cell long-term cytotoxicity and metabolic fitness. These results suggest that IL-21, through C/EBP transcription factors, drives epigenetic reprogramming of NK cells, enhancing their anti-tumor efficacy against GBM.


Assuntos
Neoplasias Encefálicas , Proteína delta de Ligação ao Facilitador CCAAT , Glioblastoma , Interleucinas , Células Matadoras Naturais , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Glioblastoma/imunologia , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/terapia , Interleucinas/genética , Interleucinas/metabolismo , Interleucinas/imunologia , Humanos , Animais , Camundongos , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Interleucina-15/genética , Interleucina-15/metabolismo , Interleucina-15/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Cell ; 42(8): 1333-1335, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39137725

RESUMO

Glioblastoma is the most common brain cancer, with a 5-year survival rate of less than 10%. This grim prognosis highlights the urgent need for novel therapeutic approaches. In this issue of Cancer Cell, Shanley et al.1 report an innovative engineering strategy to supercharge NK cell immunity against glioblastoma.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Interleucinas , Células Matadoras Naturais , Glioblastoma/imunologia , Glioblastoma/terapia , Humanos , Células Matadoras Naturais/imunologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Interleucinas/metabolismo , Interleucinas/imunologia
15.
Medicina (Kaunas) ; 60(8)2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39202515

RESUMO

Background and Objectives: Notwithstanding the major progress in the management of cancerous diseases in the last few decades, glioblastoma (GBM) remains the most aggressive brain malignancy, with a dismal prognosis, mainly due to treatment resistance and tumoral recurrence. In order to diagnose this disease and establish the optimal therapeutic approach to it, a standard tissue biopsy or a liquid biopsy can be performed, although the latter is currently less common. To date, both tissue and liquid biopsy have yielded numerous biomarkers that predict the evolution and response to treatment in GBM. However, despite all such efforts, GBM has the shortest recorded survival rates of all the primary brain malignancies. Materials and Methods: We retrospectively reviewed patients with a confirmed histopathological diagnosis of glioblastoma between June 2011 and June 2023. All the patients were treated in the Third Neurosurgical Department of the Clinical Emergency Hospital "Bagdasar-Arseni" in Bucharest, and their outcomes were analyzed and presented accordingly. Results: Out of 518 patients in our study, 222 (42.8%) were women and 296 (57.14%) were men. The most common clinical manifestations were headaches and limb paralysis, while the most frequent tumor locations were the frontal and temporal lobes. The survival rates were prolonged in patients younger than 60 years of age, in patients with gross total tumoral resection and less than 30% tumoral necrosis, as well as in those who underwent adjuvant radiotherapy. Conclusions: Despite significant advancements in relation to cancer diseases, GBM is still a field of great interest for research and in great need of new therapeutic approaches. Although the multimodal therapeutic approach can improve the prognosis, the survival rates are still short and the recurrences are constant.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/cirurgia , Glioblastoma/mortalidade , Glioblastoma/terapia , Estudos Retrospectivos , Feminino , Masculino , Pessoa de Meia-Idade , Adulto , Idoso , Neoplasias Encefálicas/cirurgia , Neoplasias Encefálicas/mortalidade , Procedimentos Neurocirúrgicos/estatística & dados numéricos , Procedimentos Neurocirúrgicos/métodos , Resultado do Tratamento , Idoso de 80 Anos ou mais , Lituânia/epidemiologia
16.
NPJ Syst Biol Appl ; 10(1): 91, 2024 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-39155294

RESUMO

Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.


Assuntos
Neoplasias Encefálicas , Simulação por Computador , Glioblastoma , Imunoterapia , Microambiente Tumoral , Glioblastoma/terapia , Glioblastoma/imunologia , Humanos , Microambiente Tumoral/imunologia , Imunoterapia/métodos , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia
17.
Nat Commun ; 15(1): 6870, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39127809

RESUMO

Current treatment outcome of patients with glioblastoma (GBM) remains poor. Following standard therapy, recurrence is universal with limited survival. Tumors from 173 GBM patients are analysed for somatic mutations to generate a personalized peptide vaccine targeting tumor-specific neoantigens. All patients were treated within the scope of an individual healing attempt. Among all vaccinated patients, including 70 treated prior to progression (primary) and 103 treated after progression (recurrent), the median overall survival from first diagnosis is 31.9 months (95% CI: 25.0-36.5). Adverse events are infrequent and are predominantly grade 1 or 2. A vaccine-induced immune response to at least one of the vaccinated peptides is detected in blood samples of 87 of 97 (90%) monitored patients. Vaccine-specific T-cell responses are durable in most patients. Significantly prolonged survival is observed for patients with multiple vaccine-induced T-cell responses (53 months) compared to those with no/low induced responses (27 months; P = 0.03). Altogether, our results highlight that the application of personalized neoantigen-targeting peptide vaccine is feasible and represents a promising potential treatment option for GBM patients.


