1
|
Jackson LR, Erickson A, Camphausen K, Krauze AV. Understanding the Immune System and Biospecimen-Based Response in Glioblastoma: A Practical Guide to Utilizing Signal Redundancy for Biomarker and Immune Signature Discovery. Curr Oncol 2024; 32:16. [PMID: 39851932 PMCID: PMC11763554 DOI: 10.3390/curroncol32010016] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 01/26/2025] Open
Abstract
Glioblastoma (GBM) is a primary central nervous system malignancy with a median survival of 15-20 months. The presence of both intra- and intertumoral heterogeneity limits understanding of biological mechanisms leading to tumor resistance, including immune escape. An attractive field of research to examine treatment resistance are immune signatures composed of cluster of differentiation (CD) markers and cytokines. CD markers are surface markers expressed on various cells throughout the body, often associated with immune cells. Cytokines are the effector molecules of the immune system. Together, CD markers and cytokines can serve as useful biomarkers to reflect immune status in patients with GBM. However, there are gaps in the understanding of the intricate interactions between GBM and the peripheral immune system and how these interactions change with standard and immune-modulating treatments. The key to understanding the true nature of these interactions is through multi-omic analysis of tumor progression and treatment response. This review aims to identify potential non-invasive blood-based biomarkers that can contribute to an immune signature through multi-omic approaches, leading to a better understanding of immune involvement in GBM.
Collapse
Affiliation(s)
| | | | | | - Andra V. Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA; (L.R.J.); (A.E.); (K.C.)
| |
Collapse
|
2
|
Murali SH, Thakar S, Chandrasekhar DV, Rajarathnam R, Aryan S. Value-Based, No-Cost-To-Patient Neurosurgery at Sri Sathya Sai Institute of Higher Medical Sciences, Bangalore: The Success Story. Neurol India 2024; 72:1054-1062. [PMID: 39428780 DOI: 10.4103/neurol-india.neurol-india-d-24-00170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/25/2024] [Indexed: 10/22/2024]
Abstract
The Sri Sathya Sai Central Trust (SSSCT) was founded in 1972 as a public charitable trust with the objectives of providing free education, healthcare, and public utility benefits without any discrimination. The neurosurgery department at the Sri Sathya Sai Institute of Higher Medical Sciences (SSSIHMS), Bangalore, one of SSSCT's many hospitals, has been offering comprehensive services with state-of-the-art facilities for a diverse range of disorders since 2001. Operating on a unique "no-cost-to-patient" model and guided by an ethos of altruism introduced by its founder Sri Sathya Sai Baba, the department has successfully provided high-quality neurosurgical care to a large number of patients. This article reviews the genesis of the department, its milestones over more than two decades, and the contributions of leaders who have played an important role in shaping the department.
Collapse
Affiliation(s)
- Sanjay Honavalli Murali
- Department of Neurosurgery, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bangalore, Karnataka, India
| | - Sumit Thakar
- Department of Neurosurgery, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bangalore, Karnataka, India
| | - D V Chandrasekhar
- Department of Neurosurgery, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bangalore, Karnataka, India
| | | | - Saritha Aryan
- Department of Neurosurgery, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bangalore, Karnataka, India
| |
Collapse
|
3
|
Łaszczych D, Czernicka A, Gostomczyk K, Szylberg Ł, Borowczak J. The role of IL-17 in the pathogenesis and treatment of glioblastoma-an update on the state of the art and future perspectives. Med Oncol 2024; 41:187. [PMID: 38918274 PMCID: PMC11199243 DOI: 10.1007/s12032-024-02434-1] [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: 05/06/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor, which, despite significant progress made in the last years in the field of neuro-oncology, remains an incurable disease. GBM has a poor prognosis with a median survival of 12-15 months, and its aggressive clinical course is related to rapid growth, extensive infiltration of adjacent tissues, resistance to chemotherapy, radiotherapy and immunotherapy, and frequent relapse. Currently, several molecular biomarkers are used in clinical practice to predict patient prognosis and response to treatment. However, due to the overall unsatisfactory efficacy of standard multimodal treatment and the remaining poor prognosis, there is an urgent need for new biomarkers and therapeutic strategies for GBM. Recent evidence suggests that GBM tumorigenesis is associated with crosstalk between cancer, immune and stromal cells mediated by various cytokines. One of the key factors involved in this process appears to be interleukin-17 (IL-17), a pro-inflammatory cytokine that is significantly upregulated in the serum and tissue of GBM patients. IL-17 plays a key role in tumorigenesis, angiogenesis, and recurrence of GBM by activating pro-oncogenic signaling pathways and promoting cell survival, proliferation, and invasion. IL-17 facilitates the immunomodulation of the tumor microenvironment by promoting immune cells infiltration and cytokine secretion. In this article we review the latest scientific reports to provide an update on the role of IL-17 role in tumorigenesis, tumor microenvironment, diagnosis, prognosis, and treatment of GBM.
Collapse
Affiliation(s)
- Dariusz Łaszczych
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland.
| | - Aleksandra Czernicka
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
| | - Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
- Department of Tumor Pathology and Pathomorphology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, dr Izabeli Romanowskiej 2 street, 85-796, Bydgoszcz, Poland
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, dr Izabeli Romanowskiej 2 street, 85-796, Bydgoszcz, Poland
| |
Collapse
|
4
|
Jarmuzek P, Defort P, Kot M, Wawrzyniak-Gramacka E, Morawin B, Zembron-Lacny A. Cytokine Profile in Development of Glioblastoma in Relation to Healthy Individuals. Int J Mol Sci 2023; 24:16206. [PMID: 38003396 PMCID: PMC10671437 DOI: 10.3390/ijms242216206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Cytokines play an essential role in the control of tumor cell development and multiplication. However, the available literature provides ambiguous data on the involvement of these proteins in the formation and progression of glioblastoma (GBM). This study was designed to evaluate the inflammatory profile and to investigate its potential for the identification of molecular signatures specific to GBM. Fifty patients aged 66.0 ± 10.56 years with newly diagnosed high-grade gliomas and 40 healthy individuals aged 71.7 ± 4.9 years were included in the study. White blood cells were found to fall within the referential ranges and were significantly higher in GBM than in healthy controls. Among immune cells, neutrophils showed the greatest changes, resulting in elevated neutrophil-to-lymphocyte ratio (NLR). The neutrophil count inversely correlated with survival time expressed by Spearman's coefficient rs = -0.359 (p = 0.010). The optimal threshold values corresponded to 2.630 × 103/µL for NLR (the area under the ROC curve AUC = 0.831, specificity 90%, sensitivity 76%, the relative risk RR = 7.875, the confidence intervals 95%CI 3.333-20.148). The most considerable changes were recorded in pro-inflammatory cytokines interleukin IL-1β, IL-6, and IL-8, which were approx. 1.5-2-fold higher, whereas tumor necrosis factor α (TNFα) and high mobility group B1 (HMGB1) were lower in GBM than healthy control (p < 0.001). The results of the ROC, AUC, and RR analysis of IL-1β, IL-6, IL-8, and IL-10 indicate their high diagnostics potential for clinical prognosis. The highest average RR was observed for IL-6 (RR = 2.923) and IL-8 (RR = 3.151), which means there is an approx. three-fold higher probability of GBM development after exceeding the cut-off values of 19.83 pg/mL for IL-6 and 10.86 pg/mL for IL-8. The high values of AUC obtained for the models NLR + IL-1β (AUC = 0.907), NLR + IL-6 (AUC = 0.908), NLR + IL-8 (AUC = 0.896), and NLR + IL-10 (AUC = 0.887) prove excellent discrimination of GBM patients from healthy individuals and may represent GBM-specific molecular signatures.
