Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Clinics (Sao Paulo) ; 78: 100212, 2023.
Article in English | MEDLINE | ID: mdl-37201304

ABSTRACT

OBJECTIVE: To explore the expression levels and clinical value of FKBP10 in lung adenocarcinoma brain metastases. DESIGN: A retrospective single-institution cohort study. PATIENTS: The perioperative records of 71 patients with lung adenocarcinoma brain metastases who underwent surgical resection at the authors' institution between November 2012 and June 2019 were retrospectively analyzed. METHODS: The authors evaluated FKBP10 expression levels using immunohistochemistry in tissue arrays of these patients. Kaplan-Meier survival curves were constructed, and a Cox proportional hazards regression model was used to identify independent prognostic biomarkers. A public database was used to detect FKBP10 expression and its clinical value in primary lung adenocarcinoma. RESULTS: The authors found that the FKBP10 protein was selectively expressed in lung adenocarcinoma brain metastases. Survival analysis showed that FKBP10 expression (p = 0.02, HR = 2.472, 95% CI [1.156, 5.289]), target therapy (p < 0.01, HR = 0.186, 95% CI [0.073, 0.477]), and radiotherapy (p = 0.006, HR = 0.330, 95% CI [0.149, 0.731]) were independent prognostic factors for survival in lung adenocarcinoma patients with brain metastases. The authors also detected FKBP10 expression in primary lung adenocarcinoma using a public database, found that FKBP10 is also selectively expressed in primary lung adenocarcinoma, and affects the overall survival and disease-free survival of patients. LIMITATIONS: The number of enrolled patients was relatively small and patients' treatment options varied. CONCLUSIONS: A combination of surgical resection, adjuvant radiotherapy, and precise target therapy may benefit the survival of selected patients with lung adenocarcinoma brain metastases. FKBP10 is a novel biomarker for lung adenocarcinoma brain metastases, which is closely associated with survival time and may serve as a potential therapeutic target.


Subject(s)
Adenocarcinoma of Lung , Brain Neoplasms , Lung Neoplasms , Humans , Adenocarcinoma of Lung/pathology , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Cohort Studies , Lung Neoplasms/pathology , Prognosis , Retrospective Studies , Tacrolimus Binding Proteins
2.
Onco Targets Ther ; 16: 179-187, 2023.
Article in English | MEDLINE | ID: mdl-36993872

ABSTRACT

Purpose: Several biomarkers, such as baseline neutrophil-to-lymphocyte ratio (NLR), have been more investigated in patients with brain metastases (BM), while their role in patients with leptomeningeal metastases (LM) has not been clarified. Considering the difference between the clinical behaviour of BM and LM, there is the need for addressing the role of these biomarkers in LM. Methods: The present study retrospectively analyzed 95 consecutive patients with LM from lung cancer who were diagnosed at the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences between January 2016 and December 2019. Baseline NLR, platelet-to-lymphocyte ratio (PLR), systemic immunoinflammation index (SII), and lymphocyte-to-monocyte ratio at diagnosis of LM were calculated based on complete blood count and correlated, along with other characteristics, with overall survival (OS) using univariate and multivariate analyses. The best cutoff values for systemic immunoinflammation biomarkers were derived using the surv_cutpoint function in R software, which optimized the significance of the split between Kaplan-Meier survival curves. Results: Median OS of patients with LM was 12 months (95% CI 9-17 months). On univariate analysis, NLR, PLR, SII, LMR, sex, smoking history, ECOG performance status (PS) scores, histological subtypes and targeted therapy were all significantly associated with OS. Only NLR (P=0.034, 95% CI 1.060-4.578) and ECOG PS scores (P=0.019, 95% CI 0.137-0.839) maintained a significant association with OS on multivariate analysis. Furthermore, patients with baseline NLR >3.57 had significantly worse OS than patients with NLR ≤3.57 (median OS 7 vs 17 months), as did patients with ECOG PS scores >2 vs ≤2 (median OS 4 vs 15 months). Conclusion: Both baseline NLR and PS scores at the time of LM diagnosis are helpful and available prognostic biomarkers for patients with LM from lung cancer.

3.
BMC Cancer ; 23(1): 104, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36717819

ABSTRACT

PURPOSE: To explore the potential pathogenesis and clinical features of second primary glioblastoma (spGBM) following first primary renal cell carcinoma (fpRCC). METHODS: Patients with spGBM after fpRCC were enrolled from our institution and the SEER dataset. Sanger sequencing, whole genome sequencing, and immunehistochemistry were used to detect molecular biomarkers. RESULTS: Four and 122 cases from our institution and the SEER dataset, respectively, were collected with an overall median age of 69 years at spGBM diagnosis following fpRCC. The median interval time between fpRCC and spGBM was 50.7 months and 4 years, for the four and 122 cases respectively. The median overall survival time was 11.2 and 6.0 months for the two datasets. In addition, spGBM patients of younger age (< 75 years) or shorter interval time (< 1 year) had favorable prognosis (p = 0.081 and 0.05, respectively). Moreover, the spGBM cases were molecularly classified as TERT only paired with TP53 mutation, PIK3CA mutation, EGFR alteration, low tumor mutation burden, and stable microsatellite status. CONCLUSIONS: This is the first study to investigate the pathogenesis and clinical features of spGBM following spRCC. We found that spGBMs are old-age related, highly malignant, and have short survival time. Moreover, they might be misdiagnosed and treated as brain metastases from RCC. Thus, the incidence of spGBMs after fpRCC is underestimated. Further studies are needed to investigate the underlying molecular mechanisms and clinical biomarkers for the development of spGBM following fpRCC.


Subject(s)
Carcinoma, Renal Cell , Glioblastoma , Kidney Neoplasms , Humans , Aged , Carcinoma, Renal Cell/pathology , Glioblastoma/pathology , Mutation , Genomics , Biomarkers, Tumor/genetics , Prognosis , Kidney Neoplasms/pathology
4.
Clinics ; 78: 100212, 2023. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1447988

ABSTRACT

Abstract Objective To explore the expression levels and clinical value of FKBP10 in lung adenocarcinoma brain metastases. Design A retrospective single-institution cohort study. Patients The perioperative records of 71 patients with lung adenocarcinoma brain metastases who underwent surgical resection at the authors' institution between November 2012 and June 2019 were retrospectively analyzed. Methods The authors evaluated FKBP10 expression levels using immunohistochemistry in tissue arrays of these patients. Kaplan-Meier survival curves were constructed, and a Cox proportional hazards regression model was used to identify independent prognostic biomarkers. A public database was used to detect FKBP10 expression and its clinical value in primary lung adenocarcinoma. Results The authors found that the FKBP10 protein was selectively expressed in lung adenocarcinoma brain metastases. Survival analysis showed that FKBP10 expression (p = 0.02, HR = 2.472, 95% CI [1.156, 5.289]), target therapy (p < 0.01, HR = 0.186, 95% CI [0.073, 0.477]), and radiotherapy (p = 0.006, HR = 0.330, 95% CI [0.149, 0.731]) were independent prognostic factors for survival in lung adenocarcinoma patients with brain metastases. The authors also detected FKBP10 expression in primary lung adenocarcinoma using a public database, found that FKBP10 is also selectively expressed in primary lung adenocarcinoma, and affects the overall survival and disease-free survival of patients. Limitations The number of enrolled patients was relatively small and patients' treatment options varied. Conclusions A combination of surgical resection, adjuvant radiotherapy, and precise target therapy may benefit the survival of selected patients with lung adenocarcinoma brain metastases. FKBP10 is a novel biomarker for lung adenocarcinoma brain metastases, which is closely associated with survival time and may serve as a potential therapeutic target.

5.
Front Oncol ; 12: 990762, 2022.
Article in English | MEDLINE | ID: mdl-36387224

ABSTRACT

Background: Systemic immune-inflammation states across the heterogeneous population of brain metastases from lung cancer are very important, especially in the context of complex brain-immune bidirectional communication. Previous studies from our team and others have shown that the L1 cell adhesion molecule (L1CAM) is deeply involved in the aggressive phenotype, immunosuppressive tumor microenvironment (TME), and metastasis during multiple malignancies, which may lead to an unfavorable outcome. However, little is known about the relationship between the L1CAM expression and the systemic immune-inflammation macroenvironment beyond the TME in brain metastases from lung cancer. Methods: Two cohorts of patients with brain metastases from lung cancer admitted to the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences, were studied in the present research. The L1CAM expression in cranial metastatic lesions by immunohistochemistry was explored in patients treated with neurosurgical resection, whereas the L1CAM expression in peripheral blood by ELISA was tested in patients treated with non-surgical antitumor management. Furthermore, based on peripheral blood cell counts in the CBC test, six systemic immune-inflammation biomarkers [neutrophil count, lymphocyte count, platelet count, systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio] were calculated. Then, the relationship between the L1CAM expression and these systemic immune-inflammation biomarkers was analyzed. In addition, these systemic immune-inflammation biomarkers were also used to compare the systemic immune-inflammation states in two cohorts of patients with brain metastases from lung cancer. Results: Positive L1CAM expressions in the metastatic brain lesions were accompanied with significantly increased peripheral platelet counts in patients treated with neurosurgical tumor resection (P < 0.05). Similarly, in patients treated with non-surgical antitumor management, L1CAM expressions in the peripheral blood were positively correlated with peripheral platelet counts (P < 0.05). In addition, patients prepared for neurosurgical tumor resection were presented with poorer systemic immune-inflammation states in comparison with the one with non-surgical antitumor management, which was characterized by a significant increase in peripheral neutrophil counts (P < 0.01), SII (P < 0.05), and NLR (P < 0.05) levels. Conclusion: The L1CAM expression in either the metastatic brain lesion or peripheral blood is positively correlated with the peripheral platelet count in patients with brain metastases from lung cancer. In addition, brain metastases that are prepared for neurosurgical tumor resection show poor systemic immune-inflammation states.

6.
Immun Inflamm Dis ; 10(10): e703, 2022 10.
Article in English | MEDLINE | ID: mdl-36169250

ABSTRACT

OBJECTIVES: Immune perturbation induced by tumor burden has been showed as the hallmark of brain tumors. To date, the vast majority of studies have focused heavily on local immune responses in the tumor microenvironment. Little is known about how the systemic immune macroenvironment is modulated by neurosurgical tumor resection in patients with brain tumors. METHOD: Medical records from patients with brain tumors admitted to the Department of Neurosurgery at the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences between January 2021 and March 2022 were retrospectively reviewed. Forty-nine patients who have lymphocyte subsets, serum immunoglobulins, C-reactive protein, and complements levels before neurosurgical tumor resection and at least once test after surgery were included into the final analysis. RESULTS: Postoperative CD3+ lymphocytes, CD4+ lymphocytes and CD4+ /CD8+ lymphocyte ratio presented bi-phasic changes, which indicated an initial decrease and a subsequent increase after neurosurgical tumor resection. Moreover, neurosurgical tumor resection induced a decrease in natural killer lymphocytes and an increase in B lymphocytes that persisted through the entire observation period after surgery. Meanwhile, significant changes in humoral immunity characterized by a decrease in immunoglobulins (IgA, IgG, and IgM) levels and an increase in the CRP level occurred after neurosurgical tumor resection. In addition, patients with postoperative infection complication had a lower preoperative CD4+ /CD8+ lymphocyte ratio. CONCLUSIONS: These findings provide evidence that either cellular immunity or humoral immunity can be remodeled by neurosurgical tumor resection, and patients with disturbed systemic immunity have increased risk of infection after surgery.


Subject(s)
Brain Neoplasms , C-Reactive Protein , Brain Neoplasms/surgery , CD4-Positive T-Lymphocytes , Humans , Immunoglobulin A , Immunoglobulin G , Immunoglobulin M , Retrospective Studies , Tumor Microenvironment
7.
Immun Inflamm Dis ; 10(10): e694, 2022 10.
Article in English | MEDLINE | ID: mdl-36169253

ABSTRACT

BACKGROUND: Systemic immune-inflammation states across the heterogeneous population of brain metastases are very important in the context of brain-immune bidirectional communication, especially among the patients needing neurosurgical resection. Four blood cell ratios based on complete blood count (CBC) test serving as prognostic biomarkers have been highlighted by previous studies, including systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR). However, the presurgical systemic immune-inflammation landscape in brain metastasis needing neurosurgical resection is limited. METHODS: Patients with brain metastases admitted to the Department of Neurosurgery at the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences between January 2016 and December 2019 were included. Based on peripheral blood cell counts in CBC test before neurosurgical resection, four systemic immune-inflammation biomarkers (SII, NLR, PLR, and LMR) were calculated. We characterized the changes of SII, NLR, PLR, and LMR in patients with brain metastasis before neurosurgical resection and the associations of these types of immune-inflammation states with patient demographics. In parallel, the corresponding data from the relative healthy populations without systemic diseases were enrolled as the control in the present study. RESULTS: Brain metastases induced systemic immune-inflammation perturbation, which was characterized by a significant increase in SII (p < .01) and NLR levels (p < .01) and a significant decrease in the LMR level (p < .01) in comparison with the healthy control group. Moreover, patients with male gender, less Karnofsky Performance Status (KPS) scores (<70), specific pathological subtypes, extracranial transfer, and history of both systemic and radiation therapy may have significant differences in one or more of these biomarkers, which indicated poorer systemic immune-inflammation states. CONCLUSIONS: This study provides evidence that brain metastasis is associated with perturbations in presurgical systemic immune-inflammation states. We should pay attention to the systemic immune-inflammation perturbations following brain metastasis in clinic, especially in the subpopulations with high risks.


Subject(s)
Brain Neoplasms , Lymphocytes , Biomarkers , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Humans , Inflammation/pathology , Lymphocytes/pathology , Male , Retrospective Studies
8.
Front Genet ; 13: 912227, 2022.
Article in English | MEDLINE | ID: mdl-35873494

ABSTRACT

Background: Studies have suggested that glioblastoma (GBM) cells originate from the subventricular zone (SVZ) and that GBM contact with the SVZ correlated with worse prognosis and higher recurrence. However, research on differentially expressed genes (DEGs) between GBM and the SVZ is lacking. Methods: We performed deep RNA sequencing on seven SVZ-involved GBMs and paired tumor-free SVZ tissues. DEGs and enrichment were assessed. We obtained GBM patient expression profiles and clinical data from the Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases. The least absolute shrinkage and selection operator Cox regression model was utilized to construct a multigene signature in the CGGA cohort. GBM patient data from TCGA cohort were used for validation. Results: We identified 137 (97 up- and 40 down-regulated) DEGs between GBM and healthy SVZ samples. Enrichment analysis revealed that DEGs were mainly enriched in immune-related terms, including humoral immune response regulation, T cell differentiation, and response to tumor necrosis factor, and the MAPK, cAMP, PPAR, PI3K-Akt, and NF-κb signaling pathways. An eight-gene (BCAT1, HPX, NNMT, TBX5, RAB42, TNFRSF19, C16orf86, and TRPC5) signature was constructed. GBM patients were stratified into two risk groups. High-risk patients showed significantly reduced overall survival compared with low-risk patients. Univariate and multivariate regression analyses indicated that the risk score level represented an independent prognostic factor. High risk score of GBM patients negatively correlated with 1p19q codeletion and IDH1 mutation. Immune infiltration analysis further showed that the high risk score was negatively correlated with activated NK cell and monocyte counts, but positively correlated with macrophage and activated dendritic cell counts and higher PD-L1 mRNA expression. Conclusion: Here, a novel gene signature based on DEGs between GBM and healthy SVZ was developed for determining GBM patient prognosis. Targeting these genes may be a therapeutic strategy for GBM.

9.
Clinics (Sao Paulo) ; 77: 100040, 2022.
Article in English | MEDLINE | ID: mdl-35525225

ABSTRACT

OBJECTIVES: Accurate prognosis assessment across the heterogeneous population of brain metastases is very important, which may facilitate clinical decision-making and appropriate stratification of future clinical trials. Previous studies have shown the L1 Cell Adhesion Molecule (L1CAM) is potentially involved in human malignancies of multiple different samples and unfavorable survival. However, no data of L1CAM are available for the brain metastases from lung adenocarcinoma, especially for the one with neurosurgical resection. METHOD: The authors investigated the L1CAM expression in cranial metastatic lesions for patients with brain metastases from lung adenocarcinoma after neurosurgical resection using tissue microarrays that were obtained from the Department of Neurosurgery at the Cancer Hospital of the Chinese Academy of Medical Sciences. Furthermore, the relationship between L1CAM expression and clinic-pathological parameters, including overall survival time, was analyzed to assess the prognostic value of L1CAM. RESULTS: L1CAM high expression was found in 62.30% of brain metastases from lung adenocarcinoma and significantly correlated with brain metastasis number (p = 0.028) and Lung-molGPA score (p = 0.042). Moreover, L1CAM expression was an independent predictor of survival for brain metastases after neurosurgical resection in a multivariate analysis. Patients with L1CAM high expression had unfavorable overall survival time (p = 0.016). In addition, the multivariate analysis also showed age and extracranial transfer were also the independent prognostic factors for this type of patient with brain metastases. CONCLUSIONS: A subset of brain metastases from lung adenocarcinoma aberrantly expresses L1CAM. L1CAM is a novel independent prognostic factor for brain metastasis from lung adenocarcinoma after neurosurgical resection.


Subject(s)
Adenocarcinoma of Lung , Brain Neoplasms , Lung Neoplasms , Neural Cell Adhesion Molecule L1 , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/surgery , Biomarkers, Tumor , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Neural Cell Adhesion Molecule L1/biosynthesis , Prognosis
10.
Transl Cancer Res ; 11(1): 63-71, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35261885

ABSTRACT

Background: Differentiating glioblastoma (GBM), brain metastases, and primary central nervous system lymphoma (PCNSL) in clinical practice is difficult. This study aimed to evaluate the diagnostic value of routine blood biomarkers in patients with GBM, brain metastases, and PCNSL and find a preoperative differential diagnostic tool for these tumors. Methods: The perioperative medical records of 70 GBM, 41 PCNSL, and 81 brain metastases patients and their preoperative blood test results were compared and analyzed, and a diagnostic model to differentiate among them established. Results: Patient age, plateletcrit, international normalized ratio (INR), and thrombin time (TT) were independently associated with differential diagnosis by multinomial logistic regression. Compared with GBM patients, brain metastases patients were significantly older (OR =1.055, 95% CI: 1.016-1.094, P=0.005) and had lower plateletcrit levels (OR =0.008, 95% CI: 0.004-0.017, P=0.027). In addition, patients with GBM had lower INR and higher TT than patients with the other two tumor types. A diagnostic model including these parameters, had an accuracy of 88.2% and 76.1% for brain metastases and GBM, respectively. Conclusions: Preoperative plateletcrit, INR, and TT may be used as inexpensive blood diagnostic biomarkers for differentiating brain metastases from other intracranial malignant tumors.

11.
Clinics ; 77: 100040, 2022. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1384616

ABSTRACT

Abstract Objectives Accurate prognosis assessment across the heterogeneous population of brain metastases is very important, which may facilitate clinical decision-making and appropriate stratification of future clinical trials. Previous studies have shown the L1 Cell Adhesion Molecule (L1CAM) is potentially involved in human malignancies of multiple different samples and unfavorable survival. However, no data of L1CAM are available for the brain metastases from lung adenocarcinoma, especially for the one with neurosurgical resection. Method The authors investigated the L1CAM expression in cranial metastatic lesions for patients with brain metastases from lung adenocarcinoma after neurosurgical resection using tissue microarrays that were obtained from the Department of Neurosurgery at the Cancer Hospital of the Chinese Academy of Medical Sciences. Furthermore, the relationship between L1CAM expression and clinic-pathological parameters, including overall survival time, was analyzed to assess the prognostic value of L1CAM. Results L1CAM high expression was found in 62.30% of brain metastases from lung adenocarcinoma and significantly correlated with brain metastasis number (p = 0.028) and Lung-molGPA score (p = 0.042). Moreover, L1CAM expression was an independent predictor of survival for brain metastases after neurosurgical resection in a multivariate analysis. Patients with L1CAM high expression had unfavorable overall survival time (p = 0.016). In addition, the multivariate analysis also showed age and extracranial transfer were also the independent prognostic factors for this type of patient with brain metastases. Conclusions A subset of brain metastases from lung adenocarcinoma aberrantly expresses L1CAM. L1CAM is a novel independent prognostic factor for brain metastasis from lung adenocarcinoma after neurosurgical resection.

12.
Immun Inflamm Dis ; 9(4): 1618-1630, 2021 12.
Article in English | MEDLINE | ID: mdl-34449972

ABSTRACT

INTRODUCTION: The prognosis of patients with glioma is dismal. It has been reported that Serpin peptidase inhibitor clade A member 3 (SERPINA3) is associated with the mobility and invasion of tumor cells. Our study was designed to explore the value of SERPINA3 messenger RNA (mRNA) expression in the biological process, prognosis, and immune significance in glioma. METHODS: We analyzed the biological functions of SERPINA3 through data from the Chinese Glioma Genome Atlas databases. Differentially expressed genes and enrichment analysis were performed and correlations between SERPINA3 expression and immune cell infiltration were analyzed. Further, we validated the expression and the survival prediction role of SERPINA3 by using tissue microarrays and RNAscope in situ hybridization in 321 gliomas. The correlations between the expression and clinical-pathological parameters as well as other biomarkers were examined. RESULTS: Univariate and multivariate regression both indicated that the level of SERPINA3 transcript represented an independent prognostic factor. High levels of SERPINA3 correlated with poor survival in patients with glioma. Expression of SERPINA3 mRNA was observed positively correlated with MCM6, IGFBP2, and FKBP10. Enrichment analysis showed SERPINA3 mainly enriched in immune-related terms and signaling pathways including MAPK, TNF, P53, PI3K-Akt, nuclear factor-κB. Immune infiltration analysis further declare the SERPINA3 expression negatively correlated with levels of Macrophages M1, native CD4+ T cell, monocytes, and Mast cell activated. And overexpression of SERPINA3 correlated with low CD4+ T cell infiltration in glioma tissues. CONCLUSIONS: SERPINA3 may play a key role in the biological process of glioma cells especially in immune suppression activities. SERPINA3 may serve as an independent survival prediction factor in glioma patients.


Subject(s)
Brain Neoplasms , Glioma , Serpins , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Glioma/genetics , Humans , Phosphatidylinositol 3-Kinases , Prognosis , Serpins/genetics
13.
Aging (Albany NY) ; 13(11): 15444-15458, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34100771

