1
|
Giordano C, Marrone L, Romano S, Della Pepa GM, Donzelli CM, Tufano M, Capasso M, Lasorsa VA, Quintavalle C, Guerri G, Martucci M, Auricchio A, Gessi M, Sala E, Olivi A, Romano MF, Gaudino S. The FKBP51s Splice Isoform Predicts Unfavorable Prognosis in Patients with Glioblastoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1296-1306. [PMID: 38651817 PMCID: PMC11097923 DOI: 10.1158/2767-9764.crc-24-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/21/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
The primary treatment for glioblastoma (GBM) is removing the tumor mass as defined by MRI. However, MRI has limited diagnostic and predictive value. Tumor-associated macrophages (TAM) are abundant in GBM tumor microenvironment (TME) and are found in peripheral blood (PB). FKBP51 expression, with its canonical and spliced isoforms, is constitutive in immune cells and aberrant in GBM. Spliced FKBP51s supports M2 polarization. To find an immunologic signature that combined with MRI could advance in diagnosis, we immunophenotyped the macrophages of TME and PB from 37 patients with GBM using FKBP51s and classical M1-M2 markers. We also determined the tumor levels of FKBP51s, PD-L1, and HLA-DR. Tumors expressing FKBP51s showed an increase in various M2 phenotypes and regulatory T cells in PB, indicating immunosuppression. Tumors expressing FKBP51s also activated STAT3 and were associated with reduced survival. Correlative studies with MRI and tumor/macrophages cocultures allowed to interpret TAMs. Tumor volume correlated with M1 infiltration of TME. Cocultures with spheroids produced M1 polarization, suggesting that M1 macrophages may infiltrate alongside cancer stem cells. Cocultures of adherent cells developed the M2 phenotype CD163/FKBP51s expressing pSTAT6, a transcription factor enabling migration and invasion. In patients with recurrences, increased counts of CD163/FKBP51s monocyte/macrophages in PB correlated with callosal infiltration and were accompanied by a concomitant decrease in TME-infiltrating M1 macrophages. PB PD-L1/FKBP51s connoted necrotic tumors. In conclusion, FKBP51s identifies a GBM subtype that significantly impairs the immune system. Moreover, FKBP51s marks PB macrophages associated with MRI features of glioma malignancy that can aid in patient monitoring. SIGNIFICANCE Our research suggests that by combining imaging with analysis of monocyte/macrophage subsets in patients with GBM, we can enhance our understanding of the disease and assist in its treatment. We discovered a similarity in the macrophage composition between the TME and PB, and through association with imaging, we could interpret macrophages. In addition, we identified a predictive biomarker that drew more attention to immune suppression of patients with GBM.
Collapse
Affiliation(s)
- Carolina Giordano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Laura Marrone
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Simona Romano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Giuseppe Maria Della Pepa
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Carlo Maria Donzelli
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Martina Tufano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Mario Capasso
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Vito Alessandro Lasorsa
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Cristina Quintavalle
- Istituto di Endocrinologia e Oncologia Sperimentale “Gaetano Salvatore” (IEOS), Consiglio Nazionale delle Ricerche (CNR), Napoli, Italia
| | - Giulia Guerri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Matia Martucci
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Annamaria Auricchio
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Marco Gessi
- UOS di Neuropatologia, UOC Anatomia Patologica, Fondazione Policlinico “A. Gemelli” IRCCS, Rome, Italy
| | - Evis Sala
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Olivi
- UOC Neurochirurgia, Istituto di Neurochirurgia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica S. Cuore, Roma, Italy
| | - Maria Fiammetta Romano
- Dipartmento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli, Federico II, Napoli, Italy
| | - Simona Gaudino
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, Universitaà Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
2
|
Won SE, Suh CH, Kim S, Park HJ, Kim KW. Summary of Key Points of the Response Assessment in Neuro-Oncology (RANO) 2.0. Korean J Radiol 2024; 25:407-411. [PMID: 38627876 PMCID: PMC11058423 DOI: 10.3348/kjr.2024.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 05/01/2024] Open
Affiliation(s)
- Sang Eun Won
- Asan Image Metrics, Clinical Trial Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sinae Kim
- Asan Image Metrics, Clinical Trial Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Wen PY, van den Bent M, Youssef G, Cloughesy TF, Ellingson BM, Weller M, Galanis E, Barboriak DP, de Groot J, Gilbert MR, Huang R, Lassman AB, Mehta M, Molinaro AM, Preusser M, Rahman R, Shankar LK, Stupp R, Villanueva-Meyer JE, Wick W, Macdonald DR, Reardon DA, Vogelbaum MA, Chang SM. RANO 2.0: Update to the Response Assessment in Neuro-Oncology Criteria for High- and Low-Grade Gliomas in Adults. J Clin Oncol 2023; 41:5187-5199. [PMID: 37774317 PMCID: PMC10860967 DOI: 10.1200/jco.23.01059] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/12/2023] [Accepted: 08/10/2023] [Indexed: 10/01/2023] Open
Abstract
PURPOSE The Response Assessment in Neuro-Oncology (RANO) criteria for high-grade gliomas (RANO-HGG) and low-grade gliomas (RANO-LGG) were developed to improve reliability of response assessment in glioma trials. Over time, some limitations of these criteria were identified, and challenges emerged regarding integrating features of the modified RANO (mRANO) or the immunotherapy RANO (iRANO) criteria. METHODS Informed by data from studies evaluating the different criteria, updates to the RANO criteria are proposed (RANO 2.0). RESULTS We recommend a standard set of criteria for both high- and low-grade gliomas, to be used for all trials regardless of the treatment modalities being evaluated. In the newly diagnosed setting, the postradiotherapy magnetic resonance imaging (MRI), rather than the postsurgical MRI, will be used as the baseline for comparison with subsequent scans. Since the incidence of pseudoprogression is high in the 12 weeks after radiotherapy, continuation of treatment and confirmation of progression during this period with a repeat MRI, or histopathologic evidence of unequivocal recurrent tumor, are required to define tumor progression. However, confirmation scans are not mandatory after this period nor for the evaluation of treatment for recurrent tumors. For treatments with a high likelihood of pseudoprogression, mandatory confirmation of progression with a repeat MRI is highly recommended. The primary measurement remains the maximum cross-sectional area of tumor (two-dimensional) but volumetric measurements are an option. For IDH wild-type glioblastoma, the nonenhancing disease will no longer be evaluated except when assessing response to antiangiogenic agents. In IDH-mutated tumors with a significant nonenhancing component, clinical trials may require evaluating both the enhancing and nonenhancing tumor components for response assessment. CONCLUSION The revised RANO 2.0 criteria refine response assessment in gliomas.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Martin van den Bent
- Department Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Gilbert Youssef
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Timothy F Cloughesy
- UCLA Brain Tumor Program, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | | | - John de Groot
- Division of Neuro-Oncology, Department of Neurosurgery, University of California, San Francisco, CA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Raymond Huang
- Division of Neuro-radiology, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Andrew B Lassman
- Division of Neuro-Oncology, Department of Neurology, Herbert Irving Comprehensive Cancer Center and Irving Institute for Clinical and Translational Research, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, NY
| | | | - Annette M Molinaro
- Division of Biomedical Statistics and Informatics, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Rifaquat Rahman
- Department of Radiation Oncology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lalitha K Shankar
- Clinical Trials Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Roger Stupp
- Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center and Departments of Neurological Surgery, Neurology and Division of Hematology/Oncology, Northwestern University, Chicago, IL
| | - Javier E Villanueva-Meyer
- Departments of Radiology and Neurosurgery, University of California San Francisco, San Francisco, CA
| | - Wolfgang Wick
- Department of Neurology Heidelberg University Hospital & Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David R Macdonald
- Departments of Clinical Neurological Sciences and Oncology (Emeritus), Western University, London, Ontario, Canada
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Michael A Vogelbaum
- Departments of Neuro-Oncology and Neurosurgery, Moffitt Cancer Center, Tampa, FL
| | - Susan M Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of California, San Francisco, CA
| |
Collapse
|
4
|
D'Alessandris QG, Martini M, Cenci T, DI Bonaventura R, Lauretti L, Stumpo V, Olivi A, Larocca LM, Pallini R, Montano N. Tailored therapy for recurrent glioblastoma: report of a personalized molecular approach. J Neurosurg Sci 2023; 67:103-107. [PMID: 32550606 DOI: 10.23736/s0390-5616.20.04943-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Failure of clinical trials with targeted therapies in glioblastoma (GBM) is probably related to the enrollment of molecularly unselected patients. In this study we report the results of a precision medicine protocol in recurrent GBM. METHODS We prospectively evaluated 34 patients with recurrent GBM. We determined the expression of vascular endothelial growth factor (VEGF), epidermal growth factor receptor variant III (EGFRvIII), and phosphatase and tensin homolog (PTEN). According to the molecular pattern we administered bevacizumab alone in patients with VEGF overexpression, absence of EGFRvIII, and normal PTEN (group A; N.=16); bevacizumab + erlotinib in patients with VEGF overexpression, expression of EGFRvIII, and normal PTEN (group B; N.=14); and bevacizumab + sirolimus in patients with VEGF overexpression and loss of PTEN, irrespective of the EGFRvIII status (group C; N.=4). We evaluated the response rate, the clinical benefit rate, the 6-month progression-free survival (PFS-6), the 12-month PFS (PFS-12) and the safety profile of the treatment. Moreover, we compared our results with the ones of EORTC 26101 trial. RESULTS Response rate was 50% in the whole cohort with the highest rate in group C (75%). Clinical benefit rate was 71% with the highest rate in group C (75%). PFS-6 was 56% in the whole cohort with the highest rate in group B (64%). PFS-12 was 21% in the whole cohort with the highest rate in group B (29%). When comparing our results with those from the combination arm of the EORTC 26101 trial we found a significantly higher PFS-6 and PFS-12 in our cohort. CONCLUSIONS The precision medicine protocol for recurrent GBM is feasible and leads to improved results if compared with studies lacking molecular selection.