Collapse
Affiliation(s)
- Pawel Jarmuzek
- Department of Nervous System Diseases, Collegium Medicum, Neurosurgery Center University Hospital, University of Zielona Gora, 65-417 Zielona Gora, Poland; (P.J.); (M.K.)
| | - Piotr Defort
- Department of Nervous System Diseases, Collegium Medicum, Neurosurgery Center University Hospital, University of Zielona Gora, 65-417 Zielona Gora, Poland; (P.J.); (M.K.)
| | - Marcin Kot
- Department of Nervous System Diseases, Collegium Medicum, Neurosurgery Center University Hospital, University of Zielona Gora, 65-417 Zielona Gora, Poland; (P.J.); (M.K.)
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, Collegium Medicum, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.W.-G.); (B.M.); (A.Z.-L.)
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, Collegium Medicum, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.W.-G.); (B.M.); (A.Z.-L.)
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, Collegium Medicum, University of Zielona Gora, 65-417 Zielona Gora, Poland; (E.W.-G.); (B.M.); (A.Z.-L.)
| |
Collapse
|
5
|
Identification of Prognostic Fatty Acid Metabolism lncRNAs and Potential Molecular Targeting Drugs in Uveal Melanoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3726351. [PMID: 36267302 PMCID: PMC9578887 DOI: 10.1155/2022/3726351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/17/2022] [Accepted: 09/24/2022] [Indexed: 11/25/2022]
Abstract
Background The aim of this study was to identify prognostic fatty acid metabolism lncRNAs and potential molecular targeting drugs in uveal melanoma through integrated bioinformatics analysis. Methods In the present study, we obtained the expression matrix of 309 FAM-mRNAs and identified 225 FAM-lncRNAs by coexpression network analysis. We then performed univariate Cox analysis, LASSO regression analysis, and cross-validation and finally obtained an optimized UVM prognosis prediction model composed of four PFAM-lncRNAs (AC104129.1, SOS1-IT1, IDI2-AS1, and DLGAP1-AS2). Results The survival curves showed that the survival time of UVM patients in the high-risk group was significantly lower than that in the low-risk group in the train cohort, test cohort, and all patients in the prognostic prediction model (P < 0.05). We further performed risk prognostic assessment, and the results showed that the risk scores of the high-risk group in the train cohort, test cohort, and all patients were significantly higher than those of the low-risk group (P < 0.05), patient survival decreased and the number of deaths increased with increasing risk scores, and AC104129.1, SOS1-IT1, and DLGAP1-AS2 were high-risk PFAM-lncRNAs, while IDI2-AS1 were low-risk PFAM-lncRNAs. Afterwards, we further verified the accuracy and the prognostic value of our model in predicting prognosis by PCA analysis and ROC curves. Conclusion We identified 24 potential molecularly targeted drugs with significant sensitivity differences between high- and low-risk UVM patients, of which 13 may be potential targeted drugs for high-risk patients. Our findings have important implications for early prediction and early clinical intervention in high-risk UVM patients.
Collapse
|
6
|
Liu XP, Jin X, Seyed Ahmadian S, Yang X, Tian SF, Cai YX, Chawla K, Snijders AM, Xia Y, van Diest PJ, Weiss WA, Mao JH, Li ZQ, Vogel H, Chang H. Clinical significance and molecular annotation of cellular morphometric subtypes in lower-grade gliomas discovered by machine learning. Neuro Oncol 2022; 25:68-81. [PMID: 35716369 PMCID: PMC9825346 DOI: 10.1093/neuonc/noac154] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Lower-grade gliomas (LGG) are heterogeneous diseases by clinical, histological, and molecular criteria. We aimed to personalize the diagnosis and therapy of LGG patients by developing and validating robust cellular morphometric subtypes (CMS) and to uncover the molecular signatures underlying these subtypes. METHODS Cellular morphometric biomarkers (CMBs) were identified with artificial intelligence technique from TCGA-LGG cohort. Consensus clustering was used to define CMS. Survival analysis was performed to assess the clinical impact of CMBs and CMS. A nomogram was constructed to predict 3- and 5-year overall survival (OS) of LGG patients. Tumor mutational burden (TMB) and immune cell infiltration between subtypes were analyzed using the Mann-Whitney U test. The double-blinded validation for important immunotherapy-related biomarkers was executed using immunohistochemistry (IHC). RESULTS We developed a machine learning (ML) pipeline to extract CMBs from whole-slide images of tissue histology; identifying and externally validating robust CMS of LGGs in multicenter cohorts. The subtypes had independent predicted OS across all three independent cohorts. In the TCGA-LGG cohort, patients within the poor-prognosis subtype responded poorly to primary and follow-up therapies. LGGs within the poor-prognosis subtype were characterized by high mutational burden, high frequencies of copy number alterations, and high levels of tumor-infiltrating lymphocytes and immune checkpoint genes. Higher levels of PD-1/PD-L1/CTLA-4 were confirmed by IHC staining. In addition, the subtypes learned from LGG demonstrate translational impact on glioblastoma (GBM). CONCLUSIONS We developed and validated a framework (CMS-ML) for CMS discovery in LGG associated with specific molecular alterations, immune microenvironment, prognosis, and treatment response.
Collapse
Affiliation(s)
| | | | - Saman Seyed Ahmadian
- Department of Pathology, Stanford University Medical Center, Stanford, California, USA
| | - Xu Yang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA,Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, California, USA,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Su-Fang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yu-Xiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Kuldeep Chawla
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA,Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Yankai Xia
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - William A Weiss
- Departments of Neurology, Neurological Surgery, and Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA,Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Zhi-Qiang Li
- Corresponding Authors: Zhi-Qiang Li, MD, PhD, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuchang District, Wuhan, Hubei 430071 China (); Hang Chang, PhD, Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA ()
| | | | - Hang Chang
- Corresponding Authors: Zhi-Qiang Li, MD, PhD, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuchang District, Wuhan, Hubei 430071 China (); Hang Chang, PhD, Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA ()
| |
Collapse
|
7
|
Shrivastava R, Gandhi P, Gothalwal R. The road-map for establishment of a prognostic molecular marker panel in glioma using liquid biopsy: current status and future directions. Clin Transl Oncol 2022; 24:1702-1714. [PMID: 35653004 DOI: 10.1007/s12094-022-02833-8] [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: 02/28/2022] [Accepted: 04/02/2022] [Indexed: 11/24/2022]
Abstract
Gliomas are primary intracranial tumors with defined molecular markers available for precise diagnosis. The prognosis of glioma is bleak as there is an overlook of the dynamic crosstalk between tumor cells and components of the microenvironment. Herein, different phases of gliomagenesis are presented with reference to the role and involvement of secreted proteomic markers at various stages of tumor initiation and development. The secreted markers of inflammatory response, namely interleukin-6, tumor necrosis factor-α, interferon-ϒ, and kynurenine, proliferation markers human telomerase reverse transcriptase and microtubule-associated-protein-Tau, and stemness marker human-mobility-group-AThook-1 are involved in glial tumor initiation and growth. Further, hypoxia and angiogenic factors, heat-shock-protein-70, endothelial-growth-factor-receptor-1 and vascular endothelial growth factor play a major role in promoting vascularization and tumor volume expansion. Eventually, molecules such as matrix-metalloprotease-7 and intercellular adhesion molecule-1 contribute to the degradation and remodeling of the extracellular matrix, ultimately leading to glioma progression. Our study delineates the roadmap to develop and evaluate a non-invasive panel of secreted biomarkers using liquid biopsy for precisely evaluating disease progression, to accomplish a clinical translation.
Collapse
Affiliation(s)
- Richa Shrivastava
- Department of Research, Bhopal Memorial Hospital and Research Centre, Raisen Bypass Road, Bhopal, M.P., 462038, India
| | - Puneet Gandhi
- Department of Research, Bhopal Memorial Hospital and Research Centre, Raisen Bypass Road, Bhopal, M.P., 462038, India.
| | - Ragini Gothalwal
- Department of Biotechnology, Barkatullah University, Bhopal, M.P., 462026, India
| |
Collapse
|
8
|
Widodo SS, Dinevska M, Furst LM, Stylli SS, Mantamadiotis T. IL-10 in glioma. Br J Cancer 2021; 125:1466-1476. [PMID: 34349251 PMCID: PMC8609023 DOI: 10.1038/s41416-021-01515-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/05/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
The prognosis for patients with glioblastoma (GBM), the most common and malignant type of primary brain tumour, is very poor, despite current standard treatments such as surgery, radiotherapy and chemotherapy. Moreover, the immunosuppressive tumour microenvironment hinders the development of effective immunotherapies for GBM. Cytokines such as interleukin-10 (IL-10) play a major role in modulating the activity of infiltrating immune cells and tumour cells in GBM, predominantly conferring an immunosuppressive action; however, in some circumstances, IL-10 can have an immunostimulatory effect. Elucidating the function of IL-10 in GBM is necessary to better strategise and improve the efficacy of immunotherapy. This review discusses the immunostimulatory and immunosuppressive roles of IL-10 in the GBM tumour microenvironment while considering IL-10-targeted treatment strategies. The molecular mechanisms that underlie the expression of IL-10 in various cell types are also outlined, and how this resulting information might provide an avenue for the improvement of immunotherapy in GBM is explored.
Collapse
Affiliation(s)
- Samuel S. Widodo
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Marija Dinevska
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Liam M. Furst
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia
| | - Stanley S. Stylli
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.416153.40000 0004 0624 1200Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC Australia
| | - Theo Mantamadiotis
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia ,grid.418025.a0000 0004 0606 5526Florey Institute of Neuroscience and Mental Health, Parkville, VIC Australia
| |
Collapse
|
9
|
Oh JH, Choi W, Ko E, Kang M, Tannenbaum A, Deasy JO. PathCNN: interpretable convolutional neural networks for survival prediction and pathway analysis applied to glioblastoma. Bioinformatics 2021; 37:i443-i450. [PMID: 34252964 PMCID: PMC8336441 DOI: 10.1093/bioinformatics/btab285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
MOTIVATION Convolutional neural networks (CNNs) have achieved great success in the areas of image processing and computer vision, handling grid-structured inputs and efficiently capturing local dependencies through multiple levels of abstraction. However, a lack of interpretability remains a key barrier to the adoption of deep neural networks, particularly in predictive modeling of disease outcomes. Moreover, because biological array data are generally represented in a non-grid structured format, CNNs cannot be applied directly. RESULTS To address these issues, we propose a novel method, called PathCNN, that constructs an interpretable CNN model on integrated multi-omics data using a newly defined pathway image. PathCNN showed promising predictive performance in differentiating between long-term survival (LTS) and non-LTS when applied to glioblastoma multiforme (GBM). The adoption of a visualization tool coupled with statistical analysis enabled the identification of plausible pathways associated with survival in GBM. In summary, PathCNN demonstrates that CNNs can be effectively applied to multi-omics data in an interpretable manner, resulting in promising predictive power while identifying key biological correlates of disease. AVAILABILITY AND IMPLEMENTATION The source code is freely available at: https://github.com/mskspi/PathCNN.
Collapse
Affiliation(s)
- Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wookjin Choi
- Department of Computer Science, Virginia State University, Petersburg, VA 23806, USA
| | - Euiseong Ko
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154, USA
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154, USA
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, New York, NY 11794, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
10
|
Ali H, Harting R, de Vries R, Ali M, Wurdinger T, Best MG. Blood-Based Biomarkers for Glioma in the Context of Gliomagenesis: A Systematic Review. Front Oncol 2021; 11:665235. [PMID: 34150629 PMCID: PMC8211985 DOI: 10.3389/fonc.2021.665235] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gliomas are the most common and aggressive tumors of the central nervous system. A robust and widely used blood-based biomarker for glioma has not yet been identified. In recent years, a plethora of new research on blood-based biomarkers for glial tumors has been published. In this review, we question which molecules, including proteins, nucleic acids, circulating cells, and metabolomics, are most promising blood-based biomarkers for glioma diagnosis, prognosis, monitoring and other purposes, and align them to the seminal processes of cancer. METHODS The Pubmed and Embase databases were systematically searched. Biomarkers were categorized in the identified biomolecules and biosources. Biomarker characteristics were assessed using the area under the curve (AUC), accuracy, sensitivity and/or specificity values and the degree of statistical significance among the assessed clinical groups was reported. RESULTS 7,919 references were identified: 3,596 in PubMed and 4,323 in Embase. Following screening of titles, abstracts and availability of full-text, 262 articles were included in the final systematic review. Panels of multiple biomarkers together consistently reached AUCs >0.8 and accuracies >80% for various purposes but especially for diagnostics. The accuracy of single biomarkers, consisting of only one measurement, was far more variable, but single microRNAs and proteins are generally more promising as compared to other biomarker types. CONCLUSION Panels of microRNAs and proteins are most promising biomarkers, while single biomarkers such as GFAP, IL-10 and individual miRNAs also hold promise. It is possible that panels are more accurate once these are involved in different, complementary cancer-related molecular pathways, because not all pathways may be dysregulated in cancer patients. As biomarkers seem to be increasingly dysregulated in patients with short survival, higher tumor grades and more pathological tumor types, it can be hypothesized that more pathways are dysregulated as the degree of malignancy of the glial tumor increases. Despite, none of the biomarkers found in the literature search seem to be currently ready for clinical implementation, and most of the studies report only preliminary application of the identified biomarkers. Hence, large-scale validation of currently identified and potential novel biomarkers to show clinical utility is warranted.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Romée Harting
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, Netherlands
| | - Meedie Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Myron G. Best
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
11
|
Rossmeisl JH, Herpai D, Quigley M, Cecere TE, Robertson JL, D'Agostino RB, Hinckley J, Tatter SB, Dickinson PJ, Debinski W. Phase I trial of convection-enhanced delivery of IL13RA2 and EPHA2 receptor targeted cytotoxins in dogs with spontaneous intracranial gliomas. Neuro Oncol 2021; 23:422-434. [PMID: 32812637 PMCID: PMC7992889 DOI: 10.1093/neuonc/noaa196] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background The interleukin-13 receptor alpha 2 (IL13RA2) and ephrin type A receptor 2 (EPHA2) are attractive therapeutic targets, being expressed in ~90% of canine and human gliomas, and absent in normal brain. Clinical trials using an earlier generation IL-13 based cytotoxin showed encouraging clinical effects in human glioma, but met with technical barriers associated with the convection-enhanced delivery (CED) method. In this study, IL-13 mutant and ephrin A1 (EFNA1)–based bacterial cytotoxins targeted to IL13RA2 and EPHA2 receptors, respectively, were administered locoregionally by CED to dogs with intracranial gliomas to evaluate their safety and preliminary efficacy. Methods In this phase I, 3 + 3 dose escalation trial, cytotoxins were infused by CED in 17 dogs with gliomas expressing IL13RA2 or EPHA2 receptors. CED was performed using a shape-fitting therapeutic planning algorithm, reflux-preventing catheters, and real-time intraoperative MRI monitoring. The primary endpoint was to determine the maximum tolerated dose of the cytotoxic cocktail in dogs with gliomas. Results Consistent intratumoral delivery of the cytotoxic cocktail was achieved, with a median target coverage of 70% (range, 40–94%). Cytotoxins were well tolerated over a dose range of 0.012–1.278 μg/mL delivered to the target volume (median, 0.099 μg/mL), with no dose limiting toxicities observed. Objective tumor responses, up to 94% tumor volume reduction, were observed in 50% (8/16) of dogs, including at least one dog in each dosing cohort >0.05 μg/mL. Conclusions This study provides preclinical data fundamental to the translation of this multireceptor targeted therapeutic approach to the human clinic.
Collapse
Affiliation(s)
- John H Rossmeisl
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Veterinary and Comparative Neurooncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia
| | - Denise Herpai
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina
| | - Mindy Quigley
- Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Thomas E Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - John L Robertson
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Veterinary and Comparative Neurooncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia
| | - Ralph B D'Agostino
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Jonathan Hinckley
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina
| | - Stephen B Tatter
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California (P.J.D.)
| | - Waldemar Debinski
- Comprehensive Cancer Center and Brain Tumor Center of Excellence of Wake Forest University, Winston-Salem, North Carolina.,Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Blacksburg, Virginia.,Department of Cancer Biology of Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
12
|
Bender DE, Schaettler MO, Sheehan KC, Johanns TM, Dunn GP. Cytokine Profiling in Plasma from Patients with Brain Tumors Versus Healthy Individuals using 2 Different Multiplex Immunoassay Platforms. Biomark Insights 2021; 16:11772719211006666. [PMID: 33854293 PMCID: PMC8013708 DOI: 10.1177/11772719211006666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/11/2021] [Indexed: 01/18/2023] Open
Abstract
We compared the performance of two 96-well multiplex immunoassay platforms in assessing plasma cytokine concentrations in patients with glioblastoma (GBM; n = 27), individuals with melanoma, breast or lung cancer metastases to the brain (n = 17), and healthy volunteers (n = 11). Assays included a bead-based fluorescence MILLIPLEX® assay/Luminex (LMX) platform and 4 planar electrochemiluminescence kits from Meso Scale Discovery (MSD). The LMX kit evaluated 21 cytokines and the 3 MSD kits evaluated 20 cytokines in total, with 19 overlapping human cytokines between platforms (GM-CSF, IFNγ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13, IL-17A, IL-21, IL-23, MIP-1α, MIP-1β, MIP-3α, TNFα). The MSD platform had lower LLoQs (lower limits of quantification) than LMX for 17/19 cytokines, and higher LLoQs for IFN-γ and IL-21. The ULoQs were higher in LMX versus MSD assays for 17/19 shared analytes, but lower than MSD for IL-17A and IL-21. With LMX, all 19 shared analytes were quantifiable in each of 55 samples. Although MSD recombinant protein standard curves indicated lower LLoQs than LMX for most cytokines, MSD detected 7/19 (37%) native analytes in <75% of samples, including 0% detection for IL-21 and 8% for IL-23. The LMX platform categorized identical samples at greater concentrations than the MSD system for most analytes (MIP-1β the sole exception), sometimes by orders of magnitude. This mismatched quantification paradigm was supported by Bland-Altman analysis. LMX identified significantly elevated levels of 10 of 19 circulating cytokines in GBM: GM-CSF, IFN-γ, IL-1β, IL-5, IL-10, IL-17A, IL-21, IL-23, MIP-1α, and MIP-3α, consistent with prior findings and confirming the utility of applying appropriate multiplex immunoassay technologies toward developing a cytokine signature profile for GBM.