Assuntos
Neoplasias Encefálicas , Vacinas Anticâncer , Glioblastoma , Medicina de Precisão , Vacinas de Subunidades Proteicas , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Glioblastoma/imunologia , Glioblastoma/terapia , Medicina de Precisão/métodos , Vacinas de Subunidades Proteicas/imunologia , Vacinas de Subunidades Proteicas/uso terapêutico , Linfócitos T/imunologia , Resultado do Tratamento
18.
ACS Nano ; 18(34): 23001-23013, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39150454

RESUMO

The currently available immune checkpoint therapy shows a disappointing therapeutic efficacy for glioblastoma multiforme (GBM), and it is of great importance to discover better immune checkpoints and develop innovative targeting strategies. The discovered metabolic immune checkpoint ecto-5-nucleotidase (CD73) in a tumor contributes to its immune evasion due to the dysregulation of extracellular adenosine (ADO), which significantly inhibits the function of antitumor T cells and increases the activity of immunosuppressive cells. Herein, we drastically inhibit the expression of CD73 to reduce the production of ADO by using versatile Au@Cu2-xSe nanoparticles (ACS NPs). ACS NPs can decrease the expression of CD73 by alleviating the tumor hypoxia through their Fenton-like reaction to weaken the ADO-driven immunosuppression for enhancing antitumor T cell infiltration and activity of GBM. The copper ions (Cu2+) released from ACS NPs can chelate with disulfide, leading to the formation of cytotoxic bis(N,N-diethyldithiocarbamate)-copper complex (CuET), which can be combined with radiotherapy to recruit more antitumor T cells to infiltrate into the tumor site. Based on the inhibition of CD73 to promote the infiltration and activity of antitumor T cells, a cascade of enhancing GBM immunotherapy effects can be achieved. The significant increase in CD8+ T and CD4+ T cells within the tumor and the memory T cells in the spleen effectively reduces tumor size by 92%, which demonstrates the excellent efficacy of immunotherapy achieved by a combination of metabolic immune checkpoint CD73 inhibition with chemoradiotherapy. This work demonstrates that modulation of CD73-mediated tumor immunosuppression is an important strategy of improving the outcome of GBM immunotherapy.


Assuntos
5'-Nucleotidase , Glioblastoma , Imunoterapia , Glioblastoma/terapia , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/tratamento farmacológico , 5'-Nucleotidase/metabolismo , 5'-Nucleotidase/antagonistas & inibidores , Animais , Humanos , Camundongos , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Cobre/química , Cobre/farmacologia , Ouro/química , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Nanopartículas Metálicas/química , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/antagonistas & inibidores , Adenosina/química , Adenosina/farmacologia
19.
Ann Ital Chir ; 95(4): 416-434, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39186332

RESUMO

AIM: Intraventricular glioblastomas (IVGBMs) are rare tumors within the central nervous system characterized by unique challenges in diagnosis and management due to their location within the ventricular system. Despite their rarity, these tumors necessitate comprehensive study to refine diagnostic approaches and optimize therapeutic strategies. METHODS: A systematic review was conducted using PubMed, Scopus, Web of Science, and Google Scholar databases to identify relevant literature published up to January 2024. Inclusion criteria encompassed studies in English focusing on clinical characteristics, radiological features, pathology, and treatment of IVGBM. Data synthesis and analysis followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS: Twenty-four articles met the inclusion criteria, comprising 47 patients with IVGBM. The median age was 47 years, with a male predominance (32 males, 15 females). Common symptoms included increased intracranial pressure and seizures. Tumors predominantly affected the lateral ventricles (body and trigone). Surgical resection (subtotal or gross total) was the primary treatment approach, with adjuvant therapies (radiotherapy, chemotherapy) administered postoperatively. CONCLUSIONS: IVGBM present distinct diagnostic and therapeutic challenges due to their ventricular location. Current treatments primarily involve surgical resection followed by adjuvant therapies, though outcomes remain guarded. Further research is needed to enhance understanding and management of this rare glioblastoma subset.


Assuntos
Neoplasias do Ventrículo Cerebral , Glioblastoma , Humanos , Glioblastoma/terapia , Terapia Combinada , Neoplasias do Ventrículo Cerebral/terapia , Neoplasias do Ventrículo Cerebral/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade
20.
Front Immunol ; 15: 1404861, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39192978

RESUMO

Background: Glioblastoma (GBM) is a poor prognosis grade 4 glioma. After surgical resection, the standard therapy consists of concurrent radiotherapy (RT) and temozolomide (TMZ) followed by TMZ alone. Our previous data on melanoma patients showed that Dendritic Cell vaccination (DCvax) could increase the amount of intratumoral-activated cytotoxic T lymphocytes. Methods: This is a single-arm, monocentric, phase II trial in two steps according to Simon's design. The trial aims to evaluate progression-free survival (PFS) at three months and the safety of a DCvax integrated with standard therapy in resected GBM patients. DCvax administration begins after completion of RT-CTwith weekly administrations for 4 weeks, then is alternated monthly with TMZ cycles. The primary endpoints are PFS at three months and safety. One of the secondary objectives is to evaluate the immune response both in vitro and in vivo (DTH skin test). Results: By December 2022, the first pre-planned step of the study was concluded with the enrollment, treatment and follow up of 9 evaluable patients. Two patients had progressed within three months after leukapheresis, but none had experienced DCvax-related G3-4 toxicities Five patients experienced a positive DTH test towards KLH and one of these also towards autologous tumor homogenate. The median PFS from leukapheresis was 11.3 months and 12.2 months from surgery. Conclusions: This combination therapy is well-tolerated, and the two endpoints required for the first step have been achieved. Therefore, the study will proceed to enroll the remaining 19 patients. (Eudract number: 2020-003755-15 https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-003755-15/IT).


Assuntos
Neoplasias Encefálicas , Vacinas Anticâncer , Células Dendríticas , Glioblastoma , Humanos , Glioblastoma/terapia , Glioblastoma/imunologia , Glioblastoma/mortalidade , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Células Dendríticas/imunologia , Células Dendríticas/transplante , Pessoa de Meia-Idade , Feminino , Masculino , Adulto , Idoso , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Temozolomida/uso terapêutico , Temozolomida/administração & dosagem , Intervalo Livre de Progressão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA