1
|
Yadav VK, Sharma S, Maurya S, Singh RK, Saini J, Jain P, Patir R, Ahlawat S, Das S, Vaishya S, Agarwal S, Singh A, Gupta RK. Presence of Fragmented Intratumoral Thrombosed Microvasculature in the Necrotic and Peri-Necrotic Regions on SWI Differentiates IDH Wild-Type Glioblastoma From IDH Mutant Grade 4 Astrocytoma. J Magn Reson Imaging 2025. [PMID: 39781627 DOI: 10.1002/jmri.29695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Isocitrate dehydrogenase (IDH) wild-type (IDHwt) glioblastomas (GB) are more aggressive and have a poorer prognosis than IDH mutant (IDHmt) tumors, emphasizing the need for accurate preoperative differentiation. However, a distinct imaging biomarker for differentiation mostly lacking. Intratumoral thrombosis has been reported as a histopathological biomarker for GB. PURPOSE To evaluate the fragmented intratumoral thrombosed microvasculature (FTV) signs on susceptibility-weighted imaging (SWI) for distinguishing IDHwt and IDHmt tumors. STUDY TYPE Retrospective. SUBJECTS Ninety-seven treatment-naïve patients with histopathologically confirmed IDHwt GB (54 males, 26 females) and IDHmt grade 4 astrocytoma (13 males, 4 females). FIELD STRENGTH/SEQUENCE 3-T, SWI, fluid-attenuated-inversion-recovery (FLAIR), T1-weighted, T2-weighted, PD-weighted, post-contrast T1-weighted and dynamic-contrast-enhanced (DCE)-MRI. ASSESSMENT SWI data were evaluated by three experienced neuroradiologists (S.S., 11 years; J.S., 15 years; R.K.G., 40 years of experience), who assessed FTV presence in necrotic and peri-necrotic regions. FTV was identified as intratumoral susceptibility signal having minimal or no interslice connections. Quantitative DCE-MRI parameters were derived using first-pass-analysis and extended Tofts model. FLAIR abnormal, contrast-enhancing, and necrotic regions were segmented using in-house developed U-Net architecture. STATISTICAL TESTS Fleiss' Kappa, Cohen's Kappa, Shapiro-Wilk test, t tests or Mann-Whitney U test, receiver-operating characteristic (ROC) analysis, confusion matrix. A P-value <0.05 was considered statistically significant. RESULTS Fleiss' kappa test provided 91% inter-rater agreement, and Cohen's kappa provided intrarater agreement ranged from 81% to 97%. The raters' accuracy in distinguishing IDHwt from IDHmt ranged from 92% to 94%. Some of the quantitative DCE-MRI parameters (CBV, Ve, and Ktrans) provided statistically significant differences in differentiating IDHwt and IDHmt. Ktrans demonstrated 80.3% sensitivity and 81.2% specificity, with ROC analysis showing an AUC of 0.77. DATA CONCLUSION FTV signs in necrotic and peri-necrotic regions on SWI demonstrated a high accuracy in distinguishing IDHwt from IDHmt. Qualitative assessment of FTV signs showed almost perfect inter-rater and intrarater agreement. Quantitative DCE-MRI metrics also showed statistically significant differentiation of IDHwt and IDHmt. PLAIN LANGUAGE SUMMARY This study demonstrates that preoperative imaging, particularly the visualization of the fragmented thrombosed vasculature (FTV) sign on susceptibility-weighted imaging (SWI), effectively differentiates isocitrate dehydrogenase (IDH) wild-type (IDHwt) glioblastoma (GB) from IDH mutant (IDHmt) grade 4 astrocytomas. Over 90% of IDHwt GB patients displayed the FTV sign, a specific imaging biomarker absent in IDHmt cases. Perfusion parameters such as cerebral blood volume, Ve, and Ktrans were elevated in IDHwt gliomas, reflecting distinct vascular profiles. SWI offers a noninvasive and accurate diagnostic method, overcoming limitations of histopathology. Despite limitations like unequal sample sizes and retrospective analysis, this study underscores the clinical potential of SWI in improving glioma characterization and aiding treatment planning. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Shalini Sharma
- Department of Radiology, Fortis Memorial Research Institute, Gurugram, India
| | - Satyajit Maurya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Rakesh K Singh
- Department of Radiology, Fortis Memorial Research Institute, Gurugram, India
| | - Jitendra Saini
- Department of the Neuroimaging and Interventional Radiology, NIMHANS, Bengaluru, India
| | - Preeti Jain
- Department of Pathology, Agilus-Fortis Memorial Research Institute, Gurugram, India
| | - Rana Patir
- Department of Neurosurgery, Fortis Memorial Research Institute, Gurugram, India
| | - Sunita Ahlawat
- Department of Pathology, Agilus-Fortis Memorial Research Institute, Gurugram, India
| | - Sumanta Das
- Department of the Neuroimaging and Interventional Radiology, NIMHANS, Bengaluru, India
| | - Sandeep Vaishya
- Department of Neurosurgery, Fortis Memorial Research Institute, Gurugram, India
| | - Sumeet Agarwal
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
- Yardi School of Artificial Intelligence, Indian Institute of Technology Delhi, New Delhi, India
| | - Anup Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, India
- Yardi School of Artificial Intelligence, Indian Institute of Technology Delhi, New Delhi, India
| | - Rakesh K Gupta
- Department of Radiology, Fortis Memorial Research Institute, Gurugram, India
| |
Collapse
|
2
|
Min Z, Guo Y, Ning L. Paromomycin targets HDAC1-mediated SUMOylation and IGF1R translocation in glioblastoma. Front Pharmacol 2024; 15:1490878. [PMID: 39723246 PMCID: PMC11668589 DOI: 10.3389/fphar.2024.1490878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/08/2024] [Indexed: 12/28/2024] Open
Abstract
Objective This study investigates the effects of Paromomycin on SUMOylation-related pathways in glioblastoma (GBM), specifically targeting HDAC1 inhibition. Methods Using TCGA and GTEx datasets, we identified SUMOylation-related genes associated with GBM prognosis. Molecular docking analysis suggested Paromomycin as a potential HDAC1 inhibitor. In vitro assays on U-251MG GBM cells were performed to assess Paromomycin's effects on cell viability, SUMOylation gene expression, and IGF1R translocation using CCK8 assays, qRT-PCR, and immunofluorescence. Results Paromomycin treatment led to a dose-dependent reduction in GBM cell viability, colony formation, and migration. It modulated SUMO1 expression and decreased IGF1R nuclear translocation, an effect reversible by the HDAC1 inhibitor Trochostatin A (TSA), suggesting Paromomycin's involvement in SUMO1-regulated pathways. Conclusion This study highlights Paromomycin's potential as a therapeutic agent for GBM by targeting HDAC1-mediated SUMOylation pathways and influencing IGF1R translocation, warranting further investigation for its clinical application.
Collapse
|
3
|
Luo Y, Liu R, Zhang H, Wang H, Yin H, Tian G, Wang B, Yan Y, Ding Z, Dai J, Niu L, Yuan G, Pan Y. Amantadine against glioma via ROS-mediated apoptosis and autophagy arrest. Cell Death Dis 2024; 15:834. [PMID: 39548081 PMCID: PMC11568115 DOI: 10.1038/s41419-024-07228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Glioma is a common primary nervous system malignant tumor with poor overall cure rate and low survival rate, yet successful treatment still remains a challenge. Here, we demonstrated that amantadine (AMT) exhibits the powerful anti-glioma effect by promoting apoptosis and autophagy in vivo and in vitro. Mechanistically, amantadine induces a large amount of reactive oxygen species (ROS) accumulation in glioma cells, and then triggers apoptosis by destroying mitochondria. In addition, amantadine induces the initiation of autophagy and inhibits the fusion of autophagosome and lysosome, consequently performing an anti-glioma role. Taken together, our findings suggest that amantadine could be a promising anti-glioma drug that inhibits glioma cells by inducing apoptosis and autophagy, which may provide a novel potential treatment option for patients.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - He Zhang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hongyu Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hang Yin
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guopeng Tian
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bo Wang
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yunji Yan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zilin Ding
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Junqiang Dai
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Liang Niu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
4
|
Zang Y, Feng L, Zheng F, Shi X, Chen X. Clinicopathological and radiological characteristics of false-positive and false-negative results in T2-FLAIR mismatch sign of IDH-mutated gliomas. Clin Neurol Neurosurg 2024; 246:108579. [PMID: 39395280 DOI: 10.1016/j.clineuro.2024.108579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/16/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
PURPOSE To explore the clinicopathological and radiological characteristics associated with false-positive and false-negative results in the identification of isocitrate dehydrogenase (IDH) mutations in gliomas using the T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign. METHODS In 1515 patients with cerebral gliomas, tumor location, restricted diffusion using diffusion-weighted imaging, and the T2-FLAIR mismatch sign were retrospectively analyzed using preoperative magnetic resonance imaging. Moreover, both the false-positive and false-negative results of the T2-FLAIR mismatch sign were obtained. Univariate and multivariate logistic analyses were performed to evaluate the risk factors associated with false-positive and false-negative results. RESULTS The overall false-positive rate was 3.5 % (53/1515), and its independent risk factors were the patient's age (adjusted odds ratio [OR], 0.977; 95 % confidence interval [CI], 0.957, 0.997; P = 0.027) and non-restricted diffusion (adjusted OR, 1.968; 95 % CI, 1.060, 3.652; P = 0.032). The overall false-negative rate was 39.7 % (602/1515); its independent risk factors were the patient's age (adjusted OR, 1.022; 95 % CI, 1.005, 1.038; P = 0.008), 1p/19q co-deletion (adjusted OR, 3.334; 95 % CI, 1.913, 5.810; P < 0.001), and telomerase reverse transcriptase promoter mutation (adjusted OR, 2.004; 95 % CI, 1.181, 3.402; P = 0.010). For the mismatch sign in idiopathic IDH, the area under the receiver operating characteristic curve (AUC) was 0.602. The combined AUC for the T2-FLAIR mismatch sign and risk factors was 0.871. CONCLUSIONS Clinicopathological and radiological characteristics can lead to the misinterpretation of IDH status in gliomas based on the T2-FLAIR mismatch sign. However, this can be avoided if careful attention is paid.
Collapse
Affiliation(s)
- Yuying Zang
- Department of Radiology, The Affiliated Children's Hospital, Capital Institute of Pediatrics, Beijing, China; Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Limei Feng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Fei Zheng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Radiology, Peking University people' hospital, Peking University, Beijing, China.
| | - Xinyao Shi
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xuzhu Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Nakase T, Guerra GA, Ostrom QT, Ge T, Melin BS, Wrensch M, Wiencke JK, Jenkins RB, Eckel-Passow JE, Bondy ML, Francis SS, Kachuri L. Genome-wide polygenic risk scores predict risk of glioma and molecular subtypes. Neuro Oncol 2024; 26:1933-1944. [PMID: 38916140 PMCID: PMC11448969 DOI: 10.1093/neuonc/noae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Polygenic risk scores (PRS) aggregate the contribution of many risk variants to provide a personalized genetic susceptibility profile. Since sample sizes of glioma genome-wide association studies (GWAS) remain modest, there is a need to efficiently capture genetic risk using available data. METHODS We applied a method based on continuous shrinkage priors (PRS-CS) to model the joint effects of over 1 million common variants on disease risk and compared this to an approach (PRS-CT) that only selects a limited set of independent variants that reach genome-wide significance (P < 5 × 10-8). PRS models were trained using GWAS stratified by histological (10 346 cases and 14 687 controls) and molecular subtype (2632 cases and 2445 controls), and validated in 2 independent cohorts. RESULTS PRS-CS was generally more predictive than PRS-CT with a median increase in explained variance (R2) of 24% (interquartile range = 11-30%) across glioma subtypes. Improvements were pronounced for glioblastoma (GBM), with PRS-CS yielding larger odds ratios (OR) per standard deviation (SD) (OR = 1.93, P = 2.0 × 10-54 vs. OR = 1.83, P = 9.4 × 10-50) and higher explained variance (R2 = 2.82% vs. R2 = 2.56%). Individuals in the 80th percentile of the PRS-CS distribution had a significantly higher risk of GBM (0.107%) at age 60 compared to those with average PRS (0.046%, P = 2.4 × 10-12). Lifetime absolute risk reached 1.18% for glioma and 0.76% for IDH wildtype tumors for individuals in the 95th PRS percentile. PRS-CS augmented the classification of IDH mutation status in cases when added to demographic factors (AUC = 0.839 vs. AUC = 0.895, PΔAUC = 6.8 × 10-9). CONCLUSIONS Genome-wide PRS has the potential to enhance the detection of high-risk individuals and help distinguish between prognostic glioma subtypes.