Collapse
Affiliation(s)
- Diane Elizabeth Bender
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Maximilian O Schaettler
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathleen Cf Sheehan
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tanner M Johanns
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA.,The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
13
|
Wang JJ, Wang H, Zhu BL, Wang X, Qian YH, Xie L, Wang WJ, Zhu J, Chen XY, Wang JM, Ding ZL. Development of a prognostic model of glioma based on immune-related genes. Oncol Lett 2020; 21:116. [PMID: 33376548 PMCID: PMC7751470 DOI: 10.3892/ol.2020.12377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Glioma is the most common type of primary brain cancer, and the prognosis of most patients with glioma, and particularly that of patients with glioblastoma, is poor. Tumor immunity serves an important role in the development of glioma. However, immunotherapy for glioma has not been completely successful, and thus, comprehensive examination of the immune-related genes (IRGs) of glioma is required. In the present study, differentially expressed genes (DEGs) and differentially expressed IRGs (DEIRGs) were identified using the edgeR package. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was used for functional enrichment analysis of DEIRGs. Survival-associated IRGs were selected via univariate Cox regression analysis. A The Cancer Genome Atlas prognostic model and GSE43378 validation model were established using lasso-penalized Cox regression analysis. Based on the median risk score value, patients were divided into high-risk and low-risk groups for clinical analysis. Receiver operating characteristic curve and nomogram analyses were used to assess the accuracy of the models. Reverse transcription-quantitative PCR was performed to measure the expression levels of relevant genes, such as cyclin-dependent kinase 4 (CDK4), interleukin 24 (IL24), NADPH oxidase 4 (NOX4), bone morphogenetic protein 2 (BMP2) and baculoviral IAP repeat containing 5 (BIRC5). A total of 3,238 DEGs, including 1,950 upregulated and 1,288 downregulated DEGs, and 97 DEIRGs, including 60 upregulated and 37 downregulated DEIRGs, were identified. ‘Neuroactive ligand-receptor interaction’ and ‘Cytokine-cytokine receptor interaction’ were the most significantly enriched pathways according to KEGG pathway analysis. A prognostic model and a validation prognostic model were created for glioma, including 15 survival-associated IRGs (FCER1G, NOX4, TRIM5, SOCS1, APOBEC3C, BIRC5, VIM, TNC, BMP2, CMTM3, IL24, JAG1, CALCRL, HNF4G and CDK4). Furthermore, multivariate Cox regression analysis results suggested that age, high WHO Grade by histopathology, wild type isocitrate dehydrogenase 1 and high risk score were independently associated with poor overall survival. The infiltration of B cells, CD8+ T cells, dendritic cells, macrophages and neutrophils was positively associated with the prognostic risk score. In the present study, several clinically significant survival-associated IRGs were identified, and a prognosis evaluation model of glioma was established.
Collapse
Affiliation(s)
- Jing-Jing Wang
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Han Wang
- Department of Oncology, Jining Cancer Hospital, Jining, Shandong 272000, P.R. China
| | - Bao-Long Zhu
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Xiang Wang
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Yong-Hong Qian
- Department of Radio-Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Lei Xie
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Wen-Jie Wang
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jie Zhu
- Department of Oncology, Changzhou Traditional Chinese Medical Hospital, Changzhou, Jiangsu, 213003, P.R. China
| | - Xing-Yu Chen
- Department of General Surgery, Taizhou Fourth People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Jing-Mei Wang
- Department of Geriatrics, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310002, P.R. China
| | - Zhi-Liang Ding
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
14
|
Urbanavičiūtė R, Skauminas K, Skiriutė D. The Evaluation of AREG, MMP-2, CHI3L1, GFAP, and OPN Serum Combined Value in Astrocytic Glioma Patients' Diagnosis and Prognosis. Brain Sci 2020; 10:brainsci10110872. [PMID: 33227903 PMCID: PMC7699177 DOI: 10.3390/brainsci10110872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Gliomas account for approximately 70% of primary brain tumors in adults. Of all gliomas, grade IV astrocytoma, also called glioblastoma, has the poorest overall survival, with <5% of patients surviving five years after diagnosis. Due to the aggressiveness, lethal nature, and impaired surgical accessibility of the tumor, early diagnosis of the tumor and, in addition, prediction of the patient's survival time are important. We hypothesize that combining the protein level values of highly recognizable glioblastoma serum biomarkers could help to achieve higher specificity and sensitivity in predicting glioma patient outcome as compared to single markers. The aim of this study was to select the most promising astrocytoma patient overall survival prediction variables from five secretory proteins-glial fibrillary acidic protein (GFAP), matrix metalloproteinase-2 (MMP-2), chitinase 3-like 1 (CHI3L1), osteopontin (OPN), and amphiregulin (AREG)-combining them with routinely used tumor markers to create a Patient Survival Score calculation tool. The study group consisted of 70 astrocytoma patients and 31 healthy controls. We demonstrated that integrating serum CHI3L1 and OPN protein level values and tumor isocitrate dehydrogenase 1 IDH1 mutational status into one parameter could predict low-grade astrocytoma patients' two-year survival with 93.8% accuracy.
Collapse
|
15
|
Linhares P, Carvalho B, Vaz R, Costa BM. Glioblastoma: Is There Any Blood Biomarker with True Clinical Relevance? Int J Mol Sci 2020; 21:E5809. [PMID: 32823572 PMCID: PMC7461098 DOI: 10.3390/ijms21165809] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most frequent malignant primary brain tumor in adults, characterized by a highly aggressive, inflammatory and angiogenic phenotype. It is a remarkably heterogeneous tumor at several levels, including histopathologically, radiographically and genetically. The 2016 update of the WHO Classification of Tumours of the Central Nervous System highlighted molecular parameters as paramount features for the diagnosis, namely IDH1/2 mutations that distinguish primary and secondary GBM. An ideal biomarker is a molecule that can be detected/quantified through simple non- or minimally invasive methods with the potential to assess cancer risk; promote early diagnosis; increase grading accuracy; and monitor disease evolution and treatment response, as well as fundamentally being restricted to one aspect. Blood-based biomarkers are particularly attractive due to their easy access and have been widely used for various cancer types. A number of serum biomarkers with multiple utilities for glioma have been reported that could classify glioma grades more precisely and provide prognostic value among these patients. At present, screening for gliomas has no clinical relevance. This is because of the low incidence, the lack of sensitive biomarkers in plasma, and the observation that gliomas may develop apparently de novo within few weeks or months. To the best of our knowledge, there is no routine use of a serum biomarker for clinical follow-up. The purpose of this paper is to review the serum biomarkers described in the literature related to glioblastoma and their possible relationship with clinical features.
Collapse
Affiliation(s)
- Paulo Linhares
- Neurosurgery Department, Centro Hospitalar São João, Alameda Prof Hernani Monteiro, 4200–319 Porto, Portugal; (P.L.); (R.V.)
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine, University of Oporto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bruno Carvalho
- Neurosurgery Department, Centro Hospitalar São João, Alameda Prof Hernani Monteiro, 4200–319 Porto, Portugal; (P.L.); (R.V.)
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine, University of Oporto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui Vaz
- Neurosurgery Department, Centro Hospitalar São João, Alameda Prof Hernani Monteiro, 4200–319 Porto, Portugal; (P.L.); (R.V.)
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine, University of Oporto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, 4710-057 Braga, Portugal
| |
Collapse
|
16
|
Sproull M, Mathen P, Miller CA, Mackey M, Cooley T, Smart D, Shankavaram U, Camphausen K. A Serum Proteomic Signature Predicting Survival in Patients with Glioblastoma. ACTA ACUST UNITED AC 2019; 4. [PMID: 33884377 DOI: 10.16966/2576-5833.117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Purpose Glioblastoma (GBM) is the most common form of brain tumor and has a uniformly poor prognosis. Development of prognostic biomarkers in easily accessible serum samples have the potential to improve the outcomes of patients with GBM through personalized therapy planning. Material/Methods In this study pre-treatment serum samples from 30 patients newly diagnosed with GBM were evaluated using a 40-protein multiplex ELISA platform. Analysis of potentially relevant gene targets using The Cancer Genome Atlas database was done using the Glioblastoma Bio Discovery Portal (GBM-BioDP). A ten-biomarker subgroup of clinically relevant molecules was selected using a functional grouping analysis of the 40 plex genes with two genes selected from each group on the basis of degree of variance, lack of co-linearity with other biomarkers and clinical interest. A Multivariate Cox proportional hazard approach was used to analyze the relationship between overall survival (OS), gene expression, and resection status as covariates. Results Thirty of 40 of the MSD molecules mapped to known genes within TCGA and separated the patient cohort into two main clusters centered predominantly around a grouping of classical and proneural versus the mesenchymal subtype as classified by Verhaak. Using the values for the 30 proteins in a prognostic index (PI) demonstrated that patients in the entire cohort with a PI below the median lived longer than those patients with a PI above the median (HR 1.8, p=0.001) even when stratified by both age and MGMT status. This finding was also consistent within each Verhaak subclass and highly significant (range p=0.0001-0.011). Additionally, a subset of ten proteins including, CRP, SAA, VCAM1, VEGF, MDC, TNFA, IL7, IL8, IL10, IL16 were found to have prognostic value within the TCGA database and a positive correlation with overall survival in GBM patients who had received gross tumor resection followed by conventional radiation therapy and temozolomide treatment concurrent with the addition of valproic acid. Conclusion These findings demonstrate that proteomic approaches to the development of prognostic assays for treatment of GBM may hold potential clinical value.
Collapse
Affiliation(s)
- Mary Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | - Peter Mathen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | | | - Megan Mackey
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | - Teresa Cooley
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | - Deedee Smart
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland USA
| |
Collapse
|
17
|
Peng H, Deng Y, Wang L, Cheng Y, Xu Y, Liao J, Wu H. Identification of Potential Biomarkers with Diagnostic Value in Pituitary Adenomas Using Prediction Analysis for Microarrays Method. J Mol Neurosci 2019; 69:399-410. [PMID: 31280474 DOI: 10.1007/s12031-019-01369-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/25/2019] [Indexed: 01/17/2023]
Abstract
Pituitary adenomas are the most common intrasellar tumors. Patients should be identified at an early stage so that effective treatment can be implemented. The study aims at detecting the potential biomarkers with diagnostic value of pituitary adenomas. Using a total of seven gene expression profiles (GEPs) of the datasets from the Gene Expression Omnibus (GEO) database, we first screened 1980 significant differentially expressed genes (DEGs). Then, we employed the prediction analysis for microarray (PAM) algorithm to identify 340 significant DEGs able to differ pituitary tumor from normal samples, which include 208 upregulated DEGs and 132 downregulated DEGs. DAVID database was used to carry out the enrichment analysis on Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathways. We found that upregulated candidates were enriched in protein folding and metabolic pathways. Downregulated DEGs saw a significant enrichment in insulin receptor signaling pathway and hedgehog signaling pathway. Based on the protein-protein interaction (PPI) network as well as module analysis, we determined ten hub genes including PHLPP, ENO2, ACTR1A, EHHADH, EHMT2, FOXO1, DLD, CCT2, CSNK1D, and CETN2 that could be potential biomarkers with diagnostic value in pituitary adenomas. In conclusion, the study contributes to reliable and potential molecular biomarkers with diagnostic value. Moreover, these potential biomarkers may be used for prognosis and new therapeutic targets for the pituitary adenomas.
Collapse
Affiliation(s)
- Hu Peng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China.,Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yue Deng
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Longhao Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Yin Cheng
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yaping Xu
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jianchun Liao
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Hao Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China. .,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China.
| |
Collapse
|
18
|
Establishing a many-cytokine signature via multivariate anomaly detection. Sci Rep 2019; 9:9684. [PMID: 31273258 PMCID: PMC6609612 DOI: 10.1038/s41598-019-46097-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/24/2019] [Indexed: 02/02/2023] Open
Abstract
Establishing a cytokine signature associated to some medical condition is an important task in immunology. Increasingly, large numbers of cytokines are used for signatures, via lists of reference ranges for each individual cytokine or ratios of cytokines. Here we argue that this common approach has weaknesses, especially when many different cytokines are analysed. Instead, we propose that establishing signatures can be framed as a multivariate anomaly detection problem, and hence exploit the many statistical methods available for this. In this framework, whether or not a given subject’s profile matches the cytokine signature of some condition is determined by whether or not the profile is typical of reference samples of that condition, as judged by an anomaly detection algorithm. We examine previously published cytokine data sets associated to pregnancy complications, brain tumours, and rheumatoid arthritis, as well as normal healthy control samples, and test the performance of a range of anomaly detection algorithms on these data, identifying the best performing methods. Finally, we suggest that this anomaly detection approach could be adopted more widely for general multi-biomarker signatures.
Collapse
|
19
|
Koper OM, Kamińska J, Sawicki K, Reszeć J, Rutkowski R, Jadeszko M, Mariak Z, Dymicka-Piekarska V, Kemona H. Cerebrospinal fluid and serum IL-8, CCL2, and ICAM-1 concentrations in astrocytic brain tumor patients. Ir J Med Sci 2017; 187:767-775. [PMID: 29086194 DOI: 10.1007/s11845-017-1695-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/04/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND The aim of the study was the evaluation of serum and CSF concentrations of CCL2, IL-8, and sICAM-1 in patients with astrocytic tumors as compared to a group of non-tumoral patients. METHODS Chemokine concentrations were measured using the ELISA method. RESULTS Regardless of the parameter tested and the patient group (brain tumor or non-tumoral patients), statistical differences (P < 0.05) were found between concentrations obtained in CSF compared to values obtained in serum for all proteins tested. CSF IL-8 concentrations were significantly elevated in CNS tumor patients as compared to non-tumoral individuals (P = 0.000); serum CCL2 and sICAM-1 concentrations were significantly decreased in CNS tumors in comparison with the comparative group (P = 0.002 and P = 0.026, respectively). Among proteins tested in the serum, a higher area under the ROC curve (AUC) revealed CCL2 compared to sICAM-1 in differentiating subjects with CNS brain tumors from non-tumoral subjects. AUC for CSF IL-8 was higher than for its index (CSF IL-8/serum IL-8). CONCLUSIONS For individual biomarkers (IL-8 and CCL2, sICAM-1), measured in CNS brain tumor patients, the appropriate material, respectively CSF or serum, should be chosen and quantitatively tested. Increased cerebrospinal fluid IL-8 with decreased serum CCL2 create a pattern of biomarkers, which may be helpful in the management of CNS astrocytic brain tumors.
Collapse
Affiliation(s)
- O M Koper
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, ul. Waszyngtona 15A, 15-269, Białystok, Poland.
| | - J Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, ul. Waszyngtona 15A, 15-269, Białystok, Poland
| | - K Sawicki
- Department of Neurosurgery, Clinical Hospital of the Medical University of Bialystok, Białystok, Poland
| | - J Reszeć
- Department of Pathomorphology, Medical University of Bialystok, Białystok, Poland
| | - R Rutkowski
- Department of Neurosurgery, Clinical Hospital of the Medical University of Bialystok, Białystok, Poland
| | - M Jadeszko
- Department of Neurosurgery, Clinical Hospital of the Medical University of Bialystok, Białystok, Poland
| | - Z Mariak
- Department of Neurosurgery, Clinical Hospital of the Medical University of Bialystok, Białystok, Poland
| | - V Dymicka-Piekarska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, ul. Waszyngtona 15A, 15-269, Białystok, Poland
| | - H Kemona
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, ul. Waszyngtona 15A, 15-269, Białystok, Poland
| |
Collapse
|
20
|
Schwartzbaum J, Wang M, Root E, Pietrzak M, Rempala GA, Huang RP, Johannesen TB, Grimsrud TK. A nested case-control study of 277 prediagnostic serum cytokines and glioma. PLoS One 2017; 12:e0178705. [PMID: 28594935 PMCID: PMC5464586 DOI: 10.1371/journal.pone.0178705] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/17/2017] [Indexed: 01/08/2023] Open
Abstract
Recent research shows bidirectional communication between the normal brain and the peripheral immune system. Glioma is a primary brain tumor characterized by systemic immunosuppression. To better understand gliomagenesis, we evaluated associations between 277 prediagnostic serum cytokines and glioma. We used glioma (n = 487) and matched control (n = 487) specimens from the Janus Serum Bank Cohort in Oslo, Norway. Conditional logistic regression allowed us to identify those cytokines that were individually associated with glioma. Next, we used heat maps to compare case to control Pearson correlation matrices of 12 cytokines modeled in an in silico study of the interaction between the microenvironment and the tumor. We did the same for case-control correlation matrices of lasso-selected cytokines and all 277 cytokines in the data set. Cytokines related to glioma risk (P ≤ .05) more than 10 years before diagnosis are sIL10RB, VEGF, beta-Catenin and CCL22. LIF was associated with decreased glioma risk within five years before glioma diagnosis (odds ratio (OR) = 0.47, 95% confidence interval (CI) = 0.23, 0.94). After adjustment for cytokines above, the previously observed interaction between IL4 and sIL4RA persisted (> 20 years before diagnosis, OR = 1.72, 95% CI = 1.20, 2.47). In addition, during this period, case correlations among 12 cytokines were weaker than were those among controls. This pattern was also observed among 30 lasso- selected cytokines and all 277 cytokines. We identified four cytokines and one interaction term that were independently related to glioma risk. We have documented prediagnostic changes in serum cytokine levels that may reflect the presence of a preclinical tumor.
Collapse
Affiliation(s)
- Judith Schwartzbaum
- Division of Epidemiology, College of Public Health, Ohio State University, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Min Wang
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
- Department of Biomedical Informatics, Ohio State University, Columbus, Ohio, United States of America
| | - Elisabeth Root
- Department of Geography, Ohio State University, Columbus, Ohio, United States of America
| | - Maciej Pietrzak
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
- Division of Biostatistics, College of Public Health, Ohio State University, Columbus, Ohio, United States of America
| | - Grzegorz A. Rempala
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
- Division of Biostatistics, College of Public Health, Ohio State University, Columbus, Ohio, United States of America
| | - Ruo-Pan Huang
- RayBiotech, Inc., Norcross, Georgia, United States of America
- RayBiotech, Inc. Guangzhou, China
| | | | - Tom K. Grimsrud
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| |
Collapse
|
21
|
Paul Y, Mondal B, Patil V, Somasundaram K. DNA methylation signatures for 2016 WHO classification subtypes of diffuse gliomas. Clin Epigenetics 2017; 9:32. [PMID: 28392842 PMCID: PMC5379538 DOI: 10.1186/s13148-017-0331-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/20/2017] [Indexed: 01/11/2023] Open
Abstract
Background Glioma is the most common of all primary brain tumors with poor prognosis and high mortality. The 2016 World Health Organization classification of the tumors of central nervous system uses molecular parameters in addition to histology to redefine many tumor entities. The new classification scheme divides diffuse gliomas into low-grade glioma (LGG) and glioblastoma (GBM) as per histology. LGGs are further divided into isocitrate dehydrogenase (IDH) wild type or mutant, which is further classified into either oligodendroglioma that harbors 1p/19q codeletion or diffuse astrocytoma that has an intact 1p/19q loci but enriched for ATRX loss and TP53 mutation. GBMs are divided into IDH wild type that corresponds to primary or de novo GBMs and IDH mutant that corresponds to secondary or progressive GBMs. To make the 2016 WHO subtypes of diffuse gliomas more robust, we carried out Prediction Analysis of Microarrays (PAM) to develop DNA methylation signatures for these subtypes. Results In this study, we applied PAM on a training set of diffuse gliomas derived from The Cancer Genome Atlas (TCGA) and identified DNA methylation signatures to classify LGG IDH wild type from LGG IDH mutant, LGG IDH mutant with 1p/19q codeletion from LGG IDH mutant with intact 1p/19q loci and GBM IDH wild type from GBM IDH mutant with an accuracy of 99–100%. The signatures were validated using the test set of diffuse glioma samples derived from TCGA with an accuracy of 96 to 99%. In addition, we also carried out additional validation of all three signatures using independent LGG and GBM cohorts. Further, the methylation signatures identified a fraction of samples as discordant, which were found to have molecular and clinical features typical of the subtype as identified by methylation signatures. Conclusions Thus, we identified methylation signatures that classified different subtypes of diffuse glioma accurately and propose that these signatures could complement 2016 WHO classification scheme of diffuse glioma. Electronic supplementary material The online version of this article (doi:10.1186/s13148-017-0331-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yashna Paul
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012 India
| | - Baisakhi Mondal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012 India
| | - Vikas Patil
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012 India
| | - Kumaravel Somasundaram
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012 India
| |
Collapse
|
22
|
Chen MH, Lu C, Sun J, Chen XD, Dai JX, Cai JY, Chen XL. Diagnostic and prognostic value of serum vitronectin levels in human glioma. J Neurol Sci 2016; 371:54-59. [DOI: 10.1016/j.jns.2016.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 09/30/2016] [Accepted: 10/14/2016] [Indexed: 12/01/2022]
|
23
|
Anderson B. Previously Undiagnosed Malignant Brain Tumor Discovered During Examination of a Patient Seeking Chiropractic Care. J Chiropr Med 2016; 15:42-6. [DOI: 10.1016/j.jcm.2016.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 12/02/2015] [Accepted: 12/15/2015] [Indexed: 11/29/2022] Open
|
24
|
Fabbri E, Brognara E, Montagner G, Ghimenton C, Eccher A, Cantù C, Khalil S, Bezzerri V, Provezza L, Bianchi N, Finotti A, Borgatti M, Moretto G, Chilosi M, Cabrini G, Gambari R. Regulation of IL-8 gene expression in gliomas by microRNA miR-93. BMC Cancer 2015; 15:661. [PMID: 26449498 PMCID: PMC4598972 DOI: 10.1186/s12885-015-1659-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 09/28/2015] [Indexed: 12/23/2022] Open
Abstract
Background Different strategies have been proposed to target neoangiogenesis in gliomas, besides those targeting Vascular Endothelial Growth Factor (VEGF). The chemokine Interleukin-8 (IL-8) has been shown to possess both tumorigenic and proangiogenic properties. Although different pathways of induction of IL-8 gene expression have been already elucidated, few data are available on its post-transcriptional regulation in gliomas. Methods Here we investigated the role of the microRNA miR-93 on the expression levels of IL-8 and other pro-inflammatory genes by RT-qPCR and Bio-Plex analysis. We used different disease model systems, including clinical samples from glioma patients and two glioma cell lines, U251 and T98G. Results IL-8 and VEGF transcripts are highly expressed in low and high grade gliomas in respect to reference healthy brain; miR-93 expression is also increased and inversely correlated with transcription of IL-8 and VEGF genes. Computational analysis showed the presence of miR-93 consensus sequences in the 3′UTR region of both VEGF and IL-8 mRNAs, predicting possible interaction with miR-93 and suggesting a potential regulatory role of this microRNA. In vitro transfection with pre-miR-93 and antagomiR-93 inversely modulated VEGF and IL-8 gene expression and protein release when the glioma cell line U251 was considered. Similar data were obtained on IL-8 gene regulation in the other glioma cell line analyzed, T98G. The effect of pre-miR-93 and antagomiR-93 in U251 cells has been extended to the secretion of a panel of cytokines, chemokines and growth factors, which consolidated the concept of a role of miR-93 in IL-8 and VEGF gene expression and evidenced a potential regulatory role also for MCP-1 and PDGF (also involved in angiogenesis). Conclusion In conclusion, our results suggest an increasing role of miR-93 in regulating the level of expression of several genes involved in the angiogenesis of gliomas. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1659-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Enrica Fabbri
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Eleonora Brognara
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Giulia Montagner
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Claudio Ghimenton
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Albino Eccher
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Cinzia Cantù
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Susanna Khalil
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Valentino Bezzerri
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Lisa Provezza
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Nicoletta Bianchi
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| | - Giuseppe Moretto
- Department of Neurosciences, University-Hospital of Verona, P.le A Stefani n.1, Verona, 37126, Italy.
| | - Marco Chilosi
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Giulio Cabrini
- Department of Pathology and Diagnostics, Laboratory of Molecular Pathology, University-Hospital of Verona, P.le A Stefani n.1, 37126, Verona, Italy.
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara n.74, 44121, Ferrara, Italy.
| |
Collapse
|