1
|
Bakas S, Vollmuth P, Galldiks N, Booth TC, Aerts HJWL, Bi WL, Wiestler B, Tiwari P, Pati S, Baid U, Calabrese E, Lohmann P, Nowosielski M, Jain R, Colen R, Ismail M, Rasool G, Lupo JM, Akbari H, Tonn JC, Macdonald D, Vogelbaum M, Chang SM, Davatzikos C, Villanueva-Meyer JE, Huang RY. Artificial Intelligence for Response Assessment in Neuro Oncology (AI-RANO), part 2: recommendations for standardisation, validation, and good clinical practice. Lancet Oncol 2024; 25:e589-e601. [PMID: 39481415 DOI: 10.1016/s1470-2045(24)00315-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 11/02/2024]
Abstract
Technological advancements have enabled the extended investigation, development, and application of computational approaches in various domains, including health care. A burgeoning number of diagnostic, predictive, prognostic, and monitoring biomarkers are continuously being explored to improve clinical decision making in neuro-oncology. These advancements describe the increasing incorporation of artificial intelligence (AI) algorithms, including the use of radiomics. However, the broad applicability and clinical translation of AI are restricted by concerns about generalisability, reproducibility, scalability, and validation. This Policy Review intends to serve as the leading resource of recommendations for the standardisation and good clinical practice of AI approaches in health care, particularly in neuro-oncology. To this end, we investigate the repeatability, reproducibility, and stability of AI in response assessment in neuro-oncology in studies on factors affecting such computational approaches, and in publicly available open-source data and computational software tools facilitating these goals. The pathway for standardisation and validation of these approaches is discussed with the view of trustworthy AI enabling the next generation of clinical trials. We conclude with an outlook on the future of AI-enabled neuro-oncology.
Collapse
Affiliation(s)
- Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Neurological Surgery, School of Medicine, Indiana University, Indianapolis, IN, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA; Department of Computer Science, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, IN, USA.
| | - Philipp Vollmuth
- Division for Computational Radiology and Clinical AI, Clinic for Neuroradiology, University Hospital Bonn, Bonn, Germany; Faculty of Medicine, University of Bonn, Bonn, Germany; Division for Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Hugo J W L Aerts
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Artificial Intelligence in Medicine Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA; Radiology and Nuclear Medicine, Maastricht University, Maastricht, Netherlands
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedikt Wiestler
- Department of Neuroradiology, University Hospital, Technical University of Munich, Munich, Germany
| | - Pallavi Tiwari
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Sarthak Pati
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA
| | - Ujjwal Baid
- Department of Pathology & Laboratory Medicine, Division of Computational Pathology, Indiana University, Indianopolis, IN, USA; Department of Radiology & Imaging Sciences, School of Medicine, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianopolis, IN, USA
| | - Evan Calabrese
- Department of Radiology, School of Medicine, Duke University, Durham, NC, USA
| | - Philipp Lohmann
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Martha Nowosielski
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Rajan Jain
- Department of Radiology and Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Rivka Colen
- Department of Radiology, Neuroradiology Division, Center for Artificial Intelligence Innovation in Medical Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marwa Ismail
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Ghulam Rasool
- Department of Machine Learning, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Joerg C Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany
| | | | - Michael Vogelbaum
- Department of Neuro-Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; Department of Neurosurgery, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA; H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Susan M Chang
- Department of Neurological Surgery, Division of Neuro-Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Artificial Intelligence for Integrated Diagnostics and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Wilcox JA, Chukwueke UN, Ahn MJ, Aizer AA, Bale TA, Brandsma D, Brastianos PK, Chang S, Daras M, Forsyth P, Garzia L, Glantz M, Oliva ICG, Kumthekar P, Le Rhun E, Nagpal S, O'Brien B, Pentsova E, Lee EQ, Remsik J, Rudà R, Smalley I, Taylor MD, Weller M, Wefel J, Yang JT, Young RJ, Wen PY, Boire AA. Leptomeningeal metastases from solid tumors: A Society for Neuro-Oncology and American Society of Clinical Oncology consensus review on clinical management and future directions. Neuro Oncol 2024; 26:1781-1804. [PMID: 38902944 PMCID: PMC11449070 DOI: 10.1093/neuonc/noae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Indexed: 06/22/2024] Open
Abstract
Leptomeningeal metastases (LM) are increasingly becoming recognized as a treatable, yet generally incurable, complication of advanced cancer. As modern cancer therapeutics have prolonged the lives of patients with metastatic cancer, specifically in patients with parenchymal brain metastases, treatment options, and clinical research protocols for patients with LM from solid tumors have similarly evolved to improve survival within specific populations. Recent expansions in clinical investigation, early diagnosis, and drug development have given rise to new unanswered questions. These include leptomeningeal metastasis biology and preferred animal modeling, epidemiology in the modern cancer population, ensuring validation and accessibility of newer leptomeningeal metastasis diagnostics, best clinical practices with multimodality treatment options, clinical trial design and standardization of response assessments, and avenues worthy of further research. An international group of multi-disciplinary experts in the research and management of LM, supported by the Society for Neuro-Oncology and American Society of Clinical Oncology, were assembled to reach a consensus opinion on these pressing topics and provide a roadmap for future directions. Our hope is that these recommendations will accelerate collaboration and progress in the field of LM and serve as a platform for further discussion and patient advocacy.
Collapse
Affiliation(s)
- Jessica A Wilcox
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ugonma N Chukwueke
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ayal A Aizer
- Department of Radiation Oncology, Brigham and Women's Hospital / Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Tejus A Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dieta Brandsma
- Department of Neuro-Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Priscilla K Brastianos
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of San Francisco California, San Francisco, California, USA
| | - Mariza Daras
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Peter Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Livia Garzia
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Glantz
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Priya Kumthekar
- The Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Emilie Le Rhun
- Departments of Neurology and Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Seema Nagpal
- Division of Neuro-Oncology, Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Barbara O'Brien
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elena Pentsova
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Eudocia Quant Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jan Remsik
- Laboratory for Immunology of Metastatic Ecosystems, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University and City of Health and Science Hospital, Turin, Italy
- Department of Neurology, Castelfranco Veneto and Treviso Hospitals, Castelfranco Veneto, Italy
| | - Inna Smalley
- Department of Tumor Biology, The Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Michael D Taylor
- Division of Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, USA
- Neuro-oncology Research Program, Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jeffrey Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonathan T Yang
- Department of Radiation Oncology, Department of Radiation Oncology, New York University School of Medicine, New York, New York, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Adrienne A Boire
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Neurology, Brain Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
3
|
Zamanian M, Abedi I, Danazadeh F, Amouheidari A, Shahreza BO. Post-chemo-radiotherapy response and pseudo-progression evaluation on glioma cell types by multi-parametric magnetic resonance imaging: a prospective study. BMC Med Imaging 2023; 23:176. [PMID: 37932656 PMCID: PMC10626695 DOI: 10.1186/s12880-023-01135-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND We focused on Differentiated pseudoprogression (PPN) of progression (PN) and the response to radiotherapy (RT) or chemoradiotherapy (CRT) using diffusion and metabolic imaging. METHODS Seventy-five patients with glioma were included in this prospective study (approved by the Iranian Registry of Clinical Trials (IRCT) (IRCT20230904059352N1) in September 2023). Contrast-enhanced lesion volume (CELV), non-enhanced lesion volume (NELV), necrotic tumor volume (NTV), and quantitative values of apparent diffusion coefficient (ADC) and magnetic resonance spectroscopy (Cho/Cr, Cho/NAA and NAA/Cr) were calculated by a neuroradiologist using a semi-automatic method. All patients were followed at one and six months after CRT. RESULTS The results of the study showed statistically significant changes before and six months after RT-CRT for M-CELV in all glioma types (𝑝 < 0.05). In glioma cell types, the changes in M-ADC, M-Cho/Cr, and Cho/NAA indices for PN were incremental and greater for PPN patients. M-NAA/Cr ratio decreased after six months which was significant only on PN for GBM, and Epn (𝑝 < 0.05). A significant difference was observed between diffusion indices, metabolic ratios, and CELV changes after six months in all types (𝑝 < 0.05). None of the patients were suspected PPN one month after treatment. The DWI/ADC indices had higher sensitivity and specificity (98.25% and 96.57%, respectively). CONCLUSION The results of the present study showed that ADC values and Cho/Cr and Cho/NAA ratios can be used to differentiate between patients with PPN and PN, although ADC is more sensitive and specific.
Collapse
Affiliation(s)
- Maryam Zamanian
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Iraj Abedi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Fatemeh Danazadeh
- Department of Radiology, School of Paramedicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | |
Collapse
|
4
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
5
|
Altinoz MA, Ozpinar A, Hacker E, Ozpinar A. Combining locoregional CAR-T cells, autologous + allogeneic tumor lysate vaccination and levamisole in treatment of glioblastoma. Immunopharmacol Immunotoxicol 2022; 44:797-808. [PMID: 35670420 DOI: 10.1080/08923973.2022.2086136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain malignancy and harbors a microenvironment limiting immune cells activity. CAR-T cells are being tested in the treatment of cancers and there exist reports which demonstrate dramatic regression of multicentric GBMs following intrathecal treatment with CAR-T cells. In this article, a triple approach for immune treatment of GBM is proposed. First, GBM tumor specimens for each patient will be saved and cultured to obtain tumor lysates. Then, levamisole will be applied, which possesses immunostimulating, anti-glycolytic, and anti-angiogenic features. Following priming the immune system, GBM patients will be injected with lysates of their own tumor cells plus lysates from a GBM cell line, U251. After 3 months of this treatment, CAR-T cells (transduced with IL13Rα2-CAR) will be applied via intratumoral approach. As such, genetically-modified and native immunocytes may 'meet' in the vicinity of deeply-invading tumor cells and demonstrate greater efficacy via cell-cell interactions. By this, a self-propagating cyclic process - a cancer-immunity cycle - may be initiated to eradicate cancer cells.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Biochemistry, Acibadem University, Istanbul, Turkey
| | - Alp Ozpinar
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily Hacker
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aysel Ozpinar
- Department of Biochemistry, Acibadem University, Istanbul, Turkey
| |
Collapse
|
6
|
Quality of Life and Cognitive Function Evaluations and Interventions for Patients with Brain Metastases in the Radiation Oncology Clinic. Cancers (Basel) 2022; 14:cancers14174301. [PMID: 36077835 PMCID: PMC9454858 DOI: 10.3390/cancers14174301] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Brain metastases (BMs) are the most common brain malignancy and are projected to increase in incidence over the coming decades. Historically, brain metastasis studies have focused on improving survival outcomes, but recently, the importance of evaluating health-related quality of life (HRQOL) and cognitive function has gained recognition. Although there is a myriad of validated HRQOL and cognitive assessments available in the radiation oncology clinic, there is an urgent need to identify tools tailored to patients with BMs and to adopt a uniform set of tests that measure HRQOL and cognition. This review presents various assessments for measuring HRQOL and cognitive function, current recommendations to improve standardization, and treatments known to preserve HRQOL and cognitive function. Abstract Brain metastases (BMs) account for a disproportionately high percentage of cancer morbidity and mortality. Historically, studies have focused on improving survival outcomes, and recent radiation oncology clinical trials have incorporated HRQOL and cognitive assessments. We are now equipped with a battery of assessments in the radiation oncology clinic, but there is a lack of consensus regarding how to incorporate them in modern clinical practice. Herein, we present validated assessments for BM patients, current recommendations for future clinical studies, and treatment advances that have improved HRQOL and cognitive outcomes for BM patients.