Collapse
Affiliation(s)
- Quintino G D'Alessandris
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Maurizio Martini
- Department of Pathology, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Tonia Cenci
- Department of Pathology, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Rina DI Bonaventura
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Liverana Lauretti
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Vittorio Stumpo
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Alessandro Olivi
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Luigi M Larocca
- Department of Pathology, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Roberto Pallini
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Nicola Montano
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy -
| |
Collapse
|
5
|
Pálsson S, Cerri S, Poulsen HS, Urup T, Law I, Van Leemput K. Predicting survival of glioblastoma from automatic whole-brain and tumor segmentation of MR images. Sci Rep 2022; 12:19744. [PMID: 36396681 PMCID: PMC9671967 DOI: 10.1038/s41598-022-19223-3] [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] [Received: 09/24/2021] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Survival prediction models can potentially be used to guide treatment of glioblastoma patients. However, currently available MR imaging biomarkers holding prognostic information are often challenging to interpret, have difficulties generalizing across data acquisitions, or are only applicable to pre-operative MR data. In this paper we aim to address these issues by introducing novel imaging features that can be automatically computed from MR images and fed into machine learning models to predict patient survival. The features we propose have a direct anatomical-functional interpretation: They measure the deformation caused by the tumor on the surrounding brain structures, comparing the shape of various structures in the patient's brain to their expected shape in healthy individuals. To obtain the required segmentations, we use an automatic method that is contrast-adaptive and robust to missing modalities, making the features generalizable across scanners and imaging protocols. Since the features we propose do not depend on characteristics of the tumor region itself, they are also applicable to post-operative images, which have been much less studied in the context of survival prediction. Using experiments involving both pre- and post-operative data, we show that the proposed features carry prognostic value in terms of overall- and progression-free survival, over and above that of conventional non-imaging features.
Collapse
Affiliation(s)
- Sveinn Pálsson
- grid.5170.30000 0001 2181 8870Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Stefano Cerri
- grid.5170.30000 0001 2181 8870Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Hans Skovgaard Poulsen
- grid.475435.4Department of Oncology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Urup
- grid.475435.4Department of Oncology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Ian Law
- grid.475435.4Department of Clinical Physiology, Nuclear Medicine and PET, Center of Diagnostic Investigation, Rigshospitalet, Copenhagen, Denmark
| | - Koen Van Leemput
- grid.5170.30000 0001 2181 8870Department of Health Technology, Technical University of Denmark, Lyngby, Denmark ,grid.32224.350000 0004 0386 9924Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
6
|
The Next Frontier in Health Disparities—A Closer Look at Exploring Sex Differences in Glioma Data and Omics Analysis, from Bench to Bedside and Back. Biomolecules 2022; 12:biom12091203. [PMID: 36139042 PMCID: PMC9496358 DOI: 10.3390/biom12091203] [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: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Sex differences are increasingly being explored and reported in oncology, and glioma is no exception. As potentially meaningful sex differences are uncovered, existing gender-derived disparities mirror data generated in retrospective and prospective trials, real-world large-scale data sets, and bench work involving animals and cell lines. The resulting disparities at the data level are wide-ranging, potentially resulting in both adverse outcomes and failure to identify and exploit therapeutic benefits. We set out to analyze the literature on women’s data disparities in glioma by exploring the origins of data in this area to understand the representation of women in study samples and omics analyses. Given the current emphasis on inclusive study design and research, we wanted to explore if sex bias continues to exist in present-day data sets and how sex differences in data may impact conclusions derived from large-scale data sets, omics, biospecimen analysis, novel interventions, and standard of care management.
Collapse
|
7
|
Takei J, Fukasawa N, Tanaka T, Yamamoto Y, Tamura R, Sasaki H, Akasaki Y, Kamata Y, Murahashi M, Shimoda M, Murayama Y. Impact of Neoadjuvant Bevacizumab on Neuroradiographic Response and Histological Findings Related to Tumor Stemness and the Hypoxic Tumor Microenvironment in Glioblastoma: Paired Comparison Between Newly Diagnosed and Recurrent Glioblastomas. Front Oncol 2022; 12:898614. [PMID: 35785200 PMCID: PMC9247463 DOI: 10.3389/fonc.2022.898614] [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: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 12/03/2022] Open
Abstract
Background Previously, we reported that bevacizumab (Bev) produces histological and neuroradiographic alterations including changes in tumor oxygenation, induction of an immunosupportive tumor microenvironment, and inhibition of stemness. To confirm how those effects vary during Bev therapy, paired samples from the same patients with newly diagnosed glioblastoma (GBM) who received preoperative neoadjuvant Bev (neoBev) were investigated with immunohistochemistry before and after recurrence. Methods Eighteen samples from nine patients with newly diagnosed GBM who received preoperative neoBev followed by surgery and chemoradiotherapy and then autopsy or salvage surgery after recurrence were investigated. The expression of carbonic anhydrase 9 (CA9), hypoxia-inducible factor-1 alpha (HIF-1α), nestin, and Forkhead box M1 (FOXM1) was evaluated with immunohistochemistry. For comparison between neoBev and recurrent tumors, we divided the present cohort into two groups based on neuroradiographic response: good and poor responders (GR and PR, respectively) to Bev were defined by the tumor regression rate on T1-weighted images with gadolinium enhancement (T1Gd) and fluid-attenuated inversion recovery images. Patterns of recurrence after Bev therapy were classified as cT1 flare-up and T2-diffuse/T2-circumscribed. Furthermore, we explored the possibility of utilizing FOXM1 as a biomarker of survival in this cohort. Results A characteristic “pseudo-papillary”-like structure containing round-shaped tumor cells clustered adjacent to blood vessels surrounded by spindle-shaped tumor cells was seen only in recurrent tumors. Tumor cells at the outer part of the “pseudo-papillary” structure were CA9-positive (CA9+)/HIF-1α+, whereas cells at the inner part of this structure were CA9−/HIF-1α+ and nestin+/FOXM1+. CA9 and HIF-1α expression was lower in T1Gd-GR and decreased in the “T2-circumscribed/T2-diffuse” pattern compared with the “T1 flare-up” pattern, suggesting that tumor oxygenation was frequently observed in T1Gd-GR in initial tumors and in the “T2-circumscribed/T2-diffuse” pattern in recurrent tumors. FOXM1 low-expression tumors tended to have a better prognosis than that of FOXM1 high-expression tumors. Conclusion A “pseudo-papillary” structure was seen in recurrent GBM after anti-vascular endothelial growth factor therapy. Bev may contribute to tumor oxygenation, leading to inhibition of stemness and correlation with a neuroimaging response during Bev therapy. FOXM1 may play a role as a biomarker of survival during Bev therapy.
Collapse
Affiliation(s)
- Jun Takei
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Nei Fukasawa
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
- Department of Neurosurgery, Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Japan
- *Correspondence: Toshihide Tanaka,
| | - Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine Daisan Hospital, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Mutsunori Murahashi
- Division of Oncology, Research Center for Medical Sciences, Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Lim YC, Jensen KE, Aguilar-Morante D, Vardouli L, Vitting-Seerup K, Gimple RC, Wu Q, Pedersen H, Elbaek KJ, Gromova I, Ihnatko R, Kristensen BW, Petersen JK, Skjoth-Rasmussen J, Flavahan W, Rich JN, Hamerlik P. Non-metabolic functions of phosphofructokinase-1 orchestrate tumor cellular invasion and genome maintenance under bevacizumab therapy. Neuro Oncol 2022; 25:248-260. [PMID: 35608632 PMCID: PMC9925708 DOI: 10.1093/neuonc/noac135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly lethal malignancy for which neoangiogenesis serves as a defining hallmark. The anti-VEGF antibody, bevacizumab, has been approved for the treatment of recurrent GBM, but resistance is universal. METHODS We analyzed expression data of GBM patients treated with bevacizumab to discover potential resistance mechanisms. Patient-derived xenografts (PDXs) and cultures were interrogated for effects of phosphofructokinase-1, muscle isoform (PFKM) loss on tumor cell motility, migration, and invasion through genetic and pharmacologic targeting. RESULTS We identified PFKM as a driver of bevacizumab resistance. PFKM functions dichotomize based on subcellular location: cytosolic PFKM interacted with KIF11, a tubular motor protein, to promote tumor invasion, whereas nuclear PFKM safeguarded genomic stability of tumor cells through interaction with NBS1. Leveraging differential transcriptional profiling, bupivacaine phenocopied genetic targeting of PFKM, and enhanced efficacy of bevacizumab in preclinical GBM models in vivo. CONCLUSION PFKM drives novel molecular pathways in GBM, offering a translational path to a novel therapeutic paradigm.
Collapse
Affiliation(s)
| | | | | | | | - Kristoffer Vitting-Seerup
- Danish Cancer Society, Denmark,Department of Health Technology, Danish Technical University, Denmark
| | - Ryan C Gimple
- Department of Medicine, Division of Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | | | - Robert Ihnatko
- Institute of Pathology, University Medical Center, Goettingen University, Germany
| | | | - Jeanette K Petersen
- Department of Pathology, Odense University Hospital, Denmark,Department of Clinical Research, University of Southern Denmark, Denmark
| | | | - William Flavahan
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeremy N Rich
- Corresponding Author: Jeremy Rich, MD, MHS, MBA, UPMC Cancer Pavilion, 5150 Centre Avenue, 5th Floor Pittsburgh, PA 15232; Tel: 4126233364 ()
| | - Petra Hamerlik
- Corresponding Author: Petra Hamerlik, MSc, PhD, Brain Tumor Biology, Danish Cancer Society Research Center, Strandboulevarden 49, 2100 Copenhagen, Denmark; Tel: 35257413 ()
| |
Collapse
|
9
|
Moon HH, Park JE, Kim YH, Kim JH, Kim HS. Contrast enhancing pattern on pre-treatment MRI predicts response to anti-angiogenic treatment in recurrent glioblastoma: comparison of bevacizumab and temozolomide treatment. J Neurooncol 2022; 157:405-415. [PMID: 35275335 DOI: 10.1007/s11060-022-03980-2] [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: 01/06/2022] [Accepted: 03/04/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate the value of the contrast enhancing pattern on pre-treatment MRI for predicting the response to anti-angiogenic treatment in patients with IDH-wild type recurrent glioblastoma. METHODS This retrospective study enrolled 65 patients with IDH wild-type recurrent glioblastoma who received standard therapy and then received either bevacizumab (46 patients) or temozolomide (19 patients) as a secondary treatment. The contrast enhancing pattern on pre-treatment MRI was visually analyzed and dichotomized into contrast enhancing lesion (CEL) dominant and non-enhancing lesion (NEL) dominant types. Quantitative volumetric analysis was used to support the dichotomization. The Kaplan-Meier method and Cox proportional hazards regression analysis were used to stratify progression free survival (PFS) according to the treatment in the entire patients, CEL dominant group, and NEL dominant group. RESULTS In all patients, the PFS of those treated with bevacizumab was not significantly different from those treated with temozolomide (log-rank test, P = 0.96). When the contrast enhancing pattern was considered, bevacizumab was associated with longer PFS in the CEL dominant group (P = 0.031), whereas temozolomide showed longer PFS in the NEL dominant group (P = 0.022). Quantitative analysis revealed mean values for the proportion of solid-enhancing tumor of 13.7% for the CEL dominant group and 4.3% for the NEL dominant group. CONCLUSION Patients with the CEL dominant type showed a better treatment response to bevacizumab, whereas NEL dominant types showed a better response to temozolomide. The contrast enhancing pattern on pre-treatment MRI can be used to stratify patients with IDH wild-type recurrent glioblastoma according to the effect of anti-angiogenic treatment.
Collapse
Affiliation(s)
- Hye Hyeon Moon
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea.
| | - Young-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 43 Olympic-ro 88, Songpa-Gu, Seoul, 05505, South Korea
| |
Collapse
|
10
|
Gaudino S, Marziali G, Giordano C, Gigli R, Varcasia G, Magnani F, Chiesa S, Balducci M, Costantini AM, Della Pepa GM, Olivi A, Russo R, Colosimo C. Regorafenib in Glioblastoma Recurrence: How to Deal With MR Imaging Treatments Changes. FRONTIERS IN RADIOLOGY 2022; 1:790456. [PMID: 37492166 PMCID: PMC10365006 DOI: 10.3389/fradi.2021.790456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/29/2021] [Indexed: 07/27/2023]
Abstract
The treatment of recurrent high-grade gliomas remains a major challenge of daily neuro-oncology practice, and imaging findings of new therapies may be challenging. Regorafenib is a multi-kinase inhibitor that has recently been introduced into clinical practice to treat recurrent glioblastoma, bringing with it a novel panel of MRI imaging findings. On the basis of the few data in the literature and on our personal experience, we have identified the main MRI changes during regorafenib therapy, and then, we defined two different patterns, trying to create a simple summary line of the main changes of pathological tissue during therapy. We named these patterns, respectively, pattern A (less frequent, similar to classical progression disease) and pattern B (more frequent, with decreased diffusivity and decrease contrast-enhancement). We have also reported MR changes concerning signal intensity on T1-weighted and T2-weighted images, SWI, and perfusion imaging, derived from the literature (small series or case reports) and from our clinical experience. The clinical implication of these imaging modifications remains to be defined, taking into account that we are still at the dawn in the evaluation of such imaging modifications.
