Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Sci Rep ; 14(1): 7984, 2024 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-38575630

RESUMEN

The extent of surgical resection is an important prognostic factor in the treatment of patients with glioblastoma. Optical coherence tomography (OCT) imaging is one of the adjunctive methods available to achieve the maximal surgical resection. In this study, the tumor margins were visualized with the OCT image obtained from a murine glioma model. A commercialized human glioblastoma cell line (U-87) was employed to develop the orthotopic murine glioma model. A swept-source OCT (SS-OCT) system of 1300 nm was used for three-dimensional imaging. Based on the OCT intensity signal, which was obtained via accumulation of each A-scan data, an en-face optical attenuation coefficient (OAC) map was drawn. Due to the limited working distance of the focused beam, OAC values decrease with depth, and using the OAC difference in the superficial area was chosen to outline the tumor boundary, presenting a challenge in analyzing the tumor margin along the depth direction. To overcome this and enable three-dimensional tumor margin detection, we converted the en-face OAC map into an en-face difference map with x- and y-directions and computed the normalized absolute difference (NAD) at each depth to construct a volumetric NAD map, which was compared with the corresponding H&E-stained image. The proposed method successfully revealed the tumor margin along the peripheral boundaries as well as the margin depth. We believe this method can serve as a useful adjunct in glioma surgery, with further studies necessary for real-world practical applications.


Asunto(s)
Glioblastoma , Glioma , Humanos , Animales , Ratones , Glioblastoma/diagnóstico por imagen , Tomografía de Coherencia Óptica/métodos , NAD , Glioma/patología , Imagenología Tridimensional
2.
Biomed Pharmacother ; 173: 115790, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38431436

RESUMEN

BACKGROUND: Although PD-1 blockade is effective for treating several types of cancer, the efficacy of this agent in glioblastoma is largely limited. To overcome non-responders and the immunosuppressive tumor microenvironment, combinational immunotherapeutic strategies with anti-PD-1 need to be considered. Here, we developed IL-12-secreting mesenchymal stem cells (MSC_IL-12) with glioblastoma tropism and evaluated the therapeutic effects of anti-PD-1, MSC_IL-12, and their combination against glioblastoma. METHODS: Therapeutic responses were evaluated using an immunocompetent mouse orthotopic model. Tumor-infiltrating lymphocytes (TILs) were analyzed using immunofluorescent imaging. Single-cell transcriptome was obtained from mouse brains after treatments. RESULTS: Anti-PD-1 and MSC_IL-12 showed complete tumor remission in 25.0% (4/16) and 23.1% (3/13) of glioblastoma-implanted mice, respectively, and their combination yielded synergistic antitumor efficacy indicated by 50.0% (6/12) of complete tumor remission. Analyses of TILs revealed that anti-PD-1 increased CD8+ T cells, while MSC_IL-12 led to infiltration of CD4+ T cells and NK cells. Both therapies reduced frequencies of Tregs. All these aspects observed in each monotherapy group were superimposed in the combination group. Notably, no tumor growth was observed upon rechallenge in cured mice, indicating long-term immunity against glioblastoma provoked by the therapies. Single-cell RNA-seq data confirmed these results and revealed that the combined treatment led to immune-favorable tumor microenvironment-CD4+, CD8+ T cells, effector memory T cells, and activated microglia were increased, whereas exhausted T cells, Tregs, and M2 polarized microglia were reduced. CONCLUSION: Anti-PD-1 and MSC_IL-12 monotherapies show long-term therapeutic responses, and their combination further enhances antitumor efficacy against glioblastoma via inducing immune-favorable tumor microenvironment.


Asunto(s)
Glioblastoma , Células Madre Mesenquimatosas , Animales , Ratones , Glioblastoma/patología , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Inmunoterapia/métodos , Interleucina-12 , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/patología , Microambiente Tumoral
3.
Cancer Lett ; 588: 216714, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38369003

RESUMEN

Glioblastoma, the most prevalent malignant primary brain tumor, presents substantial treatment challenges because of its inherent aggressiveness and limited therapeutic options. Lymphopenia, defined as reduced peripheral blood lymphocyte count, commonly occurs as a consequence of the disease and its treatment. Recent studies have associated lymphopenia with a poor prognosis. Factors that contribute to lymphopenia include radiotherapy, chemotherapy, and the tumor itself. Patients who are female, older, using dexamethasone, or receiving higher doses of radiation therapy are particularly vulnerable to this condition. Several preclinical studies have explored the use of interleukin-7, a crucial cytokine for lymphocyte homeostasis, to restore lymphocyte counts and potentially rebuild the immune system to combat glioblastoma cells. With the development of recombinant interleukin-7 for prolonged activity in the body, various clinical trials are underway to explore this treatment in patients with glioblastoma. Our study provides a comprehensive summary of the incidence of lymphopenia, its potential biological background, and the associated clinical risk factors. Furthermore, we reviewed several clinical trials using IL-7 cytokine therapy in glioblastoma patients. We propose IL-7 as a promising immunotherapeutic strategy for glioblastoma treatment. We are optimistic that our study will enhance understanding of the complex interplay between lymphopenia and glioblastoma and will pave the way for the development of more effective treatment modalities.


Asunto(s)
Glioblastoma , Linfopenia , Humanos , Femenino , Masculino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Interleucina-7/uso terapéutico , Interleucina-7/farmacología , Linfopenia/etiología , Linfopenia/patología , Linfocitos , Inmunoterapia/efectos adversos
4.
Brain Tumor Res Treat ; 12(1): 23-39, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38317486

RESUMEN

BACKGROUND: Glioma is caused by multiple genomic alterations. The evolving classification of gliomas emphasizes the significance of molecular testing. Next generation sequencing (NGS) offers the assessment of parallel combinations of multiple genetic alterations and identifying actionable mutations that guide treatment. This study comprehensively analyzed glioma patients using multi-gene NGS panels, providing powerful insights to inform diagnostic classification and targeted therapies. METHODS: We conducted a targeted panel-based NGS analysis on formalin-fixed and paraffin-embedded nucleic acids extracted from a total of 147 glioma patients. These samples underwent amplicon capture-based library preparation and sequenced using the Oncomine Comprehensive Assay platform. The resulting sequencing data were then analyzed using the bioinformatics tools. RESULTS: A total of 301 mutations, were found in 132 out of 147 tumors (89.8%). These mutations were in 68 different genes. In 62 tumor samples (42.2%), copy number variations (CNVs) with gene amplifications occurred in 25 genes. Moreover, 25 tumor samples (17.0%) showed gene fusions in 6 genes and intragenic deletion in a gene. Our analysis identified actionable targets in several genes, including 11 with mutations, 8 with CNVs, and 3 with gene fusions and intragenic deletion. These findings could impact FDA-approved therapies, NCCN guideline-based treatments, and clinical trials. CONCLUSION: We analyzed precisely diagnosing the classification of gliomas, detailing the frequency and co-occurrence of genetic alterations and identifying genetic alterations with potential therapeutic targets by NGS-based molecular analysis. The high-throughput NGS analysis is an efficient and powerful tool to comprehensively support molecular testing in neurooncology.

5.
J Neurol Surg B Skull Base ; 85(1): 21-27, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38327514

RESUMEN

Objectives The authors applied surgical techniques acquired during the use of endoscopic combined transseptal/transnasal approach to reduce approach-related morbidity and improve sinonasal outcomes. Study Design This is a retrospective cohort study of a prospectively collected database. Setting The study setting involves a tertiary referral center. Participants A total of 86 patients who underwent endoscopic endonasal transsphenoidal surgery for newly diagnosed pituitary adenomas from April 2018 to March 2021 were included. Patients treated via the combined transseptal/transnasal approach served as the study group ( n = 18); those treated via the bilateral transnasal approach comprised the control group ( n = 68). From the control group, propensity score matching (PSM) analysis was further performed to account for potential confounders and selection bias. Main Outcome Measures Paired analysis was performed for pre- and 6-month-postoperative time points in study group, control group, and PSM control group. Olfactory function was evaluated by Connecticut Chemosensory Clinical Research Center (CCCRC) test, Cross-Cultural Smell Identification Test (CCSIT), and sinonasal outcomes were assessed by Sino-Nasal Outcome Test-22 (SNOT-22). Results In the study group, CCCRC ( p = 0.517) and CCSIT ( p = 0.497) did not show any significant difference before and after surgery. There was some improvement in the symptom score of SNOT-22, but it was not statistically significant ( p = 0.115). In the control group adjusted with PSM, a significant decrease in olfaction ( p = 0.047) was observed using CCCRC. The CCSIT score was also decreased but not significant ( p = 0.163). Also, there was no difference in the improvement of SNOT-22 ( p = 0.781). Conclusion Our new surgical method preserves olfactory function without compromising surgical outcomes.

6.
Cancer Cell Int ; 24(1): 36, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38238738

RESUMEN

BACKGROUND: Although meningioma is the most common primary brain tumor, treatments rely on surgery and radiotherapy, and recurrent meningiomas have no standard therapeutic options due to a lack of clinically relevant research models. Current meningioma cell lines or organoids cannot reflect biological features of patient tumors since they undergo transformation along culture and consist of only tumor cells without microenvironment. We aim to establish patient-derived meningioma organoids (MNOs) preserving diverse cell types representative of the tumor microenvironment. METHODS: The biological features of MNOs were evaluated using WST, LDH, and collagen-based 3D invasion assays. Cellular identities in MNOs were confirmed by immunohistochemistry (IHC). Genetic alteration profiles of MNOs and their corresponding parental tumors were obtained by whole-exome sequencing. RESULTS: MNOs were established from four patients with meningioma (two grade 1 and two grade 2) at a 100% succession rate. Exclusion of enzymatic dissociation-reaggregation steps endowed MNOs with original histology and tumor microenvironment. In addition, we used a liquid media culture system instead of embedding samples into Matrigel, resulting in an easy-to-handle, cost-efficient, and time-saving system. MNOs maintained their functionality and morphology after long-term culture (> 9 wk) and repeated cryopreserving-recovery cycles. The similarities between MNOs and their corresponding parental tumors were confirmed by both IHC and whole-exome sequencing. As a representative application, we utilized MNOs in drug screening, and mifepristone, an antagonist of progesterone receptor, showed prominent antitumor efficacy with respect to viability, invasiveness, and protein expression. CONCLUSION: Taken together, our MNO model overcame limitations of previous meningioma models and showed superior resemblance to parental tumors. Thus, our model could facilitate translational research identifying and selecting drugs for meningioma in the era of precision medicine.

7.
J Neurooncol ; 165(2): 321-328, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37964132

RESUMEN

PURPOSE: Recently, reduced-dose whole-brain radiotherapy (WBRT) has been used to treat primary central nervous system lymphoma (PCNSL). However, whether reduced-dose WBRT is also an acceptable option for curative or salvage purposes has not yet been reported. We analyzed the clinical outcomes of patients with PCNSL who received radiotherapy for curative or salvage purposes and compared the clinical outcomes according to the WBRT dose. METHODS: A total of 66 patients were divided into two groups: those treated with 30 Gy (2 Gy per fraction) or less WBRT (low-dose WBRT, n = 34) and those treated with more than 30 Gy WBRT (high-dose WBRT, n = 32). The median WBRT dose was 25.2 and 49.6 Gy in low-dose and high-dose WBRT groups, respectively. The median total radiotherapy dose, including the boost dose, was 50 Gy (range, 36.0-55.8 Gy). RESULTS: The 3-year overall survival and progression-free survival were 77.8% and 29.8%, respectively. Intracranial relapse occurred in 31 patients (47.0%) at a median of 27 months after RT. Overall survival and progression-free survival did not differ between the two groups. The 3-year intracranial disease control rate did not differ between the two groups (35.2% vs. 41.6%, p = 0.300). Grade 3 or higher neurological toxicities were observed in six patients, of whom five were in the high-dose WBRT group. CONCLUSION: Reduced-dose WBRT in curative and salvage treatments for PCNSL had no significant negative effect on the intracranial disease control rate or survival. Therefore, without impaired efficacy, use of reduced-dose WBRT appears promising for reduction of neurotoxicity.


Asunto(s)
Neoplasias Encefálicas , Neoplasias del Sistema Nervioso Central , Linfoma , Humanos , Neoplasias del Sistema Nervioso Central/patología , Linfoma/patología , Recurrencia Local de Neoplasia , Neoplasias Encefálicas/radioterapia , Encéfalo/patología , Irradiación Craneana/efectos adversos
8.
Cancer Lett ; 571: 216335, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37544475