Collapse
|
7
|
Advancements in Oncology with Artificial Intelligence—A Review Article. Cancers (Basel) 2022; 14:cancers14051349. [PMID: 35267657 PMCID: PMC8909088 DOI: 10.3390/cancers14051349] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary With the advancement of artificial intelligence, including machine learning, the field of oncology has seen promising results in cancer detection and classification, epigenetics, drug discovery, and prognostication. In this review, we describe what artificial intelligence is and its function, as well as comprehensively summarize its evolution and role in breast, colorectal, and central nervous system cancers. Understanding the origin and current accomplishments might be essential to improve the quality, accuracy, generalizability, cost-effectiveness, and reliability of artificial intelligence models that can be used in worldwide clinical practice. Students and researchers in the medical field will benefit from a deeper understanding of how to use integrative AI in oncology for innovation and research. Abstract Well-trained machine learning (ML) and artificial intelligence (AI) systems can provide clinicians with therapeutic assistance, potentially increasing efficiency and improving efficacy. ML has demonstrated high accuracy in oncology-related diagnostic imaging, including screening mammography interpretation, colon polyp detection, glioma classification, and grading. By utilizing ML techniques, the manual steps of detecting and segmenting lesions are greatly reduced. ML-based tumor imaging analysis is independent of the experience level of evaluating physicians, and the results are expected to be more standardized and accurate. One of the biggest challenges is its generalizability worldwide. The current detection and screening methods for colon polyps and breast cancer have a vast amount of data, so they are ideal areas for studying the global standardization of artificial intelligence. Central nervous system cancers are rare and have poor prognoses based on current management standards. ML offers the prospect of unraveling undiscovered features from routinely acquired neuroimaging for improving treatment planning, prognostication, monitoring, and response assessment of CNS tumors such as gliomas. By studying AI in such rare cancer types, standard management methods may be improved by augmenting personalized/precision medicine. This review aims to provide clinicians and medical researchers with a basic understanding of how ML works and its role in oncology, especially in breast cancer, colorectal cancer, and primary and metastatic brain cancer. Understanding AI basics, current achievements, and future challenges are crucial in advancing the use of AI in oncology.
Collapse
|
8
|
Rahman R, Ventz S, McDunn J, Louv B, Reyes-Rivera I, Polley MYC, Merchant F, Abrey LE, Allen JE, Aguilar LK, Aguilar-Cordova E, Arons D, Tanner K, Bagley S, Khasraw M, Cloughesy T, Wen PY, Alexander BM, Trippa L. Leveraging external data in the design and analysis of clinical trials in neuro-oncology. Lancet Oncol 2021; 22:e456-e465. [PMID: 34592195 PMCID: PMC8893120 DOI: 10.1016/s1470-2045(21)00488-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 01/20/2023]
Abstract
Integration of external control data, with patient-level information, in clinical trials has the potential to accelerate the development of new treatments in neuro-oncology by contextualising single-arm studies and improving decision making (eg, early stopping decisions). Based on a series of presentations at the 2020 Clinical Trials Think Tank hosted by the Society of Neuro-Oncology, we provide an overview on the use of external control data representative of the standard of care in the design and analysis of clinical trials. High-quality patient-level records, rigorous methods, and validation analyses are necessary to effectively leverage external data. We review study designs, statistical methods, risks, and potential distortions in using external data from completed trials and real-world data, as well as data sources, data sharing models, ongoing work, and applications in glioblastoma.
Collapse
Affiliation(s)
- Rifaquat Rahman
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA.
| | - Steffen Ventz
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Jon McDunn
- Project Data Sphere, Morrisville, NC, USA
| | - Bill Louv
- Project Data Sphere, Morrisville, NC, USA
| | | | - Mei-Yin C Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | - David Arons
- National Brain Tumor Society, Newton, MA, USA
| | - Kirk Tanner
- National Brain Tumor Society, Newton, MA, USA
| | - Stephen Bagley
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mustafa Khasraw
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA; Foundation Medicine, Cambridge, MA, USA
| | - Lorenzo Trippa
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T H Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
9
|
Toward the Next Generation of High-Grade Glioma Clinical Trials in the Era of Precision Medicine. Cancer J 2021; 27:410-415. [PMID: 34570456 DOI: 10.1097/ppo.0000000000000549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT In the era of precision medicine, there is a desire to harness our improved understanding of genomic and molecular underpinnings of gliomas to develop therapies that can be tailored to individual patients and tumors. With the rapid development of novel therapies, there has been a growing need to develop smart clinical trials that are designed to efficiently test promising agents, identify therapies likely to benefit patients, and discard ineffective therapies. We review clinical trial design in gliomas and developments designed to address the unique challenges of precision medicine. To provide an overview of this topic, we examine considerations for endpoints and response assessment, biomarkers, and novel clinical trial designs such as adaptive platform trials in the testing of new therapies for glioma patients.
Collapse
|
10
|
Hughes KL, O'Neal CM, Andrews BJ, Westrup AM, Battiste JD, Glenn CA. A systematic review of the utility of amino acid PET in assessing treatment response to bevacizumab in recurrent high-grade glioma. Neurooncol Adv 2021; 3:vdab003. [PMID: 34409294 PMCID: PMC8369430 DOI: 10.1093/noajnl/vdab003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background. Currently, bevacizumab (BEV), an antiangiogenic agent, is used as an adjunctive therapy to re-irradiation and surgery in patients with recurrent high-grade gliomas (rHGG). BEV has shown to decrease enhancement on MRI, but it is often unclear if these changes are due to tumor response to BEV or treatment-induced changes in the blood brain barrier. Preliminary studies show that amino acid PET can aid in distinguishing these changes on MRI. Methods. The authors performed a systematic review of PubMed and Embase through July 2020 with the search terms ‘bevacizumab’ or ‘Avastin’ and ‘recurrent glioma’ and ‘PET,’ yielding 38 papers, with 14 meeting inclusion criteria. Results. Thirteen out of fourteen studies included in this review used static PET and three studies used dynamic PET to evaluate the use of BEV in rHGG. Six studies used the amino acid tracer [18F]FET, four studies used [11C]MET, and four studies used [18F]FDOPA. Conclusion. [18F]FET, [11C]MET, and [18F]FDOPA PET in combination with MRI have shown promising results for improving accuracy in diagnosing tumor recurrence, detecting early treatment failure, and distinguishing between tumor progression and treatment-induced changes in patients with rHGG treated with BEV.
Collapse
Affiliation(s)
- Kendall L Hughes
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Christen M O'Neal
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bethany J Andrews
- Department of Neurosurgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Alison M Westrup
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - James D Battiste
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Chad A Glenn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
11
|
Abstract
Clinical trials play a critical role in discovering new treatments, but the path to regulatory approval can be cumbersome and time consuming. Efforts to increase the efficiency and interpretability of clinical trials within the neuro-oncology community have focused on standardization of response assessment, development of consensus guidelines for clinical trial conduct, decentralization of clinical trials, removal of barriers to clinical trial accrual, and re-examination of patient eligibility criteria.
Collapse
Affiliation(s)
- Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Roth P, Pace A, Le Rhun E, Weller M, Ay C, Cohen-Jonathan Moyal E, Coomans M, Giusti R, Jordan K, Nishikawa R, Winkler F, Hong JT, Ruda R, Villà S, Taphoorn MJB, Wick W, Preusser M. Neurological and vascular complications of primary and secondary brain tumours: EANO-ESMO Clinical Practice Guidelines for prophylaxis, diagnosis, treatment and follow-up. Ann Oncol 2021; 32:171-182. [PMID: 33246022 DOI: 10.1016/j.annonc.2020.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 02/08/2023] Open
Affiliation(s)
- P Roth
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - A Pace
- Neuroncology Unit, IRCCS Regina Elena Cancer Institute, Rome, Italy
| | - E Le Rhun
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland; Université Lille, U-1192, Lille, France; Inserm, U-1192, Lille, France; Centre Hospitalier Universitaire CHU, Lille, General and Stereotaxic Neurosurgery Service, Lille, France; Oscar Lambret Center, Breast Cancer Department, Lille, France
| | - M Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - C Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center Vienna, Vienna, Austria
| | - E Cohen-Jonathan Moyal
- Radiation Oncology Department, Institut Claudius Regaud, Université Paul Sabatier, Toulouse, France; Institut Universitaire du Cancer de Toulouse IUCT Oncopole, Toulouse, France
| | - M Coomans
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - R Giusti
- Medical Oncology Unit, Azienda Ospedaliero Universitaria Sant'Andrea, Rome, Italy
| | - K Jordan
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - R Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - F Winkler
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - J T Hong
- Department of Neurosurgery, Eunpyeong St. Mary's Hospital, Seoul, The Catholic University of Korea, Republic of Korea
| | - R Ruda
- Department of Neuro-Oncology, City of Health and Science and University of Turin, Turin, Italy
| | - S Villà
- Catalan Institute of Oncology, HU Germans Trias, Badalona, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M J B Taphoorn
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands; Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands
| | - W Wick
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - M Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW This article highlights important aspects of the evaluation, diagnosis, and treatment of adult gliomas, including lower-grade astrocytomas and oligodendrogliomas, glioblastomas, and ependymomas. RECENT FINDINGS The appropriate initial evaluation and accurate diagnosis of gliomas require an understanding of the spectrum of clinical and radiographic presentations. Recent advances in the understanding of distinct molecular prognostic subtypes have led to major revisions in the diagnostic classification of gliomas. Integration of these new diagnostic and molecular classifications is an important part of the modern management of gliomas and facilitates better understanding and interpretation of the efficacy of different therapies in specific glioma subtypes. SUMMARY The management of adult gliomas is a multidisciplinary endeavor. However, despite recent molecular and treatment advances, the majority of diffuse gliomas remain incurable, and efforts aimed at the development and testing of new therapies in clinical trials are ongoing.
Collapse
|
14
|
Erker C, Tamrazi B, Poussaint TY, Mueller S, Mata-Mbemba D, Franceschi E, Brandes AA, Rao A, Haworth KB, Wen PY, Goldman S, Vezina G, MacDonald TJ, Dunkel IJ, Morgan PS, Jaspan T, Prados MD, Warren KE. Response assessment in paediatric high-grade glioma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2020; 21:e317-e329. [PMID: 32502458 DOI: 10.1016/s1470-2045(20)30173-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/27/2022]
Abstract
Response criteria for paediatric high-grade glioma vary historically and across different cooperative groups. The Response Assessment in Neuro-Oncology working group developed response criteria for adult high-grade glioma, but these were not created to meet the unique challenges in children with the disease. The Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group, consisting of an international panel of paediatric and adult neuro-oncologists, clinicians, radiologists, radiation oncologists, and neurosurgeons, was established to address issues and unique challenges in assessing response in children with CNS tumours. We established a subcommittee to develop response assessment criteria for paediatric high-grade glioma. Current practice and literature were reviewed to identify major challenges in assessing the response of paediatric high-grade gliomas to various treatments. For areas in which scientific investigation was scarce, consensus was reached through an iterative process. RAPNO response assessment recommendations include the use of MRI of the brain and the spine, assessment of clinical status, and the use of corticosteroids or antiangiogenics. Imaging standards for brain and spine are defined. Compared with the recommendations for the management of adult high-grade glioma, for paediatrics there is inclusion of diffusion-weighted imaging and a higher reliance on T2-weighted fluid-attenuated inversion recovery. Consensus recommendations and response definitions have been established and, similar to other RAPNO recommendations, prospective validation in clinical trials is warranted.
Collapse
Affiliation(s)
- Craig Erker
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Dalhousie University and IWK Health Centre, Halifax, NS, Canada.
| | - Benita Tamrazi
- Department of Radiology, Keck School of Medicine, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Daddy Mata-Mbemba
- Department of Diagnostic Imaging, Dalhousie University and IWK Health Centre, Halifax, NS, Canada
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Alba A Brandes
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Arvind Rao
- Departments of Computational Medicine and Bioinformatics and Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kellie B Haworth
- Division of Neuro-Oncology, Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stewart Goldman
- Department of Haematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Ira J Dunkel
- Department of Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul S Morgan
- Department of Medical Physics and Clinical Engineering, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Tim Jaspan
- Department of Radiology, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Katherine E Warren
- Department of Pediatric Oncology, Dana- Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
15
|
Snyder JM, Pawloski JA, Poisson LM. Developing Real-world Evidence-Ready Datasets: Time for Clinician Engagement. Curr Oncol Rep 2020; 22:45. [PMID: 32297007 PMCID: PMC8214868 DOI: 10.1007/s11912-020-00904-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE OF REVIEW Real-world data (RWD) applications in healthcare that support learning health systems and pragmatic clinical trials are gaining momentum, largely due to legislation supporting real-world evidence (RWE) for drug approvals. Clinical notes are thought to be the cornerstone of RWD applications, particularly for conditions with limited effective treatments, extrapolation of treatments from other conditions, or heterogenous disease biology and clinical phenotypes. RECENT FINDINGS Here, we discuss current issues in applying RWD captured at the point-of-care and provide a framework for clinicians to engage in RWD collection. To achieve clinically meaningful results, RWD must be reliably captured using consistent terminology in the description of our patients. RWD complements traditional clinical trials and research by informing the generalizability of results, generating new hypotheses, and creating a large data network for scientific discovery. Effective clinician engagement in the development of RWD applications is necessary for continued progress in the field.
Collapse
Affiliation(s)
- James M Snyder
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Jacob A Pawloski
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Laila M Poisson
- Department of Public Health Sciences, Hermelin Brain Tumor Center, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| |
Collapse
|
16
|
Suh CH, Kim HS, Jung SC, Choi CG, Kim SJ, Kim KW. Optimized Image-Based Surrogate Endpoints in Targeted Therapies for Glioblastoma: A Systematic Review and Meta-Analysis of Phase III Randomized Controlled Trials. Korean J Radiol 2020; 21:471-482. [PMID: 32193895 PMCID: PMC7082650 DOI: 10.3348/kjr.2019.0839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/19/2019] [Indexed: 01/11/2023] Open
Abstract
Objective We aimed to determine the optimized image-based surrogate endpoints (IBSEs) in targeted therapies for glioblastoma through a systematic review and meta-analysis of phase III randomized controlled trials (RCTs). Materials and Methods A systematic search of OVID-MEDLINE and EMBASE for phase III RCTs on glioblastoma was performed in December 2017. Data on overall survival (OS) and IBSEs, including progression-free survival (PFS), 6-month PFS (6moPFS), 12-month PFS (12moPFS), median PFS, and objective response rate (ORR) were extracted. Weighted linear regression analysis for the hazard ratio for OS and the hazard ratios or odds ratios for IBSEs was performed. The associations between IBSEs and OS were evaluated. Subgroup analyses according to disease stage (newly diagnosed glioblastoma versus recurrent glioblastoma), types of test treatment, and types of response assessment criteria were performed. Results Twenty-three phase III RCTs published between 2000 and 2017, including 8387 patients, met the inclusion criteria. OS showed strong correlations with PFS (standardized β coefficient [R] = 0.719), 6moPFS (R = 0.647), and 12moPFS (R = 0.638). OS showed no correlations with median PFS and ORR. In subgroup analysis according to types of therapies, PFS showed the highest correlations with OS in targeted therapies for cell cycle pathways (R = 0.913) and growth factor receptors and their downstream pathways (R = 0.962). 12moPFS showed the highest correlation with OS in antiangiogenic therapy (R = 0.821). The response assessment in neuro-oncology criteria provided higher correlation coefficients between OS and IBSEs than the Macdonald criteria. Conclusion Overall, PFS is an optimized IBSE in targeted therapies for glioblastoma; however, 12moPFS is optimal in antiangiogenic therapy.
Collapse
Affiliation(s)
- Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea.
| | - Seung Chai Jung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Choong Gon Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Joon Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
17
|
Abstract
Impaired neurocognitive function is an increasingly recognized morbidity in patients who have cancer. Cancer treatments, psychosocial stressors, and the malignancy itself can alter brain function. The mechanisms by which this occurs are under active investigation. Although there is a growing appreciation of its prevalence and causes, there remain limited therapeutic options for the treatment of neurocognitive dysfunction in this population. A persistent scientific and clinical effort to understand its mechanisms and impact is critical to the care of oncology patients.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 0431, Houston, TX 77030, USA.
| | - Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 0431, Houston, TX 77030, USA
| |
Collapse
|
18
|
Noll KR, Bradshaw ME, Parsons MW, Dawson EL, Rexer J, Wefel JS. Monitoring of Neurocognitive Function in the Care of Patients with Brain Tumors. Curr Treat Options Neurol 2019; 21:33. [PMID: 31250277 DOI: 10.1007/s11940-019-0573-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW A detailed characterization of the nature of neurocognitive impairment in patients with brain tumors is provided, as well as considerations for clinical practice regarding neuropsychological assessment throughout the disease course. RECENT FINDINGS Neurocognitive impairment is common in patients with brain tumors and may result from the tumor itself, as a consequence of treatment, including surgery, chemotherapy, and radiation, or in association with supportive care medications (e.g., anticonvulsant and pain medications). Serial surveillance of neurocognitive functioning in this population can facilitate medical decision-making and inform recommendations to improve patient daily functioning and quality of life. Neuropsychological assessment is increasingly recognized as a critical component of the multidisciplinary care of patients with brain tumors and has already had practice-changing effects. Further understanding of genetic risk factors for neurocognitive decline along with the development of novel assessment and intervention strategies may further enhance functioning and general well-being in this patient population.
Collapse
Affiliation(s)
- Kyle R Noll
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Mariana E Bradshaw
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Michael W Parsons
- Department of Neuro-Oncology, Psychology Assessment Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Erica L Dawson
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Jennie Rexer
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA. .,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Optimizing Neuro-Oncology Imaging: A Review of Deep Learning Approaches for Glioma Imaging. Cancers (Basel) 2019; 11:cancers11060829. [PMID: 31207930 PMCID: PMC6627902 DOI: 10.3390/cancers11060829] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/06/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023] Open
Abstract
Radiographic assessment with magnetic resonance imaging (MRI) is widely used to characterize gliomas, which represent 80% of all primary malignant brain tumors. Unfortunately, glioma biology is marked by heterogeneous angiogenesis, cellular proliferation, cellular invasion, and apoptosis. This translates into varying degrees of enhancement, edema, and necrosis, making reliable imaging assessment challenging. Deep learning, a subset of machine learning artificial intelligence, has gained traction as a method, which has seen effective employment in solving image-based problems, including those in medical imaging. This review seeks to summarize current deep learning applications used in the field of glioma detection and outcome prediction and will focus on (1) pre- and post-operative tumor segmentation, (2) genetic characterization of tissue, and (3) prognostication. We demonstrate that deep learning methods of segmenting, characterizing, grading, and predicting survival in gliomas are promising opportunities that may enhance both research and clinical activities.
Collapse
|
20
|
Boydell E, Marinari E, Migliorini D, Dietrich PY, Patrikidou A, Dutoit V. Exploratory Study of the Effect of IMA950/Poly-ICLC Vaccination on Response to Bevacizumab in Relapsing High-Grade Glioma Patients. Cancers (Basel) 2019; 11:E464. [PMID: 30986995 PMCID: PMC6520681 DOI: 10.3390/cancers11040464] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 11/17/2022] Open
Abstract
Immunotherapy, including therapeutic vaccines, is increasingly being developed for patients with high-grade glioma, and combinations of immunotherapies and synergy with standard of care are being investigated. In this regard, bevacizumab (BEV) has been shown to synergize with immunotherapy in preclinical studies of glioma and in other tumour entities. Here, we conducted a post-hoc exploratory study to evaluate the effect of the IMA950/poly-ICLC peptide vaccine on subsequent BEV administration in high-grade glioma patients. 16 IMA950-vaccinated and 40 non-vaccinated patients were included. At initial diagnosis, patients benefited from surgery and chemoradiation. At first or subsequent recurrence, patients received 10mg/kg of BEV every 2-3 weeks. Primary endpoints were overall survival (OS) and progression-free survival (PFS) from BEV initiation. IMA950-vaccinated patients did not show improved response to BEV as compared to non-vaccinated patients: there was no difference in median PFS (2.6 vs. 4.2 months for vaccinated and control patients, respectively, p = 0.50) nor in median OS (7.8 vs. 10.0 months for vaccinated and control patients, respectively, p = 0.69). In conclusion, potential synergy of BEV and therapeutic vaccines, when administered sequentially, has yet to be established in the clinical setting of GBM recurrence. Potential synergy of concomitant administration should be tested in future trials.