Collapse
Affiliation(s)
- Taishi Nakase
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, USA
| | - Geno A Guerra
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Quinn T Ostrom
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Beatrice S Melin
- Department of Diagnostics and Intervention, Oncology Umeå University, Umeå, Sweden
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Melissa L Bondy
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Stephen S Francis
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Stefan H, Bösebeck F, Rössler K. Brain tumor-associated epilepsies in adulthood: Current state of diagnostic and individual treatment options. Seizure 2024:S1059-1311(24)00161-4. [PMID: 38910076 DOI: 10.1016/j.seizure.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Brain tumors are one of the most frequent causes of structural epilepsy and set a major burden on treatment costs and the social integrity of patients. Although promising oncological treatment strategies are already available, epileptological treatment is often intractable and requires lifelong epileptological care. Therefore, treatment strategies must be adapted to age-related needs, and specific aspects of late-onset epilepsy (LOE) must be considered. The practical implementation of individual decisions from tumor boards and the current state of the art in scientific knowledge about pathological mechanisms, modern diagnostic procedures and biomarkers, and patient-individualized treatment options into practical epileptological disease management is a prerequisite. This narrative review focuses on the current work progress regarding pathogenesis, diagnosis, and therapy. Exemplarily, interdisciplinary approaches for optimized individualized therapy will be discussed, emphasizing the combination of neurological-epileptological and oncological perspectives.
Collapse
Affiliation(s)
- Hermann Stefan
- Department of Neurology, Biomagnetism, University Hospital Erlangen, Germany; Private Practice, 50, Allee am Röthelheimpark, Erlangen, Germany.
| | - Frank Bösebeck
- AGAPLESION Diakonieklinikum Rotenburg, Neurological Clinic - Epilepsy Center, Rotenburg, Germany
| | - Karl Rössler
- Medizinische Universität Wien, Klinik für Neurochirurgie, Wien, Austria
| |
Collapse
|
7
|
Nakase T, Guerra G, Ostrom QT, Ge T, Melin B, Wrensch M, Wiencke JK, Jenkins RB, Eckel-Passow JE, Bondy ML, Francis SS, Kachuri L. Genome-wide Polygenic Risk Scores Predict Risk of Glioma and Molecular Subtypes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.10.24301112. [PMID: 38260701 PMCID: PMC10802631 DOI: 10.1101/2024.01.10.24301112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Polygenic risk scores (PRS) aggregate the contribution of many risk variants to provide a personalized genetic susceptibility profile. Since sample sizes of glioma genome-wide association studies (GWAS) remain modest, there is a need to find efficient ways of capturing genetic risk factors using available germline data. Methods We developed a novel PRS (PRS-CS) that uses continuous shrinkage priors to model the joint effects of over 1 million polymorphisms on disease risk and compared it to an approach (PRS-CT) that selects a limited set of independent variants that reach genome-wide significance (P<5×10-8). PRS models were trained using GWAS results stratified by histological (10,346 cases, 14,687 controls) and molecular subtype (2,632 cases, 2,445 controls), and validated in two independent cohorts. Results PRS-CS was consistently more predictive than PRS-CT across glioma subtypes with an average increase in explained variance (R2) of 21%. Improvements were particularly pronounced for glioblastoma tumors, with PRS-CS yielding larger effect sizes (odds ratio (OR)=1.93, P=2.0×10-54 vs. OR=1.83, P=9.4×10-50) and higher explained variance (R2=2.82% vs. R2=2.56%). Individuals in the 95th percentile of the PRS-CS distribution had a 3-fold higher lifetime absolute risk of IDH mutant (0.63%) and IDH wildtype (0.76%) glioma relative to individuals with average PRS. PRS-CS also showed high classification accuracy for IDH mutation status among cases (AUC=0.895). Conclusions Our novel genome-wide PRS may improve the identification of high-risk individuals and help distinguish between prognostic glioma subtypes, increasing the potential clinical utility of germline genetics in glioma patient management.
Collapse
Affiliation(s)
- Taishi Nakase
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Geno Guerra
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Quinn T. Ostrom
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology Umeå University, Umeå, Sweden
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - John K. Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Robert B. Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Melissa L. Bondy
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen S. Francis
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Linda Kachuri
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Li J, Guo Q, Xing R. Construction and validation of an immune infiltration-related risk model for predicting prognosis and immunotherapy response in low grade glioma. BMC Cancer 2023; 23:727. [PMID: 37543576 PMCID: PMC10403952 DOI: 10.1186/s12885-023-11222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/25/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Low grade glioma (LGG) is considered a heterogeneous tumor with highly variable survival and limited efficacy of immunotherapy. To identify high-risk subsets and apply immunotherapy effectively in LGG, the status and function of immune infiltration in the glioma microenvironment must be explored. METHODS Four independent glioma cohorts comprising 1,853 patients were enrolled for bioinformatics analysis. We used ConsensusClusterPlus to cluster patients into four different immune subtypes based on immune infiltration. The immune-infiltration signature (IIS) was constructed by LASSO regression analysis. Somatic mutation and copy number variation (CNV) analyses were performed to explore genomic and transcriptomic traits in the high- and low- risk groups. The correlation between response to programmed cell death 1 (PD-1) blockade and the IIS risk score was confirmed in an in vivo glioma model. RESULTS Patients were clustered into four different immune subtypes based on immune infiltration, and the high immune infiltration subtype was associated with worse survival in LGG. The high immune infiltration subtype had stronger inflammatory response, immune response and immune cell chemotaxis. The IIS, consisting of EMP3, IQGAP2, METTL7B, SLC1A6 and TNFRSF11B, could predict LGG malignant progression, which was validated with internal clinical samples. M2 macrophage infiltration positively correlated with the IIS risk score. The high-risk group had significantly more somatic mutations and CNVs. The IIS risk score was related to immunomodulatory molecules and could predict immunotherapy clinical benefit. In vivo, immunotherapy-sensitive glioma model exhibited higher IIS risk score and more infiltration of immune cells, especially M2 macrophages. The IIS risk score was decreased in an immunotherapy-sensitive glioma model after anti-PD1 immunotherapy. CONCLUSION Different immune subtypes of LGG had unique immune cell infiltration characteristics, and the high immune infiltration subtype was associated with immunosuppressive signaling pathways. A novel IIS prognostic model based on immune infiltration status was constructed for immunophenotypic classification, risk stratification, prognostication and immunotherapy response prediction in LGG.
Collapse
Affiliation(s)
- Jinna Li
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Qing Guo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110000, China.
| | - Rui Xing
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
9
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
10
|
Xue H, Han Z, Li H, Li X, Jia D, Qi M, Zhang H, Zhang K, Gong J, Wang H, Feng Z, Ni S, Han B, Li G. Application of Intraoperative Rapid Molecular Diagnosis in Precision Surgery for Glioma: Mimic the World Health Organization CNS5 Integrated Diagnosis. Neurosurgery 2023; 92:762-771. [PMID: 36607719 PMCID: PMC10508407 DOI: 10.1227/neu.0000000000002260] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/22/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND With the advent of the molecular era, the diagnosis and treatment systems of glioma have also changed. A single histological type cannot be used for prognosis grade. Only by combining molecular diagnosis can precision medicine be realized. OBJECTIVE To develop an automatic integrated gene detection system (AIGS) for intraoperative detection in glioma and to explore its positive role in intraoperative diagnosis and treatment. METHODS We analyzed the isocitrate dehydrogenase 1 (IDH1) mutation status of 105 glioma samples and evaluated the product's potential value for diagnosis; 37 glioma samples were detected intraoperatively to evaluate the feasibility of using the product in an actual situation. A blinding method was used to evaluate the effect of the detection technology on the accuracy of intraoperative histopathological diagnosis by pathologists. We also reviewed the current research status in the field of intraoperative molecular diagnosis. RESULTS Compared with next-generation sequencing, the accuracy of AIGS in detecting IDH1 was 100% for 105 samples and 37 intraoperative samples. The blind diagnostic results were compared between the 2 groups, and the molecular information provided by AIGS increased the intraoperative diagnostic accuracy of glioma by 16.2%. Using the technical advantages of multipoint synchronous detection, we determined the tumor molecular margins for 5 IDH-positive patients and achieved accurate resection at the molecular level. CONCLUSION AIGS can quickly and accurately provide molecular information during surgery. This methodology not only improves the accuracy of intraoperative pathological diagnosis but also provides an important molecular basis for determining tumor margins to facilitate precision surgery.
Collapse
Affiliation(s)
- Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Shandong, China
| | - Zhe Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Shandong, China
| | - Haiyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Xueen Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Deze Jia
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Mei Qi
- Department of Pathology, Shandong University Qilu Hospital, Shandong, China
| | - Hui Zhang
- Shandong Key Laboratory of Brain Function Remodeling, Shandong, China
| | - Kailiang Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Jie Gong
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Hongwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Zichao Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Bo Han
- Department of Pathology, Shandong University Qilu Hospital, Shandong, China
- Department of Pathology, Shandong University School of Basic Medical Sciences, Shandong, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Shandong, China
- Shandong Key Laboratory of Brain Function Remodeling, Shandong, China
| |
Collapse
|
11
|
Chiu FY, Yen Y. Imaging biomarkers for clinical applications in neuro-oncology: current status and future perspectives. Biomark Res 2023; 11:35. [PMID: 36991494 DOI: 10.1186/s40364-023-00476-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Biomarker discovery and development are popular for detecting the subtle diseases. However, biomarkers are needed to be validated and approved, and even fewer are ever used clinically. Imaging biomarkers have a crucial role in the treatment of cancer patients because they provide objective information on tumor biology, the tumor's habitat, and the tumor's signature in the environment. Tumor changes in response to an intervention complement molecular and genomic translational diagnosis as well as quantitative information. Neuro-oncology has become more prominent in diagnostics and targeted therapies. The classification of tumors has been actively updated, and drug discovery, and delivery in nanoimmunotherapies are advancing in the field of target therapy research. It is important that biomarkers and diagnostic implements be developed and used to assess the prognosis or late effects of long-term survivors. An improved realization of cancer biology has transformed its management with an increasing emphasis on a personalized approach in precision medicine. In the first part, we discuss the biomarker categories in relation to the courses of a disease and specific clinical contexts, including that patients and specimens should both directly reflect the target population and intended use. In the second part, we present the CT perfusion approach that provides quantitative and qualitative data that has been successfully applied to the clinical diagnosis, treatment and application. Furthermore, the novel and promising multiparametric MR imageing approach will provide deeper insights regarding the tumor microenvironment in the immune response. Additionally, we briefly remark new tactics based on MRI and PET for converging on imaging biomarkers combined with applications of bioinformatics in artificial intelligence. In the third part, we briefly address new approaches based on theranostics in precision medicine. These sophisticated techniques merge achievable standardizations into an applicatory apparatus for primarily a diagnostic implementation and tracking radioactive drugs to identify and to deliver therapies in an individualized medicine paradigm. In this article, we describe the critical principles for imaging biomarker characterization and discuss the current status of CT, MRI and PET in finiding imaging biomarkers of early disease.
Collapse
Affiliation(s)
- Fang-Ying Chiu
- Center for Cancer Translational Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Center for Brain and Neurobiology Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Teaching and Research Headquarters for Sustainable Development Goals, Tzu Chi University, Hualien City, 970374, Taiwan.
| | - Yun Yen
- Center for Cancer Translational Research, Tzu Chi University, Hualien City, 970374, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei City, 110301, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City, 110301, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei City, 110301, Taiwan.
- Cancer Center, Taipei Municipal WanFang Hospital, Taipei City, 116081, Taiwan.
| |
Collapse
|
12
|
Vanhauwaert D, Pinson H, Vanschoenbeek K, Dedeurwaerdere F, De Gendt C, Boterberg T, De Vleeschouwer S. Cancer Registration, Molecular Marker Status, and Adherence to the WHO 2016 Classification of Pathology Reports for Glioma Diagnosed during 2017-2019 in Belgium. Pathobiology 2023; 90:365-376. [PMID: 36702113 DOI: 10.1159/000529320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION The objective of this study was to cross-check and, if necessary, adjust registered ICD-O-3 topography and morphology codes with the findings in pathology reports available at the Belgian Cancer Registry (BCR) for glioma patients. Additionally, integration of molecular markers in the pathological diagnosis and concordance with WHO 2016 classification is investigated. METHODS Since information regarding molecular tests and corresponding conclusions are not available as structured data at population level, a manual screening of all pseudonymized pathology reports available at the BCR for registered glioma patients (2017-2019) was conducted. ICD-O-3 morphology and topography codes from the BCR database (based on information as provided by hospital oncological care programmes and pathology laboratories), were, at tumour level, cross-checked with the data from the pathology reports and, if needed, specified or corrected. Relevant molecular markers (IDH1/2, 1p19q codeletion, promoter region of the MGMT gene [MGMTp]) were manually extracted from the pathology reports. RESULTS In 95.3% of gliomas, the ICD-O-3 morphology code was correct. Non-specific topography codes were specified in 9.3%, while 3.3% of specific codes were corrected. The IDH status was known in 75.2% of astrocytic tumours. The rate of correct integrated diagnoses varied from 47.6% to 56.4% among different gliomas. MGMTp methylation status was available in 32.2% of glioblastomas. CONCLUSION Both the integration of molecular markers in the conclusion of the pathology reports and the delivery of those reports to the BCR can be improved. The availability of distinct ICD-O-3 codes for each molecularly defined tumour entity within the WHO classification would increase the consistency of cancer registration, facilitate population level research and international benchmarking.
Collapse
Affiliation(s)
| | - Harry Pinson
- Department of Neurosurgery, Ghent University Hospital, Ghent, Belgium
| | | | | | | | - Tom Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Steven De Vleeschouwer
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
- Department Neurosciences and Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Romano A, Palizzi S, Romano A, Moltoni G, Di Napoli A, Maccioni F, Bozzao A. Diffusion Weighted Imaging in Neuro-Oncology: Diagnosis, Post-Treatment Changes, and Advanced Sequences-An Updated Review. Cancers (Basel) 2023; 15:cancers15030618. [PMID: 36765575 PMCID: PMC9913305 DOI: 10.3390/cancers15030618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
DWI is an imaging technique commonly used for the assessment of acute ischemia, inflammatory disorders, and CNS neoplasia. It has several benefits since it is a quick, easily replicable sequence that is widely used on many standard scanners. In addition to its normal clinical purpose, DWI offers crucial functional and physiological information regarding brain neoplasia and the surrounding milieu. A narrative review of the literature was conducted based on the PubMed database with the purpose of investigating the potential role of DWI in the neuro-oncology field. A total of 179 articles were included in the study.
Collapse
Affiliation(s)
- Andrea Romano
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
| | - Serena Palizzi
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
| | - Allegra Romano
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
| | - Giulia Moltoni
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
- Correspondence: ; Tel.: +39-3347906958
| | - Alberto Di Napoli
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Francesca Maccioni
- Department of Radiology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Alessandro Bozzao
- NESMOS Department, U.O.C. Neuroradiology, “Sant’Andrea” University Hospital, 00189 Rome, Italy
| |
Collapse
|
14
|
Sharma N, Mallela AN, Shi DD, Tang LW, Abou-Al-Shaar H, Gersey ZC, Zhang X, McBrayer SK, Abdullah KG. Isocitrate dehydrogenase mutations in gliomas: A review of current understanding and trials. Neurooncol Adv 2023; 5:vdad053. [PMID: 37287696 PMCID: PMC10243983 DOI: 10.1093/noajnl/vdad053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Isocitrate dehydrogenase (IDH) is a key enzyme in normal metabolism and homeostasis. However, mutant forms of IDH are also defining features of a subset of diffuse gliomas. In this review, we highlight current techniques targeting IDH-mutated gliomas and summarize current and completed clinical trials exploring these strategies. We discuss clinical data from peptide vaccines, mutant IDH (mIDH) inhibitors, and PARP inhibitors. Peptide vaccines have the unique advantage of targeting the specific epitope of a patient's tumor, inducing a highly tumor-specific CD4+ T-cell response. mIDH-inhibitors, on the other hand, specifically target mutant IDH proteins in cancer cell metabolism and thus help halt gliomagenesis. We also explore PARP inhibitors and their role in treating diffuse gliomas, which exploit IDH-mutant diffuse gliomas by allowing the persistence of unrepaired DNA complexes. We summarize various completed and current trials targeting IDH1 and IDH2 mutations in diffuse gliomas. Therapies targeting mutant IDH have significant promise in treating progressive or recurrent IDH-mutant gliomas and may significantly change treatment paradigms in the next decade.
Collapse
Affiliation(s)
- Nikhil Sharma
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Arka N Mallela
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diana D Shi
- Harvard Radiation Oncology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Lilly W Tang
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussam Abou-Al-Shaar
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zachary C Gersey
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Xiaoran Zhang
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Samuel K McBrayer
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Wang Y, Zhang J, Luo C, Yao Y, Qin G, Wu J. Predictive models and survival analysis of postoperative mental health disturbances in adult glioma patients. Front Oncol 2023; 13:1153455. [PMID: 37152011 PMCID: PMC10160603 DOI: 10.3389/fonc.2023.1153455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
Background and Objectives Patients with primary malignant brain tumors may experience mental health disturbances that can significantly affect their daily life. This study aims to identify risk factors and generate predictive models for postoperative mental health disturbances (PMHDs) in adult glioma patients in accordance with different clinical periods; additionally, survival analyses will be performed. Methods This longitudinal cohort study included 2,243 adult patients (age at diagnosis ≥ 18 years) with nonrecurrent glioma who were pathologically diagnosed and had undergone initial surgical resection. Six indicators of distress, sadness, fear, irritability, mood and enjoyment of life, ranging from 0-10, were selected to assess PMHDs in glioma patients in the third month after surgery, mainly referring to the M.D. Anderson Symptom Inventory Brain Tumor Module (MDASI-BT). Factor analysis (FA) was applied on these indicators to divide participants into PMHD and control groups based on composite factor scores. Survival analyses were performed, and separate logistic regression models were formulated for preoperative and postoperative factors predicting PMHDs. Results A total of 2,243 adult glioma patients were included in this study. Based on factor analysis results, 300 glioma patients had PMHDs in the third postoperative month, and the remaining 1,943 were controls. Candidate predictors for PMHDs in the preoperative model were associated with age, clinical symptoms (intracranial space-occupying lesion, muscle weakness and memory deterioration), and tumor location (corpus callosum, basal ganglia and brainstem), whereas age, clinical symptoms (nausea and memory deterioration), tumor location (basal ganglia and brainstem), hospitalization days, WHO grade 4, postoperative chemotherapy or radiotherapy and postoperative Karnofsky Performance Scale (KPS) served as important factors in the postoperative model. In addition, the median overall survival (OS) time for glioma patients with PMHDs was 19 months, compared to 13 months for glioblastoma, IDH-wild type (GBM) patients with PMHDs. Conclusion The risk factors for PMHDs were identified. These findings may provide new insights into predicting the probability of PMHD occurrence in glioma patients in addition to aiding effective early intervention and improving prognosis based on different clinical stages.
Collapse
Affiliation(s)
- Yi Wang
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Municipal Health Commission, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Municipal Health Commission, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
| | - Ye Yao
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
- *Correspondence: Ye Yao, ; Jinsong Wu,
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neurosurgical Institute, Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai Municipal Health Commission, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- *Correspondence: Ye Yao, ; Jinsong Wu,
| |
Collapse
|
16
|
Beyond Imaging and Genetic Signature in Glioblastoma: Radiogenomic Holistic Approach in Neuro-Oncology. Biomedicines 2022; 10:biomedicines10123205. [PMID: 36551961 PMCID: PMC9775324 DOI: 10.3390/biomedicines10123205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor exhibiting rapid and infiltrative growth, with less than 10% of patients surviving over 5 years, despite aggressive and multimodal treatments. The poor prognosis and the lack of effective pharmacological treatments are imputable to a remarkable histological and molecular heterogeneity of GBM, which has led, to date, to the failure of precision oncology and targeted therapies. Identification of molecular biomarkers is a paradigm for comprehensive and tailored treatments; nevertheless, biopsy sampling has proved to be invasive and limited. Radiogenomics is an emerging translational field of research aiming to study the correlation between radiographic signature and underlying gene expression. Although a research field still under development, not yet incorporated into routine clinical practice, it promises to be a useful non-invasive tool for future personalized/adaptive neuro-oncology. This review provides an up-to-date summary of the recent advancements in the use of magnetic resonance imaging (MRI) radiogenomics for the assessment of molecular markers of interest in GBM regarding prognosis and response to treatments, for monitoring recurrence, also providing insights into the potential efficacy of such an approach for survival prognostication. Despite a high sensitivity and specificity in almost all studies, accuracy, reproducibility and clinical value of radiomic features are the Achilles heel of this newborn tool. Looking into the future, investigators' efforts should be directed towards standardization and a disciplined approach to data collection, algorithms, and statistical analysis.
Collapse
|
17
|
Xu Q, Xu QQ, Shi N, Dong LN, Zhu H, Xu K. A multitask classification framework based on vision transformer for predicting molecular expressions of glioma. Eur J Radiol 2022; 157:110560. [DOI: 10.1016/j.ejrad.2022.110560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
|
18
|
Guo H, Liu J, Hu J, Zhang H, Zhao W, Gao M, Zhang Y, Yang G, Cui Y. Diagnostic performance of gliomas grading and IDH status decoding A comparison between 3D amide proton transfer APT and four diffusion-weighted MRI models. J Magn Reson Imaging 2022; 56:1834-1844. [PMID: 35488516 PMCID: PMC9790544 DOI: 10.1002/jmri.28211] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The focus of neuro-oncology research has changed from histopathologic grading to molecular characteristics, and medical imaging routinely follows this change. PURPOSE To compare the diagnostic performance of amide proton transfer (APT) and four diffusion models in gliomas grading and isocitrate dehydrogenase (IDH) genotype. STUDY TYPE Prospective. POPULATION A total of 62 participants (37 males, 25 females; mean age, 52 ± 13 years) whose IDH genotypes were mutant in 6 of 14 grade II gliomas, 8 of 20 of grade III gliomas, and 4 of 28 grade IV gliomas. FIELD STRENGTH/SEQUENCE APT imaging using sampling perfection with application optimized contrasts by using different flip angle evolutions (SPACE) and DWI with q-space Cartesian grid sampling were acquired at 3 T. ASSESSMENT The ability of diffusion kurtosis imaging, diffusion kurtosis imaging, neurite orientation dispersion and density imaging (NODDI), mean apparent propagator (MAP), and APT imaging for glioma grade and IDH status were assessed, with histopathological grade and genetic testing used as a reference standard. Regions of interest (ROIs) were drawn by two neuroradiologists after consensus. STATISTICAL TESTS T-test and Mann-Whitney U test; one-way analysis of variance (ANOVA); receiver operating curve (ROC) and area under the curve (AUC); DeLong test. P value < 0.05 was considered statistically significant. RESULTS Compared with IDH-mutant gliomas, IDH-wildtype gliomas showed a significantly higher mean, 5th-percentile (APT5 ), and 95th-percentile from APTw, the 95th-percentile value of axial, mean, and radial diffusivity from DKI, and 95th-percentile value of isotropic volume fraction from NODDI, and no significantly different parameters from DTI and MAP (P = 0.075-0.998). The combined APT model showed a significantly wider area under the curve (AUC 0.870) for IDH status, when compared with DKI and NODDI. APT5 was significantly different between two of the three groups (glioma II vs. glioma III vs. glioma IV: 1.35 ± 0.75 vs. 2.09 ± 0.93 vs. 2.71 ± 0.81). DATA CONCLUSION APT has higher diagnostic accuracy than DTI, DKI, MAP, and NODDI in glioma IDH genotype. APT5 can effectively identify both tumor grading and IDH genotyping, making it a promising biomarker for glioma classification. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Hu Guo
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Road, ChangshaHunan410011China
| | - Jun Liu
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Road, ChangshaHunan410011China,Department of Radiology Quality Control CenterHunan ProvinceChangsha410011China
| | - JunJiao Hu
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Road, ChangshaHunan410011China
| | - HuiTing Zhang
- MR Scientific Marketing, Siemens Healthineers Ltd.Wuhan430071China
| | - Wei Zhao
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Road, ChangshaHunan410011China
| | - Min Gao
- Department of RadiologyThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Road, ChangshaHunan410011China
| | - Yi Zhang
- Department of Biomedical EngineeringCollege of Biomedical Engineering & Instrument Science, Zhejiang UniversityHangzhouZhejiangChina
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic ResonanceSchool of Physics and Electronic, East China Normal UniversityShanghaiChina
| | - Yan Cui
- Department of NeurosurgeryThe Second Xiangya Hospital, Central South UniversityNo. 139 Middle Renmin Rd, ChangshaHunan Province410011P.R. China
| |
Collapse
|
19
|
Ma L, Li G, Yang T, Zhang L, Wang X, Xu X, Ni H. An inhibitor of BRD4, GNE987, inhibits the growth of glioblastoma cells by targeting C-Myc and S100A16. Cancer Chemother Pharmacol 2022; 90:431-444. [PMID: 36224471 PMCID: PMC9637061 DOI: 10.1007/s00280-022-04483-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Among children, glioblastomas (GBMs) are a relatively common type of brain tumor. BRD4 expression was elevated in GBM and negatively correlated with the prognosis of glioma. We investigated the anti-GBM effects of a novel BRD4 inhibitor GNE987. METHODS We evaluated the anti-tumor effect of GNE987 in vitro and in vivo by Western blot, CCK8, flow cytometry detection, clone formation, the size of xenografts, and Ki67 immunohistochemical staining, and combined ChIP-seq with RNA-seq techniques to find its anti-tumor mechanism. RESULTS In vitro experiments showed that GNE987 significantly degraded BRD4, inhibited the proliferation of GBM cells, blocked the cell cycle, and induced apoptosis. Similarly, in vivo experiments, GNE987 also inhibited GBM growth as seen from the size of xenografts and Ki67 immunohistochemical staining. Based on Western blotting, GNE987 can significantly reduce the protein level of C-Myc; meanwhile, we combined ChIP-seq with RNA-seq techniques to confirm that GNE987 downregulated the transcription of S100A16 by disturbing H3K27Ac. Furthermore, we validated that S100A16 is indispensable in GBM growth. CONCLUSION GNE987 may be effective against GBM that targets C-Myc expression and influences S100A16 transcription through downregulation of BRD4.
Collapse
Affiliation(s)
- Liya Ma
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
- Medical College of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Tianquan Yang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Li Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Xinxin Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Xiaowen Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Hong Ni
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China.
| |
Collapse
|
20
|
Luo Y, Tian G, Fang X, Bai S, Yuan G, Pan Y. Ferroptosis and Its Potential Role in Glioma: From Molecular Mechanisms to Therapeutic Opportunities. Antioxidants (Basel) 2022; 11:2123. [PMID: 36358495 PMCID: PMC9686959 DOI: 10.3390/antiox11112123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/20/2022] [Accepted: 10/26/2022] [Indexed: 09/29/2023] Open
Abstract
Glioma is the most common intracranial malignant tumor, and the current main standard treatment option is a combination of tumor surgical resection, chemotherapy and radiotherapy. Due to the terribly poor five-year survival rate of patients with gliomas and the high recurrence rate of gliomas, some new and efficient therapeutic strategies are expected. Recently, ferroptosis, as a new form of cell death, has played a significant role in the treatment of gliomas. Specifically, studies have revealed key processes of ferroptosis, including iron overload in cells, occurrence of lipid peroxidation, inactivation of cysteine/glutathione antiporter system Xc- (xCT) and glutathione peroxidase 4 (GPX4). In the present review, we summarized the molecular mechanisms of ferroptosis and introduced the application and challenges of ferroptosis in the development and treatment of gliomas. Moreover, we highlighted the therapeutic opportunities of manipulating ferroptosis to improve glioma treatments, which may improve the clinical outcome.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guopeng Tian
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Xiang Fang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Shengwei Bai
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guoqiang Yuan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yawen Pan
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
21
|
Muacevic A, Adler JR. Clinical, Therapeutic, and Prognostic Experience in Patients With Glioblastoma. Cureus 2022; 14:e29856. [PMID: 36381939 PMCID: PMC9635935 DOI: 10.7759/cureus.29856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Glioblastoma (GB) represents the most aggressive type of glioma with a poor prognosis despite the therapies used. As of today, data availability for therapeutic and prognosis experiences is limited. The cornerstone for this study is to create a framework overview of Mexico´s experience throughout 17 years of research. METHODS Retrospective analysis from 2000 to 2017 including patients with a histological diagnosis of GB was performed. Data were collected from the ABC Medical Center and the Neurology and Neurosurgery National Institute. RESULTS One hundred and thirty-seven patients were included with a mean age of 54 years. Histological diagnosis was made in all patients, of which 58.1% had a total resection, 31.6% had a partial resection, and 10.3% of them underwent biopsy. In all cases, patients received treatment under the following conditions: 10 patients were treated exclusively with stereotactic radiotherapy (RT). In 55 patients, a combination of RT and TMZ was used, the other 40 patients received RT plus CBP. Eighteen patients RT added to nitrosourea medication and lastly, 14 patients received a combination of RT/TMZ and Bevacizumab, a monoclonal antibody that inhibits the formation of blood vessels (BVZ). The progression-free survival (PFS) and overall survival (OS) were higher in the RT/TMZ/BVZ group (16.5 to 22.9 months) and the RT/TMZ group (11 to 17 months), the prognostic parameters included: Isocitrate dehydrogenase 1 mutation (IDH1), usage of BVZ and TMZ in the PLS and OS, considering as well, age range (<70 years) as a favorable prognostic factor. CONCLUSIONS GB represents the most frequent intracranial neoplasia. Combined fractionated stereotactic RT added to Temozolomide and Bevacizumab received in our population reports favorable and superior results compared to the ones described in the literature. Further studies are necessary to know the biological behavior of our population.
Collapse
|
22
|
Chen S, Jiang J, Shen A, Miao Y, Cao Y, Zhang Y, Cong P, Gao P. Rewired Metabolism of Amino Acids and Its Roles in Glioma Pathology. Metabolites 2022; 12:918. [PMID: 36295820 PMCID: PMC9611130 DOI: 10.3390/metabo12100918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Amino acids (AAs) are indispensable building blocks of diverse bio-macromolecules as well as functional regulators for various metabolic processes. The fact that cancer cells live with a voracious appetite for specific AAs has been widely recognized. Glioma is one of the most lethal malignancies occurring in the central nervous system. The reprogrammed metabolism of AAs benefits glioma proliferation, signal transduction, epigenetic modification, and stress tolerance. Metabolic alteration of specific AAs also contributes to glioma immune escape and chemoresistance. For clinical consideration, fluctuations in the concentrations of AAs observed in specific body fluids provides opportunities to develop new diagnosis and prognosis markers. This review aimed at providing an extra dimension to understanding glioma pathology with respect to the rewired AA metabolism. A deep insight into the relevant fields will help to pave a new way for new therapeutic target identification and valuable biomarker development.
Collapse
Affiliation(s)
- Sirui Chen
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Jingjing Jiang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ao Shen
- HE University, Shenyang 110163, China
| | - Ying Miao
- E&M College, Shenyang Aerospace University, Shenyang 110136, China
| | - Yunfeng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Ying Zhang
- Internal Medicine Department, Dalian Public Health Clinical Center, Dalian 116033, China
| | - Peiyu Cong
- Neurosurgery Department, Affiliated Dalian Municipal Central Hospital of Dalian Medical University, Dalian 116022, China
| | - Peng Gao
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
23
|
Gong Y, Wei S, Wei Y, Chen Y, Cui J, Yu Y, Lin X, Yan H, Qin H, Yi L. IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review). Oncol Lett 2022; 24:278. [PMID: 35814829 PMCID: PMC9260733 DOI: 10.3892/ol.2022.13398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022] Open
Abstract
As the risk of harmful environmental exposure is increasing, it is important to find suitable targets for the diagnosis and treatment of the diseases caused. Isocitrate dehydrogenase 2 (IDH2) is an enzyme located in the mitochondria; it plays an important role in numerous cell processes, including maintaining redox homeostasis, participating in the tricarboxylic acid cycle and indirectly taking part in the transmission of the oxidative respiratory chain. IDH2 mutations promote progression in acute myeloid leukemia, glioma and other diseases. The present review mainly summarizes the role and mechanism of IDH2 with regard to the biological effects, such as the mitophagy and apoptosis of animal or human cells, caused by environmental pollution such as radiation, heavy metals and other environmental exposure factors. The possible mechanisms of these biological effects are described in terms of IDH2 expression, reduced nicotine adenine dinucleotide phosphate content and reactive oxygen species level, among other variables. The impact of environmental pollution on human health is increasingly attracting attention. IDH2 may therefore become useful as a potential diagnostic and therapeutic target for environmental exposure-induced diseases.
Collapse
Affiliation(s)
- Ya Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuan Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yong Chen
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Cui
- Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yue Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong Yan
- Pediatric Intensive Care Unit, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
24
|
DNA methylation-regulated YTHDF2 correlates with cell migration and immune cell infiltration in glioma. Aging (Albany NY) 2022; 14:7774-7793. [PMID: 35661004 DOI: 10.18632/aging.204104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Glioma is a lethal malignant brain tumor, it comprises about 80% of all malignant brain tumours. Mounting evidence has reported that YTHDF2 plays a significant role in the cancer progression. However, the effects of YTHDF2 on the prognosis of low-grade gliomas (LGGs) and its correlation with tumor immune infiltration are unclear. The present study was designed to determine the biological functions of YTHDF2 in glioma and to evaluate the association of YTHDF2 expression with glioma progression. METHODS Clinical data on patients with glioma were obtained from The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), the Gene Expression Omnibus (GEO), as well as the Rembrandt and Gravendeel databases. The correlations among YTHDF2 expression, pathological characteristics, glioma progression and clinical outcome were evaluated. In addition, the correlation of YTHDF2 expression with immune cell infiltration was analyzed too. RESULTS We found that YTHDF2 was significantly up-regulated in LGGs which correlated with tumor grade and poor prognosis. Interestingly, we showed that YTHDF2 expression in LGG was associated with copy number variation, DNA hypomethylation, and induced transcription factor YY1. Besides, KEGG pathway analysis shows that YTHDF2 mainly participates in the immune response and oncogenic signaling pathway. Additionally, YTHDF2 is positively associated with diverse immune cells infiltration, immune cells, and multiple immune checkpoint molecules. Finally, we confirmed that YTHDF2 was highly expressed in LGGs tissues and correlated with the tumor grade with immunohistochemistry assay. More importantly, our results demonstrated that YTHDF2 was elevated in GBM cells. Knockdown of YTHDF2 significantly inhibits the proliferation and migration of GBM cells. CONCLUSION YTHDF2 correlates with glioma progression and immune cell infiltration, suggesting that YTHDF2 may be a useful prognostic biomarker for glioma.
Collapse
|
25
|
Wu W, Wang Y, Xiang J, Li X, Wahafu A, Yu X, Bai X, Yan G, Wang C, Wang N, Du C, Xie W, Wang M, Wang J. A Novel Multi-Omics Analysis Model for Diagnosis and Survival Prediction of Lower-Grade Glioma Patients. Front Oncol 2022; 12:729002. [PMID: 35646656 PMCID: PMC9133344 DOI: 10.3389/fonc.2022.729002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 03/24/2022] [Indexed: 01/13/2023] Open
Abstract
Background Lower-grade gliomas (LGGs) are characterized by remarkable genetic heterogeneity and different clinical outcomes. Classification of LGGs is improved by the development of molecular stratification markers including IDH mutation and 1p/19q chromosomal integrity, which are used as a hallmark of survival and therapy sensitivity of LGG patients. However, the reproducibility and sensitivity of the current classification remain ambiguous. This study aimed to construct more accurate risk-stratification approaches. Methods According to bioinformatics, the sequencing profiles of methylation and transcription and imaging data derived from LGG patients were analyzed and developed predictable risk score and radiomics score. Moreover, the performance of predictable models was further validated. Results In this study, we determined a cluster of 6 genes that were correlated with IDH mutation/1p19q co-deletion status. Risk score model was calculated based on 6 genes and showed gratifying sensitivity and specificity for survival prediction and therapy response of LGG patients. Furthermore, a radiomics risk score model was established to noninvasively assist judgment of risk score in pre-surgery. Taken together, a predictable nomogram that combined transcriptional signatures and clinical characteristics was established and validated to be preferable to the histopathological classification. Our novel multi-omics nomograms showed a satisfying performance. To establish a user-friendly application, the nomogram was further developed into a web-based platform: https://drw576223193.shinyapps.io/Nomo/, which could be used as a supporting method in addition to the current histopathological-based classification of gliomas. Conclusions Our novel multi-omics nomograms showed the satisfying performance of LGG patients and assisted clinicians to draw up individualized clinical management.
Collapse
Affiliation(s)
- Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianyang Xiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaodong Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Alafate Wahafu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Bai
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ge Yan
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chunbao Wang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Changwang Du
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanfu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
26
|
Johnson GW, Han RH, Smyth MD, Leuthardt EC, Kim AH. Laser Interstitial Thermal Therapy in Grade 2/3 IDH1/2 Mutant Gliomas: A Preliminary Report and Literature Review. Curr Oncol 2022; 29:2550-2563. [PMID: 35448183 PMCID: PMC9028957 DOI: 10.3390/curroncol29040209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022] Open
Abstract
Laser interstitial thermal therapy (LITT) has become an increasingly utilized alternative to surgical resection for the treatment of glioma in patients. However, treatment outcomes in isocitrate dehydrogenase 1 and 2 (IDH1/2) mutant glioma, specifically, have not been reported. The objective of this study was to characterize a single institution’s cohort of IDH1/2 mutant grade 2/3 glioma patients treated with LITT. We collected data on patient presentation, radiographic features, tumor molecular profile, complications, and outcomes. We calculated progression-free survival (PFS) and tested factors for significant association with longer PFS. Overall, 22.7% of our cohort experienced progression at a median follow up of 1.8 years. The three- and five-year estimates of PFS were 72.5% and 54.4%, respectively. This is the first study to characterize outcomes in patients with IDH1/2 mutant glioma after LITT. Our results suggest that LITT is an effective treatment option for IDH1/2 mutant glioma.
Collapse
Affiliation(s)
- Gabrielle W. Johnson
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (G.W.J.); (R.H.H.); (E.C.L.)
| | - Rowland H. Han
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (G.W.J.); (R.H.H.); (E.C.L.)
| | - Matthew D. Smyth
- Department of Neurosurgery, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA;
| | - Eric C. Leuthardt
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (G.W.J.); (R.H.H.); (E.C.L.)
- Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Albert H. Kim
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (G.W.J.); (R.H.H.); (E.C.L.)
- Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Correspondence:
| |
Collapse
|
27
|
Sethi MK, Downs M, Shao C, Hackett WE, Phillips JJ, Zaia J. In-Depth Matrisome and Glycoproteomic Analysis of Human Brain Glioblastoma Versus Control Tissue. Mol Cell Proteomics 2022; 21:100216. [PMID: 35202840 PMCID: PMC8957055 DOI: 10.1016/j.mcpro.2022.100216] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor. The extracellular matrix, also known as the matrisome, helps determine glioma invasion, adhesion, and growth. Little attention, however, has been paid to glycosylation of the extracellular matrix components that constitute the majority of glycosylated protein mass and presumed biological properties. To acquire a comprehensive understanding of the biological functions of the matrisome and its components, including proteoglycans (PGs) and glycosaminoglycans (GAGs), in GBM tumorigenesis, and to identify potential biomarker candidates, we studied the alterations of GAGs, including heparan sulfate (HS) and chondroitin sulfate (CS), the core proteins of PGs, and other glycosylated matrisomal proteins in GBM subtypes versus control human brain tissue samples. We scrutinized the proteomics data to acquire in-depth site-specific glycoproteomic profiles of the GBM subtypes that will assist in identifying specific glycosylation changes in GBM. We observed an increase in CS 6-O sulfation and a decrease in HS 6-O sulfation, accompanied by an increase in unsulfated CS and HS disaccharides in GBM versus control samples. Several core matrisome proteins, including PGs (decorin, biglycan, agrin, prolargin, glypican-1, and chondroitin sulfate proteoglycan 4), tenascin, fibronectin, hyaluronan link protein 1 and 2, laminins, and collagens, were differentially regulated in GBM versus controls. Interestingly, a higher degree of collagen hydroxyprolination was also observed for GBM versus controls. Further, two PGs, chondroitin sulfate proteoglycan 4 and agrin, were significantly lower, about 6-fold for isocitrate dehydrogenase-mutant, compared to the WT GBM samples. Differential regulation of O-glycopeptides for PGs, including brevican, neurocan, and versican, was observed for GBM subtypes versus controls. Moreover, an increase in levels of glycosyltransferase and glycosidase enzymes was observed for GBM when compared to control samples. We also report distinct protein, peptide, and glycopeptide features for GBM subtypes comparisons. Taken together, our study informs understanding of the alterations to key matrisomal molecules that occur during GBM development. (Data are available via ProteomeXchange with identifier PXD028931, and the peaks project file is available at Zenodo with DOI 10.5281/zenodo.5911810).
Collapse
Affiliation(s)
- Manveen K Sethi
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - Margaret Downs
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - Chun Shao
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - William E Hackett
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California, USA; Division of Neuropathology, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Joseph Zaia
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
28
|
León-García MC, Silva-Gaona OG, Hernández-Ortiz M, Vargas-Ortiz K, Ramírez-Emiliano J, Garay-Sevilla ME, Encarnación-Guevara S, Pérez-Vázquez V. Curcumin Prevents the Glycation of Tricarboxylic Acid Cycle and Cell Respiration Proteins in the Heart of Mice Fed with a High-Fructose Diet. Curr Pharm Des 2022; 28:1769-1778. [PMID: 35362381 DOI: 10.2174/1381612828666220331160501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND A high fructose diet (HFD) induces protein glycation. The latter is related to a higher risk of cardiovascular disease. Curcumin is a natural pleiotropic compound that may possess antiglycant properties. OBJECTIVE To analyze the effect of curcumin on the content of glycated proteins in the hearts of 6-week-old mice fed with a HFD for 15 weeks. METHODS Mice were allocated in four groups (n = 6/group): a control group that received a standard diet (CT); a group that received 30% w/v fructose in water (F); a group that received 0.75% w/w curcumin supplemented in food (C); a group that received 30% w/v fructose in water and 0.75% w/w curcumin supplemented in food (F+C). The content of glycated proteins in the heart was determined by Western Blot (whereas the spots were detected by 2D-PAGE) using anti-AGE and anti-CML antibodies. A densitometric analysis was performed using the ImageLab software. Glycated proteins were identified by MALDI-TOF-MS, and an ontological analysis was performed in terms of biological processes and molecular function based on the STRING and DAVID databases. RESULTS Fourteen glycated protein spots were detected, two of them with anti-AGE and the other 12 with anti-CML. In total, eleven glycated proteins were identified, out of which three had decreased glycation levels due to curcumin exposure. The identified proteins participate in processes such as cellular respiration, oxidative phosphorylation, lipid metabolism, carbohydrate metabolism, the tricarboxylic acid cycle (TAC), and the organization of intermediate filaments. CONCLUSIONS Curcumin decreased the fructose-induced glycation level of the ACO2, NDUFS7, and DLAT proteins.
Collapse
Affiliation(s)
- María Cristina León-García
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| | - Oscar Gerardo Silva-Gaona
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| | | | - Katya Vargas-Ortiz
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| | - Joel Ramírez-Emiliano
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| | - Ma Eugenia Garay-Sevilla
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| | | | - Victoriano Pérez-Vázquez
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato, México
| |
Collapse
|
29
|
Löhr M, Härtig W, Schulze A, Kroiß M, Sbiera S, Lapa C, Mages B, Strobel S, Hundt JE, Bohnert S, Kircher S, Janaki-Raman S, Monoranu CM. SOAT1: A Suitable Target for Therapy in High-Grade Astrocytic Glioma? Int J Mol Sci 2022; 23:ijms23073726. [PMID: 35409086 PMCID: PMC8998855 DOI: 10.3390/ijms23073726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
Targeting molecular alterations as an effective treatment for isocitrate dehydrogenase-wildtype glioblastoma (GBM) patients has not yet been established. Sterol-O-Acyl Transferase 1 (SOAT1), a key enzyme in the conversion of endoplasmic reticulum cholesterol to esters for storage in lipid droplets (LD), serves as a target for the orphan drug mitotane to treat adrenocortical carcinoma. Inhibition of SOAT1 also suppresses GBM growth. Here, we refined SOAT1-expression in GBM and IDH-mutant astrocytoma, CNS WHO grade 4 (HGA), and assessed the distribution of LD in these tumors. Twenty-seven GBM and three HGA specimens were evaluated by multiple GFAP, Iba1, IDH1 R132H, and SOAT1 immunofluorescence labeling as well as Oil Red O staining. To a small extent SOAT1 was expressed by tumor cells in both tumor entities. In contrast, strong expression was observed in glioma-associated macrophages. Triple immunofluorescence labeling revealed, for the first time, evidence for SOAT1 colocalization with Iba1 and IDH1 R132H, respectively. Furthermore, a notable difference in the amount of LD between GBM and HGA was observed. Therefore, SOAT1 suppression might be a therapeutic option to target GBM and HGA growth and invasiveness. In addition, the high expression in cells related to neuroinflammation could be beneficial for a concomitant suppression of protumoral microglia/macrophages.
Collapse
Affiliation(s)
- Mario Löhr
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany;
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany;
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Matthias Kroiß
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany;
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Silviu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| | - Bianca Mages
- Institute for Anatomy, University of Leipzig, 04103 Leipzig, Germany;
| | - Sabrina Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
| | | | - Simone Bohnert
- Institute of Forensic Medicine, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Stefan Kircher
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
| | - Sudha Janaki-Raman
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
- Correspondence: ; Tel.: +49-931-3181184
| |
Collapse
|
30
|
Cindil E, Sendur HN, Cerit MN, Erdogan N, Celebi F, Dag N, Celtikci E, Inan A, Oner Y, Tali T. Prediction of IDH Mutation Status in High-grade Gliomas Using DWI and High T1-weight DSC-MRI. Acad Radiol 2022; 29 Suppl 3:S52-S62. [PMID: 33685792 DOI: 10.1016/j.acra.2021.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/24/2021] [Accepted: 02/03/2021] [Indexed: 01/09/2023]
Abstract
RATIONALE AND OBJECTIVES We aimed to evaluate the diagnostic performance of diffusion-weighted imaging (DWI) and dynamic susceptibility contrast-enhanced (DSC) magnetic resonance imaging (MRI) parameters in the noninvasive prediction of the isocitrate dehydrogenase (IDH) mutation status in high-grade gliomas (HGGs). MATERIALS AND METHODS A total of 58 patients with histopathologically proved HGGs were included in this retrospective study. All patients underwent multiparametric MRI on 3-T, including DSC-MRI and DWI before surgery. The mean apparent diffusion coefficient (ADC), relative maximum cerebral blood volume (rCBV), and percentage signal recovery (PSR) of the tumor core were measured and compared depending on the IDH mutation status and tumor grade. The Mann-Whitney U test was used to detect statistically significant differences in parameters between IDH-mutant-type (IDH-m-type) and IDH-wild-type (IDH-w-type) HGGs. Receiver operating characteristic curve (ROC) analysis was performed to evaluate the diagnostic performance. RESULTS The rCBV was significantly higher, and the PSR value was significantly lower in IDH-w-type tumors than in the IDH-m group (p = 0.002 and <0.001, respectively).The ADC value in IDH-w-type tumors was significantly lower compared with the one in IDH-m types (p = 0.023), but remarkable overlaps were found between the groups. The PSR showed the best diagnostic performance with an AUC of 0.938 and with an accuracy rate of 0.87 at the optimal cutoff value of 86.85. The combination of the PSR and the rCBV for the identification of the IDH mutation status increased the discrimination ability at the AUC level of 0.955. In terms of each tumor grade, the PSR and rCBV showed significant differences between the IDH-m and IDH-w groups (p ≤0.001). CONCLUSION The rCBV and PSR from DSC-MRI may be feasible noninvasive imaging parameters for predicting the IDH mutation status in HGGs. The standardization of the imaging protocol is indispensable to the utility of DSC perfusion MRI in wider clinical usage.
Collapse
|
31
|
Gong S, Wu C, Köhler F, Meixensberger J, Schopow N, Kallendrusch S. Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase Family: Novel Prognostic Biomarkers and Tumor Microenvironment Regulators for Lower-Grade Glioma. Front Cell Neurosci 2022; 16:838548. [PMID: 35250490 PMCID: PMC8894330 DOI: 10.3389/fncel.2022.838548] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Lower-grade glioma (LGG) is a group of tumors arising from the cells of the central nervous system. Although various therapy interventions are used, the prognosis remains different. Novel biomarkers are needed for the prognosis of disease and novel therapeutic strategies in LGG. The procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD) family contains three members and is related to multiple cancers, yet it was not investigated in LGG. Data from the Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) cohorts were used to analyze the role of PLOD in LGG. As the PLOD family is involved in processes, such as tumor formation and cancer metastasis, we focused on its relationship to the tumor microenvironment (TME) in LGG. A high expression of the PLOD family relates to poor prognosis and high infiltration of immune cells within the TME. The expression level of the PLOD family might become a novel biomarker for prognosis and is a potential target for individual treatment decisions in LGG.
Collapse
Affiliation(s)
- Siming Gong
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Changwu Wu
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
- *Correspondence: Changwu Wu,
| | | | | | - Nikolas Schopow
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
- Department of Orthopedics, Trauma and Plastic Surgery, Sarcoma Center, University Hospital Leipzig, Leipzig, Germany
| | - Sonja Kallendrusch
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
- Department of Medicine, Health and Medical University Potsdam, Potsdam, Germany
| |
Collapse
|
32
|
Nyati KK, Kishimoto T. Recent Advances in the Role of Arid5a in Immune Diseases and Cancer. Front Immunol 2022; 12:827611. [PMID: 35126382 PMCID: PMC8809363 DOI: 10.3389/fimmu.2021.827611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/31/2021] [Indexed: 12/09/2022] Open
Abstract
AT-rich interactive domain 5a (Arid5a) is a nucleic acid binding protein. In this review, we highlight recent advances in the association of Arid5a with inflammation and human diseases. Arid5a is known as a protein that performs dual functions. In in vitro and in vivo studies, it was found that an inflammation-dependent increase in Arid5a expression mediates both transcriptional and post-transcriptional regulatory effects that are implicated in immune regulation and cellular homeostasis. A series of publications demonstrated that inhibiting Arid5a augmented several processes, such as preventing septic shock, experimental autoimmune encephalomyelitis, acute lung injury, invasion and metastasis, immune evasion, epithelial-to-mesenchymal transition, and the M1-like tumor-associated macrophage (TAM) to M2-like TAM transition. In addition, Arid5a controls adipogenesis and obesity in mice to maintain metabolic homeostasis. Taken together, recent progress indicates that Arid5a exhibits multifaceted, both beneficial and detrimental, roles in health and disease and suggest the relevance of Arid5a as a potential therapeutic target.
Collapse
|
33
|
Nyati KK, Kishimoto T. The emerging role of Arid5a in cancer: A new target for tumors. Genes Dis 2022. [DOI: 10.1016/j.gendis.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
34
|
Nguyen TTT, Shang E, Westhoff MA, Karpel-Massler G, Siegelin MD. Methodological Approaches for Assessing Metabolomic Changes in Glioblastomas. Methods Mol Biol 2022; 2445:305-328. [PMID: 34973000 DOI: 10.1007/978-1-0716-2071-7_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glioblastoma (GBM), a highly malignant primary brain tumor, inevitably leads to death. In the last decade, a variety of novel molecular characteristics of GBMs were unraveled. The identification of the mutation in the IDH1 and less commonly IDH2 gene was surprising and ever since has nurtured research in the field of GBM metabolism. While initially thought that mutated IDH1 were to act as a loss of function mutation it became clear that it conferred the production of an oncometabolite that in turn substantially reprograms GBM metabolism. While mutated IDH1 represents truly the tip of the iceberg, there are numerous other related observations in GBM that are of significant interest to the field, including the notion that oxidative metabolism appears to play a more critical role than believed earlier. Metabolic zoning is another important hallmark of GBM since it was found that the infiltrative margin that drives GBM progression reveals enrichment of fatty acid derivatives. Consistently, fatty acid metabolism appears to be a novel therapeutic target for GBM. How metabolism in GBM intersects is another pivotal issue that appears to be important for its progression and response and resistance to therapies. In this review, we will summarize some of the most relevant findings related to GBM metabolism and cell death and how these observations are influencing the field. We will provide current approaches that are applied in the field to measure metabolomic changes in GBM models, including the detection of unlabeled and labeled metabolites as well as extracellular flux analysis.
Collapse
Affiliation(s)
- Trang T T Nguyen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, NY, USA
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
35
|
Xu G, Seng Z, Zhang M, Qu J. Angiomotin-like 1 plays a tumor-promoting role in glioma by enhancing the activation of YAP1 signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:2500-2511. [PMID: 34480788 DOI: 10.1002/tox.23363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
Angiomotin-like 1 (AMOTL1) is reportedly a pivotal tumor-associated protein that is strongly associated with the tumorigenesis of multiple malignant tumors. However, the issue of whether AMOTL1 plays a role in the tumorigenesis of glioma remains unclear. The aim of this work was to explore the possible relationship between AMOTL1 and glioma progression. Results demonstrated that high levels of AMOTL1 in glioma tissues were associated with a reduced survival rate in patients with glioma. Cellular functional assays revealed that silencing of AMOTL1 in glioma cell lines substantially decreased cell proliferation and invasion and increased cell apoptosis. Further investigation revealed that silencing of AMOTL1 inhibited the activation of yes-associated protein 1 (YAP1) and decreased the expression of YAP1 target genes. Reactivation of YAP1 reversed AMOTL1-silencing-induced antitumor effects, whereas inhibition of YAP1 abolished AMOTL1-overexpression-induced tumor-promoting effects in glioma cells. Silencing of AMOTL1 also retarded the growth of glioma cell-derived xenograft tumors in vivo. In conclusion, these findings suggested that AMOTL1 may exert a tumor-promoting function in glioma by enhancing the activation of YAP1 signaling. This work suggested AMOTL1 as a potential target for the development of antiglioma therapy.
Collapse
Affiliation(s)
- Gang Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiyuan Seng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqiang Qu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
36
|
Aurora kinase A inhibition reverses the Warburg effect and elicits unique metabolic vulnerabilities in glioblastoma. Nat Commun 2021; 12:5203. [PMID: 34471141 PMCID: PMC8410792 DOI: 10.1038/s41467-021-25501-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Aurora kinase A (AURKA) has emerged as a drug target for glioblastoma (GBM). However, resistance to therapy remains a critical issue. By integration of transcriptome, chromatin immunoprecipitation sequencing (CHIP-seq), Assay for Transposase-Accessible Chromatin sequencing (ATAC-seq), proteomic and metabolite screening followed by carbon tracing and extracellular flux analyses we show that genetic and pharmacological AURKA inhibition elicits metabolic reprogramming mediated by inhibition of MYC targets and concomitant activation of Peroxisome Proliferator Activated Receptor Alpha (PPARA) signaling. While glycolysis is suppressed by AURKA inhibition, we note an increase in the oxygen consumption rate fueled by enhanced fatty acid oxidation (FAO), which was accompanied by an increase of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α). Combining AURKA inhibitors with inhibitors of FAO extends overall survival in orthotopic GBM PDX models. Taken together, these data suggest that simultaneous targeting of oxidative metabolism and AURKAi might be a potential novel therapy against recalcitrant malignancies. Glioblastoma patients are treated with Aurora kinase A (AURKA) inhibitors but resistance can occur. Here, the authors show that AURKA inhibition induces metabolic reprogramming, which leads to increased mitochondrial activity and inhibition of oxidative metabolism sensitizes glioblastoma cells to AURKA inhibition.
Collapse
|
37
|
Murnyak B, Huang LE. Association of TP53 Alteration with Tissue Specificity and Patient Outcome of IDH1-Mutant Glioma. Cells 2021; 10:2116. [PMID: 34440884 PMCID: PMC8394030 DOI: 10.3390/cells10082116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 12/20/2022] Open
Abstract
Since the initial discovery of recurrent isocitrate dehydrogenase 1 (IDH1) mutations at Arg132 in glioma, IDH1 hotspot mutations have been identified in cholangiocarcinoma, chondrosarcoma, leukemia, and various other types of cancer of sporadic incidence. Studies in glioma and leukemia have helped promote the theory that IDH1 mutations are an oncogenic event that drives tumorigenesis in general. Through bioinformatic analysis of more than 45,000 human pan-cancer samples from three independent datasets, we show here that IDH1 mutations are rare events in human cancer but are exclusively prevalent in WHO grade II and grade III (lower-grade) glioma. Interestingly, alterations in the tumor-suppressor gene TP53 (tumor protein p53) co-occur significantly with IDH1 mutations and show a tendency of exclusivity to IDH2 mutations. The co-occurrence of IDH1 mutation and TP53 alteration is widespread in glioma, particularly in those harboring IDH1R132H, IDH1R132G, and IDH1R132S, whereas co-occurrence of IDH1R132C and TP53 alteration can be found sporadically in other cancer types. In keeping with the importance of p53 in tumor suppression, TP53 status is an independent predictor of overall survival irrespective of histological and molecular subgroups in lower-grade glioma. Together, these results indicate tissue specificity of IDH1 hotspot mutation and TP53 alteration and the importance of TP53 status as a predictor of patient outcome in lower-grade glioma.
Collapse
Affiliation(s)
- Balazs Murnyak
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT 84132, USA;
| | - L. Eric Huang
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, Salt Lake City, UT 84132, USA;
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
38
|
Shah AS, Sylvester PT, Yahanda AT, Vellimana AK, Dunn GP, Evans J, Rich KM, Dowling JL, Leuthardt EC, Dacey RG, Kim AH, Grubb RL, Zipfel GJ, Oswood M, Jensen RL, Sutherland GR, Cahill DP, Abram SR, Honeycutt J, Shah M, Tao Y, Chicoine MR. Intraoperative MRI for newly diagnosed supratentorial glioblastoma: a multicenter-registry comparative study to conventional surgery. J Neurosurg 2021; 135:505-514. [PMID: 33035996 DOI: 10.3171/2020.6.jns19287] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/04/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Intraoperative MRI (iMRI) is used in the surgical treatment of glioblastoma, with uncertain effects on outcomes. The authors evaluated the impact of iMRI on extent of resection (EOR) and overall survival (OS) while controlling for other known and suspected predictors. METHODS A multicenter retrospective cohort of 640 adult patients with newly diagnosed supratentorial glioblastoma who underwent resection was evaluated. iMRI was performed in 332/640 cases (51.9%). Reviews of MRI features and tumor volumetric analysis were performed on a subsample of cases (n = 286; 110 non-iMRI, 176 iMRI) from a single institution. RESULTS The median age was 60.0 years (mean 58.5 years, range 20.5-86.3 years). The median OS was 17.0 months (95% CI 15.6-18.4 months). Gross-total resection (GTR) was achieved in 403/640 cases (63.0%). Kaplan-Meier analysis of 286 cases with volumetric analysis for EOR (grouped into 100%, 95%-99%, 80%-94%, and 50%-79%) showed longer OS for 100% EOR compared to all other groups (p < 0.01). Additional resection after iMRI was performed in 104/122 cases (85.2%) with initial subtotal resection (STR), leading to a 6.3% mean increase in EOR and a 2.2-cm3 mean decrease in tumor volume. For iMRI cases with volumetric analysis, the GTR rate increased from 54/176 (30.7%) on iMRI to 126/176 (71.5%) postoperatively. The EOR was significantly higher in the iMRI group for intended GTR and STR groups (p = 0.02 and p < 0.01, respectively). Predictors of GTR on multivariate logistic regression included iMRI use and intended GTR. Predictors of shorter OS on multivariate Cox regression included older age, STR, isocitrate dehydrogenase 1 (IDH1) wild type, no O 6-methylguanine DNA methyltransferase (MGMT) methylation, and no Stupp therapy. iMRI was a significant predictor of OS on univariate (HR 0.82, 95% CI 0.69-0.98; p = 0.03) but not multivariate analyses. Use of iMRI was not associated with an increased rate of new permanent neurological deficits. CONCLUSIONS GTR increased OS for patients with newly diagnosed glioblastoma after adjusting for other prognostic factors. iMRI increased EOR and GTR rate and was a significant predictor of GTR on multivariate analysis; however, iMRI was not an independent predictor of OS. Additional supporting evidence is needed to determine the clinical benefit of iMRI in the management of glioblastoma.
Collapse
Affiliation(s)
- Amar S Shah
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Peter T Sylvester
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Alexander T Yahanda
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ananth K Vellimana
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gavin P Dunn
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - John Evans
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Keith M Rich
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua L Dowling
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Eric C Leuthardt
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ralph G Dacey
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Albert H Kim
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert L Grubb
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory J Zipfel
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Oswood
- 2Department of Radiology, University of Minnesota, Minneapolis, Minnesota
- 3Allina Health, Minneapolis, Minnesota
| | - Randy L Jensen
- 4Department of Neurosurgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Garnette R Sutherland
- 5Department of Clinical Sciences and Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Daniel P Cahill
- 6Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Steven R Abram
- 7Department of Neurosurgery, St. Thomas Hospital, Nashville, Tennessee
| | - John Honeycutt
- 8Department of Neurosurgery, Cook Children's Hospital, Fort Worth, Texas; and
| | - Mitesh Shah
- 9Department of Neurological Surgery, Goodman Campbell and Indiana University, Indianapolis, Indiana
| | - Yu Tao
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Michael R Chicoine
- 1Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
39
|
Cheng Q, Duan W, He S, Li C, Cao H, Liu K, Ye W, Yuan B, Xia Z. Multi-Omics Data Integration Analysis of an Immune-Related Gene Signature in LGG Patients With Epilepsy. Front Cell Dev Biol 2021; 9:686909. [PMID: 34336837 PMCID: PMC8322853 DOI: 10.3389/fcell.2021.686909] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background The tumor immune microenvironment significantly affects tumor occurrence, progression, and prognosis, but its impact on the prognosis of low-grade glioma (LGG) patients with epilepsy has not been reported. Hence, the purpose of this study is to explore its effect on LGG patients with epilepsy. Methods The data of LGG patients derived from the TCGA database. The level of immune cell infiltration and the proportion of 22 immune cells were evaluated by ESTIMATE and CIBERSORT algorithms, respectively. The Cox and LASSO regression analysis was adopted to determine the DEGs, and further established the clustering and risk score models. The association between genomic alterations and risk score was investigated using CNV and somatic mutation data. GSVA was adopted to identify the immunological pathways, immune infiltration and inflammatory profiles related to the signature genes. The Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and GDSC database were used to predict the patient’s response to immunotherapy and chemotherapy, respectively. Results The prognosis of LGG patients with epilepsy was associated with the immune score. Three prognostic DEGs (ABCC3, PDPN, and INA) were screened out. The expression of signature genes was regulated by DNA methylation. The clustering and risk score models could stratify glioma patients into distinct prognosis groups. The risk score was an independent predictor in prognosis, with a high risk-score indicating a poor prognosis, more malignant clinicopathological and genomic aberration features. The nomogram had the better predictive ability. Patients at high risk had a higher level of macrophage infiltration and increased inflammatory activities associated with T cells and macrophages. While the higher percentage of NK CD56bright cell and more active inflammatory activity associated with B cell were present in the low-risk patients. The signature genes participated in the regulation of immune-related pathways, such as IL6-JAK-STAT3 signaling, IFN-α response, IFN-γ response, and TNFA-signaling-via-NFKB pathways. The high-risk patients were more likely to benefit from anti-PD1 and temozolomide (TMZ) treatment. Conclusion An immune-related gene signature was established based on ABCC3, PDPN, and INA, which can be used to predict the prognosis, immune infiltration status, immunotherapy and chemotherapy response of LGG patients with epilepsy.
Collapse
Affiliation(s)
- Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Duan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Shiqing He
- Department of Neurosurgery, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Chen Li
- Department of Rehabilitation Medicine, Hunan Provincial People's Hospital, Hunan Normal University, Changsha, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Kun Liu
- Department of Cerebrovascular Surgery, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Yuan
- Department of Cerebrovascular Surgery, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Changsha Medical University, Changsha, China
| |
Collapse
|
40
|
Ludwig N, Rao A, Sandlesh P, Yerneni SS, Swain AD, Bullock KM, Hansen KM, Zhang X, Jaman E, Allen J, Krueger K, Hong CS, Banks WA, Whiteside TL, Amankulor NM. Characterization of systemic immunosuppression by IDH mutant glioma small extracellular vesicles. Neuro Oncol 2021; 24:197-209. [PMID: 34254643 DOI: 10.1093/neuonc/noab153] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gliomas are the most common primary brain tumors and are universally fatal. Mutations in the isocitrate dehydrogenase genes (IDH1 and IDH2) define a distinct glioma subtype associated with an immunosuppressive tumor microenvironment. Mechanisms underlying systemic immunosuppression in IDH mutant (mutIDH) gliomas are largely unknown. Here, we define genotype-specific local and systemic tumor immunomodulatory functions of tumor-derived glioma exosomes (TEX). METHODS TEX produced by human and murine wildtype and mutant IDH glioma cells (wtIDH and mutIDH, respectively) were isolated by size exclusion chromatography (SEC). TEX morphology, size, quantity, molecular profiles and biodistribution were characterized. TEX were injected into naive and tumor-bearing mice, and the local and systemic immune microenvironment composition was characterized. RESULTS Using in vitro and in vivo glioma models, we show that mutIDH TEX are more numerous, possess distinct morphological features and are more immunosuppressive than wtIDH TEX. mutIDH TEX cargo mimics their parental cells, and induces systemic immune suppression in naive and tumor-bearing mice. TEX derived from mutIDH gliomas and injected into wtIDH tumor-bearing mice reduce tumor-infiltrating effector lymphocytes, dendritic cells and macrophages, and increase circulating monocytes. Astonishingly, mutIDH TEX injected into brain tumor-bearing syngeneic mice accelerate tumor growth and increase mortality compared with wtIDH TEX. CONCLUSIONS Targeting of mutIDH TEX represents a novel therapeutic approach in gliomas.
Collapse
Affiliation(s)
- Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Aparna Rao
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Poorva Sandlesh
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Alexander D Swain
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - Kristin M Bullock
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Kim M Hansen
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Xiaoran Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Emade Jaman
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jordan Allen
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Katharine Krueger
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Chang-Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Nduka M Amankulor
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Departments of Immunology and Otolaryngology, Pittsburgh, USA
| |
Collapse
|
41
|
Characterization of ferroptosis signature to evaluate the predict prognosis and immunotherapy in glioblastoma. Aging (Albany NY) 2021; 13:17655-17672. [PMID: 34244461 PMCID: PMC8312442 DOI: 10.18632/aging.203257] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/19/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common type of brain cancer with poor survival outcomes and unsatisfactory response to current therapeutic strategies. Recent studies have demonstrated that ferroptosis-related genes (FRGs) are linked with the occurrence and development of GBM and may become promising biological indicators in GBM therapy. METHODS We systematically assessed the relationship between FRGs expression profiles and prognosis in glioma patients based on the Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) datasets to establish a risk score model according to the gene signature of multiple survival-associated DEGs. Further, the differences between the tumor microenvironment score, immune cell infiltration, immune checkpoint expression levels, and drug sensitivity in the high- and low-risk group are analyzed through a variety of algorithms in R software. RESULTS GBM patients were divided into two subgroups (high- and low-risk) according to the established risk score model. Patients in the high-risk group showed significantly reduced overall survival compared with those in the low-risk group. Also, we found that the high-risk group showed higher ImmuneScore and StromalScore, while different subgroups have significant differences in immune cell infiltration, immune checkpoint expression levels, and drug sensitivity. In summary, we developed and validated an FRGs risk model, which served as an independent prognostic indicator for GBM. Besides, the two subgroups divided by the model have significant differences, which provides novel insights for further studies as well as the personalized treatment of patients.
Collapse
|
42
|
Zhao K, Sun G, Wang Q, Xue Z, Liu G, Xia Y, Yao A, Zhao Y, You N, Yang C, Xu B. The Diagnostic Value of Conventional MRI and CT Features in the Identification of the IDH1-Mutant and 1p/19q Co-Deletion in WHO Grade II Gliomas. Acad Radiol 2021; 28:e189-e198. [PMID: 32359929 DOI: 10.1016/j.acra.2020.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022]
Abstract
RATIONALE AND OBJECTIVES The classification of patients based on pathology and molecular features is important for improving WHO grade II glioma patient prognosis, especially for the initially diagnosed patients. Less invasive and more convenient methods for the prediction of the pathological type and gene status are desired. MATERIALS AND METHODS This study investigates the ability to use conventional magnetic resonance imaging (MRI) and computed tomography (CT) features for determining the Isocitrate Dehydrogenase (IDH)-mutant and 1p/19q-codeletion status, through a retrospective review of information obtained from 189 WHO grade II glioma patients. Diffuse astrocytoma (IDH-mutant), Diffuse astrocytoma (IDH- wildtype) and Oligodendroglioma (IDH-mutant and 1p/19q co-deletion) were included in this cohort. All patients were divided into IDH-mutant group and IDH-wildtype group according to the IDH R132H mutation status. Moreover, all patients were divided into 1p/19q co-deletion group and 1p/19q non-codeletion group according to the 1p and 19q chromosome status. Patients underwent pre-operative CT and MRI scans, followed by operation and histopathological analyses, including immunohistochemistry and polymerase chain reaction analysis for IDH mutants, and fluorescence capillary electrophoresis analysis for the 1p/19q co-deletion. The χ2 test, logistical regression and receiver operating characteristic curve analysis were conducted for statistical analysis. RESULTS IDH-mutant group patients exhibited a higher calcification frequency (25.2% vs 2.4%, p = 0.006) and lower frequency of T1 enhancement (20.4% vs 38.1%, p = 0.028) comparing patients in IDH-wildtype group, while 1p/19q co-deletion group patients exhibited a higher calcification frequency (46.67% vs 2.6%, p < 0.001) and lower homogenous signal frequency in T2WI (12.0% vs 31.6%, p = 0.014), sharp lesion margins (14.7% vs 43.0%, p = 0.010), T2/fluid attenuated inversion recovery mismatch signs (22.7% vs 50.9%, p = 0.001), and subventricular zone involvement (64.0% vs 15.8%, p = 0.021) comparing patients in 1p/19q non-codeletion group. According to the results of receiver operating characteristic analysis, these features were observed to have certain diagnostic abilities, especially with regard to combination parameters, which had a high diagnostic capability, with an area under the curve of 0.848. CONCLUSION Conventional MRI and CT features, which still represent the most convenient and widely used predictive method, might be a promising noninvasive predictor for differentiating between varied WHO grade II gliomas. Patients with calcification and T1 nonenhancement are more likely to be IDH-mutant. Moreover, patients with noncalcification, homogenous signal, sharp lesion margins, subventricular zone involvement on T2 and T2/fluid attenuated inversion recovery mismatch signs are more likely to be 1p/19q non-codeletion.