RESUMEN

Glioblastoma is the most common brain malignancy with devastating prognosis. Numerous clinical trials using various target therapeutic agents have failed and recent clinical trials using check point inhibitors also failed to provide survival benefits for glioblastoma patients. Adoptive T cell transfer is suggested as a novel therapeutic approach that has exhibited promise in preliminary clinical studies. However, the clinical outcomes are inconsistent, and there are several limitations of current adoptive T cell transfer strategies for glioblastoma treatment. As an alternative cell therapy, gamma-delta (γδ) T cells have been recently introduced for several cancers including glioblastoma. Since the leading role of γδ T cells is immune surveillance by recognizing a broad range of ligands including stress molecules, phosphoantigens, or lipid antigens, recent studies have suggested the potential benefits of γδ T cell transfer against glioblastomas. However, γδ T cells, as a small subset (1-5%) of T cells in human peripheral blood, are relatively unknown compared to conventional alpha-beta (αß) T cells. In this context, our study introduced γδ T cells as an alternative and novel option to overcome several challenges regarding immune cell therapy in glioblastoma treatment. We described the unique characteristics and advantages of γδ T cells compared to conventional αß T cells and summarize several recent preclinical studies using human gamma-delta T cell therapy for glioblastomas. Finally, we suggested future direction of human γδ T cell therapy for glioblastomas.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patología , Receptores de Antígenos de Linfocitos T gamma-delta , Neoplasias Encefálicas/patología , Inmunoterapia Adoptiva , Tratamiento Basado en Trasplante de Células y Tejidos
9.
J Pathol Transl Med ; 57(4): 217-231, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37460396

RESUMEN

BACKGROUND: The metastatic brain tumor is the most common brain tumor. The aim of this study was to demonstrate the clinicopathological and molecular pathologic features of brain metastases (BM). METHODS: A total of 269 patients were diagnosed with BM through surgical resection at Seoul St. Mary's Hospital from January 2010 to March 2020. We reviewed the clinicopathological features and molecular status of primary and metastatic brain tissues using immunohistochemistry and molecular pathology results. RESULTS: Among 269 patients, 139 males and 130 females were included. The median age of primary tumor was 58 years (range, 13 to 87 years) and 86 patients (32.0%) had BM at initial presentation. Median BM free interval was 28.0 months (range, 1 to 286 months). The most frequent primary site was lung 46.5% (125/269), and followed by breast 15.6% (42/269), colorectum 10.0% (27/269). Epidermal growth factor receptor (EGFR) mutation was found in 50.8% (32/63) and 58.0% (40/69) of lung primary and BM, respectively. In both breast primary and breast cancer with BM, luminal B was the most frequent subtype at 37.9% (11/29) and 42.9% (18/42), respectively, followed by human epidermal growth factor receptor 2 with 31.0% (9/29) and 33.3% (14/42). Triple-negative was 20.7% (6/29) and 16.7% (7/42), and luminal A was 10.3% (3/29) and 7.1% (3/42) of breast primary and BM, respectively. In colorectal primary and colorectal cancer with BM, KRAS mutation was found in 76.9% (10/13) and 66.7% (2/3), respectively. CONCLUSIONS: We report the clinicopathological and molecular pathologic features of BM that can provide useful information for understanding the pathogenesis of metastasis and for clinical trials based on the tumor's molecular pathology.

10.
J Clin Med ; 12(14)2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37510895

RESUMEN

PURPOSE: The immune responses of natural killer (NK) cells against cancer cells vary by patient. Killer Ig-like receptors (KIRs), which are some of the major receptors involved in regulating NK cell activity for killing cancer cells, have significant genetic variation. Numerous studies have suggested a potential association between the genetic variation of KIR genes and the risk of development or prognosis of various cancer types. However, an association between genetic variations of KIR genes and glioblastoma (GB) remains uncertain. We sought to evaluate the association of genetic variations of KIRs and their ligand genes with the risk of GB development in Koreans. METHODS: A case-control study was performed to identify the odds ratios (ORs) of KIR genes and Classes A, B, and, C of the human leukocyte antigen (HLA) for GB. The GB group was comprised of 77 patients with newly diagnosed IDH-wildtype GB at our institution, and the control group consisted of 200 healthy Korean volunteers. RESULTS: There was no significant difference in the frequency of KIR genes and KIR haplotypes between the GB and control groups. Genetic variations of KIR-2DL1, 3DL1, and 3DS1 with their ligand genes (HLA-C2, HLA-Bw4/6, and Bw4, respectively) had effects on the risk of GB in Korean patients. The frequency of KIR-2DL1 with HLA-C2 (OR 2.05, CI 1.19-3.52, p = 0.009), the frequency of KIR-3DL1 without HLA-Bw4 (80I) (OR 8.36, CI 4.06-17.18, p < 0.001), and the frequency of KIR-3DL1 with Bw6 (OR 4.54, CI 2.55-8.09, p < 0.001) in the GB group were higher than in the control group. In addition, the frequency of KIR-2DL1 without HLA-C2 (OR 0.44, CI 0.26-0.75, p = 0.003), the frequency of KIR-3DL1 with HLA-Bw4 (80T) (OR 0.13, CI 0.06-0.27, p < 0.001), the frequency of KIR-3DL1 without Bw6 (OR 0.27, CI 0.15-0.49, p < 0.001), and the frequency of KIR-3DS1 with Bw4 (80I) (OR 0.03, CI 0.00-0.50, p < 0.001) in the GB group were lower than in the control group. CONCLUSIONS: This study suggests that genetic variations of KIRs and their ligand genes may affect GB development in the Korean population. Further investigations are needed to demonstrate the different immune responses for GB cells according to genetic variations of KIR genes and their ligand genes.