Collapse
Affiliation(s)
- Emma Boydell
- Laboratory of Tumour Immunology and Department of Oncology, Geneva University Hospital, 1211 Geneva, Switzerland.
- Translational Research Center for Oncohaematology, Department of Internal Medicine Specialties, University of Geneva, 1211 Geneva, Switzerland.
| | - Eliana Marinari
- Laboratory of Tumour Immunology and Department of Oncology, Geneva University Hospital, 1211 Geneva, Switzerland.
- Translational Research Center for Oncohaematology, Department of Internal Medicine Specialties, University of Geneva, 1211 Geneva, Switzerland.
| | - Denis Migliorini
- Laboratory of Tumour Immunology and Department of Oncology, Geneva University Hospital, 1211 Geneva, Switzerland.
- Translational Research Center for Oncohaematology, Department of Internal Medicine Specialties, University of Geneva, 1211 Geneva, Switzerland.
- Department of Oncology, Clinical Research Unit, Dr Dubois Ferrière Dinu Lipatti Research Foundation, Geneva University Hospital, 1211 Geneva, Switzerland.
| | - Pierre-Yves Dietrich
- Translational Research Center for Oncohaematology, Department of Internal Medicine Specialties, University of Geneva, 1211 Geneva, Switzerland.
- Department of Oncology, Clinical Research Unit, Dr Dubois Ferrière Dinu Lipatti Research Foundation, Geneva University Hospital, 1211 Geneva, Switzerland.
| | - Anna Patrikidou
- Department of Oncology, Clinical Research Unit, Dr Dubois Ferrière Dinu Lipatti Research Foundation, Geneva University Hospital, 1211 Geneva, Switzerland.
| | - Valérie Dutoit
- Laboratory of Tumour Immunology and Department of Oncology, Geneva University Hospital, 1211 Geneva, Switzerland.
- Translational Research Center for Oncohaematology, Department of Internal Medicine Specialties, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
21
|
Chinnaiyan P, Won M, Wen PY, Rojiani AM, Werner-Wasik M, Shih HA, Ashby LS, Michael Yu HH, Stieber VW, Malone SC, Fiveash JB, Mohile NA, Ahluwalia MS, Wendland MM, Stella PJ, Kee AY, Mehta MP. A randomized phase II study of everolimus in combination with chemoradiation in newly diagnosed glioblastoma: results of NRG Oncology RTOG 0913. Neuro Oncol 2019; 20:666-673. [PMID: 29126203 DOI: 10.1093/neuonc/nox209] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background This phase II study was designed to determine the efficacy of the mammalian target of rapamycin (mTOR) inhibitor everolimus administered daily with conventional radiation therapy and chemotherapy in patients with newly diagnosed glioblastoma. Methods Patients were randomized to radiation therapy with concurrent and adjuvant temozolomide with or without daily everolimus (10 mg). The primary endpoint was progression-free survival (PFS) and the secondary endpoints were overall survival (OS) and treatment-related toxicities. Results A total of 171 patients were randomized and deemed eligible for this study. Patients randomized to receive everolimus experienced a significant increase in both grade 4 toxicities, including lymphopenia and thrombocytopenia, and treatment-related deaths. There was no significant difference in PFS between patients randomized to everolimus compared with control (median PFS time: 8.2 vs 10.2 mo, respectively; P = 0.79). OS for patients randomized to receive everolimus was inferior to that for control patients (median survival time: 16.5 vs 21.2 mo, respectively; P = 0.008). A similar trend was observed in both O6-methylguanine-DNA-methyltransferase promoter hypermethylated and unmethylated tumors. Conclusion Combining everolimus with conventional chemoradiation leads to increased treatment-related toxicities and does not improve PFS in patients with newly diagnosed glioblastoma. Although the median survival time in patients receiving everolimus was comparable to contemporary studies, it was inferior to the control in this randomized study.
Collapse
Affiliation(s)
| | - Minhee Won
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania, USA
| | - Patrick Y Wen
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | - Amyn M Rojiani
- Augusta University-Medical College of Georgia, Augusta, Georgia, USA
| | | | - Helen A Shih
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lynn S Ashby
- Barrow Neurological Institute accruals under Arizona Oncology Services Foundation, Phoenix, Arizona, USA
| | | | - Volker W Stieber
- Novant Health Forsyth Regional Cancer Center accruals under Southeast Cancer Control Consortium, Inc, CCOP, Goldsboro, North Carolina, USA
| | - Shawn C Malone
- The Ottawa Hospital Regional Cancer Centre, Ottawa, Ontario, Canada
| | - John B Fiveash
- University of Alabama at Birmingham Medical Center, Birmingham, Alabama, USA
| | | | | | | | - Philip J Stella
- Saint Joseph Mercy Hospital accruals under Michigan Cancer Research Consortium CCOP, Ypsilanti, Michigan, USA
| | - Andrew Y Kee
- Legacy Health Systems accruals under Mayo Clinic, Portland, Oregon, USA
| | | |
Collapse
|
22
|
Shergalis A, Bankhead A, Luesakul U, Muangsin N, Neamati N. Current Challenges and Opportunities in Treating Glioblastoma. Pharmacol Rev 2018; 70:412-445. [PMID: 29669750 PMCID: PMC5907910 DOI: 10.1124/pr.117.014944] [Citation(s) in RCA: 504] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common and aggressive primary brain tumor, has a high mortality rate despite extensive efforts to develop new treatments. GBM exhibits both intra- and intertumor heterogeneity, lending to resistance and eventual tumor recurrence. Large-scale genomic and proteomic analysis of GBM tumors has uncovered potential drug targets. Effective and “druggable” targets must be validated to embark on a robust medicinal chemistry campaign culminating in the discovery of clinical candidates. Here, we review recent developments in GBM drug discovery and delivery. To identify GBM drug targets, we performed extensive bioinformatics analysis using data from The Cancer Genome Atlas project. We discovered 20 genes, BOC, CLEC4GP1, ELOVL6, EREG, ESR2, FDCSP, FURIN, FUT8-AS1, GZMB, IRX3, LITAF, NDEL1, NKX3-1, PODNL1, PTPRN, QSOX1, SEMA4F, TH, VEGFC, and C20orf166AS1 that are overexpressed in a subpopulation of GBM patients and correlate with poor survival outcomes. Importantly, nine of these genes exhibit higher expression in GBM versus low-grade glioma and may be involved in disease progression. In this review, we discuss these proteins in the context of GBM disease progression. We also conducted computational multi-parameter optimization to assess the blood-brain barrier (BBB) permeability of small molecules in clinical trials for GBM treatment. Drug delivery in the context of GBM is particularly challenging because the BBB hinders small molecule transport. Therefore, we discuss novel drug delivery methods, including nanoparticles and prodrugs. Given the aggressive nature of GBM and the complexity of targeting the central nervous system, effective treatment options are a major unmet medical need. Identification and validation of biomarkers and drug targets associated with GBM disease progression present an exciting opportunity to improve treatment of this devastating disease.
Collapse
Affiliation(s)
- Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Armand Bankhead
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Urarika Luesakul
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nongnuj Muangsin
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| |
Collapse
|
23
|
Luo L, Chi H, Ling J. MiR-124-3p suppresses glioma aggressiveness via targeting of Fra-2. Pathol Res Pract 2018; 214:1825-1834. [PMID: 30243808 DOI: 10.1016/j.prp.2018.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Abstract
Malignant glioma is the most common and deadly primary brain tumor in adults. However, the mechanisms underlying the malignancy of glioma remain unclear. In the present study, we found that Fos-related antigen-2 (Fra-2) was overexpressed in most glioma cells, and knockdown of Fra-2 prevented cell proliferation, migration, and invasion. Mechanistically, Fra-2 silencing led to a significant reduction in cell-cycle drivers (Cyclin D1 and Cyclin E1), one invasion-associated gene (MMP9), the mesenchymal marker (Vimentin), and induction of the epithelial marker (E-cadherin). Further study confirmed that miR-124-3p decreased the expression of Fra-2 via directly targeting the 3'-UTR, and transfection with miR-124-3p in glioma cells inhibited expression of the above cell-cycle and EMT promoters. Phenotypic experiments also showed that overexpression of Fra-2 weakened the inhibitory effects of miR-124-3p on the proliferation, migration, and invasion of glioma cells. In addition, Fra-2 knockdown impaired the malignant phenotypes enhanced by miR-124-3p inhibition, which suggested a crucial role for the miR-124-3p/Fra-2 pathway in glioma development. Consistently, high expression of Fra-2 was closely associated with low miR-124-3p level and indicated a poor prognosis in patients with glioma. In conclusion, this study indicates the existence of an aberrant miR-124-3p/Fra-2 pathway that results in glioma aggressiveness, which suggests novel therapeutic opportunities for this fatal disease.
Collapse
Affiliation(s)
- Lifei Luo
- Clinical Laboratory, Enze Hospital, Taizhou Enze Medical Center, Luqiao 318050, China
| | - Hongbo Chi
- Clinical Laboratory, Enze Hospital, Taizhou Enze Medical Center, Luqiao 318050, China
| | - Jie Ling
- Clinical Laboratory, Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical University, Huangyan 318020, China.
| |
Collapse
|
24
|
Habets EJJ, Taphoorn MJB, Klein M, Vissers T, Dirven L. The level of reporting of neurocognitive outcomes in randomised controlled trials of brain tumour patients: A systematic review. Eur J Cancer 2018; 100:104-125. [PMID: 30014880 DOI: 10.1016/j.ejca.2018.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 05/18/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Neurocognitive impairment is frequently present in brain tumour patients and is therefore considered an important outcome in brain tumour research. To use neurocognitive outcomes (NCO) in clinical decision-making, neurocognitive evidence should be of sufficiently high quality. We aimed to investigate the level of neurocognitive functioning reporting in randomised controlled trials (RCTs) in brain tumour patients. METHODS We conducted a systematic literature search in several databases up to August 2017. Of the selected relevant RCTs, the following data were retrieved: basic trial demographics and NCO characteristics, quality of NCO reporting and risk of bias. We also analysed studies that should impact clinical decision-making based on their quality of reporting. RESULTS We identified 65 RCTs, of which NCO was the primary end-point in 14 (22%). Important methodological limitations were related to the documentation of statistical approaches for dealing with missing data and to discussing limitations and generalisability issues uniquely related to the NCO components. Risk of bias was high regarding blinding of personnel and incomplete outcome data. Twenty RCTs (31%), eight with NCO as primary end-point and 12 as secondary end-point, satisfied a sufficient number of criteria to be classified as 'high-quality' NCO evidence. Most of these studies did contribute to clinical decision-making. CONCLUSION Investigators involved in brain tumour research should give attention to methodological challenges related to NCO reporting as identified in this review, as 'high-quality' reporting of NCO evidence can be of value in clinical decision-making.
Collapse
Affiliation(s)
- Esther J J Habets
- Department of Neurology, Haaglanden Medical Center, Lijnbaan 32, 2501 CK, The Hague, The Netherlands.
| | - Martin J B Taphoorn
- Department of Neurology, Haaglanden Medical Center, Lijnbaan 32, 2501 CK, The Hague, The Netherlands; Department of Neurology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Martin Klein
- Department of Medical Psychology, VU University Medical Center, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Thomas Vissers
- Medical Library, Haaglanden Medical Center, Lijnbaan 32, 2501 CK, The Hague, The Netherlands
| | - Linda Dirven
- Department of Neurology, Haaglanden Medical Center, Lijnbaan 32, 2501 CK, The Hague, The Netherlands; Department of Neurology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
25
|
Ellingson BM, Wen PY, Cloughesy TF. Evidence and context of use for contrast enhancement as a surrogate of disease burden and treatment response in malignant glioma. Neuro Oncol 2018; 20:457-471. [PMID: 29040703 PMCID: PMC5909663 DOI: 10.1093/neuonc/nox193] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The use of contrast enhancement within the brain on CT or MRI has been the gold standard for diagnosis and therapeutic response assessment in malignant gliomas for decades. The use of contrast enhancing tumor size, however, remains controversial as a tool for accurately diagnosing and assessing treatment efficacy in malignant gliomas, particularly in the current, quickly evolving therapeutic landscape. The current article consolidates overwhelming evidence from hundreds of studies in the field of neuro-oncology, providing the necessary evidence base and specific contexts of use for consideration of contrast enhancing tumor size as an appropriate surrogate biomarker for disease burden and as a tool for measuring treatment response in malignant glioma, including glioblastoma.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- UCLA Brain Research Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Physics in Medicine and Biology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA, University of California Los Angeles, Los Angeles, California
| | - Patrick Y Wen
- Department of Neurooncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
26
|
Rechallenge with bevacizumab in patients with glioblastoma progressing off therapy. J Neurooncol 2018; 138:141-145. [DOI: 10.1007/s11060-018-2780-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/26/2018] [Indexed: 01/05/2023]
|
27
|
Dongas J, Asahina AT, Bacchi S, Patel S. Magnetic Resonance Perfusion Imaging in the Diagnosis of High-Grade Glioma Progression and Treatment-Related Changes: A Systematic Review. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/ojmn.2018.83024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Wefel JS, Parsons MW, Gondi V, Brown PD. Neurocognitive aspects of brain metastasis. HANDBOOK OF CLINICAL NEUROLOGY 2018; 149:155-165. [PMID: 29307352 DOI: 10.1016/b978-0-12-811161-1.00012-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Brain metastases are common, occurring in approximately 20% of cancer patients. One of the biggest concerns for these patients and their families is neurocognitive decline. Neurocognitive issues in this patient population are complex and many patients have neurocognitive impairment due to systemic therapies even before they develop brain metastases. The development of brain metastases as well as the treatment of these tumors can cause decline in neurocognitive function. Diffuse treatments such as whole-brain radiotherapy are more frequently associated with neurocognitive decline than focal interventions such as radiosurgery, surgical resection, and implantable chemotherapy wafers. For patients with brain metastases treatment decisions require a multidisciplinary approach, balancing many factors including the neurocognitive impact of treatment and the disease process itself. Finally, to continue to advance the field there needs to be continued utilization, both off and on clinical trial, of performance-based clinical outcome assessments (i.e., neurocognitive tests) to objectively assess and measure the neurocognitive outcomes of these patients.
Collapse
Affiliation(s)
- Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| | - Michael W Parsons
- Section of Neuropsychology, Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, OH, United States
| | - Vinai Gondi
- Brain and Spine Tumor Center, Northwestern Medicine Cancer Center Warrenville and Northwestern Medicine Chicago Proton Center, Warrenville, IL, United States
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
29
|
Wen PY, Chang SM, Van den Bent MJ, Vogelbaum MA, Macdonald DR, Lee EQ. Response Assessment in Neuro-Oncology Clinical Trials. J Clin Oncol 2017; 35:2439-2449. [PMID: 28640707 PMCID: PMC5516482 DOI: 10.1200/jco.2017.72.7511] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Development of novel therapies for CNS tumors requires reliable assessment of response and progression. This requirement has been particularly challenging in neuro-oncology for which contrast enhancement serves as an imperfect surrogate for tumor volume and is influenced by agents that affect vascular permeability, such as antiangiogenic therapies. In addition, most tumors have a nonenhancing component that can be difficult to accurately quantify. To improve the response assessment in neuro-oncology and to standardize the criteria that are used for different CNS tumors, the Response Assessment in Neuro-Oncology (RANO) working group was established. This multidisciplinary international working group consists of neuro-oncologists, medical oncologists, neuroradiologists, neurosurgeons, radiation oncologists, neuropsychologists, and experts in clinical outcomes assessments, working in collaboration with government and industry to enhance the interpretation of clinical trials. The RANO working group was originally created to update response criteria for high- and low-grade gliomas and to address such issues as pseudoresponse and nonenhancing tumor progression from antiangiogenic therapies, and pseudoprogression from radiochemotherapy. RANO has expanded to include working groups that are focused on other tumors, including brain metastases, leptomeningeal metastases, spine tumors, pediatric brain tumors, and meningiomas, as well as other clinical trial end points, such as clinical outcomes assessments, seizures, corticosteroid use, and positron emission tomography imaging. In an effort to standardize the measurement of neurologic function for clinical assessment, the Neurologic Assessment in Neuro-Oncology scale was drafted. Born out of a workshop conducted by the Jumpstarting Brain Tumor Drug Development Coalition and the US Food and Drug Administration, a standardized brain tumor imaging protocol now exists to reduce variability and improve reliability. Efforts by RANO have been widely accepted and are increasingly being used in neuro-oncology trials, although additional refinements will be needed.
Collapse
Affiliation(s)
- Patrick Y. Wen
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Susan M. Chang
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Martin J. Van den Bent
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Michael A. Vogelbaum
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - David R. Macdonald
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| | - Eudocia Q. Lee
- Patrick Y. Wen and Eudocia Q. Lee, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Susan M. Chang, University of California, San Francisco, San Francisco, CA; Michael A. Vogelbaum, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH; Martin J. Van den Bent, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands; and David R. Macdonald, London Regional Cancer Program, Western University, London, Ontario, Canada
| |
Collapse
|
30
|
Dysregulation of Fra1 expression by Wnt/β-catenin signalling promotes glioma aggressiveness through epithelial-mesenchymal transition. Biosci Rep 2017; 37:BSR20160643. [PMID: 28232512 PMCID: PMC5469333 DOI: 10.1042/bsr20160643] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/12/2017] [Accepted: 02/22/2017] [Indexed: 01/08/2023] Open
Abstract
Aberrant expression of Fos-related antigen-1 (Fra1) is commonly elevated in various malignant cancers and is strongly implicated in invasion and metastasis. However, the molecular mechanisms underlying its dysregulation in human glioma remain poorly understood. In the present study, we demonstrate that up-regulation of Fra1 plays a crucial role in the glioma aggressiveness and epithelial–mesenchymal transition (EMT) activated by Wnt/β-catenin signal pathway. In glioma cells, activation of Wnt/β-catenin signalling by Wnt3a administration obviously induced EMT and directly activated the transcription of Fra1. Phenotype experiments revealed that up-regulation of Fra1 induced by Wnt/β-catenin signalling drove the EMT of glioma cells. Furthermore, it was found that the cisplatin resistance acquired by Wnt/β-catenin signalling activation depended on increased expression of Fra1. Analysis of clinical specimens verified a positive correlation between Fra1 and β-catenin as well as a poor prognosis in glioma patients with double-high expressions of them. These findings indicate that an aberrant Wnt/β-catenin signalling leads to the EMT and drug resistance of glioma via Fra1 induction, which suggests novel therapeutic strategies for the malignant disease.
Collapse
|
31
|
Ediebah DE, Reijneveld JC, Taphoorn MJB, Coens C, Zikos E, Aaronson NK, Heimans JJ, Bottomley A, Klein M. Impact of neurocognitive deficits on patient-proxy agreement regarding health-related quality of life in low-grade glioma patients. Qual Life Res 2016; 26:869-880. [PMID: 27744512 PMCID: PMC5334398 DOI: 10.1007/s11136-016-1426-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Clinical trials in glioma patients with neurocognitive deficits face challenges due to lacking or unreliable patient self-reports on their health-related quality of life (HRQOL). Patient-proxy data could help solve this issue. We determined whether patient-proxy concordance levels were affected by patients' neurocognitive functioning. METHODS Patient and patient-by-proxy HRQOL ratings were assessed via SF-36 and EORTC QLQ-BN20, respectively, in 246 patients. Data on neurocognitive functioning were collected on a subgroup of 195 patients. Patient-proxy agreement was measured using the Bland-Altman limit of agreement, the mean difference, the concordance correlation coefficient (CCC), and the percentage difference (PD, ±0, 5, or 10 points). We defined patients to be cognitively impaired (n = 66) or cognitively intact (n = 129) based on their neurocognitive performance. RESULTS Patients rated their physical function and general health to be better than their proxies did, while at the same time, patients reported more visual disorders, communication deficits, itchy skin, and problems with bladder control. The cognitively impaired subgroup reported poorer physical functioning, more visual disorders, headaches, itchy skin, and issues with bladder control. In the cognitively intact group, no statistical significant differences were observed between patients and proxies. Not surprisingly, Bland-Altman plots revealed a high agreement between the patient and patient-by-proxy rating in all HRQOL domains ranging from 95 to 99 %. The CCC was fairly high in all HRQOL domains (0.37-0.80), and the percentage of perfect agreement (PD ± 0) ranged from 8.5 to 76.8 %. In the cognitively impaired patients, the mean difference between patients and proxies was overall larger, and accordingly, agreement based on Bland-Altman plots was lower. CONCLUSIONS The level of agreement between patient and patient-by-proxy ratings of low-grade glioma patients' HRQOL is generally high. However, patient-proxy agreement is lower in patients with neurocognitive deficits than in patients without neurocognitive deficits.
Collapse
Affiliation(s)
- Divine E Ediebah
- Quality of Life Department, European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Jaap C Reijneveld
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Martin J B Taphoorn
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurology, Medical Center Haaglanden, The Hague, The Netherlands
| | - Corneel Coens
- Quality of Life Department, European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Efstathios Zikos
- Quality of Life Department, European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Neil K Aaronson
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan J Heimans
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Andrew Bottomley
- Quality of Life Department, European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Martin Klein
- Department of Medical Psychology - B7D349, VU University Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | | |
Collapse
|
32
|
Huang RY, Wen PY. Response Assessment in Neuro-Oncology Criteria and Clinical Endpoints. Magn Reson Imaging Clin N Am 2016; 24:705-718. [PMID: 27742111 DOI: 10.1016/j.mric.2016.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Response Assessment in Neuro-Oncology (RANO) Working Group is an international multidisciplinary group whose goal is to improve response criteria and define endpoints for neuro-oncology trials. The RANO criteria for high-grade gliomas attempt to address the issues of pseudoprogression, pseudoresponse, and nonenhancing tumor progression. Incorporation of advanced MR imaging may eventually help improve the ability of these criteria to define enhancing and nonenhancing disease better. The RANO group has also developed criteria for neurologic response and evaluation of patients receiving immunologic therapies. RANO criteria have been developed for brain metastases and are in progress for meningiomas, leptomeningeal disease, spinal tumors, and pediatric tumors.
Collapse
Affiliation(s)
- Raymond Y Huang
- Division of Neuroradiology, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Patrick Y Wen
- Division of Neuro-Oncology, Department of Neurology, Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
33
|
Levin VA, Tonge PJ, Gallo JM, Birtwistle MR, Dar AC, Iavarone A, Paddison PJ, Heffron TP, Elmquist WF, Lachowicz JE, Johnson TW, White FM, Sul J, Smith QR, Shen W, Sarkaria JN, Samala R, Wen PY, Berry DA, Petter RC. CNS Anticancer Drug Discovery and Development Conference White Paper. Neuro Oncol 2016; 17 Suppl 6:vi1-26. [PMID: 26403167 DOI: 10.1093/neuonc/nov169] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Following the first CNS Anticancer Drug Discovery and Development Conference, the speakers from the first 4 sessions and organizers of the conference created this White Paper hoping to stimulate more and better CNS anticancer drug discovery and development. The first part of the White Paper reviews, comments, and, in some cases, expands on the 4 session areas critical to new drug development: pharmacological challenges, recent drug approaches, drug targets and discovery, and clinical paths. Following this concise review of the science and clinical aspects of new CNS anticancer drug discovery and development, we discuss, under the rubric "Accelerating Drug Discovery and Development for Brain Tumors," further reasons why the pharmaceutical industry and academia have failed to develop new anticancer drugs for CNS malignancies and what it will take to change the current status quo and develop the drugs so desperately needed by our patients with malignant CNS tumors. While this White Paper is not a formal roadmap to that end, it should be an educational guide to clinicians and scientists to help move a stagnant field forward.
Collapse
Affiliation(s)
- Victor A Levin
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Peter J Tonge
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - James M Gallo
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Marc R Birtwistle
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Arvin C Dar
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Antonio Iavarone
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Patrick J Paddison
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Timothy P Heffron
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - William F Elmquist
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Jean E Lachowicz
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Ted W Johnson
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Forest M White
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Joohee Sul
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Quentin R Smith
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Wang Shen
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Jann N Sarkaria
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Ramakrishna Samala
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Patrick Y Wen
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Donald A Berry
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| | - Russell C Petter
- Kaiser Permanente, Redwood City, California, USA (V.A.L.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (V.A.L.); University of California, San Francisco, CA, USA (V.A.L.); SUNY Stony Brook University, Stony Brook, NY, USA (P.J.T.); Icahn School of Medicine at Mount Sinai, New York, NY, USA (J.M.G., M.R.B., A.C.D.); Columbia University Institute for Cancer Genetics, New York, NY, USA (A.I.); Fred Hutchinson Cancer Research Center, Seattle, WA, USA (P.J.P.); Genentech, Inc., South San Francisco, CA, USA (T.P.H.); University of Minnesota School of Pharmacy, Minneapolis, MN, USA (W.F.E.); Angiochem, Inc., Montreal, Quebec, Canada (J.E.L.); Pfizer Oncology, San Diego, CA, USA (T.W.J.); Massachusetts Institute of Technology, Cambridge, MA, USA (F.M.W.); US Food and Drug Administration, Silver Spring, MD, USA (J.S.); Texas Tech University School of Pharmacy, Amarillo, TX, USA (Q.R.S., R.S.); NewGen Therapeutics, Inc., Menlo Park, CA, USA (W.S.); Mayo Clinic, Rochester, MN, USA (J.N.S.); Dana-Farber Cancer Institute, Boston, MA, USA (P.Y.W.); University of Texas MD Anderson Cancer Center, Houston, TX, USA (D.A.B.); Celgene Avilomics Research, Bedford, MA, USA (R.C.P.)
| |
Collapse
|
34
|
Kleesiek J, Petersen J, Döring M, Maier-Hein K, Köthe U, Wick W, Hamprecht FA, Bendszus M, Biller A. Virtual Raters for Reproducible and Objective Assessments in Radiology. Sci Rep 2016; 6:25007. [PMID: 27118379 PMCID: PMC4846987 DOI: 10.1038/srep25007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/06/2016] [Indexed: 11/09/2022] Open
Abstract
Volumetric measurements in radiologic images are important for monitoring tumor growth and treatment response. To make these more reproducible and objective we introduce the concept of virtual raters (VRs). A virtual rater is obtained by combining knowledge of machine-learning algorithms trained with past annotations of multiple human raters with the instantaneous rating of one human expert. Thus, he is virtually guided by several experts. To evaluate the approach we perform experiments with multi-channel magnetic resonance imaging (MRI) data sets. Next to gross tumor volume (GTV) we also investigate subcategories like edema, contrast-enhancing and non-enhancing tumor. The first data set consists of N = 71 longitudinal follow-up scans of 15 patients suffering from glioblastoma (GB). The second data set comprises N = 30 scans of low- and high-grade gliomas. For comparison we computed Pearson Correlation, Intra-class Correlation Coefficient (ICC) and Dice score. Virtual raters always lead to an improvement w.r.t. inter- and intra-rater agreement. Comparing the 2D Response Assessment in Neuro-Oncology (RANO) measurements to the volumetric measurements of the virtual raters results in one-third of the cases in a deviating rating. Hence, we believe that our approach will have an impact on the evaluation of clinical studies as well as on routine imaging diagnostics.
Collapse
Affiliation(s)
- Jens Kleesiek
- University of Heidelberg, Department of Neuroradiology, Heidelberg, Germany.,German Cancer Research Center, Junior Group Medical Image Computing, Heidelberg, Germany.,University of Heidelberg, HCI/IWR, Heidelberg, Germany.,German Cancer Research Center, Division of Radiology, Heidelberg, Germany
| | - Jens Petersen
- University of Heidelberg, Department of Neuroradiology, Heidelberg, Germany.,German Cancer Research Center, Junior Group Medical Image Computing, Heidelberg, Germany
| | - Markus Döring
- University of Heidelberg, Department of Neuroradiology, Heidelberg, Germany
| | - Klaus Maier-Hein
- German Cancer Research Center, Junior Group Medical Image Computing, Heidelberg, Germany
| | - Ullrich Köthe
- University of Heidelberg, HCI/IWR, Heidelberg, Germany
| | - Wolfgang Wick
- University of Heidelberg, Department of Neurology, Heidelberg, Germany
| | | | - Martin Bendszus
- University of Heidelberg, Department of Neuroradiology, Heidelberg, Germany
| | - Armin Biller
- University of Heidelberg, Department of Neuroradiology, Heidelberg, Germany.,German Cancer Research Center, Division of Radiology, Heidelberg, Germany
| |
Collapse
|
35
|
Wolf J, Campos B, Bruckner T, Vogt L, Unterberg A, Ahmadi R. Evaluation of neuropsychological outcome and “quality of life” after glioma surgery. Langenbecks Arch Surg 2016; 401:541-9. [DOI: 10.1007/s00423-016-1403-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/07/2016] [Indexed: 11/30/2022]
|
36
|
Response Assessment and Magnetic Resonance Imaging Issues for Clinical Trials Involving High-Grade Gliomas. Top Magn Reson Imaging 2016; 24:127-36. [PMID: 26049816 DOI: 10.1097/rmr.0000000000000054] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There exist multiple challenges associated with the current response assessment criteria for high-grade gliomas, including the uncertain role of changes in nonenhancing T2 hyperintensity, and the phenomena of pseudoresponse and pseudoprogression in the setting of antiangiogenic and chemoradiation therapies, respectively. Advanced physiological magnetic resonance imaging (MRI), including diffusion and perfusion (dynamic susceptibility contrast MRI and dynamic contrast-enhanced MRI) sensitive techniques for overcoming response assessment challenges, has been proposed, with their own potential advantages and inherent shortcomings. Measurement variability exists for conventional and advanced MRI techniques, necessitating the standardization of image acquisition parameters in order to establish the utility of these imaging methods in multicenter trials for high-grade gliomas. This review chapter highlights the important features of MRI in clinical brain tumor trials, focusing on the current state of response assessment in brain tumors, advanced imaging techniques that may provide additional value for determining response, and imaging issues to be considered for multicenter trials.
Collapse
|
37
|
Brandes AA, Finocchiaro G, Zagonel V, Reni M, Caserta C, Fabi A, Clavarezza M, Maiello E, Eoli M, Lombardi G, Monteforte M, Proietti E, Agati R, Eusebi V, Franceschi E. AVAREG: a phase II, randomized, noncomparative study of fotemustine or bevacizumab for patients with recurrent glioblastoma. Neuro Oncol 2016; 18:1304-12. [PMID: 26951379 DOI: 10.1093/neuonc/now035] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/07/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Few prospective studies have assessed the role of bevacizumab and included a control arm with standard treatments for recurrent glioblastoma. We conducted a noncomparative phase II trial (AVAREG) to examine the efficacy of bevacizumab or fotemustine in this setting. METHODS Eligible patients were randomized 2:1 to receive bevacizumab (10 mg/kg every 2 weeks) or fotemustine (75 mg/m(2) on days 1, 8, and 15, then 100 mg/m(2) every 3 weeks after a 35-day interval). The primary endpoint was 6-month overall survival (OS) rate (OS-6). No formal efficacy comparison was made between the treatment arms. RESULTS Ninety-one patients were enrolled (bevacizumab n = 59; fotemustine n = 32). Median age was 57 years (range, 28-78 y), and patients had Eastern Cooperative Oncology Group performance status of 0 (n = 42), 1 (n = 35), or 2 (n = 14). OS-6 rate was 62.1% (95% confidence interval [CI], 48.4-74.5) with bevacizumab and 73.3% (95% CI, 54.1-87.7) with fotemustine. OS-6 rates were lower in bevacizumab-treated patients with MGMT promoter methylated tumors than in those with unmethylated tumors (50% and 85%, respectively), but higher in fotemustine-treated patients (87.5% and 50%, respectively). OS rates at 9 months were 37.9% (95% CI, 25.5-51.6) and 46.7% (95% CI, 28.3-65.7) with bevacizumab and fotemustine, respectively, and median OS was 7.3 months (95% CI, 5.8-9.2) and 8.7 months (95% CI, 6.3-15.4), respectively. Toxicity was as expected with the 2 agents. CONCLUSION Single-agent bevacizumab may have a role in patients with recurrent glioblastoma.
Collapse
Affiliation(s)
- Alba A Brandes
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Gaetano Finocchiaro
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Vittorina Zagonel
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Michele Reni
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Claudia Caserta
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Alessandra Fabi
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Matteo Clavarezza
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Evaristo Maiello
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Marica Eoli
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Giuseppe Lombardi
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Marta Monteforte
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Emanuela Proietti
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Raffaele Agati
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Vincenzo Eusebi
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| | - Enrico Franceschi
- Department of Medical Oncology, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Bologna, Italy (A.A.B., E.F.); Molecular Neuro-Oncology Unit, IRCCS Foundation Carlo Besta, Milan, Italy (G.F., M.E.); Department of Clinical and Experimental Oncology, Medical Oncology, Veneto Institute of Oncology- IRCCS Padua, Italy (V.Z., G.L.); Department of Medical Oncology, IRCCS San Raffaele, Milan, Italy (M.R.); Oncology Department, Santa Maria Hospital, Terni, Italy (C.C.); Department of Medical Oncology, Regina Elena National Cancer Institute, Rome, Italy (A.F.); E.O. Ospedale Galliera, Genova, Italy (M.C.); Oncology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy (E.M.); OPIS, Palazzo Aliprandi, Desio MB, Italy (M.M.); Roche S.p.A. Medical Affairs and CO, Monza, Italy (E.P.); Neuroradiology Department, Bellaria-Maggiore Hospital, Azienda USL-IRCCS Institute of Neurological Sciences, Ospedale Bellaria, Bologna, Italy (R.A.); Department of Biomedical and Neuromotor Science, University of Bologna, Section of Anatomic Pathology M. Malpighi-Bellaria Hospital, Bologna, Italy (V.E.)
| |
Collapse
|
38
|
Helfer JL, Wen PY, Blakeley J, Gilbert MR, Armstrong TS. Report of the Jumpstarting Brain Tumor Drug Development Coalition and FDA clinical trials clinical outcome assessment endpoints workshop (October 15, 2014, Bethesda MD). Neuro Oncol 2016; 18 Suppl 2:ii26-ii36. [PMID: 26989130 PMCID: PMC4795997 DOI: 10.1093/neuonc/nov270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Abstract
On October 15, 2014, a workshop was held on the use of clinical outcome assessments in clinical trials for high-grade glioma of the brain. This workshop was sponsored by the Jumpstarting Brain Tumor Drug Development Coalition, consisting of the National Brain Tumor Society, the Society for Neuro-Oncology, the Musella Foundation for Brain Tumor Research and Information, and Accelerate Brain Cancer Cure. It was planned and carried out with participation from the US Food and Drug Administration. The workshop also included stakeholders from all aspects of the brain tumor community, including clinicians, researchers, industry, clinical research organizations, patients and patient advocates, and the National Cancer Institute. This report summarizes the presentations and discussions of that workshop and the proposals that emerged to move the field forward and toward greater inclusion of these endpoints in future clinical trials for high-grade gliomas.
Collapse
Affiliation(s)
- Jennifer L Helfer
- National Brain Tumor Society, Newton, Massachusetts (J.L.H.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Johns Hopkins University, Brain Cancer Program, Baltimore, Maryland (J.B.); Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland (M.R.G.); The University of Texas Health Science Center at Houston, School of Nursing, Department of Family Health, Houston, Texas (T.S.A.)
| | - Patrick Y Wen
- National Brain Tumor Society, Newton, Massachusetts (J.L.H.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Johns Hopkins University, Brain Cancer Program, Baltimore, Maryland (J.B.); Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland (M.R.G.); The University of Texas Health Science Center at Houston, School of Nursing, Department of Family Health, Houston, Texas (T.S.A.)
| | - Jaishri Blakeley
- National Brain Tumor Society, Newton, Massachusetts (J.L.H.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Johns Hopkins University, Brain Cancer Program, Baltimore, Maryland (J.B.); Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland (M.R.G.); The University of Texas Health Science Center at Houston, School of Nursing, Department of Family Health, Houston, Texas (T.S.A.)
| | - Mark R Gilbert
- National Brain Tumor Society, Newton, Massachusetts (J.L.H.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Johns Hopkins University, Brain Cancer Program, Baltimore, Maryland (J.B.); Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland (M.R.G.); The University of Texas Health Science Center at Houston, School of Nursing, Department of Family Health, Houston, Texas (T.S.A.)
| | - Terri S Armstrong
- National Brain Tumor Society, Newton, Massachusetts (J.L.H.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Johns Hopkins University, Brain Cancer Program, Baltimore, Maryland (J.B.); Center for Cancer Research, Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland (M.R.G.); The University of Texas Health Science Center at Houston, School of Nursing, Department of Family Health, Houston, Texas (T.S.A.)
| |
Collapse
|
39
|
Dirven L, Koekkoek JA, Reijneveld JC, Taphoorn MJ. Health-related quality of life in brain tumor patients: as an endpoint in clinical trials and its value in clinical care. ACTA ACUST UNITED AC 2016. [DOI: 10.1080/23809000.2016.1136793] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
40
|
Wang J, Yan Z, Liu X, Che S, Wang C, Yao W. Alpinetin targets glioma stem cells by suppressing Notch pathway. Tumour Biol 2016; 37:9243-8. [DOI: 10.1007/s13277-016-4827-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/08/2016] [Indexed: 12/26/2022] Open
|
41
|
LIU JIN, ZHANG KE, ZHEN YONGZHAN, WEI JIE, HU GANG, GAO JUNLING, TIAN YANXIA, LIN YAJUN. Antitumor activity of rhein lysinate against human glioma U87 cells in vitro and in vivo. Oncol Rep 2015; 35:1711-7. [DOI: 10.3892/or.2015.4518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/12/2015] [Indexed: 11/06/2022] Open
|
42
|
Wen Q, Jalilian L, Lupo JM, Li Y, Roy R, Molinaro AM, Chang SM, Prados M, Butowski N, Clarke J, Nelson SJ. Association of Diffusion and Anatomic Imaging Parameters with Survival for Patients with Newly Diagnosed Glioblastoma Participating in Two Different Clinical Trials. Transl Oncol 2015; 8:446-55. [PMID: 26692525 PMCID: PMC4700297 DOI: 10.1016/j.tranon.2015.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/14/2015] [Accepted: 10/02/2015] [Indexed: 01/20/2023] Open
Abstract
PURPOSE: To evaluate the time course and association with survival of anatomic lesion volumes and diffusion imaging parameters for patients with newly diagnosed glioblastoma who were treated with radiation and concurrently with either temozolomide and enzastaurin (TMZ+enza cohort) or temozolomide, erlotonib, and bevaciumab (TMZ+erl+bev cohort). MATERIALS AND METHODS: Regions of interest corresponding to the contrast-enhancing and hyperintense lesions on T2-weighted images were generated. Diffusion-weighted images were processed to provide maps of apparent diffusion coefficient, fractional anisotropy, and longitudinal and radial eigenvalues. Histograms of diffusion values were generated and summary statistics calculated. Cox proportional hazards models were employed to assess the association of representative imaging parameters with survival with adjustments for age, Karnofsky performance status, and extent of resection. RESULTS: Although progression-free survival was significantly longer for the TMZ+erl+bev cohort (12.8 vs 7.3 months), there was no significant difference in overall survival between the two populations (17.0 vs 17.8 months). The median contrast-enhancing lesion volumes decreased from 6.3 to 1.9 cm3 from baseline to the postradiotherapy scan for patients in the TMZ+enza cohort and from 2.8 to 0.9cm3 for the TMZ+erl+bev cohort. Changes in the T2 lesion volumes were only significant for the latter cohort (26.5 to 11.9 cm3). The median apparent diffusion coefficient and related diffusion parameters were significantly increased for the TMZ+enza cohort (1054 to 1225 μm2/s). More of the anatomic parameters were associated with survival for the TMZ+enza cohort, whereas more diffusion parameters were associated with survival for the TMZ+erl+bev cohort. CONCLUSION: The early changes in anatomic and diffusion imaging parameters and their association with survival reflected differences in the mechanisms of action of the treatments that were being given. This suggests that integrating diffusion metrics and anatomic lesion volumes into the Response Assessment in Neuro-Oncology criteria would assist in interpreting treatment-induced changes and predicting outcome in patients with newly diagnosed glioblastoma who are receiving such combination treatments.
Collapse
Affiliation(s)
- Qiuting Wen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States
| | - Laleh Jalilian
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States
| | - Ritu Roy
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA United States
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA United States
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA United States; UCSF/UCB Joint Graduate Group in Bioengineering, University of California, San Francisco, San Francisco, CA United States; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA United States.
| |
Collapse
|
43
|
Chang SM, Wen PY, Vogelbaum MA, Macdonald DR, van den Bent MJ. Response Assessment in Neuro-Oncology (RANO): more than imaging criteria for malignant glioma. Neurooncol Pract 2015; 2:205-209. [PMID: 31386074 PMCID: PMC6664617 DOI: 10.1093/nop/npv037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Indexed: 12/12/2022] Open
Abstract
The introduction of antiangiogenic therapies for the treatment of malignant glioma and the effect of these agents on standard imaging studies were the stimuli for forming a small group of investigators to critically evaluate the limitations of the Macdonald criteria in assessing response to treatment. The initial goal of this group was to highlight the challenges in accurately determining the efficacy of therapeutic interventions for malignant glioma and to develop new criteria that could be implemented in clinical care as well as in the design and conduct of clinical trials. This initial Response Assessment in Neuro-Oncology (RANO) effort started in 2008 and over the last 7 years, it has expanded to include a critical review of response assessment across several tumor types as well as endpoint selection and trial design to improve outcome criteria for neuro-oncological trials. In this paper, we review the overarching principles of the RANO initiative and the efforts to date. We also highlight the diverse and expanding efforts of the multidisciplinary groups of investigators who have volunteered their time as part of this endeavor.
Collapse
Affiliation(s)
- Susan M. Chang
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California (S.M.C.); Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (P.Y.W.); Rose Ella Burkhardt Brain Tumor and NeuroOncology Center, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (M.A.V.); Medical Oncology, London Regional Cancer Program, Western University, London, ON, Canada (D.R.M.); Dept Neuro-oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands (M.J.v.d.B.)
| | - Patrick Y. Wen
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California (S.M.C.); Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (P.Y.W.); Rose Ella Burkhardt Brain Tumor and NeuroOncology Center, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (M.A.V.); Medical Oncology, London Regional Cancer Program, Western University, London, ON, Canada (D.R.M.); Dept Neuro-oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands (M.J.v.d.B.)
| | - Michael A. Vogelbaum
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California (S.M.C.); Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (P.Y.W.); Rose Ella Burkhardt Brain Tumor and NeuroOncology Center, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (M.A.V.); Medical Oncology, London Regional Cancer Program, Western University, London, ON, Canada (D.R.M.); Dept Neuro-oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands (M.J.v.d.B.)
| | - David R. Macdonald
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California (S.M.C.); Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (P.Y.W.); Rose Ella Burkhardt Brain Tumor and NeuroOncology Center, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (M.A.V.); Medical Oncology, London Regional Cancer Program, Western University, London, ON, Canada (D.R.M.); Dept Neuro-oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands (M.J.v.d.B.)
| | - Martin J. van den Bent
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California (S.M.C.); Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (P.Y.W.); Rose Ella Burkhardt Brain Tumor and NeuroOncology Center, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (M.A.V.); Medical Oncology, London Regional Cancer Program, Western University, London, ON, Canada (D.R.M.); Dept Neuro-oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands (M.J.v.d.B.)
| |
Collapse
|
44
|
Affiliation(s)
- Sanne B Schagen
- The Netherlands Cancer Institute, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
| | - Jeffrey S Wefel
- The Netherlands Cancer Institute, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
| |
Collapse
|
45
|
Yang D. Standardized MRI assessment of high-grade glioma response: a review of the essential elements and pitfalls of the RANO criteria. Neurooncol Pract 2015; 3:59-67. [PMID: 31579522 DOI: 10.1093/nop/npv023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Indexed: 01/01/2023] Open
Abstract
Accurately evaluating response in the treatment of high-grade gliomas presents considerable challenges. This review looks at the advancements made in response criteria while critically outlining remaining weaknesses, and directs our vision toward promising endpoints to come. The 2010 guidelines from the Response Assessment in Neuro-Oncology (RANO) working group have enhanced interpretation of clinical trials involving novel treatments for high-grade glioma. Yet, while the criteria are considered clinically applicable to high-grade glioma trials, as well as reasonably accurate and reproducible, RANO lacks sufficient detail for consistent implementation in certain aspects and leaves some issues from the original Macdonald guidelines unresolved. To provide the most accurate assessment of response to therapeutic intervention currently possible, it is essential that trial oncologists and radiologists not only have a solid understanding of RANO guidelines, but also proper insight into the inherent limitations of the criteria. With the expectation of improved data collection as a standard, the author anticipates that the next high-grade glioma response criteria updates will incorporate advanced MRI methods and quantitative tumor volume measurements, availing a more accurate interpretation of response in the future.
Collapse
Affiliation(s)
- Dewen Yang
- ICON Medical Imaging, 2800 Kelly Road, Warrington, PA 18976
| |
Collapse
|
46
|
Gilbert MR, Armstrong TS, Pope WB, van den Bent MJ, Wen PY. Facing the future of brain tumor clinical research. Clin Cancer Res 2015; 20:5591-600. [PMID: 25398842 DOI: 10.1158/1078-0432.ccr-14-0835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This edition of CCR Focus provides critical reviews of several important areas in the field, including the application of findings from genomic investigations of brain tumors to improve diagnosis, clinical trial design, and ultimately optimizing individual patient treatment. Another article is a critical review provided by experts in the field that discusses the recent clinical trials using angiogenesis inhibitors, possible explanations for the results, and how to move forward. There is a concise discussion of the application of immunotherapy to brain tumors by key investigators in this field, reflecting the potential opportunities as well as the disease-specific challenges. Finally, leading pediatric brain tumor investigators provide an overview of the field and insights about the recent seminal discoveries in two pediatric brain tumors, supporting the paradigm that laboratory investigations lead to more precise diagnosis, prognosis, and ultimately better treatment. Herein, an overview of the recent advances and challenges in the area of clinical and translational brain tumor research is provided to set the stage for the contributions that follow.
Collapse
Affiliation(s)
- Mark R Gilbert
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Terri S Armstrong
- University of Texas Health Science Center School of Nursing and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Whitney B Pope
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | |
Collapse
|
47
|
Ellingson BM, Wen PY, van den Bent MJ, Cloughesy TF. Pros and cons of current brain tumor imaging. Neuro Oncol 2015; 16 Suppl 7:vii2-11. [PMID: 25313235 DOI: 10.1093/neuonc/nou224] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Over the past 20 years, very few agents have been approved for the treatment of brain tumors. Recent studies have highlighted some of the challenges in assessing activity in novel agents for the treatment of brain tumors. This paper reviews some of the key challenges related to assessment of tumor response to therapy in adult high-grade gliomas and discusses the strengths and limitations of imaging-based endpoints. Although overall survival is considered the "gold standard" endpoint in the field of oncology, progression-free survival and response rate are endpoints that hold great value in neuro-oncology. Particular focus is given to advancements made since the January 2006 Brain Tumor Endpoints Workshop, including the development of Response Assessment in Neuro-Oncology criteria, the value of T2/fluid-attenuated inversion recovery, use of objective response rates and progression-free survival in clinical trials, and the evaluation of pseudoprogression, pseudoresponse, and inflammatory response in radiographic images.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Patrick Y Wen
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Martin J van den Bent
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| | - Timothy F Cloughesy
- Department of Radiological Sciences (B.M.E.), Department of Biomedical Physics, David Geffen School of Medicine at UCLA (B.M.E.); Department of Bioengineering, Henry Samueli School of Engineering and Applied Science at UCLA (B.M.E.); Brain Research Institute, David Geffen School of Medicine at UCLA (B.M.E., T.F.C.); UCLA Neuro-Oncology Program, David Geffen School of Medicine at UCLA, Los Angeles, California (B.M.E., T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, Massachusetts (P.Y.W.); Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands (M.J.v.d.B.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (T.F.C.)
| |
Collapse
|
48
|
Xie K, Liu CY, Hasso AN, Crow RW. Visual field changes as an early indicator of glioblastoma multiforme progression: two cases of functional vision changes before MRI detection. Clin Ophthalmol 2015; 9:1041-7. [PMID: 26089636 PMCID: PMC4468993 DOI: 10.2147/opth.s79723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma multiforme is an aggressive tumor associated with a high rate of recurrence even after maximal therapy. In a disease with poor prognosis and rapid deterioration, early detection of tumor progression is necessary to make timely treatment decisions or to initiate end of life care. We identify two cases where Humphrey visual field testing predated magnetic resonance imaging and positron emission tomography findings of tumor progression by months in glioblastoma multiforme. New or worsening visual field defects may indicate signs of tumor progression in glioblastoma multiforme and should prompt further investigation.
Collapse
Affiliation(s)
- Kate Xie
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, USA
| | - Catherine Y Liu
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, USA
| | - Anton N Hasso
- Department of Radiological Sciences, University of California Irvine Medical Center, Orange, CA, USA
| | - Robert Wade Crow
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
49
|
Abstract
Glioblastoma (GBM) is the most common adult primary brain neoplasm. Despite advances in treatment, GBM continues to be associated with considerable morbidity and mortality as compared with other malignancies. Standard treatment for GBM results in survival of 12.9 months (95% CI: 12.3-13.7 months) with a median progression-free survival of 7.2 months (95% CI: 6.4-8.2 months) in a modern GBM cohort. These aggressive tumors recur and treatment for recurrent GBM continues to have very poor outcomes. Prior to the use of bevacizumab, monoclonal antibody to VEGF, 6-month progression-free survival in clinical trials for recurrent GBM ranged from 9 to 15%. Trials utilizing bevacizumab and its subsequent US FDA approval have given more hope to recurrent GBM and this concise review discusses bevacizumab in recurrent GBM. This review focuses on time-to-event outcomes (overall survival, progression-free survival and 6-month progression-free survival) in clinical trials utilizing bevacizumab for the treatment of recurrent GBM. For this review, we have chosen to focus primarily on Phase II clinical trials that have been published and available in the literature (PubMed). While we focused primarily on time-to-event variables, toxicity and safety of bevacizumab is very important and this agent can be associated with serious life-threatening toxicities. We have included a general section of toxicities but for a more lengthy review please see the excellent study by Odia and colleagues.
Collapse
Affiliation(s)
- Ashley Ghiaseddin
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, PO Box 3624, Durham, NC 27710, USA
| | | |
Collapse
|
50
|
Abstract
The treatment of glial brain tumors begins with surgery, and standard adjuvant treatment at the end of the past millennium for high-grade glioma and high-risk low-grade glioma was radiotherapy and chemotherapy was given at recurrence. However, over the past 10 years much has changed regarding the role of chemotherapy in gliomas and it is now clear that chemotherapy has a role in the treatment of almost all newly diagnosed diffuse gliomas (WHO grade II-IV). This is the result of several prospective studies that showed survival benefit after combined chemoradiotherapy with temozolomide in glioblastoma (WHO grade IV) or after procarbazine, CCNU (lomustine) and vincristine chemotherapy in diffuse low-grade (WHO grade II) and anaplastic (WHO grade III) glioma. The current standard of treatment for diffuse gliomas is described in this overview and in addition some attention is given to targeted therapies.
Collapse
Affiliation(s)
- Walter Taal
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Jacoline EC Bromberg
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Martin J van den Bent
- Department of Neurology/Neuro-Oncology, Erasmus MC Cancer Institute, Erasmus MC University Medical Center, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| |
Collapse
|