ABSTRACT

BACKGROUND: Diffuse gliomas are the most common malignant brain tumors, and immune checkpoint inhibitors have limited therapeutic effects against this cancer. Three oncogenic pathways are altered in diffuse gliomas: the RTK/Ras/PI3K/AKT signaling, TP53, and RB pathways. Although these pathways may affect the tumor immune microenvironment, their association with immunotherapy biomarkers remains unclear. METHODS: We used copy number variation and mutation data to stratify patients with specific oncogenic signaling alterations, and evaluated their correlation with predictive immunotherapy biomarkers, including tumor mutation burden (TMB), immune cytolytic activity (CYT), tumor purity, and tumor-infiltrating CD8+ T cells. Immune checkpoint expression and interferon-γ signaling activity were also compared in these samples. RESULTS: We identified differentially expressed genes in three distinct oncogenic pathways. Gene ontology analysis of these genes revealed the involvement of RTK/Ras/PI3K/AKT-associated genes in immune and inflammatory responses. Moreover, significantly elevated TMB, CYT, and numbers of CD8+ T cells and decreased tumor purity were correlated with altered RTK/Ras/PI3K/AKT signaling. Single cell sequencing also confirmed that this tumor subgroup had increased immune checkpoint expression and interferon-γ signaling activity. Immune phenotyping based on the presence of CD274 and TMB or CD274 and CD8 T+ cells indicated that tumors with altered RTK/Ras/PI3K/AKT pathways represent a beneficial subtype and are associated with improved survival. CONCLUSION: Altered RTK/Ras/PI3K/AKT signaling and immunotherapy biomarkers are strongly correlated in gliomas. Gliomas with altered expression of RTK/Ras/PI3K/AKT pathway components may be sensitive to immunotherapy. A combination of small-molecule kinase inhibitors and immunotherapy is proposed for this subgroup of tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Glioma/immunology , Glioma/therapy , Immunotherapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ras Proteins/metabolism , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Male , Middle Aged , Multivariate Analysis , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Survival Analysis , Treatment Outcome
14.
World Neurosurg ; 152: e62-e70, 2021 08.
Article in English | MEDLINE | ID: mdl-33940259

ABSTRACT

OBJECTIVE: Surgical treatment of advanced intracranial and extracranial communicating skull base tumors is challenging, especially for the reconstruction of the large composite defect left by tumor resection. The aim of the study is to evaluate the utility of the free flap reconstruction of the defects resulting from radical resection of these tumors in a single institution. METHODS: The clinical data of 17 consecutive patients who underwent free flap reconstruction for defect left by salvage resection of advanced intracranial and extracranial communicating tumors from 2013 to 2019 were retrospectively collected and analyzed. RESULTS: There were 5 squamous cell carcinomas, 4 adenoid cystic carcinomas, 2 basal cell carcinomas, 2 meningiomas, 1 anaplastic hemangiopericytoma, 1 pleomorphic adenoma, 1 osteosarcoma, and 1 chondrosarcoma. All patients had recurrent neoplasms, 2 of whom had pulmonary metastasis. A modified radical cervical dissection was performed in 6 patients. The anterolateral thigh myocutaneous flap and rectus abdominis myocutaneous flap were used in 15 patients (88.2%) and 2 patients (11.8%), respectively. Complications were seen in 3 of 17 patients (17.6%) with 1 total flap loss. The median progression-free survival duration was 31 months. The 3- and 5-year progression-free survival rates were 0.47 and 0.24, respectively. The mean overall survival duration was 66 months. The 3- and 5-year overall survival rates were 0.85 and 0.68, respectively. CONCLUSIONS: Free flap transfer is a safe and effective method with acceptable complications, useful for reconstruction of large composite skull base defects after salvage resection of advanced intracranial and extracranial communicating tumors. The functional and cosmetic results are satisfying.


Subject(s)
Brain Neoplasms/surgery , Free Tissue Flaps/transplantation , Plastic Surgery Procedures/methods , Salvage Therapy/methods , Skull Base Neoplasms/surgery , Skull Base/surgery , Adult , Aged , Brain Neoplasms/diagnostic imaging , Female , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , Skull Base/diagnostic imaging , Skull Base Neoplasms/diagnostic imaging , Treatment Outcome
15.
J Biomed Sci ; 28(1): 13, 2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33557829

ABSTRACT

BACKGROUND: Although the availability of therapeutic options including temozolomide, radiotherapy and some target agents following neurosurgery, the prognosis of glioma patients remains poor. Thus, there is an urgent need to explore possible targets for clinical treatment of this disease. METHODS: Tissue microarrays and immunohistochemistry were used to detect FKBP10, Hsp47, p-AKT (Ser473), p-CREB (Ser133) and PCNA expression in glioma tissues and xenografts. CCK-8 tests, colony formation assays and xenograft model were performed to test proliferation ability of FKBP10 in glioma cells in vitro and in vivo. Quantitative reverse transcriptase-PCR, western-blotting, GST-pull down, co-immunoprecipitation and confocal-immunofluorescence staining assay were used to explore the molecular mechanism underlying the functions of overexpressed FKBP10 in glioma cells. RESULTS: FKBP10 was highly expressed in glioma tissues and its expression was positively correlates with grade, poor prognosis. FKBP10-knockdown suppressed glioma cell proliferation in vitro and subcutaneous/orthotopic xenograft tumor growth in vivo. Silencing of FKBP10 reduced p-AKT (Ser473), p-CREB (Ser133), PCNA mRNA and PCNA protein expression in glioma cells. FKBP10 interacting with Hsp47 enhanced the proliferation ability of glioma cells via AKT-CREB-PCNA cascade. In addition, correlation between these molecules were also found in xenograft tumor and glioma tissues. CONCLUSIONS: We showed for the first time that FKBP10 is overexpressed in glioma and involved in proliferation of glioma cells by interacting with Hsp47 and activating AKT-CREB-PCNA signaling pathways. Our findings suggest that inhibition of FKBP10 related signaling might offer a potential therapeutic option for glioma patients.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/physiopathology , Tacrolimus Binding Proteins/genetics , Glioma/genetics , Heterografts , Humans , Immunohistochemistry , Tacrolimus Binding Proteins/metabolism , Tissue Array Analysis
16.
Oncol Lett ; 20(3): 2788-2796, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782596

ABSTRACT

S phase kinase-associated protein 2 (SKP2), a substrate recognizing protein, serves an important role in promoting cell cycle progression through ubiquitination and degradation of cell cycle inhibitors. In the present study, the clinical significance of SKP2 in gliomas was studied; 395 glioma specimens and 20 non-neoplastic tissues were collected and immunohistochemical analysis was performed. χ2 test was used to assess the associations between SKP2 expression and clinical parameters. Overall survival (OS) curves were plotted according to the Kaplan-Meier method. In the tested glioma samples, SKP2 expression was mainly observed in glioblastomas (GBMs). Survival analysis demonstrated that the overall survival time of the high SKP2 expression group was lower compared with the low SKP2 expression group (median OS, 10.04 months vs. 16.50 months; P=0.003). Moreover, SKP2 was independently associated with an unfavorable prognosis in GBMs. In addition, the expression of SKP2 was associated with the expression of phosphorylated retinoblastoma protein and the epidermal growth factor receptor. A combination of SKP2 expression along with isocitrate dehydrogenase 1 (IDH1) mutations and telomerase reverse transcriptase (TERT) promoter mutations was used to classify glioma patients for survival analysis. Patients with low SKP2 expression, IDH1 mutation and wild-type TERT promoter demonstrated the longest survival time. The findings of the present study, indicate that SKP2 is a potential prognostic biomarker in patients with GBMs.

17.
Front Oncol ; 10: 669, 2020.
Article in English | MEDLINE | ID: mdl-32528873

ABSTRACT

Temozolomide (TMZ) is considered a standard chemotherapeutic agent for glioblastoma (GBM). Characterizing the biological molecules and signaling pathways involved in TMZ sensitivity would be helpful for selecting therapeutic schemes and evaluating prognosis for GBM. Thus, in the present study, we selected 34 glioma cell lines paired with specific IC50 values of TMZ obtained from CancerRxGene and RNA-seq data downloaded from the Cancer Cell Line Encyclopedia to identify genes related to TMZ sensitivity. The results showed that 1,373 genes were related to the response of GBM cells to TMZ. Biological function analysis indicated that epithelial-mesenchymal transition, Wnt signaling, and immune response were the most significantly activated functions in TMZ-resistant cell lines. Additionally, negative regulation of telomere maintenance via telomerase was enriched in TMZ-sensitive glioma cell lines. We also preliminarily observed a synergistic effect of combination treatment comprising TMZ and a telomerase inhibitor in vitro. We identified six genes (MROH8, BET1, PTPRN2, STC1, NKX3-1, and ARMC10) using the random survival forests variable hunting algorithm based on the minimum error rate of the gene combination and constructed a gene expression signature. The signature was strongly related to GBM clinical characteristics and exhibited good prognosis accuracy for both The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) datasets. Patients in the high score group had a shorter survival time than those in the low score group (11.2 vs. 22.2 months, hazard ratio = 7.31, p = 4.59e-11) of the TCGA dataset. The CGGA dataset was selected as a validation group with 40 patients in the high score set and 43 patients in the low score set (12.5 vs. 28.8 months, hazard ratio = 3.42, p = 8.61e-5). Moreover, the signature showed a better prognostic value than MGMT promoter methylation in both datasets. We also developed a nomogram for clinical use that integrated the TMZ response signature and four other risk factors to individually predict patient survival after TMZ chemotherapy. Overall, our study provides promising therapeutic targets and potential guidance for adjuvant therapy of GBM.

18.
J Cancer ; 10(23): 5654-5660, 2019.
Article in English | MEDLINE | ID: mdl-31737102

ABSTRACT

Various hematological markers are associated with survival in patients with glioblastomas (GBMs), as they reflect inflammation and nutrition status. However, single markers are insufficient for predicting prognosis in GBM, and a comprehensive scoring system is needed. In this study, we developed a simple, inexpensive, and non-invasive scoring system, referred to as the Sanbo Scoring System (SSS), to predict survival in patients with GBMs. Patients with GBM were retrospectively assigned to two independent cohorts at Sanbo Brain Hospital and National Cancer Center/Cancer Hospital. Clinical records, including age, routine blood tests, biochemistry and coagulation examinations, and IDH-1 status, were collected. In total, 274 and 87 patients with GBMs at Sanbo Brain Hospital and National Cancer Center/Cancer Hospital were included as derivation and validation cohorts, retrospectively. We developed the SSS based on data for the derivation cohort, i.e., age, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), albumin-to-globulin ratio (AGR), and fibrinogen levels. These patients were divided into three groups that differed with respect to age, inflammation-nutrition status, and overall survival (p < 0.001), i.e., SSS 0, 1, and 2. NLR, PLR, and fibrinogen levels were lower and AGR was higher in the SSS 2 group than in the other groups, indicating better inflammation and nutrition statuses. Additionally, the longest overall survival was observed in this group. A multivariate analysis showed that SSS was an independent prognostic factor. The validation cohort supported all the results. SSS was a simple, non-invasive, and effective scoring system, and independently predicted survival in GBMs.

19.
Biosci Rep ; 39(6)2019 06 28.
Article in English | MEDLINE | ID: mdl-31138764

ABSTRACT

The prognosis of patients with glioblastoma (GBM) is dismal. It has been reported that Insulin-like growth factor (IGF) binding protein 2 (IGFBP2) is associated with the mobility and invasion of tumor cells. We investigated the expression of IGFBP2 mRNA in GBMs and its clinical relevance, using tissue microarrays and RNAscope in situ hybridization in 180 GBMs and 13 normal or edematous tissues. The correlations between the expression and clinical pathological parameters as well as some other biomarkers were analyzed. Overexpression of IGFBP2 mRNA was observed in 23.9% of tumors tested. No expression of IGFBP2 mRNA was detected in normal or edematous tissues. Kaplan-Meier survival analysis showed that the survival time of all the patients with high IGFBP2 tumors had shorter survival than those with low IGFBP2 (P<0.01). Univariate regression and multivariate regression both indicated that the expression of IGFBP2 transcript level was an independent prognostic factor (P=0.008 and 0.007, respectively). Furthermore, expression of IGFBP2 mRNA was related to the occurrence of isocitrate dehydrogenase 1 (IDH1) mutation, high heat shock protein 27 (Hsp27) expression and telomerase reverse transcriptase (TERT) promoter mutation (TERTp+) (P=0.013, 0.015 and 0.016, respectively), and patients with TERTp+/IGFBP2high showed the shortest survival. In conclusion, IGFBP2 mRNA expression status is an independent prognostic biomarker in GBMs, and the combination of IGFBP2 mRNA and TERTp status might serve as a prognostic indicator in patients with GBM.


Subject(s)
Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Neoplastic , Glioblastoma , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Biomarkers, Tumor/genetics , Disease-Free Survival , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Survival Rate
20.
J Neuroinflammation ; 15(1): 145, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29764444

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors have been shown to promote antitumor immunity and achieve durable tumor remissions. However, certain tumors are refractory to current immunotherapy. These negative results encouraged us to uncover other therapeutic targets and strategies. PTPN2 (protein tyrosine phosphatase, non-receptor type 2) has been newly identified as an immunotherapy target. Loss of PTPN2 sensitizes the tumor to immunotherapy via IFNγ signaling. METHODS: Here, we investigated the relationship between PTPN2 mRNA levels and clinical characteristics in gliomas. RNA-seq data of a cohort of 325 patients with glioma were available from the Chinese Glioma Genome Atlas and 671 from The Cancer Genome Atlas. R language, GraphPad Prism 5, and SPSS 22.0 were used to analyze data and draw figures. RESULTS: PTPN2 transcript levels increased significantly with higher grades of glioma and in isocitrate dehydrogenase (IDH) wild-type and mesenchymal subtype gliomas. A comprehensive biological analysis was conducted, which indicated a crucial role of PTPN2 in the immune and inflammation responses in gliomas. Specifically, PTPN2 was positively associated with HCK, LCK, MHC II, and STAT1 but negatively related to IgG and interferon. Moreover, canonical correlation analysis showed a positive correlation of PTPN2 with infiltrating immune cells, such as macrophages, neutrophils, and CD8+ T cells. Clinically, higher levels of PTPN2 were associated with a worse overall survival both in patients with gliomas and glioblastomas. CONCLUSION: PTPN2 expression level was increased in glioblastomas and associated with gliomas of the IDH wild-type and mesenchymal subtype. There was a close correlation between PTPN2 and the immune response and inflammatory activity in gliomas. Our results show that PTPN2 is a promising immunotherapy target and may provide additional treatment strategies.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , RNA, Messenger/metabolism , Area Under Curve , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cohort Studies , Correlation of Data , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic/immunology , Glioma/immunology , Glioma/mortality , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Macrophages/pathology , Male , Mutation/genetics , Neutrophils/pathology , Proto-Oncogene Proteins c-hck/metabolism , STAT1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...