Collapse
|
43
|
Cho U, Yang SH, Yoo C. Estimation of the occurrence rates of IDH1 and IDH2 mutations in gliomas and the reconsideration of IDH-wildtype anaplastic astrocytomas: an institutional experience. J Int Med Res 2021; 49:3000605211019258. [PMID: 34162262 PMCID: PMC8236789 DOI: 10.1177/03000605211019258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives Most diffuse gliomas are reported to harbor isocitrate dehydrogenase (IDH) mutations. However, when these mutations are tested in clinical practice, the results are often negative. Methods This study examined the frequency of IDH1 and IDH2 mutations in gliomas classified according to the revised 2016 World Health Organization classification, and investigated their prognostic relevance. We tested 87 gliomas for IDH1 and IDH2 mutations using the peptide nucleic acid clamp method. Results IDH1 mutations were observed in 42% of diffuse astrocytomas, 23% of anaplastic astrocytomas, all oligodendrogliomas and anaplastic oligodendrogliomas, and 17% of glioblastomas. An IDH2 mutation was identified in one case of diffuse astrocytoma. In the survival analysis of diffuse astrocytic tumors, patients with IDH1/2-wildtype anaplastic astrocytomas tended to have a poor prognosis, similar to that of glioblastomas. Conclusions IDH2 mutations were infrequent in gliomas. In anaplastic astrocytomas, the frequency of IDH1/2-wildtype was relatively high, and the prognosis of patients with this type of tumor was very similar to that of those with glioblastomas. It may therefore be necessary to reconsider the classification and treatment strategies for IDH1/2-wildtype anaplastic astrocytomas.
Collapse
Affiliation(s)
- Uiju Cho
- Department of Pathology, St. Vincent?s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Changyoung Yoo
- Department of Pathology, St. Vincent?s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
44
|
Ma S, Rudra S, Campian JL, Chheda MG, Johanns TM, Ansstas G, Abraham CD, Chicoine MR, Leuthardt EC, Dowling JL, Dunn GP, Kim AH, Huang J. Salvage therapies for radiation-relapsed isocitrate dehydrogenase-mutant astrocytoma and 1p/19q codeleted oligodendroglioma. Neurooncol Adv 2021; 3:vdab081. [PMID: 34345818 PMCID: PMC8324173 DOI: 10.1093/noajnl/vdab081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Background Optimal management for recurrent IDH-mutant glioma after radiation therapy (RT) is not well-defined. This study assesses practice patterns for managing recurrent IDH-mutant astrocytoma (Astro) and 1p/19q codeleted oligodendroglioma (Oligo) after RT and surveys their clinical outcomes after different salvage approaches. Methods Ninety-four recurrent Astro or Oligo patients after RT who received salvage systemic therapy (SST) between 2001 and 2019 at a tertiary cancer center were retrospectively analyzed. SST was defined as either alkylating chemotherapy (AC) or nonalkylating therapy (non-AC). Overall survival (OS) and progression-free survival (PFS) were calculated using the Kaplan-Meier method from the start of SST. Multivariable analysis (MVA) was conducted using Cox regression analysis. Results Recurrent Oligo (n = 35) had significantly higher PFS (median: 3.1 vs 0.8 years, respectively, P = .002) and OS (median: 6.3 vs 1.5 years, respectively, P < .001) than Astro (n = 59). Overall, 90% of recurrences were local. Eight-three percent received AC as the first-line SST; 50% received salvage surgery before SST; approximately 50% with local failure >2 years after prior RT received reirradiation. On MVA, non-AC was associated with worse OS for both Oligo and Astro; salvage surgery was associated with improved PFS and OS for Astro; early reirradiation was associated with improved PFS for Astro. Conclusions Recurrent radiation-relapsed IDH-mutant gliomas represent a heterogeneous group with variable treatment approaches. Surgery, AC, and reirradiation remain the mainstay of salvage options for retreatment.
Collapse
Affiliation(s)
- Sirui Ma
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Soumon Rudra
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jian L Campian
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Milan G Chheda
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tanner M Johanns
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - George Ansstas
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Christopher D Abraham
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael R Chicoine
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Eric C Leuthardt
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joshua L Dowling
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jiayi Huang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA.,Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
45
|
Rao D, Yu C, Sheng J, Lv E, Huang W. The Emerging Roles of circFOXO3 in Cancer. Front Cell Dev Biol 2021; 9:659417. [PMID: 34150756 PMCID: PMC8213346 DOI: 10.3389/fcell.2021.659417] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
Circular RNAs (circRNAs) are a class of endogenous non-coding RNAs which are mainly formed by reverse splicing of precursor mRNAs. They are relatively stable and resistant to RNase R because of their covalently closed structure without 5' caps or 3' poly-adenylated tails. CircRNAs are widely expressed in eukaryotic cells and show tissue, timing, and disease specificity. Recent studies have found that circRNAs play an important role in many diseases. In particular, they affect the proliferation, invasion and prognosis of cancer by regulating gene expression. CircRNA Forkhead box O3 (circFOXO3) is a circRNA confirmed to be abnormally expressed in a variety of cancers, including prostate cancer, hepatocellular carcinoma, glioblastoma, bladder cancer, and breast cancer, etc. At present, the feature of circFOXO3 as a molecular sponge is widely studied to promote or inhibit the development of cancers. However, the diverse functions of circFOXO3 have not been fully understood. Hence, it is important to review the roles of circFOXO3 in cancers. This review has summarized and discussed the roles and molecular mechanism of circFOXO3 and its target genes in these cancers, which can help to enrich our understanding to the functions of circRNAs and carry out subsequent researches on circFOXO3.
Collapse
Affiliation(s)
- Dean Rao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengpeng Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Sheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Enjun Lv
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Li X, Sun C, Chen J, Ma JF, Pan YH. ERK-CREB pathway is involved in HSPB8-mediated glioma cell growth and metastatic properties. Exp Mol Pathol 2021:104653. [PMID: 34043982 DOI: 10.1016/j.yexmp.2021.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/14/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the mechanism of HSPB8 (heat shock protein beta-8) in the growth and metastatic properties of glioma cells. METHODS HSPB8 expression in glioma tissue and cell was detected via Western blotting. Then, glioma U87 and U251 cell lines were divided into Mock group, Control siRNA group, HSPB8 siRNA-1 group and HSPB8 siRNA-2 group. Cell proliferation was detected using MTT assay, while its invasion, migration and apoptosis were determined by Transwell, wound-healing and flow cytometry, respectively. The expression of HSPB8 and ERK-CREB pathway-related molecules were also measured by Western blotting. Xenograft models were constructed on nude mice, and accordingly, the growth curve of subcutaneous xenograft was prepared. RESULTS In glioma tissues, HSPB8 expression was upregulated with the increasing grade of glioma. Besides, glioma cells in the HSPB8 siRNA-1 group and HSPB8 siRNA-2 group manifested the significant enhancement in apoptotic rates and reductions in its proliferation, migration and invasion compared to those in the Mock group, meanwhile, the expression of HSPB8, p-ERK1/2/ERK1/2 and p-CREB/CREB were downregulated. On the other hand, the tumor growth in the nude mice of Ad-HSPB8 shRNA-1 group and Ad-HSPB8 shRNA-2 group was retarded significantly, with an acute decrease in the tumor weight. CONCLUSION Silencing HSPB8 can inhibit the malignant features, while facilitate the apoptosis of glioma cells, with inactivation of ERK-CREB pathway.
Collapse
Affiliation(s)
- Xia Li
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Cui Sun
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jing Chen
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Ji-Fen Ma
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yi-Heng Pan
- Center for Diagnosis and Treatment of Neuro-oncology Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China.
| |
Collapse
|
47
|
Zhou Q, Zhou J, Fan J. Expression and Prognostic Value of ARID5A and its Correlation With Tumor-Infiltrating Immune Cells in Glioma. Front Oncol 2021; 11:638803. [PMID: 34094918 PMCID: PMC8172138 DOI: 10.3389/fonc.2021.638803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/30/2021] [Indexed: 12/18/2022] Open
Abstract
AT-rich interaction domain 5A (ARID5A) is a member of the ARID family with a function that has been linked to autoimmune as well as inflammatory diseases. Some ARID family members are involved in the initiation and progression of human cancers. However, the function of ARID5A in glioma remains unknown. In this study, ARID5A expression levels were analyzed using the Gene Expression Profiling Interactive Analysis (GEPIA) database. Subsequently, the relationship between ARID5A expression and the clinical characteristics of glioma patients was evaluated using the Chinese Glioma Genome Atlas (CGGA) database and The Cancer Genome Atlas (TCGA) database. The prognostic value of ARID5A in glioma was estimated by Kaplan-Meier analysis and the receiver operating characteristic (ROC) curve analysis. Gene ontology (GO) analysis and gene set enrichment analysis (GSEA) were performed for functional prediction. The Tumor Immune Estimation Resource (TIMER) database was used to analyze the relationship between ARID5A and immune cell infiltration in glioma. Our results demonstrate that the expression of ARID5A was upregulated in glioma compared with that in nontumor brain tissues. High expression of ARID5A is associated with poor prognosis in glioma. We found that the expression of ARID5A was significantly upregulated with an increase in tumor malignancy. GO analysis revealed that co-expression genes of ARID5A are significantly involved in some important functions in glioma, and GSEA showed that multiple cancer-associated and immune-associated signaling pathways are enriched in the high ARID5A expression group. TIMER database indicated that ARID5A is correlated with tumor-infiltrating immune cells in glioma. Collectively, these findings indicate that ARID5A may be a potential prognostic biomarker and is correlated with immune infiltration in glioma.
Collapse
Affiliation(s)
| | | | - Jingyi Fan
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Rizzo A, Ricci AD, Brandi G. Ivosidenib in IDH-mutant cholangiocarcinoma: where do we stand? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1915126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Alessandro Rizzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
49
|
IDH inhibitors in advanced cholangiocarcinoma: Another arrow in the quiver? Cancer Treat Res Commun 2021; 27:100356. [PMID: 33799004 DOI: 10.1016/j.ctarc.2021.100356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinomas (CCAs) are a heterogenous group of hepatobiliary tumors with poor prognosis and limited therapeutic options. In the last decade, the advent of genomic profiling has led to the identification of several putative actionable aberrations in CCAs, and genomic characterization is playing an increasing role in the management of these malignancies. Thus, a wide number of targetable mutations are currently under investigation, and early studies on this approach in CCAs have been recently presented or published. Among these, isocitrate dehydrogenase (IDH) mutations have been reported in approximately 15-20% of intrahepatic cholangiocarcinoma (iCCA) patients, while these aberrations are considered to be less frequent in perihilar CCA (pCCA), distal CCA (dCCA), and gallbladder cancer. Of note, the recent findings of the ClarIDHy phase III trial add to mounting evidence showing the potential advantages of molecularly targeted therapies in CCA, on the basis of a benefit in previously treated IDH1-mutant patients receiving ivosidenib versus placebo. However, although the results of this trial showed a statistically significant improvement in progression-free survival and overall survival for IDH-mutant CCAs treated with ivosidenib, several questions regarding the real impact of IDH inhibitors in this setting remain open. In this review, we will provide an overview on the biological rationale behind the use of IDH inhibitors in CCA patients and current clinical implications of these molecularly targeted agents. The recently published results of the ClarIDHy - as well as ongoing clinical trials in this setting - are highlighted and critically discussed.
Collapse
|
50
|
Abstract
Metabolic reprogramming is an important characteristics of glioma, the most common form of malignant brain tumor. In this chapter, we aim to discuss some of the recently discovered metabolic alterations in glioma, including the dysregulated TCA cycle, amino acid, nucleotide, and lipid metabolism. We have also detailed some of the metabolomic applications in gliomas, particularly the analyses of body fluids and tissues of glioma patients. With new improvement of the technology, metabolomics will become a powerful tool to discover truly meaningful biomarkers for clinical applications in gliomas. Metabolomic studies of gliomas will also facilitate a better understanding of the molecular targets/pathways and the development of new therapeutic treatments for this devastating disease.
Collapse
|