11.
Clin Neurol Neurosurg ; 228: 107708, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37043844

RESUMEN

BACKGROUND: The prognostic significance of the presence of cystic features in patients with newly diagnosed glioblastoma (GB) is highly controversial. The purpose of this study was to determine whether cystic GB patients have a more favorable prognosis compared to non-cystic GB patients. METHODS: The records of all GB patients diagnosed between August 2008 and December 2020 at Seoul St. Mary's Hospital were reviewed retrospectively. Out of 254 GB patients, we excluded patients with a confirmed isocitrate dehydrogenase (IDH) mutation or an unknown IDH mutation status. A total of 145 patients met our eligibility criteria. RESULTS: Of the 145 patients we analyzed, 16 patients were classified as the cystic group, and 129 patients were classified into the non-cystic group. As there was a significant difference in the extent of resection between the two groups, 32 patients were matched according to propensity score matching. A Kaplan-Meier survival curve of the two groups indicated that the cystic group had better survival than the non-cystic group (28.6 months versus 18.8 months, respectively; p = 0.055). On multivariate analysis, the presence of cystic features (hazard ratio [HR]: 0.40, 95% confidence interval [CI]: 0.17-0.91, p = 0.029) was significantly related with a longer OS. Longer OS was also related with well-known prognostic factors, such as grossly total resection (HR: 0.05, CI: 0.01-0.31, respectively; p = 0.001) and lower European Cooperative Oncology Group (ECOG) score (HR: 3.67, CI: 1.56-9.02, respectively; p = 0.003). CONCLUSION: Our results suggest that the presence of cystic features could be an independent prognostic factor suggesting better survival in GB patients. Further larger and prospective studies to validate our findings are needed.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/diagnóstico , Glioblastoma/genética , Glioblastoma/cirugía , Isocitrato Deshidrogenasa/genética , Estudios Retrospectivos , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/cirugía , Estudios Prospectivos , Pronóstico , Mutación
12.
Cancer Med ; 12(6): 6778-6787, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36583472

RESUMEN

PURPOSE: Addressing lymphopenia in cancer patients has been suggested as a novel immunotherapeutic strategy. As interleukin-7 (IL-7) is necessary for proliferation of lymphocytes and to increase total lymphocyte count (TLC), IL-7 therapy has been attempted in various cancers. Here, we describe the clinical results of treatment of recurrent glioblastoma (GBM) with a long-acting engineered version of recombinant human IL-7 (rhIL-7-hyFc). METHODS: This prospective case series based on compassionate use was approved by the Ministry of Food and Drug Safety in South Korea. Primary outcomes were safety profile and TLC. Secondary outcomes were overall survival (OS) and progression-free survival (PFS). RESULTS: Among the 18 patients enrolled, 10 received rhIL-7-hyFc with temozolomide, 5 received rhIL-7-hyFc with bevacizumab, 1 received rhIL-7-hyFc with PCV chemotherapy, and 2 received rhIL-7-hyFc alone. Mean TLC of the enrolled patients after the first rhIL-7-hyFc treatment increased significantly from 1131 cells/mm3 (330-2989) at baseline to 4356 cells/mm3 (661-22,661). Higher TLCs were maintained while rhIL-7-hyFc was repeatedly administered. Median OS and PFS were 378 days (107-864 days) and 231 days (55-726 days), respectively. CONCLUSION: Our study reports that IL-7 immunotherapy can restore and maintain TLC during treatment with various salvage chemotherapies in recurrent GBM patients without serious toxicity.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Linfopenia , Humanos , Interleucina-7/uso terapéutico , Glioblastoma/tratamiento farmacológico , Ensayos de Uso Compasivo , Terapia Recuperativa/métodos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Linfopenia/etiología , Factores Inmunológicos/uso terapéutico
13.
Oncoimmunology ; 11(1): 2138152, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36338147

RESUMEN

Adoptive transfer of γδ T cells is a novel immunotherapeutic approach to glioblastoma. Few recent studies have shown the efficacy of γδ T cells against glioblastoma, but no previous studies have identified the ligand-receptor interactions between γδ T cells and glioblastoma cells. Here, we identify those ligand-receptor interactions and provide a basis for using γδ T cells to treat glioblastoma. Vγ9Vδ2 T cells were generated from peripheral blood mononuclear cells of healthy donors using artificial antigen presenting cells. MICA, ULBP, PVR and Nectin-2 expression in 10 patient-derived glioblastoma (PDG) cells were analyzed. The in vitro cytokine secretion from the γδ T cells and their cytotoxicity toward the PDG cells were also analyzed. The in vivo anti-tumor effects were evaluated using a U87 orthotopic xenograft glioblastoma model. Expression of ligands and cytotoxicity of the γδ T cells varied among the PDG cells. IFN-γ and Granzyme B secretion levels were significantly higher when γδ Tcells were co-cultured with high-susceptible PDG cells than when they were co-cultured with low-susceptible PDG cells. Cytotoxicity correlated significantly with the expression levels of DNAM-1 ligands of the PDG cells. Blocking DNAM-1 resulted in a decrease in γδ T cell-mediated cytotoxicity and cytokine secretion. Intratumoral injection of γδ T cells showed anti-tumor effects in an orthotopic mouse model. Allogenic γδ T cells showed potent anti-tumor effects on glioblastoma in a DNAM-1 axis dependent manner. Our findings will facilitate the development of clinical strategies using γδ T cells for glioblastoma treatment.


Asunto(s)
Glioblastoma , Ratones , Animales , Humanos , Glioblastoma/terapia , Receptores de Antígenos de Linfocitos T gamma-delta , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Ligandos , Linfocitos T , Citocinas
14.
Brain Tumor Res Treat ; 10(3): 135-143, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35929110

RESUMEN

Over the last two decades, numerous studies have investigated the presence of human cytomegalovirus (CMV) within glioblastoma or gliomas; however, the results are severely conflicting. While a few researchers have suggested the potential benefits of cytotoxic T lymphocyte or dendritic cell-based vaccines for recurrent or newly diagnosed glioblastoma patients, several studies did not at all agree with the existence of CMV in glioblastoma cells. In this review, we summarized the conflicting results and issues about the detection of CMV in glioblastoma or glioma patients. We also provided the clinical data of published and unpublished clinical trials using CMV-specific immunotherapy for glioblastomas.

15.
Cells ; 11(6)2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35326480

RESUMEN

The aim of this study was to validate the use of human brain organoids (hBOs) to investigate the therapeutic potential and mechanism of human-neural-crest-derived nasal turbinate stem cells (hNTSCs) in models of Alzheimer's disease (AD). We generated hBOs from human induced pluripotent stem cells, investigated their characteristics according to neuronal markers and electrophysiological features, and then evaluated the protective effect of hNTSCs against amyloid-ß peptide (Aß1-42) neurotoxic activity in vitro in hBOs and in vivo in a mouse model of AD. Treatment of hBOs with Aß1-42 induced neuronal cell death concomitant with decreased expression of neuronal markers, which was suppressed by hNTSCs cocultured under Aß1-42 exposure. Cytokine array showed a significantly decreased level of osteopontin (OPN) in hBOs with hNTSC coculture compared with hBOs only in the presence of Aß1-42. Silencing OPN via siRNA suppressed Aß-induced neuronal cell death in cell culture. Notably, compared with PBS, hNTSC transplantation significantly enhanced performance on the Morris water maze, with reduced levels of OPN after transplantation in a mouse model of AD. These findings reveal that hBO models are useful to evaluate the therapeutic effect and mechanism of stem cells for application in treating AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Síndromes de Neurotoxicidad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Organoides/metabolismo , Osteopontina , Cornetes Nasales/metabolismo
16.
Oncol Lett ; 23(4): 114, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35251345

RESUMEN

Procarbazine, lomustine and vincristine (PCV) chemotherapy is considered a salvage option for adult glioma; however, its significant toxicities frequently lead to dose reduction or discontinuation in patients with recurrent glioma. The current study evaluated the safety and efficacy of modified procarbazine and lomustine (PC) chemotherapy that omits vincristine and reduces the lomustine dose compared with those of conventional PCV chemotherapy. Using electronic medical records, all patients with adult recurrent glioma who received PC or PCV chemotherapy between 2009 and 2020 at Seoul St. Mary's Hospital or St. Vincent's Hospital were examined retrospectively. A total of 59 patients met the eligibility criteria. Among them, 15 patients received modified PC chemotherapy (PC group) and 44 patients received PCV chemotherapy (PCV group). The PC group presented a significantly lower hematology toxicity (anemia, 6.7 vs. 45.5%, P=0.02; thrombocytopenia 20.0 vs. 70.4%, P<0.001). Additionally, the clinical impacts of PC chemotherapy, including delay of a cycle, dose reduction, discontinuation of drug(s) or total cessation of chemotherapy, were significantly less frequent compared with the PCV group (26.7 vs. 68.2%, P=0.012). The overall survival of the PC group was also significantly longer than that of PCV group (396 vs. 232 days, P=0.042), while there was no significant difference in progression-free survival between the two groups (284.5 vs. 131 days, P=0.077). The results suggested that modified PC chemotherapy may be an alternative chemotherapeutic regimen with tolerable toxicity and without loss of clinical efficacy in patients with recurrent adult glioma. Further prospective and larger studies are required to validate our findings.

17.
Laryngoscope ; 132(5): 965-972, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35112725

RESUMEN

OBJECTIVES: We estimated volume changes in the posterior bony wall of the sphenoid sinus, as well as alterations in nasal function (including olfactory function and subjective symptoms), after sphenoid mucosal repositioning using the endoscopic endonasal transsphenoidal approach (EETSA). METHODS: During 2010 and 2021, 13 patients underwent sphenoid mucosal repositioning during EETSA, while 24 patients (the control group) did not. Pre- and postoperative paranasal sinus computed tomography and the Mimics program were used to evaluate three-dimensional changes in the posterior wall of the sphenoid sinus. All patients underwent the Connecticut Chemosensory Clinical Research Center (CCCRC) test, the Cross-Cultural Smell Identification Test (CCSIT), Nasal Obstruction Symptoms Evaluation (NOSE), the Sino-Nasal Outcome Test-20 (SNOT-20), and visual analog scale (VAS) evaluation. RESULTS: The increase in the volume of the posterior wall of the sphenoid sinus after surgery was objectively smaller in the sphenoid mucosal repositioning group than in the control group (P = .046). However, this did not affect olfactory function (as revealed by the CCCRC test or the CCSIT) or subjective symptoms (as revealed by the NOSE, SNOT-20, and VAS scores) (all P > .05). CONCLUSION: Surgical closure via sphenoid mucosal repositioning during EETSA reduces the volume of the posterior wall of the sphenoid sinus and facilitates re-operation. We suggest that sphenoid mucosal repositioning is appropriate during EETSA. LEVEL OF EVIDENCE: 4 Laryngoscope, 132:965-972, 2022.


Asunto(s)
Obstrucción Nasal , Neoplasias de la Base del Cráneo , Endoscopía/métodos , Humanos , Estudios Retrospectivos , Neoplasias de la Base del Cráneo/cirugía , Hueso Esfenoides/diagnóstico por imagen , Hueso Esfenoides/cirugía , Seno Esfenoidal/diagnóstico por imagen , Seno Esfenoidal/cirugía
18.
Clin Neurol Neurosurg ; 212: 107068, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34847484