Collapse
Affiliation(s)
- Simona Gaudino
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
| | - Giammaria Marziali
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Carolina Giordano
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Riccardo Gigli
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Varcasia
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesca Magnani
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Silvia Chiesa
- Department of Diagnostic Imaging, Oncological Radiotherapy, and Hematology, UOC di Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mario Balducci
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy, and Hematology, UOC di Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandro Maria Costantini
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, Rome, Italy
| | - Alessandro Olivi
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, Rome, Italy
| | - Rosellina Russo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Cesare Colosimo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
| |
Collapse
|
11
|
Volumetric study reveals the relationship between outcome and early radiographic response during bevacizumab-containing chemoradiotherapy for unresectable glioblastoma. J Neurooncol 2021; 154:187-196. [PMID: 34322829 PMCID: PMC8437873 DOI: 10.1007/s11060-021-03812-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/16/2021] [Indexed: 01/14/2023]
Abstract
PURPOSE Although we have shown the clinical benefit of bevacizumab (BEV) in the treatment of unresectable newly diagnosed glioblastomas (nd-GBM), the relationship between early radiographic response and survival outcome remains unclear. We performed a volumetric study of early radiographic responses in nd-GBM treated with BEV. METHODS Twenty-two patients with unresectable nd-GBM treated with BEV during concurrent temozolomide radiotherapy were analyzed. An experienced neuroradiologist interpreted early responses on fluid-attenuated inversion recovery (FLAIR) and gadolinium-enhanced T1-weighted images (GdT1WI). Volumetric changes were evaluated using diffusion-weighted imaging (DWI) and GdT1WI according to the Response Assessment in Neuro-Oncology (RANO) criteria. The results were categorized into improved (complete response [CR] or partial response [PR]) or non-improved (stable disease [SD] or progressive disease [PD]) groups; outcomes were compared using Kaplan-Meier analysis. RESULTS The volumetric GdT1WI improvement was a significant predictive factor for overall survival (OS) prolongation (p = 0.0093, median OS: 24.7 vs. 13.6 months); however, FLAIR and DWI images were not predictive. The threshold for the neuroradiologist's interpretation of improvement in GdT1WI was nearly 20% of volume reduction, which was lesser than 50%, the definition of PR applied in the RANO criteria. However, even less stringent neuroradiologist interpretation could successfully predict OS prolongation (improved vs. non-improved: p = 0.0067, median OS: 17.6 vs. 8.3 months). Significant impact of OS on the early response in volumetric GdT1WI was observed within the cut-off range of 20-50% (20%, p = 0.0315; 30%, p = 0.087; 40%, p = 0.0456). CONCLUSIONS Early response during BEV-containing chemoradiation can be a predictive indicator of patient outcome in unresectable nd-GBM.
Collapse
|
12
|
Cho SJ, Kim HS, Suh CH, Park JE. Radiological Recurrence Patterns after Bevacizumab Treatment of Recurrent High-Grade Glioma: A Systematic Review and Meta-Analysis. Korean J Radiol 2020; 21:908-918. [PMID: 32524791 PMCID: PMC7289701 DOI: 10.3348/kjr.2019.0898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 03/05/2020] [Indexed: 12/28/2022] Open
Abstract
Objective To categorize the radiological patterns of recurrence after bevacizumab treatment and to derive the pooled proportions of patients with recurrent malignant glioma showing the different radiological patterns. Materials and Methods A systematic literature search in the Ovid-MEDLINE and EMBASE databases was performed to identify studies reporting radiological recurrence patterns in patients with recurrent malignant glioma after bevacizumab treatment failure until April 10, 2019. The pooled proportions according to radiological recurrence patterns (geographically local versus non-local recurrence) and predominant tumor portions (enhancing tumor versus non-enhancing tumor) after bevacizumab treatment were calculated. Subgroup and meta-regression analyses were also performed. Results The systematic review and meta-analysis included 17 articles. The pooled proportions were 38.3% (95% confidence interval [CI], 30.6–46.1%) for a geographical radiologic pattern of non-local recurrence and 34.2% (95% CI, 27.3–41.5%) for a non-enhancing tumor-predominant recurrence pattern. In the subgroup analysis, the pooled proportion of non-local recurrence in the patients treated with bevacizumab only was slightly higher than that in patients treated with the combination with cytotoxic chemotherapy (34.9% [95% CI, 22.8–49.4%] versus 22.5% [95% CI, 9.5–44.6%]). Conclusion A substantial proportion of high-grade glioma patients show non-local or non-enhancing radiologic patterns of recurrence after bevacizumab treatment, which may provide insight into surrogate endpoints for treatment failure in clinical trials of recurrent high-grade glioma.
Collapse
Affiliation(s)
- Se Jin Cho
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
13
|
Hanna C, Lawrie TA, Rogozińska E, Kernohan A, Jefferies S, Bulbeck H, Ali UM, Robinson T, Grant R. Treatment of newly diagnosed glioblastoma in the elderly: a network meta-analysis. Cochrane Database Syst Rev 2020; 3:CD013261. [PMID: 32202316 PMCID: PMC7086476 DOI: 10.1002/14651858.cd013261.pub2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND A glioblastoma is a fatal type of brain tumour for which the standard of care is maximum surgical resection followed by chemoradiotherapy, when possible. Age is an important consideration in this disease, as older age is associated with shorter survival and a higher risk of treatment-related toxicity. OBJECTIVES To determine the most effective and best-tolerated approaches for the treatment of elderly people with newly diagnosed glioblastoma. To summarise current evidence for the incremental resource use, utilities, costs and cost-effectiveness associated with these approaches. SEARCH METHODS We searched electronic databases including the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE and Embase to 3 April 2019, and the NHS Economic Evaluation Database (EED) up to database closure. We handsearched clinical trial registries and selected neuro-oncology society conference proceedings from the past five years. SELECTION CRITERIA Randomised trials (RCTs) of treatments for glioblastoma in elderly people. We defined 'elderly' as 70+ years but included studies defining 'elderly' as over 65+ years if so reported. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods for study selection and data extraction. Where sufficient data were available, treatment options were compared in a network meta-analysis (NMA) using Stata software (version 15.1). For outcomes with insufficient data for NMA, pairwise meta-analysis were conducted in RevMan. The GRADE approach was used to grade the evidence. MAIN RESULTS We included 12 RCTs involving approximately 1818 participants. Six were conducted exclusively among elderly people (either defined as 65 years or older or 70 years or older) with newly diagnosed glioblastoma, the other six reported data for an elderly subgroup among a broader age range of participants. Most participants were capable of self-care. Study quality was commonly undermined by lack of outcome assessor blinding and attrition. NMA was only possible for overall survival; other analyses were pair-wise meta-analyses or narrative syntheses. Seven trials contributed to the NMA for overall survival, with interventions including supportive care only (one trial arm); hypofractionated radiotherapy (RT40; four trial arms); standard radiotherapy (RT60; five trial arms); temozolomide (TMZ; three trial arms); chemoradiotherapy (CRT; three trial arms); bevacizumab with chemoradiotherapy (BEV_CRT; one trial arm); and bevacizumab with radiotherapy (BEV_RT). Compared with supportive care only, NMA evidence suggested that all treatments apart from BEV_RT prolonged survival to some extent. Overall survival High-certainty evidence shows that CRT prolongs overall survival (OS) compared with RT40 (hazard ratio (HR) 0.67, 95% confidence interval (CI) 0.56 to 0.80) and low-certainty evidence suggests that CRT may prolong overall survival compared with TMZ (TMZ versus CRT: HR 1.42, 95% CI 1.01 to 1.98). Low-certainty evidence also suggests that adding BEV to CRT may make little or no difference (BEV_CRT versus CRT: HR 0.83, 95% CrI 0.48 to 1.44). We could not compare the survival effects of CRT with different radiotherapy fractionation schedules (60 Gy/30 fractions and 40 Gy/15 fractions) due to a lack of data. When treatments were ranked according to their effects on OS, CRT ranked higher than TMZ, RT and supportive care only, with the latter ranked last. BEV plus RT was the only treatment for which there was no clear benefit in OS over supportive care only. One trial comparing tumour treating fields (TTF) plus adjuvant chemotherapy (TTF_AC) with adjuvant chemotherapy alone could not be included in the NMA as participants were randomised after receiving concomitant chemoradiotherapy, not before. Findings from the trial suggest that the intervention probably improves overall survival in this selected patient population. We were unable to perform NMA for other outcomes due to insufficient data. Pairwise analyses were conducted for the following. Quality of life Moderate-certainty narrative evidence suggests that overall, there may be little difference in QoL between TMZ and RT, except for discomfort from communication deficits, which are probably more common with RT (1 study, 306 participants, P = 0.002). Data on QoL for other comparisons were sparse, partly due to high dropout rates, and the certainty of the evidence tended to be low or very low. Progression-free survival High-certainty evidence shows that CRT increases time to disease progression compared with RT40 (HR 0.50, 95% CI 0.41 to 0.61); moderate-certainty evidence suggests that RT60 probably increases time to disease progression compared with supportive care only (HR 0.28, 95% CI 0.17 to 0.46), and that BEV_RT probably increases time to disease progression compared with RT40 alone (HR 0.46, 95% CI 0.27 to 0.78). Evidence for other treatment comparisons was of low- or very low-certainty. Severe adverse events Moderate-certainty evidence suggests that TMZ probably increases the risk of grade 3+ thromboembolic events compared with RT60 (risk ratio (RR) 2.74, 95% CI 1.26 to 5.94; participants = 373; studies = 1) and also the risk of grade 3+ neutropenia, lymphopenia, and thrombocytopenia. Moderate-certainty evidence also suggests that CRT probably increases the risk of grade 3+ neutropenia, leucopenia and thrombocytopenia compared with hypofractionated RT alone. Adding BEV to CRT probably increases the risk of thromboembolism (RR 16.63, 95% CI 1.00 to 275.42; moderate-certainty evidence). Economic evidence There is a paucity of economic evidence regarding the management of newly diagnosed glioblastoma in the elderly. Only one economic evaluation on two short course radiotherapy regimen (25 Gy versus 40 Gy) was identified and its findings were considered unreliable. AUTHORS' CONCLUSIONS For elderly people with glioblastoma who are self-caring, evidence suggests that CRT prolongs survival compared with RT and may prolong overall survival compared with TMZ alone. For those undergoing RT or TMZ therapy, there is probably little difference in QoL overall. Systemic anti-cancer treatments TMZ and BEV carry a higher risk of severe haematological and thromboembolic events and CRT is probably associated with a higher risk of these events. Current evidence provides little justification for using BEV in elderly patients outside a clinical trial setting. Whilst the novel TTF device appears promising, evidence on QoL and tolerability is needed in an elderly population. QoL and economic assessments of CRT versus TMZ and RT are needed. More high-quality economic evaluations are needed, in which a broader scope of costs (both direct and indirect) and outcomes should be included.
Collapse
Affiliation(s)
- Catherine Hanna
- University of GlasgowDepartment of OncologyBeatson West of Scotland Cancer CentreGreat Western RoadGlasgowScotlandUKG4 9DL
| | - Theresa A Lawrie
- The Evidence‐Based Medicine Consultancy Ltd3rd Floor Northgate HouseUpper Borough WallsBathUKBA1 1RG
| | - Ewelina Rogozińska
- The Evidence‐Based Medicine Consultancy Ltd3rd Floor Northgate HouseUpper Borough WallsBathUKBA1 1RG
| | - Ashleigh Kernohan
- Newcastle UniversityInstitute of Health & SocietyBaddiley‐Clark Building, Richardson RoadNewcastle upon TyneUKNE2 4AA
| | - Sarah Jefferies
- Addenbrooke's HospitalDepartment of OncologyHills RoadCambridgeUKCB2 0QQ
| | - Helen Bulbeck
- brainstrustDirector of Services4 Yvery CourtCastle RoadCowesIsle of WightUKPO31 7QG
| | - Usama M Ali
- University of OxfordNuffield Department of Population HealthRoosevelt DriveOld Road CampusOxfordOxfordshireUKOX3 7LF
| | - Tomos Robinson
- Newcastle UniversityInstitute of Health & SocietyBaddiley‐Clark Building, Richardson RoadNewcastle upon TyneUKNE2 4AA
| | - Robin Grant
- Western General HospitalEdinburgh Centre for Neuro‐Oncology (ECNO)Crewe RoadEdinburghScotlandUKEH4 2XU
| | | |
Collapse
|
14
|
Ramezani S, Vousooghi N, Joghataei MT, Chabok SY. The Role of Kinase Signaling in Resistance to Bevacizumab Therapy for Glioblastoma Multiforme. Cancer Biother Radiopharm 2020; 34:345-354. [PMID: 31411929 DOI: 10.1089/cbr.2018.2651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant primary brain tumor and is characterized by vascular hyperplasia, necrosis, and high cell proliferation. Despite current standard therapies, including surgical resection and chemoradiotherapy, GBM patients survive for only about 15 months after diagnosis. Recently, the U.S. Food and Drug Administration (FDA) has approved an antiangiogenesis medication for recurrent GBM-bevacizumab-which has improved progression-free survival in GBM patients. Although bevacizumab has resulted in significant early clinical benefit, it inescapably predisposes tumor to relapse that can be represented as an infiltrative phenotype. Fundamentally, bevacizumab antagonizes the vascular endothelial growth factor A (VEGFA), which is consistently released on both endothelial cells (ECs) and GBM cells. Actually, VEGFA inhibition on the ECs leads to the suppression of vascular progression, permeability, and the vasogenic edema. However, the consequence of the VEGFA pathway blockage on the GBM cells remains controversial. Nevertheless, a piece of evidence supports the relationship between bevacizumab application and compensatory activation of kinase signaling within GBM cells, leading to a tumor cell invasion known as the main mechanism of bevacizumab-induced tumor resistance. A complete understanding of kinase responses associated with tumor invasion in bevacizumab-resistant GBMs offers new therapeutic opportunities. Thus, this study aimed at presenting a brief overview of preclinical and clinical data of the tumor invasion and resistance induced by bevacizumab administration in GBMs, with a focus on the kinase responses during treatment. The novel therapeutic strategies to overcome this resistance by targeting protein kinases have also been summarized.
Collapse
Affiliation(s)
- Sara Ramezani
- 1Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,2Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasim Vousooghi
- 3Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,4Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran.,5Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- 6Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,7Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Yousefzadeh Chabok
- 1Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,2Guilan Road Trauma Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
15
|
Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis 2019; 23:55-74. [PMID: 31865479 DOI: 10.1007/s10456-019-09698-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Vessel co-option is a non-angiogenic mechanism of tumour vascularisation in which cancer cells utilise pre-existing blood vessels instead of inducing new blood vessel formation. Vessel co-option has been observed across a range of different tumour types, in both primary cancers and metastatic disease. Importantly, vessel co-option is now implicated as a major mechanism that mediates resistance to conventional anti-angiogenic drugs and this may help to explain the limited efficacy of this therapeutic approach in certain clinical settings. This includes the use of anti-angiogenic drugs to treat advanced-stage/metastatic disease, treatment in the adjuvant setting and the treatment of primary disease. In this article, we review the available evidence linking vessel co-option with resistance to anti-angiogenic therapy in numerous tumour types, including breast, colorectal, lung and pancreatic cancer, glioblastoma, melanoma, hepatocellular carcinoma, and renal cell carcinoma. The finding that vessel co-option is a significant mechanism of resistance to anti-angiogenic therapy may have important implications for the future of anti-cancer therapy, including (a) predicting response to anti-angiogenic drugs, (b) the need to develop therapies that target both angiogenesis and vessel co-option in tumours, and (c) predicting the response to other therapeutic modalities, including immunotherapy.
Collapse
|
16
|
Zeiner PS, Kinzig M, Divé I, Maurer GD, Filipski K, Harter PN, Senft C, Bähr O, Hattingen E, Steinbach JP, Sörgel F, Voss M, Steidl E, Ronellenfitsch MW. Regorafenib CSF Penetration, Efficacy, and MRI Patterns in Recurrent Malignant Glioma Patients. J Clin Med 2019; 8:jcm8122031. [PMID: 31766326 PMCID: PMC6947028 DOI: 10.3390/jcm8122031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/28/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The phase 2 Regorafenib in Relapsed Glioblastoma (REGOMA) trial indicated a survival benefit for patients with first recurrence of a glioblastoma when treated with the multikinase inhibitor regorafenib (REG) instead of lomustine. The aim of this retrospective study was to investigate REG penetration to cerebrospinal fluid (CSF), treatment efficacy, and effects on magnetic resonance imaging (MRI) in patients with recurrent high-grade gliomas. (2) Methods: Patients were characterized by histology, adverse events, steroid treatment, overall survival (OS), and MRI growth pattern. REG and its two active metabolites were quantified by liquid chromatography/tandem mass spectrometry in patients’ serum and CSF. (3) Results: 21 patients mainly with IDH-wildtype glioblastomas who had been treated with REG were retrospectively identified. Thirteen CFS samples collected from 3 patients of the cohort were available for pharmacokinetic testing. CSF levels of REG and its metabolites were significantly lower than in serum. Follow-up MRI was available in 19 patients and showed progressive disease (PD) in all but 2 patients. Two distinct MRI patterns were identified: 7 patients showed classic PD with progression of contrast enhancing lesions, whereas 11 patients showed a T2-dominant MRI pattern characterized by a marked reduction of contrast enhancement. Median OS was significantly better in patients with a T2-dominant growth pattern (10 vs. 27 weeks respectively, p = 0.003). Diffusion restrictions were observed in 13 patients. (4) Conclusion: REG and its metabolites were detectable in CSF. A distinct MRI pattern that might be associated with an improved OS was observed in half of the patient cohort. Treatment response in the total cohort was poor.
Collapse
Affiliation(s)
- Pia S. Zeiner
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Martina Kinzig
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
| | - Iris Divé
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Gabriele D. Maurer
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
| | - Katharina Filipski
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Patrick N. Harter
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Christian Senft
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany;
| | - Oliver Bähr
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- Department of Neurology, Klinikum Aschaffenburg-Alzenau, 63739 Aschaffenburg, Germany
| | - Elke Hattingen
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Joachim P. Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Fritz Sörgel
- IBMP—Institute for Biomedical and Pharmaceutical Research, 90562 Nürnberg-Heroldsberg, Germany; (M.K.); (F.S.)
- Institute of Pharmacology, University Duisburg-Essen, 45141 Essen, Germany
| | - Martin Voss
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Eike Steidl
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Department of Neuroradiology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany
| | - Michael W. Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (P.S.Z.); (I.D.); (G.D.M.); (O.B.); (J.P.S.); (M.V.)
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (K.F.); (P.N.H.); (E.H.); (E.S.)
- German Cancer Consortium (DKTK), 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-87711; Fax: +49-69-6301-87713
| |
Collapse
|
17
|
Roetzer T, Leskovar K, Peter N, Furtner J, Muck M, Augustin M, Lichtenegger A, Nowosielski M, Hainfellner JA, Baumann B, Woehrer A. Evaluating cellularity and structural connectivity on whole brain slides using a custom-made digital pathology pipeline. J Neurosci Methods 2019; 311:215-221. [PMID: 30365968 PMCID: PMC6269083 DOI: 10.1016/j.jneumeth.2018.10.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 11/29/2022]
Abstract
AIMS In brain research, the histopathological examination of coronar whole-brain slides provides important insights into spatial disease characteristics. Regarding brain tumor research, this enables visualization of tumor heterogeneity, infiltration patterns and the relationship with the surrounding brain parenchyma. The precise correlation between radiological imaging and post-mortem brains is of special interest. NEW METHOD We developed a wide-field slide scanner, comprising a microscope, a high-precision remotely controllable x-y-stage, a camera and a computer workstation, for automatically scanning uncommonly large formats. We analyzed whole brain slides of three patients and constructed cellularity heatmaps and fiber tract maps using a custom-made image processing pipeline. RESULTS The obtained cellularity heatmaps allow for distinguishing compact tumor (5714 ± 1786 cells/mm², mean ± standard deviation) from white matter (3581 ± 828 cells/mm²) and grey matter (2473 ± 716 cells/mm²). Compared to magnetic resonance imaging, the proposed histopathological work-up (i) reveals a larger zone of tumor infiltration around the compact tumor areas and (ii) shows how pre-existing tracts are displaced by the tumor bulk. Moreover, we highlight differences in the histological tumor growth pattern of two different radiological progression subtypes. COMPARISON WITH EXISTING METHOD(S) Compared to existing (commercial) solutions, our slide scanning solution is adaptable and cost-efficient. Moreover, we showcase potential clinical applications by mapping whole brain histology to magnetic resonance imaging. CONCLUSIONS We herein provide instructions on how to (i) construct a custom-built slide scanner capable of scanning arbitrary slide formats, (ii) automatically evaluate the cell density and (iii) perform fiber tracking on whole brain slides.
Collapse
Affiliation(s)
- Thomas Roetzer
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Konrad Leskovar
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Nadine Peter
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Martina Muck
- Institute of Neurology, Medical University of Vienna, Vienna, Austria; Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Marco Augustin
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Antonia Lichtenegger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| | - Adelheid Woehrer
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Falchetti ML, D'Alessandris QG, Pacioni S, Buccarelli M, Morgante L, Giannetti S, Lulli V, Martini M, Larocca LM, Vakana E, Stancato L, Ricci-Vitiani L, Pallini R. Glioblastoma endothelium drives bevacizumab-induced infiltrative growth via modulation of PLXDC1. Int J Cancer 2018; 144:1331-1344. [PMID: 30414187 PMCID: PMC6590500 DOI: 10.1002/ijc.31983] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/10/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022]
Abstract
Bevacizumab, a VEGF‐targeting monoclonal antibody, may trigger an infiltrative growth pattern in glioblastoma. We investigated this pattern using both a human specimen and rat models. In the human specimen, a substantial fraction of infiltrating tumor cells were located along perivascular spaces in close relationship with endothelial cells. Brain xenografts of U87MG cells treated with bevacizumab were smaller than controls (p = 0.0055; Student t‐test), however, bands of tumor cells spread through the brain farther than controls (p < 0.001; Student t‐test). Infiltrating tumor Cells exhibited tropism for vascular structures and propensity to form tubules and niches with endothelial cells. Molecularly, bevacizumab triggered an epithelial to mesenchymal transition with over‐expression of the receptor Plexin Domain Containing 1 (PLXDC1). These results were validated using brain xenografts of patient‐derived glioma stem‐like cells. Enforced expression of PLXDC1 in U87MG cells promoted brain infiltration along perivascular spaces. Importantly, PLXDC1 inhibition prevented perivascular infiltration and significantly increased the survival of bevacizumab‐treated rats. Our study indicates that bevacizumab‐induced brain infiltration is driven by vascular endothelium and depends on PLXDC1 activation of tumor cells. What's new? Bevacizumab, a VEGF‐targeting monoclonal antibody, has been observed to trigger an infiltrative growth pattern in glioblastoma as an escape mechanism. The mechanisms underlying this gliomatosis‐like growth pattern, however, remain unclear. Here, the authors found that the infiltrative growth pattern occurs mostly along perivascular spaces and relies on the over‐expression of PLXDC1 by tumor cells and on the restoration of the endothelial component of blood brain barrier. Altogether, the data show that the brain infiltration induced by bevacizumab is mainly driven by the vascular endothelium. Importantly, inhibition of PLXDC1 prevents bevacizumab‐induced infiltrative growth, resulting in significant increase of survival.
Collapse
Affiliation(s)
| | - Quintino Giorgio D'Alessandris
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simone Pacioni
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Liliana Morgante
- Institute of Anatomy and Cell Biology, Universitá Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Universitá Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Lulli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Maurizio Martini
- Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigi Maria Larocca
- Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eliza Vakana
- Discovery Research, Eli Lilly and Company, Indianapolis, IN
| | - Louis Stancato
- Discovery Research, Eli Lilly and Company, Indianapolis, IN
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH, Furtner J, Sheffield NC, Datlinger P, Peter N, Nowosielski M, Augustin M, Mischkulnig M, Ströbel T, Alpar D, Ergüner B, Senekowitsch M, Moser P, Freyschlag CF, Kerschbaumer J, Thomé C, Grams AE, Stockhammer G, Kitzwoegerer M, Oberndorfer S, Marhold F, Weis S, Trenkler J, Buchroithner J, Pichler J, Haybaeck J, Krassnig S, Mahdy Ali K, von Campe G, Payer F, Sherif C, Preiser J, Hauser T, Winkler PA, Kleindienst W, Würtz F, Brandner-Kokalj T, Stultschnig M, Schweiger S, Dieckmann K, Preusser M, Langs G, Baumann B, Knosp E, Widhalm G, Marosi C, Hainfellner JA, Woehrer A, Bock C. The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med 2018; 24:1611-1624. [PMID: 30150718 PMCID: PMC6181207 DOI: 10.1038/s41591-018-0156-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Glioblastoma is characterized by widespread genetic and transcriptional heterogeneity, yet little is known about the role of the epigenome in glioblastoma disease progression. Here, we present genome-scale maps of DNA methylation in matched primary and recurring glioblastoma tumors, using data from a highly annotated clinical cohort that was selected through a national patient registry. We demonstrate the feasibility of DNA methylation mapping in a large set of routinely collected FFPE samples, and we validate bisulfite sequencing as a multipurpose assay that allowed us to infer a range of different genetic, epigenetic, and transcriptional characteristics of the profiled tumor samples. On the basis of these data, we identified subtle differences between primary and recurring tumors, links between DNA methylation and the tumor microenvironment, and an association of epigenetic tumor heterogeneity with patient survival. In summary, this study establishes an open resource for dissecting DNA methylation heterogeneity in a genetically diverse and heterogeneous cancer, and it demonstrates the feasibility of integrating epigenomics, radiology, and digital pathology for a national cohort, thereby leveraging existing samples and data collected as part of routine clinical practice.
Collapse
Affiliation(s)
- Johanna Klughammer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Barbara Kiesel
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
| | - Thomas Roetzer
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Amelie Nemc
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Karl-Heinz Nenning
- Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research Lab, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Division of Neuroradiology and Musculoskeletal Radiology, Medical University of Vienna, Vienna, Austria
| | - Nathan C Sheffield
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Paul Datlinger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nadine Peter
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- University Medical Center, Neurology, German Cancer Research Center, Heidelberg, Germany
| | - Marco Augustin
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Mario Mischkulnig
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
| | - Thomas Ströbel
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Donat Alpar
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bekir Ergüner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Martin Senekowitsch
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Patrizia Moser
- Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Astrid E Grams
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günther Stockhammer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Melitta Kitzwoegerer
- Department of Pathology, University Hospital of St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Stefan Oberndorfer
- Department of Neurology, University Hospital of St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Franz Marhold
- Department of Neurosurgery, University Hospital of St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Serge Weis
- Department of Neuropathology, Neuromed Campus Wagner-Jauregg, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Johannes Trenkler
- Department of Neuroradiology, Neuromed Campus Wagner-Jauregg, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Johanna Buchroithner
- Department of Neurosurgery, Neuromed Campus Wagner-Jauregg, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Josef Pichler
- Department of Internal Medicine, Neuromed Campus Wagner-Jauregg, Kepler University Hospital, Johannes Kepler University of Linz, Linz, Austria
| | - Johannes Haybaeck
- Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic & Research Center for Molecular BioMedicine, Department of Neuropathology, Institute of Pathology, Medical University of Graz, Graz, Austria
- Department of Pathology, Medical Faculty, Otto-von-Guericke University of Magdeburg, Magdeburg, Germany
| | - Stefanie Krassnig
- Diagnostic & Research Center for Molecular BioMedicine, Department of Neuropathology, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Kariem Mahdy Ali
- Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Gord von Campe
- Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Franz Payer
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Camillo Sherif
- Department of Neurosurgery, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Julius Preiser
- Department of Pathology, Krankenanstalt Rudolfstiftung, Vienna, Austria
| | - Thomas Hauser
- Department of Neurosurgery, Christian-Doppler-Klinik, Paracelsus Private Medical University, Salzburg, Austria
| | - Peter A Winkler
- Department of Neurosurgery, Christian-Doppler-Klinik, Paracelsus Private Medical University, Salzburg, Austria
| | - Waltraud Kleindienst
- Department of Neurology, Christian-Doppler-Klinik, Paracelsus Private Medical University, Salzburg, Austria
| | - Franz Würtz
- Institute of Pathology, State Hospital Klagenfurt, Klagenfurt, Austria
| | | | | | - Stefan Schweiger
- Department of Neurosurgery, General Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Karin Dieckmann
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Department of Radiotherapy, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Georg Langs
- Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research Lab, Medical University of Vienna, Vienna, Austria
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Engelbert Knosp
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
| | - Christine Marosi
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Johannes A Hainfellner
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Adelheid Woehrer
- Comprehensive Cancer Center, Central Nervous System Tumor Unit, Medical University of Vienna, Vienna, Austria.
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| |
Collapse
|
20
|
Mikkelsen VE, Stensjøen AL, Granli US, Berntsen EM, Salvesen Ø, Solheim O, Torp SH. Angiogenesis and radiological tumor growth in patients with glioblastoma. BMC Cancer 2018; 18:862. [PMID: 30176826 PMCID: PMC6122710 DOI: 10.1186/s12885-018-4768-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The preoperative growth of human glioblastomas (GBMs) has been shown to vary among patients. In animal studies, angiogenesis has been linked to hypoxia and faster growth of GBM, however, its relation to the growth of human GBMs is sparsely studied. We have therefore aimed to look for associations between radiological speed of growth and microvessel density (MVD) counts of the endothelial markers vWF (Factor VIII related antigen) and CD105 (endoglin). METHODS Preoperative growth was estimated from segmented tumor volumes of two preoperative T1-weighted postcontrast magnetic resonance imaging scans taken ≥14 days apart in patients with newly diagnosed GBMs. A Gompertzian growth curve was computed from the volume data and separated the patients into two groups of either faster or slower tumor growth than expected. MVD counts of the immunohistochemical markers von Willebrand factor (vWF) (a pan-endothelial marker) and CD105 (a marker of proliferating endothelial cells) were assessed for associations with fast-growing tumors using Mann-Whitney U tests and a multivariable binary logistic regression analysis. RESULTS We found that only CD105-MVD was significantly associated with faster growth in a univariable analysis (p = 0.049). However, CD105-MVD was no longer significant when corrected for the presence of thromboses and high cellular density in a multivariable model, where the latter features were significant independent predictors of faster growth with respective odds ratios 4.2 (95% confidence interval, 1.2, 14.3), p = 0.021 and 2.6 (95% confidence interval, 1.0, 6.5), p = 0.048. CONCLUSIONS MVDs of neither endothelial marker were independently associated with faster growth, suggesting angiogenesis-independent processes contribute to faster glioblastoma growth.
Collapse
Affiliation(s)
- Vilde Elisabeth Mikkelsen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.
| | - Anne Line Stensjøen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway
| | - Unn Sophie Granli
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.,Cellular and Molecular Imaging Core Facility (CMIC), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs University Hospital, Trondheim, Norway
| | - Øyvind Salvesen
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Ole Solheim
- Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway.,National Advisory Unit for Ultrasound and Image Guided Therapy, St. Olavs University Hospital, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre Helge Torp
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.,Department of Pathology, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
21
|
Galldiks N, Dunkl V, Ceccon G, Tscherpel C, Stoffels G, Law I, Henriksen OM, Muhic A, Poulsen HS, Steger J, Bauer EK, Lohmann P, Schmidt M, Shah NJ, Fink GR, Langen KJ. Early treatment response evaluation using FET PET compared to MRI in glioblastoma patients at first progression treated with bevacizumab plus lomustine. Eur J Nucl Med Mol Imaging 2018; 45:2377-2386. [PMID: 29982845 DOI: 10.1007/s00259-018-4082-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND The goal of this prospective study was to compare the value of both conventional MRI and O-(2-18F-fluoroethyl)-L-tyrosine (FET) PET for response evaluation in glioblastoma patients treated with bevacizumab plus lomustine (BEV/LOM) at first progression. METHODS After chemoradiation with concomitant and adjuvant temozolomide, 21 IDH wild-type glioblastoma patients at first progression (age range, 33-75 years; MGMT promoter unmethylated, 81%) were treated with BEV/LOM. Contrast-enhanced MRI and FET-PET scans were performed at baseline and after 8-10 weeks. We obtained FET metabolic tumor volumes (MTV) and tumor/brain ratios. Threshold values of FET-PET parameters for treatment response were established by ROC analyses using the post-progression overall survival (OS) ≤/>9 months as the reference. MRI response assessment was based on RANO criteria. The predictive ability of FET-PET thresholds and MRI changes on early response assessment was evaluated subsequently concerning OS using uni- and multivariate survival estimates. RESULTS Early treatment response as assessed by RANO criteria was not predictive for an OS>9 months (P = 0.203), whereas relative reductions of all FET-PET parameters significantly predicted an OS>9 months (P < 0.05). The absolute MTV at follow-up enabled the most significant OS prediction (sensitivity, 85%; specificity, 88%; P = 0.001). Patients with an absolute MTV below 5 ml at follow-up survived significantly longer (12 vs. 6 months, P < 0.001), whereas early responders defined by RANO criteria lived only insignificantly longer (9 vs. 6 months; P = 0.072). The absolute MTV at follow-up remained significant in the multivariate survival analysis (P = 0.006). CONCLUSIONS FET-PET appears to be useful for identifying responders to BEV/LOM early after treatment initiation.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany. .,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany. .,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany.
| | - Veronika Dunkl
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Garry Ceccon
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Caroline Tscherpel
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Otto M Henriksen
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Aida Muhic
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hans S Poulsen
- Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jan Steger
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Elena K Bauer
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Matthias Schmidt
- Dept. of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital Cologne, Josef-Stelzmann St. 9, 50937, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Forschungszentrum Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
22
|
Kong Z, Yan C, Zhu R, Wang J, Wang Y, Wang Y, Wang R, Feng F, Ma W. Imaging biomarkers guided anti-angiogenic therapy for malignant gliomas. NEUROIMAGE-CLINICAL 2018; 20:51-60. [PMID: 30069427 PMCID: PMC6067083 DOI: 10.1016/j.nicl.2018.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022]
Abstract
Antiangiogenic therapy is a universal approach to the treatment of malignant gliomas but fails to prolong the overall survival of newly diagnosed or recurrent glioblastoma patients. Imaging biomarkers are quantitative imaging parameters capable of objectively describing biological processes, pathological changes and treatment responses in some situations and have been utilized for outcome predictions of malignant gliomas in anti-angiogenic therapy. Advanced magnetic resonance imaging techniques (including perfusion-weighted imaging and diffusion-weighted imaging), positron emission computed tomography and magnetic resonance spectroscopy are imaging techniques that can be used to acquire imaging biomarkers, including the relative cerebral blood volume (rCBV), Ktrans, and the apparent diffusion coefficient (ADC). Imaging indicators for a better prognosis when treating malignant gliomas with antiangiogenic therapy include the following: a lower pre- or post-treatment rCBV, less change in rCBV during treatment, a lower pre-treatment Ktrans, a higher vascular normalization index during treatment, less change in arterio-venous overlap during treatment, lower pre-treatment ADC values for the lower peak, smaller ADC volume changes during treatment, and metabolic changes in glucose and phenylalanine. The investigation and utilization of these imaging markers may confront challenges, but may also promote further development of anti-angiogenic therapy. Despite considerable evidence, future prospective studies are critically needed to consolidate the current data and identify novel biomarkers. Anti-angiogenic therapy only benefits specific populations of glioma patients. Advanced imaging techniques can produce quantitative imaging biomarkers. Physiological and metabolic parameter can predict outcome for anti-angiogenic therapy. Larger prospective studies are needed to provide further evidence.
Collapse
Key Words
- 18F-FDOPA, 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine
- 18F-FLT, [18F]-fluoro-3-deoxy-3-L-fluorothymidine
- ADC, apparent diffusion coefficient
- AVOL, arterio-venous overlap
- Anti-angiogenic
- BBB, blood brain barrier
- Biomarkers
- CBF, cerebral blood flow
- CBV, cerebral blood volume
- CNS, central nervous system
- CT, computed tomography
- D-2HG, D-2-hydroxypentanedioic acid
- DCE-MRI, dynamic contrast-enhanced magnetic resonance imaging
- DSC-MRI, dynamic susceptibility contrast magnetic resonance imaging
- DWI, diffusion-weighted imaging
- FDG, fluorodeoxyglucose
- FLAIR, fluid-attenuated inversion recovery
- FSE pcASL, fast spin echo pseudocontinuous artery spin labeling
- GBM, glioblastoma
- Glioma
- Imaging
- Ktrans, volume transfer constant between blood plasma and extravascular extracellular space
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- OS, overall survival
- PET, positron emission computed tomography
- PFS, progression-free survival
- PWI, perfusion-weighted imaging
- RANO, Response Assessment in Neuro-Oncology
- ROI, region of interest
- RSI, restriction spectrum imaging
- SUV, standardized uptake value
- TMZ, temozolomide
- Therapy
- VAI, vessel architectural imaging
- VEGF-A, vascular endothelial growth factor A
- VNI, vascular normalization index.
- fDMs, functional diffusion maps
- nGBM, newly diagnosed glioblastoma
- rCBF, relative cerebral blood flow
- rCBV, relative cerebral blood volume
- rGBM, recurrent glioblastoma
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Chengrui Yan
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China; Department of Neurosurgery, Peking University International Hospital, Peking University, Beijing, China
| | - Ruizhe Zhu
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Jiaru Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Yaning Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| | - Renzhi Wang
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| | - Feng Feng
- Department of Radiology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China..
| | - Wenbin Ma
- Department of Neurosurgery, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
23
|
Nandu H, Wen PY, Huang RY. Imaging in neuro-oncology. Ther Adv Neurol Disord 2018; 11:1756286418759865. [PMID: 29511385 PMCID: PMC5833173 DOI: 10.1177/1756286418759865] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/18/2018] [Indexed: 12/11/2022] Open
Abstract
Imaging plays several key roles in managing brain tumors, including diagnosis, prognosis, and treatment response assessment. Ongoing challenges remain as new therapies emerge and there are urgent needs to find accurate and clinically feasible methods to noninvasively evaluate brain tumors before and after treatment. This review aims to provide an overview of several advanced imaging modalities including magnetic resonance imaging and positron emission tomography (PET), including advances in new PET agents, and summarize several key areas of their applications, including improving the accuracy of diagnosis and addressing the challenging clinical problems such as evaluation of pseudoprogression and anti-angiogenic therapy, and rising challenges of imaging with immunotherapy.
Collapse
Affiliation(s)
- Hari Nandu
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02445, USA
| |
Collapse
|