RESUMEN

PURPOSE: The treatment of choice for chronic subdural hematoma (CSDH) has been established as burr-hole trephination with drain insertion; however, controversy remains over the best place for the drainage catheter. In this study, we compare the safety and efficacy of a subperiosteal drain (SPD) with that of a subdural drain (SDD) after one burr-hole trephination for CSDH. METHODS: This retrospective and comparative study includes all CSDH patients treated with burr-hole trephination at our institution between January 2015 and December 2019. 59 patients were treated with SPD insertion (SPD group), and 203 patients were treated with SDD insertion (SDD group). RESULTS: The median hematoma thickness of the SPD group within 24 h after surgery was significantly thicker than that of the SDD group (9.5 mm vs. 7.5 mm, p = 0.003), but the midline shifting of the SPD group did not differ from that of the SDD group (3.8 mm vs. 3.5 mm, p = 0.280). The recurrence rate in the SPD group did not differ significantly from that in the SDD group (13.2% vs. 8.5%, p = 0.351). The frequency of bleeding events after surgery also did not differ significantly (5.1% vs. 3.5% p-value = 0.636). In contrast to surgery-related morbidities, medical morbidities such as pneumonia were significantly higher in the SDD group (4.4% vs. 0.0%, p = 0.044). The all-cause mortality rates during the perioperative period did not differ between the two groups (5.1% vs. 3.4%, p = 0.848). CONCLUSION: Our findings may suggest that burr-hole trephination with SPD insertion had better surgical feasibility and fewer perioperative complications than SDD insertion. The type of anesthesia seems to be related with fewer medical complications at perioperative period. Larger, randomized clinical trials focusing not only the drain type but anesthesia type, are needed to validate our findings.


Asunto(s)
Drenaje/efectos adversos , Hematoma Subdural Crónico/cirugía , Trepanación/efectos adversos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Drenaje/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Procesos y Resultados en Atención de Salud , Periostio/cirugía , Complicaciones Posoperatorias , Estudios Retrospectivos , Espacio Subdural/cirugía , Trepanación/métodos , Adulto Joven
19.
PLoS One ; 16(12): e0260618, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34882724

RESUMEN

PURPOSE: Immune responses for cancer cells can be altered according to genetic variation of human leukocyte antigen (HLA). Association of HLA polymorphism with risk of various cancer types is well known. However, the association between HLA and glioblastoma (GBM) remains uncertain. We sought to evaluate the association of HLA polymorphism with risk of GBM development in Koreans. MATERIALS AND METHODS: A case-control study was performed to identify the odds ratios (OR) of HLA class I and II genes for GBM. The control group consisted of 142 healthy Korean volunteers, and the GBM group was 80 patients with newly diagnosed GBM at our institution. HLA class I (-A, -B, and-C) and class II (-DR, -DQ, and-DP) genotyping was performed by high-resolution polymerase chain reaction (PCR)-sequence-based typing (PCR-SBT) methods. RESULTS: There were significantly decreased frequencies of HLA-A*26:02 (OR 0.22 CI 0.05-0.98), HLA-C*08:01 (OR 0.29 CI 0.10-0.87), and HLA-DRB1*08:03 (OR 0.32 CI 0.11-0.98), while there was significantly increased frequency of HLA-C*04:01 (OR 2.29 CI 1.05-4.97). In analysis of haplotypes, the frequency of DRB1*14:05-DQB1*05:03 was significantly decreased (OR 0.22 CI 0.05-0.98). CONCLUSION: This study suggests that genetic variations of HLA may affect GBM development in Koreans. Further investigations with larger sample sizes are needed to delineate any potential role of the HLA polymorphisms in the pathogenesis of GBM development.


Asunto(s)
Pueblo Asiatico/genética , Glioblastoma/genética , Antígenos HLA-A/genética , Antígenos HLA-C/genética , Cadenas HLA-DRB1/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética , Técnicas de Genotipaje , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , República de Corea , Adulto Joven
20.
Neurooncol Adv ; 3(1): vdab098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34738083

RESUMEN

BACKGROUND: The association between height and the risk of developing primary brain malignancy remains unclear. We evaluated the association between height and risk of primary brain malignancy based on a nationwide population-based database of Koreans. METHODS: Using data from the Korean National Health Insurance System cohort, 6 833 744 people over 20 years of age that underwent regular national health examination were followed from January 2009 until the end of 2017. We documented 4771 cases of primary brain malignancy based on an ICD-10 code of C71 during the median follow-up period of 7.30 years and 49 877 983 person-years. RESULTS: When dividing the population into quartiles of height for each age group and sex, people within the highest height quartile had a significantly higher risk of brain malignancy, compared to those within the lowest height quartile (HR 1.21 CI 1.18-1.32) after adjusting for potential confounders. We also found that the risk of primary brain malignancy increased in proportion with the quartile increase in height. After analyzing subgroups based on older age (≥ 65) and sex, we found positive relationships between height and primary brain malignancy in all subgroups. CONCLUSIONS: This study is the first to suggest that height is associated with an increased risk of primary brain malignancy in the East-Asian population. Further prospective and larger studies with precise designs are needed to validate our findings.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA