1
|
Seyhan AA. Circulating Liquid Biopsy Biomarkers in Glioblastoma: Advances and Challenges. Int J Mol Sci 2024; 25:7974. [PMID: 39063215 PMCID: PMC11277426 DOI: 10.3390/ijms25147974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Gliomas, particularly glioblastoma (GBM), represent the most prevalent and aggressive tumors of the central nervous system (CNS). Despite recent treatment advancements, patient survival rates remain low. The diagnosis of GBM traditionally relies on neuroimaging methods such as magnetic resonance imaging (MRI) or computed tomography (CT) scans and postoperative confirmation via histopathological and molecular analysis. Imaging techniques struggle to differentiate between tumor progression and treatment-related changes, leading to potential misinterpretation and treatment delays. Similarly, tissue biopsies, while informative, are invasive and not suitable for monitoring ongoing treatments. These challenges have led to the emergence of liquid biopsy, particularly through blood samples, as a promising alternative for GBM diagnosis and monitoring. Presently, blood and cerebrospinal fluid (CSF) sampling offers a minimally invasive means of obtaining tumor-related information to guide therapy. The idea that blood or any biofluid tests can be used to screen many cancer types has huge potential. Tumors release various components into the bloodstream or other biofluids, including cell-free nucleic acids such as microRNAs (miRNAs), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), proteins, extracellular vesicles (EVs) or exosomes, metabolites, and other factors. These factors have been shown to cross the blood-brain barrier (BBB), presenting an opportunity for the minimally invasive monitoring of GBM as well as for the real-time assessment of distinct genetic, epigenetic, transcriptomic, proteomic, and metabolomic changes associated with brain tumors. Despite their potential, the clinical utility of liquid biopsy-based circulating biomarkers is somewhat constrained by limitations such as the absence of standardized methodologies for blood or CSF collection, analyte extraction, analysis methods, and small cohort sizes. Additionally, tissue biopsies offer more precise insights into tumor morphology and the microenvironment. Therefore, the objective of a liquid biopsy should be to complement and enhance the diagnostic accuracy and monitoring of GBM patients by providing additional information alongside traditional tissue biopsies. Moreover, utilizing a combination of diverse biomarker types may enhance clinical effectiveness compared to solely relying on one biomarker category, potentially improving diagnostic sensitivity and specificity and addressing some of the existing limitations associated with liquid biomarkers for GBM. This review presents an overview of the latest research on circulating biomarkers found in GBM blood or CSF samples, discusses their potential as diagnostic, predictive, and prognostic indicators, and discusses associated challenges and future perspectives.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
2
|
Riviere-Cazaux C, Graser CJ, Warrington AE, Hoplin MD, Andersen KM, Malik N, Palmer EA, Carlstrom LP, Dasari S, Munoz-Casabella A, Ikram S, Ghadimi K, Himes BT, Jusue-Torres I, Sarkaria JN, Meyer FB, Van Gompel JJ, Kizilbash SH, Sener U, Michor F, Campian JL, Parney IF, Burns TC. The dynamic impact of location and resection on the glioma CSF proteome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.15.24307463. [PMID: 38798641 PMCID: PMC11118641 DOI: 10.1101/2024.05.15.24307463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
While serial sampling of glioma tissue is rarely performed prior to recurrence, cerebrospinal fluid (CSF) is an underutilized longitudinal source of candidate glioma biomarkers for understanding therapeutic impacts. However, the impact of key variables to consider in longitudinal CSF samples, including anatomical location and post-surgical changes, remains unknown. To that end, pre- versus post-resection intracranial CSF samples were obtained at early (1-16 days; n=20) or delayed (86-153 days; n=11) timepoints for patients with glioma. Paired lumbar-versus-intracranial glioma CSF samples were also obtained (n=14). Using aptamer-based proteomics, we identify significant differences in the CSF proteome between lumbar, subarachnoid, and ventricular CSF. Our analysis of serial intracranial CSF samples suggests the early potential for disease monitoring and evaluation of pharmacodynamic impact of targeted therapies. Importantly, we found that resection had a significant, evolving longitudinal impact on the CSF proteome. Proteomic data are provided with individual clinical annotations as a resource for the field. One Sentence Summary Glioma cerebrospinal fluid (CSF) accessed intra-operatively and longitudinally via devices can reveal impacts of treatment and anatomical location.
Collapse
|
3
|
Sabeghi P, Zarand P, Zargham S, Golestany B, Shariat A, Chang M, Yang E, Rajagopalan P, Phung DC, Gholamrezanezhad A. Advances in Neuro-Oncological Imaging: An Update on Diagnostic Approach to Brain Tumors. Cancers (Basel) 2024; 16:576. [PMID: 38339327 PMCID: PMC10854543 DOI: 10.3390/cancers16030576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
This study delineates the pivotal role of imaging within the field of neurology, emphasizing its significance in the diagnosis, prognostication, and evaluation of treatment responses for central nervous system (CNS) tumors. A comprehensive understanding of both the capabilities and limitations inherent in emerging imaging technologies is imperative for delivering a heightened level of personalized care to individuals with neuro-oncological conditions. Ongoing research in neuro-oncological imaging endeavors to rectify some limitations of radiological modalities, aiming to augment accuracy and efficacy in the management of brain tumors. This review is dedicated to the comparison and critical examination of the latest advancements in diverse imaging modalities employed in neuro-oncology. The objective is to investigate their respective impacts on diagnosis, cancer staging, prognosis, and post-treatment monitoring. By providing a comprehensive analysis of these modalities, this review aims to contribute to the collective knowledge in the field, fostering an informed approach to neuro-oncological care. In conclusion, the outlook for neuro-oncological imaging appears promising, and sustained exploration in this domain is anticipated to yield further breakthroughs, ultimately enhancing outcomes for individuals grappling with CNS tumors.
Collapse
Affiliation(s)
- Paniz Sabeghi
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Paniz Zarand
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Sina Zargham
- Department of Basic Science, California Northstate University College of Medicine, 9700 West Taron Drive, Elk Grove, CA 95757, USA;
| | - Batis Golestany
- Division of Biomedical Sciences, Riverside School of Medicine, University of California, 900 University Ave., Riverside, CA 92521, USA;
| | - Arya Shariat
- Kaiser Permanente Los Angeles Medical Center, 4867 W Sunset Blvd, Los Angeles, CA 90027, USA;
| | - Myles Chang
- Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, CA 90089, USA;
| | - Evan Yang
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Priya Rajagopalan
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Daniel Chang Phung
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California, 1500 San Pablo St., Los Angeles, CA 90033, USA; (P.S.); (E.Y.); (P.R.); (D.C.P.)
| |
Collapse
|
4
|
I Kh Almadhoun MK, Hattab AAW. Pediatric Glioblastoma Multiforme: A Challenging Case of Rapid Growth and Clinical Deterioration in an 11-Year-Old Female Patient. Cureus 2023; 15:e47697. [PMID: 38021881 PMCID: PMC10674094 DOI: 10.7759/cureus.47697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive primary brain tumor that primarily affects adults, with cases in children being extremely rare. Gross total resection with subsequent irradiation and temozolomide, currently delivering the greatest overall survival, is the mainstay of therapy for juvenile GBM. Maximal surgical excision of the visible tumor mass has been shown to have a positive prognostic effect, but radiation concerns for growing brains and inconsistent results from different chemotherapy regimens in pediatric GBM make treatment choices for young patients challenging. Here, we report a case of GBM in an 11-year-old female child who presented with a dramatic presentation of neurologic deficits and clinical worsening due to rapid tumor growth.
Collapse
|
5
|
Ren D, Zhuang X, Lv Y, Zhang Y, Xu J, Gao F, Chen D, Wang Y. Identification of Family with Sequence Similarity 110 Member C (FAM110C) as a Candidate Diagnostic and Prognostic Biomarker for Glioma. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:2117-2127. [PMID: 37899918 PMCID: PMC10612548 DOI: 10.18502/ijph.v52i10.13850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 10/31/2023]
Abstract
Background Gliomas are the most frequent and dangerous primary cerebral tumors. Therefore, there is a need to develop molecular targets for the diagnosis and treatment for glioma. Methods In September 2020, we retrieved the expression matrix of glioblastoma (GBM) sufferers and pertinent clinical data from the TCGA (The Cancer Genome Atlas) database. Prognostic differences between various families with sequence similarity 110 member C (FAM110C) expression groups were assessed by Kaplan-Meier with log-rank test. The R platform get used to assess the accuracy of FAM110C delivery in predicting the prognosis of PDAC using a time-dependent receptor operating characteristic (ROC) curve. The delivery level of FAM110C was determined by qRT-PCR and western blot. Gene set enrichment investigated possible mechanisms between different FAM110C expression groups in GBM (GSEA). The impact of FAM110C on glioma cell movement was discovered using migration test. The drug's gene-targeting impact was validated by the CCK8 test. Results A total of 173 GBM samples were obtained from the TCGA database, with 148 including information on IDH1 mutations and 151 containing information on overall survival. The mRNA expression level of FAM110C was greater in wild-type GBM, according to qRT-PCR data. The connection between FAM110C expression and Hallmark, GO, and KEGG pathway gene sets was investigated using GSEA software. We used migration test to assess the impact of FAM110C on glioma motility in order to confirm the findings of the GSEA analysis. Conclusion FAM110C might get used as a possible diagnostic and prognostic biomarker for wild-type GBM, and its inhibition could be used to prevention and treatment wild-type GBM.
Collapse
Affiliation(s)
- Deshuai Ren
- Department of Neurosurgery, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 162400, China
| | - Xiaoyu Zhuang
- Hospital Chief Office, Tailai People’s Hospital, Qiqihar 161000, China
| | - Yanxin Lv
- Basic Medicine Department, Qiqihar Medical University, Qiqihar 161006, China
| | - Yun Zhang
- Department of Neurosurgery, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 162400, China
| | - Jiazhi Xu
- Department of Neurosurgery, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 162400, China
| | - Fengquan Gao
- Department of Neurosurgery, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 162400, China
| | - Dagang Chen
- Department of Neurosurgery, the Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 162400, China
| | - Yu Wang
- Basic Medicine Department, Qiqihar Medical University, Qiqihar 161006, China
| |
Collapse
|
6
|
García A, Cámara JA, Boullosa AM, Gustà MF, Mondragón L, Schwartz S, Casals E, Abasolo I, Bastús NG, Puntes V. Nanoceria as Safe Contrast Agents for X-ray CT Imaging. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2208. [PMID: 37570527 PMCID: PMC10421217 DOI: 10.3390/nano13152208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
Cerium oxide nanoparticles (CeO2NPs) have exceptional catalytic properties, rendering them highly effective in removing excessive reactive oxygen species (ROS) from biological environments, which is crucial in safeguarding these environments against radiation-induced damage. Additionally, the Ce atom's high Z number makes it an ideal candidate for utilisation as an X-ray imaging contrast agent. We herein show how the injection of albumin-stabilised 5 nm CeO2NPs into mice revealed substantial enhancement in X-ray contrast, reaching up to a tenfold increase at significantly lower concentrations than commercial or other proposed contrast agents. Remarkably, these NPs exhibited prolonged residence time within the target organs. Thus, upon injection into the tail vein, they exhibited efficient uptake by the liver and spleen, with 85% of the injected dose (%ID) recovered after 7 days. In the case of intratumoral administration, 99% ID of CeO2NPs remained within the tumour throughout the 7-day observation period, allowing for observation of disease dynamics. Mass spectrometry (ICP-MS) elemental analysis confirmed X-ray CT imaging observations.
Collapse
Affiliation(s)
- Ana García
- Design and Pharmacokinetics of Nanoparticles, CIBBIM-Nanomedicine, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.G.); (L.M.)
| | - Juan Antonio Cámara
- Preclinical Imaging Platform, Vall d’Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain;
| | - Ana María Boullosa
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.M.B.); (I.A.)
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
| | - Muriel F. Gustà
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
| | - Laura Mondragón
- Design and Pharmacokinetics of Nanoparticles, CIBBIM-Nanomedicine, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.G.); (L.M.)
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
| | - Simó Schwartz
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.M.B.); (I.A.)
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Eudald Casals
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China;
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.M.B.); (I.A.)
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Neus G. Bastús
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
| | - Víctor Puntes
- Design and Pharmacokinetics of Nanoparticles, CIBBIM-Nanomedicine, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.G.); (L.M.)
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain; (M.F.G.); (N.G.B.)
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
- Institut Català de Recerca i Estudis Avançats, (ICREA), P. Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
7
|
Qi D, Li J, Quarles CC, Fonkem E, Wu E. Assessment and prediction of glioblastoma therapy response: challenges and opportunities. Brain 2023; 146:1281-1298. [PMID: 36445396 PMCID: PMC10319779 DOI: 10.1093/brain/awac450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma is the most aggressive type of primary adult brain tumour. The median survival of patients with glioblastoma remains approximately 15 months, and the 5-year survival rate is <10%. Current treatment options are limited, and the standard of care has remained relatively constant since 2011. Over the last decade, a range of different treatment regimens have been investigated with very limited success. Tumour recurrence is almost inevitable with the current treatment strategies, as glioblastoma tumours are highly heterogeneous and invasive. Additionally, another challenging issue facing patients with glioblastoma is how to distinguish between tumour progression and treatment effects, especially when relying on routine diagnostic imaging techniques in the clinic. The specificity of routine imaging for identifying tumour progression early or in a timely manner is poor due to the appearance similarity of post-treatment effects. Here, we concisely describe the current status and challenges in the assessment and early prediction of therapy response and the early detection of tumour progression or recurrence. We also summarize and discuss studies of advanced approaches such as quantitative imaging, liquid biomarker discovery and machine intelligence that hold exceptional potential to aid in the therapy monitoring of this malignancy and early prediction of therapy response, which may decisively transform the conventional detection methods in the era of precision medicine.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Jing Li
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - C Chad Quarles
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ekokobe Fonkem
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- Department of Oncology and LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
8
|
Luna LP, Ahmed A, Daftaribesheli L, Deng F, Intrapiromkul J, Lanzman BA, Yedavalli V. Arterial spin labeling clinical applications for brain tumors and tumor treatment complications: A comprehensive case-based review. Neuroradiol J 2023; 36:129-141. [PMID: 35815750 PMCID: PMC10034709 DOI: 10.1177/19714009221114444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Arterial spin labeling (ASL) is a noninvasive neuroimaging technique that allows for quantifying cerebral blood flow without intravenous contrast. Various neurovascular disorders and tumors have cerebral blood flow alterations. Identifying these perfusion changes through ASL can aid in the diagnosis, especially in entities with normal structural imaging. In addition, complications of tumor treatment and tumor progression can also be monitored using ASL. In this case-based review, we demonstrate the clinical applications of ASL in diagnosing and monitoring brain tumors and treatment complications.
Collapse
Affiliation(s)
- Licia P Luna
- Russell H. Morgan Department of
Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MA, USA
| | - Amara Ahmed
- Florida State University College of
Medicine, Tallahassee, FL, USA
| | - Laleh Daftaribesheli
- Russell H. Morgan Department of
Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MA, USA
| | - Francis Deng
- Massachusetts General Hospital and
Harvard Medical School, Boston, MA, USA
| | - Jarunee Intrapiromkul
- Russell H. Morgan Department of
Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MA, USA
| | - Bryan A Lanzman
- Department of Radiology, Stanford University, California, USA
| | - Vivek Yedavalli
- Russell H. Morgan Department of
Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MA, USA
| |
Collapse
|
9
|
Hormuth DA, Farhat M, Christenson C, Curl B, Chad Quarles C, Chung C, Yankeelov TE. Opportunities for improving brain cancer treatment outcomes through imaging-based mathematical modeling of the delivery of radiotherapy and immunotherapy. Adv Drug Deliv Rev 2022; 187:114367. [PMID: 35654212 PMCID: PMC11165420 DOI: 10.1016/j.addr.2022.114367] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/01/2022]
Abstract
Immunotherapy has become a fourth pillar in the treatment of brain tumors and, when combined with radiation therapy, may improve patient outcomes and reduce the neurotoxicity. As with other combination therapies, the identification of a treatment schedule that maximizes the synergistic effect of radiation- and immune-therapy is a fundamental challenge. Mechanism-based mathematical modeling is one promising approach to systematically investigate therapeutic combinations to maximize positive outcomes within a rigorous framework. However, successful clinical translation of model-generated combinations of treatment requires patient-specific data to allow the models to be meaningfully initialized and parameterized. Quantitative imaging techniques have emerged as a promising source of high quality, spatially and temporally resolved data for the development and validation of mathematical models. In this review, we will present approaches to personalize mechanism-based modeling frameworks with patient data, and then discuss how these techniques could be leveraged to improve brain cancer outcomes through patient-specific modeling and optimization of treatment strategies.
Collapse
Affiliation(s)
- David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Maguy Farhat
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Chase Christenson
- Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Brandon Curl
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - C Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Caroline Chung
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Oncology, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Imaging Physics, MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
10
|
Li AY, Iv M. Conventional and Advanced Imaging Techniques in Post-treatment Glioma Imaging. FRONTIERS IN RADIOLOGY 2022; 2:883293. [PMID: 37492665 PMCID: PMC10365131 DOI: 10.3389/fradi.2022.883293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 07/27/2023]
Abstract
Despite decades of advancement in the diagnosis and therapy of gliomas, the most malignant primary brain tumors, the overall survival rate is still dismal, and their post-treatment imaging appearance remains very challenging to interpret. Since the limitations of conventional magnetic resonance imaging (MRI) in the distinction between recurrence and treatment effect have been recognized, a variety of advanced MR and functional imaging techniques including diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), perfusion-weighted imaging (PWI), MR spectroscopy (MRS), as well as a variety of radiotracers for single photon emission computed tomography (SPECT) and positron emission tomography (PET) have been investigated for this indication along with voxel-based and more quantitative analytical methods in recent years. Machine learning and radiomics approaches in recent years have shown promise in distinguishing between recurrence and treatment effect as well as improving prognostication in a malignancy with a very short life expectancy. This review provides a comprehensive overview of the conventional and advanced imaging techniques with the potential to differentiate recurrence from treatment effect and includes updates in the state-of-the-art in advanced imaging with a brief overview of emerging experimental techniques. A series of representative cases are provided to illustrate the synthesis of conventional and advanced imaging with the clinical context which informs the radiologic evaluation of gliomas in the post-treatment setting.
Collapse
Affiliation(s)
- Anna Y. Li
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael Iv
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
11
|
Barca C, Foray C, Zinnhardt B, Winkeler A, Herrlinger U, Grauer OM, Jacobs AH. In Vivo Quantitative Imaging of Glioma Heterogeneity Employing Positron Emission Tomography. Cancers (Basel) 2022; 14:cancers14133139. [PMID: 35804911 PMCID: PMC9264799 DOI: 10.3390/cancers14133139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma is the most common primary brain tumor, highly aggressive by being proliferative, neovascularized and invasive, heavily infiltrated by immunosuppressive glioma-associated myeloid cells (GAMs), including glioma-associated microglia/macrophages (GAMM) and myeloid-derived suppressor cells (MDSCs). Quantifying GAMs by molecular imaging could support patient selection for GAMs-targeting immunotherapy, drug target engagement and further assessment of clinical response. Magnetic resonance imaging (MRI) and amino acid positron emission tomography (PET) are clinically established imaging methods informing on tumor size, localization and secondary phenomena but remain quite limited in defining tumor heterogeneity, a key feature of glioma resistance mechanisms. The combination of different imaging modalities improved the in vivo characterization of the tumor mass by defining functionally distinct tissues probably linked to tumor regression, progression and infiltration. In-depth image validation on tracer specificity, biological function and quantification is critical for clinical decision making. The current review provides a comprehensive overview of the relevant experimental and clinical data concerning the spatiotemporal relationship between tumor cells and GAMs using PET imaging, with a special interest in the combination of amino acid and translocator protein (TSPO) PET imaging to define heterogeneity and as therapy readouts.
Collapse
Affiliation(s)
- Cristina Barca
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Correspondence: (C.B.); (A.H.J.)
| | - Claudia Foray
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Biomarkers & Translational Technologies (BTT), Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Alexandra Winkeler
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, F-91401 Orsay, France;
| | - Ulrich Herrlinger
- Division of Clinical Neuro-Oncology, Department of Neurology, University Hospital Bonn, D-53105 Bonn, Germany;
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
| | - Oliver M. Grauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, D-48149 Münster, Germany;
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging (EIMI), University of Münster, D-48149 Münster, Germany; (C.F.); (B.Z.)
- Centre of Integrated Oncology (CIO), University Hospital Bonn, D-53127 Bonn, Germany
- Department of Geriatrics with Neurology, Johanniter Hospital, D-53113 Bonn, Germany
- Correspondence: (C.B.); (A.H.J.)
| |
Collapse
|
12
|
Bernstock JD, Gary SE, Klinger N, Valdes PA, Ibn Essayed W, Olsen HE, Chagoya G, Elsayed G, Yamashita D, Schuss P, Gessler FA, Peruzzi PP, Bag A, Friedman GK. Standard clinical approaches and emerging modalities for glioblastoma imaging. Neurooncol Adv 2022; 4:vdac080. [PMID: 35821676 PMCID: PMC9268747 DOI: 10.1093/noajnl/vdac080] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary adult intracranial malignancy and carries a dismal prognosis despite an aggressive multimodal treatment regimen that consists of surgical resection, radiation, and adjuvant chemotherapy. Radiographic evaluation, largely informed by magnetic resonance imaging (MRI), is a critical component of initial diagnosis, surgical planning, and post-treatment monitoring. However, conventional MRI does not provide information regarding tumor microvasculature, necrosis, or neoangiogenesis. In addition, traditional MRI imaging can be further confounded by treatment-related effects such as pseudoprogression, radiation necrosis, and/or pseudoresponse(s) that preclude clinicians from making fully informed decisions when structuring a therapeutic approach. A myriad of novel imaging modalities have been developed to address these deficits. Herein, we provide a clinically oriented review of standard techniques for imaging GBM and highlight emerging technologies utilized in disease characterization and therapeutic development.
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Sam E Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Neil Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Hannah E Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Patrick Schuss
- Department of Neurosurgery, Unfallkrankenhaus Berlin , Berlin, Germany
| | | | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital , Memphis, TN USA
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham , AL, USA
| |
Collapse
|
13
|
Gaudino S, Marziali G, Giordano C, Gigli R, Varcasia G, Magnani F, Chiesa S, Balducci M, Costantini AM, Della Pepa GM, Olivi A, Russo R, Colosimo C. Regorafenib in Glioblastoma Recurrence: How to Deal With MR Imaging Treatments Changes. FRONTIERS IN RADIOLOGY 2022; 1:790456. [PMID: 37492166 PMCID: PMC10365006 DOI: 10.3389/fradi.2021.790456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/29/2021] [Indexed: 07/27/2023]
Abstract
The treatment of recurrent high-grade gliomas remains a major challenge of daily neuro-oncology practice, and imaging findings of new therapies may be challenging. Regorafenib is a multi-kinase inhibitor that has recently been introduced into clinical practice to treat recurrent glioblastoma, bringing with it a novel panel of MRI imaging findings. On the basis of the few data in the literature and on our personal experience, we have identified the main MRI changes during regorafenib therapy, and then, we defined two different patterns, trying to create a simple summary line of the main changes of pathological tissue during therapy. We named these patterns, respectively, pattern A (less frequent, similar to classical progression disease) and pattern B (more frequent, with decreased diffusivity and decrease contrast-enhancement). We have also reported MR changes concerning signal intensity on T1-weighted and T2-weighted images, SWI, and perfusion imaging, derived from the literature (small series or case reports) and from our clinical experience. The clinical implication of these imaging modifications remains to be defined, taking into account that we are still at the dawn in the evaluation of such imaging modifications.
Collapse
Affiliation(s)
- Simona Gaudino
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
| | - Giammaria Marziali
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Carolina Giordano
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Riccardo Gigli
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Varcasia
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesca Magnani
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Silvia Chiesa
- Department of Diagnostic Imaging, Oncological Radiotherapy, and Hematology, UOC di Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mario Balducci
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy, and Hematology, UOC di Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandro Maria Costantini
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Maria Della Pepa
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, Rome, Italy
| | - Alessandro Olivi
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
- Institute of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University, Rome, Italy
| | - Rosellina Russo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Cesare Colosimo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Institute of Radiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore of Rome, Rome, Italy
| |
Collapse
|
14
|
Ye X, Schreck KC, Ozer BH, Grossman SA. High-grade glioma therapy: adding flexibility in trial design to improve patient outcomes. Expert Rev Anticancer Ther 2022; 22:275-287. [PMID: 35130447 DOI: 10.1080/14737140.2022.2038138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Outcomes for patients with high grade gliomas have changed little over the past thirty years. This realization prompted renewed efforts to increase flexibility in the design and conduct of clinical brain tumor trials. AREAS COVERED This manuscript reviews the development of clinical trial methods, challenges and considerations of flexible clinical trial designs, approaches to improve identification and testing of active agents for high grade gliomas, and evaluation of their delivery to the central nervous system. EXPERT OPINION Flexibility can be introduced in clinical trials in several ways. Flexible designs tout smaller sample sizes, adaptive modifications, fewer control arms, and inclusion of multiple arms in one study. Unfortunately, modifications in study designs cannot address two challenges that are largely responsible for the lack of progress in treating high grade gliomas: 1) the identification of active pharmaceutical agents and 2) the delivery of these agents to brain tumor tissue in therapeutic concentrations. To improve the outcomes of patients with high grade gliomas efforts must be focused on the pre-clinical screening of drugs for activity, the ability of these agents to achieve therapeutic concentrations in non-enhancing tumors, and a willingness to introduce novel compounds in minimally pre-treated patient populations.
Collapse
Affiliation(s)
- Xiaobu Ye
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Karisa C Schreck
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Byram H Ozer
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Stuart A Grossman
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| |
Collapse
|
15
|
Ak M, Toll SA, Hein KZ, Colen RR, Khatua S. Evolving Role and Translation of Radiomics and Radiogenomics in Adult and Pediatric Neuro-Oncology. AJNR Am J Neuroradiol 2021; 43:792-801. [PMID: 34649914 DOI: 10.3174/ajnr.a7297] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022]
Abstract
Exponential technologic advancements in imaging, high-performance computing, and artificial intelligence, in addition to increasing access to vast amounts of diverse data, have revolutionized the role of imaging in medicine. Radiomics is defined as a high-throughput feature-extraction method that unlocks microscale quantitative data hidden within standard-of-care medical imaging. Radiogenomics is defined as the linkage between imaging and genomics information. Multiple radiomics and radiogenomics studies performed on conventional and advanced neuro-oncology image modalities show that they have the potential to differentiate pseudoprogression from true progression, classify tumor subgroups, and predict recurrence, survival, and mutation status with high accuracy. In this article, we outline the technical steps involved in radiomics and radiogenomics analyses with the use of artificial intelligence methods and review current applications in adult and pediatric neuro-oncology.
Collapse
Affiliation(s)
- M Ak
- From the Department of Radiology (M.A., R.R.C.), University of Pittsburgh, Pittsburgh, Pennsylvania.,Hillman Cancer Center (M.A., R.R.C.), University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - S A Toll
- Department of Hematology-Oncology (S.A.T.), Children's Hospital of Michigan, Detroit, Michigan
| | - K Z Hein
- Department of Leukemia (K.Z.H.), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - R R Colen
- From the Department of Radiology (M.A., R.R.C.), University of Pittsburgh, Pittsburgh, Pennsylvania.,Hillman Cancer Center (M.A., R.R.C.), University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - S Khatua
- Department of Pediatric Hematology-Oncology (S.K.), Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
16
|
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: 11.5] [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
|
17
|
Zaccagna F, Grist JT, Quartuccio N, Riemer F, Fraioli F, Caracò C, Halsey R, Aldalilah Y, Cunningham CH, Massoud TF, Aloj L, Gallagher FA. Imaging and treatment of brain tumors through molecular targeting: Recent clinical advances. Eur J Radiol 2021; 142:109842. [PMID: 34274843 DOI: 10.1016/j.ejrad.2021.109842] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Molecular imaging techniques have rapidly progressed over recent decades providing unprecedented in vivo characterization of metabolic pathways and molecular biomarkers. Many of these new techniques have been successfully applied in the field of neuro-oncological imaging to probe tumor biology. Targeting specific signaling or metabolic pathways could help to address several unmet clinical needs that hamper the management of patients with brain tumors. This review aims to provide an overview of the recent advances in brain tumor imaging using molecular targeting with positron emission tomography and magnetic resonance imaging, as well as the role in patient management and possible therapeutic implications.
Collapse
Affiliation(s)
- Fulvio Zaccagna
- Division of Neuroimaging, Department of Medical Imaging, University of Toronto, Toronto, Canada.
| | - James T Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom; Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom; Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Palermo, Italy
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre, University of Bergen, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Corradina Caracò
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Richard Halsey
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Yazeed Aldalilah
- Institute of Nuclear Medicine, University College London, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom; Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Charles H Cunningham
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Tarik F Massoud
- Division of Neuroimaging and Neurointervention, Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Zaman N, Dass SS, DU Parcq P, Macmahon S, Gallagher L, Thompson L, Khorashad JS, LimbÄck-Stanic C. The KDR (VEGFR-2) Genetic Polymorphism Q472H and c-KIT Polymorphism M541L Are Associated With More Aggressive Behaviour in Astrocytic Gliomas. Cancer Genomics Proteomics 2021; 17:715-727. [PMID: 33099473 DOI: 10.21873/cgp.20226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND/AIM Better diagnostic and prognostic markers are required for a more accurate diagnosis and an earlier detection of glioma progression and for suggesting better treatment strategies. This retrospective study aimed to identify actionable gene variants to define potential markers of clinical significance. MATERIALS AND METHODS 56 glioblastomas (GBM) and 44 grade 2-3 astrocytomas were profiled with next generation sequencing (NGS) as part of routine diagnostic workup and bioinformatics analysis was used for the identification of variants. CD34 immunohistochemistry (IHC) was used to measure microvessel density (MVD) and Log-rank test to compare survival and progression in the presence or absence of these variants. RESULTS Bioinformatic analysis highlighted frequently occurring variants in genes involved in angiogenesis regulation (KDR, KIT, TP53 and PIK3CA), with the most common ones being KDR (rs1870377) and KIT (rs3822214). The KDR variant was associated with increased MVD and shorter survival in GBM. We did not observe any correlation between the KIT variant and MVD; however, there was an association with tumour grade. CONCLUSION This study highlights the role of single-nucleotide variants (SNVs) that may be considered non-pathogenic and suggests the prognostic significance for survival of KIT rs3822214 and KDR rs1870377 and potential importance in planning new treatment strategies for gliomas.
Collapse
Affiliation(s)
- Niyaz Zaman
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K
| | - Serena Santhana Dass
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K
| | - Persephone DU Parcq
- Department of Cell Pathology, Imperial College Healthcare NHS Trust, London, U.K
| | - Suzanne Macmahon
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Lewis Gallagher
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Lisa Thompson
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Jamshid S Khorashad
- Department of Immunology and Inflammation, Imperial College London, London, U.K
| | - Clara LimbÄck-Stanic
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K. .,Department of Cell Pathology, Imperial College Healthcare NHS Trust, London, U.K
| |
Collapse
|
19
|
Nakata J, Isohashi K, Oka Y, Nakajima H, Morimoto S, Fujiki F, Oji Y, Tsuboi A, Kumanogoh A, Hashimoto N, Hatazawa J, Sugiyama H. Imaging Assessment of Tumor Response in the Era of Immunotherapy. Diagnostics (Basel) 2021; 11:diagnostics11061041. [PMID: 34198874 PMCID: PMC8226723 DOI: 10.3390/diagnostics11061041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Assessment of tumor response during treatment is one of the most important purposes of imaging. Before the appearance of immunotherapy, response evaluation criteria in solid tumors (RECIST) and positron emission tomography response criteria in solid tumors (PERCIST) were, respectively, the established morphologic and metabolic response criteria, and cessation of treatment was recommended when progressive disease was detected according to these criteria. However, various types of immunotherapy have been developed over the past 20 years, which show novel false positive findings on images, as well as distinct response patterns from conventional therapies. Antitumor immune response itself causes 18F-fluorodeoxyglucose (FDG) uptake in tumor sites, known as "flare phenomenon", so that positron emission tomography using FDG can no longer accurately identify remaining tumors. Furthermore, tumors often initially increase, followed by stability or decrease resulting from immunotherapy, which is called "pseudoprogression", so that progressive disease cannot be confirmed by computed tomography or magnetic resonance imaging at a single time point. As a result, neither RECIST nor PERCIST can accurately predict the response to immunotherapy, and therefore several new response criteria fixed for immunotherapy have been proposed. However, these criteria are still controversial, and also require months for response confirmation. The establishment of optimal response criteria and the development of new imaging technologies other than FDG are therefore urgently needed. In this review, we summarize the false positive images and the revision of response criteria for each immunotherapy, in order to avoid discontinuation of a truly effective immunotherapy.
Collapse
Affiliation(s)
- Jun Nakata
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Correspondence: ; Tel.: +81-6-6879-3676; Fax: +81-6-6879-3677
| | - Kayako Isohashi
- Department of Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, Takatsuki City 596-8686, Osaka, Japan;
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| | - Yusuke Oji
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (S.M.); (A.T.)
| | - Atsushi Kumanogoh
- Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City 565-0871, Osaka, Japan;
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto City 602-8566, Kyoto, Japan;
| | - Jun Hatazawa
- Department of Research Center for Nuclear Physics, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan;
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City 565-0871, Osaka, Japan; (H.N.); (F.F.); (H.S.)
| |
Collapse
|
20
|
Batsios G, Taglang C, Cao P, Gillespie AM, Najac C, Subramani E, Wilson DM, Flavell RR, Larson PEZ, Ronen SM, Viswanath P. Imaging 6-Phosphogluconolactonase Activity in Brain Tumors In Vivo Using Hyperpolarized δ-[1- 13C]gluconolactone. Front Oncol 2021; 11:589570. [PMID: 33937017 PMCID: PMC8082394 DOI: 10.3389/fonc.2021.589570] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION The pentose phosphate pathway (PPP) is essential for NADPH generation and redox homeostasis in cancer, including glioblastomas. However, the precise contribution to redox and tumor proliferation of the second PPP enzyme 6-phosphogluconolactonase (PGLS), which converts 6-phospho-δ-gluconolactone to 6-phosphogluconate (6PG), remains unclear. Furthermore, non-invasive methods of assessing PGLS activity are lacking. The goal of this study was to examine the role of PGLS in glioblastomas and assess the utility of probing PGLS activity using hyperpolarized δ-[1-13C]gluconolactone for non-invasive imaging. METHODS To interrogate the function of PGLS in redox, PGLS expression was silenced in U87, U251 and GS2 glioblastoma cells by RNA interference and levels of NADPH and reduced glutathione (GSH) measured. Clonogenicity assays were used to assess the effect of PGLS silencing on glioblastoma proliferation. Hyperpolarized δ-[1-13C]gluconolactone metabolism to 6PG was assessed in live cells treated with the chemotherapeutic agent temozolomide (TMZ) or with vehicle control. 13C 2D echo-planar spectroscopic imaging (EPSI) studies of hyperpolarized δ-[1-13C]gluconolactone metabolism were performed on rats bearing orthotopic glioblastoma tumors or tumor-free controls on a 3T spectrometer. Longitudinal 2D EPSI studies of hyperpolarized δ-[1-13C]gluconolactone metabolism and T2-weighted magnetic resonance imaging (MRI) were performed in rats bearing orthotopic U251 tumors following treatment with TMZ to examine the ability of hyperpolarized δ-[1-13C]gluconolactone to report on treatment response. RESULTS PGLS knockdown downregulated NADPH and GSH, elevated oxidative stress and inhibited clonogenicity in all models. Conversely, PGLS expression and activity and steady-state NADPH and GSH were higher in tumor tissues from rats bearing orthotopic glioblastoma xenografts relative to contralateral brain and tumor-free brain. Importantly, [1-13C]6PG production from hyperpolarized δ-[1-13C]gluconolactone was observed in live glioblastoma cells and was significantly reduced by treatment with TMZ. Furthermore, hyperpolarized δ-[1-13C]gluconolactone metabolism to [1-13C]6PG could differentiate tumor from contralateral normal brain in vivo. Notably, TMZ significantly reduced 6PG production from hyperpolarized δ-[1-13C]gluconolactone at an early timepoint prior to volumetric alterations as assessed by anatomical imaging. CONCLUSIONS Collectively, we have, for the first time, identified a role for PGLS activity in glioblastoma proliferation and validated the utility of probing PGLS activity using hyperpolarized δ-[1-13C]gluconolactone for non-invasive in vivo imaging of glioblastomas and their response to therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sabrina M. Ronen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, United States
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, United States
| |
Collapse
|
21
|
Kavya S, Reghu R. An Overview of High-grade Glioma: Current and Emerging Treatment Approaches. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666200721155514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High grade glioma is one of the severe form of tumour that progresses in the glial cells
of the brain and spinal cord. Age, gender, exposure to infections, race, ethnicity, viruses and allergens,
environmental carcinogens, diet, head injury or trauma and ionizing radiation may report
with increased glioma risk. Headache, seizure mainly generalized tonic-clonic seizure, memory
loss and altered sensorium are considered as common symptoms of glioma. Magnetic Resonance
Imaging (MRI), CT scans, neurological examinations and biopsy are considered as the diagnostic
option for glioma. Treatment for glioma mainly depended upon the tumour progression, malignancy,
cell type, age, location of tumour growth and anatomic structure. The standard treatment includes
surgery, radiation therapy and chemotherapy. Temozolomide is usually prescribed at a
dosage of 75 mg/m2 and began in combination with radiation therapy and continued daily. The primary
indicator of hepatotoxicity is the elevation of the liver profiles, i.e. the changes in any of the
liver panels may be considered to be hepatotoxic. Serum glutamic oxaloacetic transaminase (SGOT),
Serum Glutamic Pyruvic Transaminase (SGPT), Alkaline phosphatase (ALP) are rising panels
of the liver, which are elevated during toxicity. In some patients, albumin and globulin levels
may show variations. Treatment for glioma associated symptoms like seizures, depression anxiety
etc. are also mentioned along with supportive care for glioma. New trends in the treatment for glioma
are RINTEGA, an experimental immunotherapeutic agent and bevazizumab, a recombinant
monoclonal, a humanized antibody against the VEGF ligand [VEGF-A (vascular endothelial
growth factor)] in tumor cells.
Collapse
Affiliation(s)
- S.G. Kavya
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - R. Reghu
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| |
Collapse
|
22
|
The role of 11C-methionine PET in patients with negative diffusion-weighted magnetic resonance imaging: correlation with histology and molecular biomarkers in operated gliomas. Nucl Med Commun 2021; 41:696-705. [PMID: 32371671 DOI: 10.1097/mnm.0000000000001202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To compare 11C-methionine (11C-METH) PET with diffusion-weighted MRI (DWI-MRI) diagnostic accuracy and prognostic value in patients with glioma candidate to neurosurgery. METHODS We collected and analyzed data from 124 consecutive patients (n = 124) investigated during preoperative work-up. Both visual and semiquantitative parameters were utilized for image analysis. The reference standard was based on histopathology. The median follow-up was 14.3 months. RESULTS Overall, 47 high-grade gliomas (HGG) and 77 low-grade gliomas (LGG) were diagnosed. On visual assessment, sensitivity and specificity for differentiating HGG from LGG were 80.8 and 59.7% for DWI-MRI, versus 95.7 and 41.5% for 11C-METH PET, respectively. On semiquantitative analysis, the sensitivity, specificity, and area under the curve were 78.7, 71.4, and 80.4% for SUVmax, 78.7, 70.1, and 81.1% for SUVratio, and 74.5, 61, and 76.7% for MTB (metabolic tumor burden), respectively. In patients with negative DWI-MRI and IDH-wild type, SUVmax and SUVratio were higher compared to IDH-mutated (P = 0.025 and P = 0.01, respectively). In LGG, patients with 1p/19q codeletion showed higher SUVmax (P = 0.044). In all patients with negative DWI-MRI, median PFS was longer for SUVmax <3.9 (median not reached vs 34.2 months, P = 0.004), SUVratio <2.3 (median not reached vs 21.5 months, P < 0.001), and MTB <3.1 (median not reached vs 45.7 months, P = 0.05). In LGG patients with negative DWI-MRI, only SUVratio <2.3 and MTB <3.1 were associated with longer PFS (P = 0.016 and P = 0.024, respectively). CONCLUSION C-METH PET was found highly sensitive for glioma differentiation and molecular characterization. In DWI-negative patients, PET parameters correlated with molecular profile were associated with clinical outcome.
Collapse
|
23
|
The predictive value of absolute lymphocyte counts on tumor progression and pseudoprogression in patients with glioblastoma. BMC Cancer 2021; 21:285. [PMID: 33726710 PMCID: PMC7968315 DOI: 10.1186/s12885-021-08004-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/02/2021] [Indexed: 11/29/2022] Open
Abstract
Background Differentiating true glioblastoma multiforme (GBM) from pseudoprogression (PsP) remains a challenge with current standard magnetic resonance imaging (MRI). The objective of this study was to explore whether patients’ absolute lymphocyte count (ALC) levels can be utilized to predict true tumor progression and PsP. Methods Patients were considered eligible for the study if they had 1) GBM diagnosis, 2) a series of blood cell counts and clinical follow-ups, and 3) tumor progression documented by both MRI and pathology. Data analysis results include descriptive statistics, median (IQR) for continuous variables and count (%) for categorical variables, p values from Wilcoxon rank sum test or Fisher’s exact test for comparison, respectively, and Kaplan-Meier analysis for overall survival (OS). OS was defined as the time from patients’ second surgery to their time of death or last follow up if patients were still alive. Results 78 patients were included in this study. The median age was 56 years. Median ALC dropped 34.5% from baseline 1400 cells/mm3 to 917 cells/mm3 after completion of radiation therapy (RT) and temozolomide (TMZ). All study patients had undergone surgical biopsy upon MRI-documented progression. 37 had true tumor progression (47.44%) and 41 had pseudoprogression (52.56%). ALC before RT/TMZ, post RT/TMZ and at the time of MRI-documented progression did not show significant difference between patients with true progression and PsP. Although not statistically significant, this study found that patients with true progression had worse OS compared to those with PsP (Hazard Ratio [HR] 1.44, 95% CI 0.86–2.43, P = 0.178). This study also found that patients with high ALC (dichotomized by median) post-radiation had longer OS. Conclusion Our results indicate that ALC level in GBM patients before or after treatment does not have predictive value for true disease progression or pseudoprogression. Patients with true progression had worse OS compared to those who had pseudoprogression. A larger sample size that includes CD4 cell counts may be needed to evaluate the PsP predictive value of peripheral blood biomarkers.
Collapse
|
24
|
Tripathi SK, Kean R, Bongiorno E, Hooper DC, Jin YY, Wickstrom E, McCue PA, Thakur ML. Targeting VPAC1 Receptors for Imaging Glioblastoma. Mol Imaging Biol 2021; 22:293-302. [PMID: 31292914 DOI: 10.1007/s11307-019-01388-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 μCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 μCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-μm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.
Collapse
Affiliation(s)
- Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Bongiorno
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Douglas C Hooper
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiology, Laboratories of Radiopharmaceutical Research and Molecular Imaging, 1020 Locust Street, 359-JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
25
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Neuroimaging Clin N Am 2021; 31:103-120. [PMID: 33220823 DOI: 10.1016/j.nic.2020.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
26
|
Le Fèvre C, Constans JM, Chambrelant I, Antoni D, Bund C, Leroy-Freschini B, Schott R, Cebula H, Noël G. Pseudoprogression versus true progression in glioblastoma patients: A multiapproach literature review. Part 2 - Radiological features and metric markers. Crit Rev Oncol Hematol 2021; 159:103230. [PMID: 33515701 DOI: 10.1016/j.critrevonc.2021.103230] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/10/2021] [Accepted: 01/16/2021] [Indexed: 12/28/2022] Open
Abstract
After chemoradiotherapy for glioblastoma, pseudoprogression can occur and must be distinguished from true progression to correctly manage glioblastoma treatment and follow-up. Conventional treatment response assessment is evaluated via conventional MRI (contrast-enhanced T1-weighted and T2/FLAIR), which is unreliable. The emergence of advanced MRI techniques, MR spectroscopy, and PET tracers has improved pseudoprogression diagnostic accuracy. This review presents a literature review of the different imaging techniques and potential imaging biomarkers to differentiate pseudoprogression from true progression.
Collapse
Affiliation(s)
- Clara Le Fèvre
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Jean-Marc Constans
- Department of Radiology, Amiens-Picardie University Hospital, 1 rond-point du Professeur Christian Cabrol, 80054, Amiens Cedex 1, France.
| | - Isabelle Chambrelant
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Delphine Antoni
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Caroline Bund
- Department of Nuclear Medicine, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Benjamin Leroy-Freschini
- Department of Nuclear Medicine, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Roland Schott
- Departement of Medical Oncology, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| | - Hélène Cebula
- Departement of Neurosurgery, Hautepierre University Hospital, 1, avenue Molière, 67200, Strasbourg, France.
| | - Georges Noël
- Department of Radiotherapy, ICANS, Institut Cancérologie Strasbourg Europe, 17 rue Albert Calmette, 67200, Strasbourg Cedex, France.
| |
Collapse
|
27
|
Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [ 18F]DASA-23. Mol Imaging Biol 2021; 22:124-133. [PMID: 30989436 DOI: 10.1007/s11307-019-01353-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET). PROCEDURE U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6 days (n = 6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake. RESULTS Significant interactions were found between the treatment agent (n = 12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p = 0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p < 0. 0001) followed by irinotecan (p = 0. 0012), erlotinib (p = 0. 02), and 5-fluorouracil (p = 0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r = 0.44, p = 0.15). CONCLUSIONS These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.
Collapse
|
28
|
Yu G, Butler MK, Abdelmaksoud A, Pang Y, Su YT, Rae Z, Dadkhah K, Kelly MC, Song YK, Wei JS, Terabe M, Atony R, Mentges K, Theeler BJ, Penas-Prado M, Butman J, Camphausen K, Zaghloul KA, Nduom E, Quezado M, Aldape K, Armstrong TS, Gilbert MR, Gulley JL, Khan J, Wu J. Case Report: Single-Cell Transcriptomic Analysis of an Anaplastic Oligodendroglioma Post Immunotherapy. Front Oncol 2021; 10:601452. [PMID: 33520712 PMCID: PMC7841290 DOI: 10.3389/fonc.2020.601452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/20/2020] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common primary malignant brain tumor with a poor prognosis. Immune checkpoint inhibitors have been of great interest in investigation of glioma treatments. Here, we report single-cell transcriptomic analyses of two tumor areas from an oligodendroglioma taken from a patient who had multiple tumor recurrences, following several chemotherapies and radiation treatments. The patient subsequently received nivolumab and was considered have disease progression based on conventional diagnostic imaging after two cycles of treatment. He underwent a debulking surgical resection and pathological diagnosis was recurrent disease. During the surgery, tumor tissues were also collected from the enhancing and non-enhancing areas for a scRNAseq analysis to investigate the tumor microenvironment of these radiographically divergent areas. The scRNAseq analysis reveals a plethora of immune cells, suggesting that the increased mass observed on MRI may be partially a result of immune cell infiltration. The patient continued to receive immunotherapy after a short course of palliative radiation and remained free of disease progression for at least 12 months after the last surgery, suggesting a sustained response to immunotherapy. The scRNAseq analysis indicated that the radiological progression was in large part due to immune cell infiltrate and continued immunotherapy led to a positive clinical outcome in a patient who would have otherwise been admitted to hospice care with halting of immunotherapy. Our study demonstrates the potential of scRNAseq analyses in understanding the tumor microenvironment, which may assist the clinical decision-making process for challenging glioma cases following immunotherapy.
Collapse
Affiliation(s)
- Guangyang Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Madison K. Butler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Abdalla Abdelmaksoud
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ying Pang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yu-Ting Su
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zachary Rae
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Kimia Dadkhah
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Michael C. Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Institutes of Health, Bethesda, MD, United States
| | - Young K. Song
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jun S. Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Masaki Terabe
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ramya Atony
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kelly Mentges
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Brett J. Theeler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marta Penas-Prado
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - John Butman
- Diagnostic Radiology Department, The Clinical Center of the National Institutes of Health, Bethesda, MD, United States
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kareem A. Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Edjah Nduom
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Terri S. Armstrong
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mark R. Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
29
|
Viswanath P, Batsios G, Mukherjee J, Gillespie AM, Larson PEZ, Luchman HA, Phillips JJ, Costello JF, Pieper RO, Ronen SM. Non-invasive assessment of telomere maintenance mechanisms in brain tumors. Nat Commun 2021; 12:92. [PMID: 33397920 PMCID: PMC7782549 DOI: 10.1038/s41467-020-20312-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/27/2020] [Indexed: 01/29/2023] Open
Abstract
Telomere maintenance is a universal hallmark of cancer. Most tumors including low-grade oligodendrogliomas use telomerase reverse transcriptase (TERT) expression for telomere maintenance while astrocytomas use the alternative lengthening of telomeres (ALT) pathway. Although TERT and ALT are hallmarks of tumor proliferation and attractive therapeutic targets, translational methods of imaging TERT and ALT are lacking. Here we show that TERT and ALT are associated with unique 1H-magnetic resonance spectroscopy (MRS)-detectable metabolic signatures in genetically-engineered and patient-derived glioma models and patient biopsies. Importantly, we have leveraged this information to mechanistically validate hyperpolarized [1-13C]-alanine flux to pyruvate as an imaging biomarker of ALT status and hyperpolarized [1-13C]-alanine flux to lactate as an imaging biomarker of TERT status in low-grade gliomas. Collectively, we have identified metabolic biomarkers of TERT and ALT status that provide a way of integrating critical oncogenic information into non-invasive imaging modalities that can improve tumor diagnosis and treatment response monitoring.
Collapse
Affiliation(s)
- Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Joydeep Mukherjee
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Peder E Z Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - H Artee Luchman
- Department of Cell Biology and Anatomy, Arnie Charbonneau Cancer Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Joanna J Phillips
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Joseph F Costello
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Russell O Pieper
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
30
|
Winter SF, Loebel F, Loeffler J, Batchelor TT, Martinez-Lage M, Vajkoczy P, Dietrich J. Treatment-induced brain tissue necrosis: a clinical challenge in neuro-oncology. Neuro Oncol 2020; 21:1118-1130. [PMID: 30828724 DOI: 10.1093/neuonc/noz048] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/04/2018] [Accepted: 02/25/2019] [Indexed: 12/29/2022] Open
Abstract
Cancer therapy-induced adverse effects on the brain are a major challenge in neuro-oncology. Brain tissue necrosis (treatment necrosis [TN]) as a consequence of brain directed cancer therapy remains an insufficiently characterized condition with diagnostic and therapeutic difficulties and is frequently associated with significant patient morbidity. A better understanding of the underlying mechanisms, improvement of diagnostic tools, development of preventive strategies, and implementation of evidence-based therapeutic practices are pivotal to improve patient management. In this comprehensive review, we address existing challenges associated with current TN-related clinical and research practices and highlight unanswered questions and areas in need of further research with the ultimate goal to improve management of patients affected by this important neuro-oncological condition.
Collapse
Affiliation(s)
- Sebastian F Winter
- MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Charité‒Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Franziska Loebel
- Department of Neurosurgery, Charité‒Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jay Loeffler
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tracy T Batchelor
- MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Martinez-Lage
- C S Kubik Laboratory for Neuropathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité‒Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jorg Dietrich
- MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Predicting Survival in Glioblastoma Patients Using Diffusion MR Imaging Metrics-A Systematic Review. Cancers (Basel) 2020; 12:cancers12102858. [PMID: 33020420 PMCID: PMC7600641 DOI: 10.3390/cancers12102858] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Simple Summary An accurate survival analysis is crucial for disease management in glioblastoma (GBM) patients. Due to the ability of the diffusion MRI techniques of providing a quantitative assessment of GBM tumours, an ever-growing number of studies aimed at investigating the role of diffusion MRI metrics in survival prediction of GBM patients. Since the role of diffusion MRI in prediction and evaluation of survival outcomes has not been fully addressed and results are often controversial or unsatisfactory, we performed this systematic review in order to collect, summarize and evaluate all studies evaluating the role of diffusion MRI metrics in predicting survival in GBM patients. We found that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters. Abstract Despite advances in surgical and medical treatment of glioblastoma (GBM), the medium survival is about 15 months and varies significantly, with occasional longer survivors and individuals whose tumours show a significant response to therapy with respect to others. Diffusion MRI can provide a quantitative assessment of the intratumoral heterogeneity of GBM infiltration, which is of clinical significance for targeted surgery and therapy, and aimed at improving GBM patient survival. So, the aim of this systematic review is to assess the role of diffusion MRI metrics in predicting survival of patients with GBM. According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement, a systematic literature search was performed to identify original articles since 2010 that evaluated the association of diffusion MRI metrics with overall survival (OS) and progression-free survival (PFS). The quality of the included studies was evaluated using the QUIPS tool. A total of 52 articles were selected. The most examined metrics were associated with the standard Diffusion Weighted Imaging (DWI) (34 studies) and Diffusion Tensor Imaging (DTI) models (17 studies). Our findings showed that quantitative diffusion MRI metrics provide useful information for predicting survival outcomes in GBM patients, mainly in combination with other clinical and multimodality imaging parameters.
Collapse
|
32
|
Raza IJ, Tingate CA, Gkolia P, Romero L, Tee JW, Hunn MK. Blood Biomarkers of Glioma in Response Assessment Including Pseudoprogression and Other Treatment Effects: A Systematic Review. Front Oncol 2020; 10:1191. [PMID: 32923382 PMCID: PMC7456864 DOI: 10.3389/fonc.2020.01191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/12/2020] [Indexed: 12/21/2022] Open
Abstract
Imaging-based monitoring of disease burden in glioma patients is frequently confounded by treatment effects. Circulating biomarkers could theoretically augment imaging-based response monitoring. This systematic review aimed to present and evaluate evidence for differential expression and diagnostic accuracy of circulating biomarkers with respect to outcomes of tumor response, progression, stable disease, and treatment effects (pseudoprogression, radionecrosis, pseudoresponse, and pseudolesions) in patients undergoing treatment for World Health Organization grades II-IV diffuse astrocytic and oligodendroglial tumors. MEDLINE, EMBASE, Web Of Science, and SCOPUS databases were searched until August 18, 2019, for observational or diagnostic studies on multiple circulating biomarker types: extracellular vesicles, circulating nucleic acids, circulating tumor cells, circulating proteins, and metabolites, angiogenesis related cells, immune cells, and other cell lines. Methodological quality of included studies was assessed using an adapted Quality Assessment of Diagnostic Accuracy Studies-2 tool, and level of evidence (IA-IVD) for individual biomarkers was evaluated using an adapted framework from the National Comprehensive Cancer Network guidelines on evaluating tumor marker utility. Of 13,202 unique records, 58 studies met the inclusion criteria. One hundred thirty-three distinct biomarkers were identified in a total of 1,853 patients across various treatment modalities. Fifteen markers for response, progression, or stable disease and five markers for pseudoprogression or radionecrosis reached level IB. No biomarkers reached level IA. Only five studies contained data for diagnostic accuracy measures. Overall methodological quality of included studies was low. While extensive data on biomarker dysregulation in varying response categories were reported, no biomarkers ready for clinical application were identified. Further assay refinement and evaluation in larger cohorts with diagnostic accuracy study designs are required. PROSPERO Registration: CRD42018110658.
Collapse
Affiliation(s)
- Istafa J Raza
- Department of Neurosurgery, The Alfred Hospital, Melbourne, VIC, Australia
| | - Campbell A Tingate
- Department of Neurosurgery, The Alfred Hospital, Melbourne, VIC, Australia
| | - Panagiota Gkolia
- Department of Neurosurgery, The Alfred Hospital, Melbourne, VIC, Australia
| | - Lorena Romero
- The Ian Potter Library, The Alfred Hospital, Melbourne, VIC, Australia
| | - Jin W Tee
- Department of Neurosurgery, The Alfred Hospital, Melbourne, VIC, Australia.,Department of Surgery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Martin K Hunn
- Department of Neurosurgery, The Alfred Hospital, Melbourne, VIC, Australia.,Department of Surgery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
33
|
The Role of RNA and DNA Aptamers in Glioblastoma Diagnosis and Therapy: A Systematic Review of the Literature. Cancers (Basel) 2020; 12:cancers12082173. [PMID: 32764266 PMCID: PMC7463716 DOI: 10.3390/cancers12082173] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor of the central nervous system in adults. Despite advances in surgical and medical neuro-oncology, the median survival is about 15 months. For this reason, initial diagnosis, prognosis, and targeted therapy of GBM represent very attractive areas of study. Aptamers are short three-dimensional structures of single-stranded nucleic acids (RNA or DNA), identified by an in vitro process, named systematic evolution of ligands by exponential enrichment (SELEX), starting from a partially random oligonucleotide library. They bind to a molecular target with high affinity and specificity and can be easily modified to optimize binding affinity and selectivity. Thanks to their properties (low immunogenicity and toxicity, long stability, and low production variability), a large number of aptamers have been selected against GBM biomarkers and provide specific imaging agents and therapeutics to improve the diagnosis and treatment of GBM. However, the use of aptamers in GBM diagnosis and treatment still represents an underdeveloped topic, mainly due to limited literature in the research world. On these bases, we performed a systematic review aimed at summarizing current knowledge on the new promising DNA and RNA aptamer-based molecules for GBM diagnosis and treatment. Thirty-eight studies from 2000 were included and investigated. Seventeen involved the use of aptamers for GBM diagnosis and 21 for GBM therapy. Our findings showed that a number of DNA and RNA aptamers are promising diagnostic and therapeutic tools for GBM management.
Collapse
|
34
|
Abdalla G, Hammam A, Anjari M, D'Arco DF, Bisdas DS. Glioma surveillance imaging: current strategies, shortcomings, challenges and outlook. BJR Open 2020; 2:20200009. [PMID: 33178973 PMCID: PMC7594888 DOI: 10.1259/bjro.20200009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/11/2023] Open
Abstract
Inaccurate assessment of surveillance imaging to assess response to glioma therapy may have life-changing consequences. Varied management plans including chemotherapy, radiotherapy or immunotherapy may all contribute to heterogeneous post-treatment appearances and the overlap between the morphological features of pseudoprogression, pseudoresponse and radiation necrosis can make their discrimination very challenging. Therefore, there has been a drive to develop objective strategies for post-treatment assessment of brain gliomas. This review discusses the most important of these approaches such as the RANO "Response Assessment in Neuro-Oncology", iRANO "Immunotherapy Response Assessment in Neuro-Oncology" and RAPNO "Response Assessment in Paediatric Neuro-Oncology" models. In addition to these systematic approaches for glioma surveillance, the relatively limited information provided by conventional imaging modalities alone has motivated the development of novel advanced magnetic resonance (MR) and metabolic imaging methods for further discrimination between viable tumour and treatment induced changes. Multiple clinical trials and meta-analyses have investigated the diagnostic performance of these novel techniques in the follow up of brain gliomas, including both single modality descriptive studies and comparative imaging assessment. In this manuscript, we review the literature and discuss the promises and pitfalls of frequently studied modalities in glioma surveillance imaging, including MR perfusion, MR diffusion and MR spectroscopy. In addition, we evaluate other promising MR techniques such as chemical exchange saturation transfer as well as fludeoxyglucose and non-FDG positron emission tomography techniques.
Collapse
Affiliation(s)
- Gehad Abdalla
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Ahmed Hammam
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Mustafa Anjari
- Department of Neuroradiology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Dr. Felice D'Arco
- Department of Neuroradiology, Great Ormond Street Hospital for Children, London, UK
| | | |
Collapse
|
35
|
Khan L, Soliman H, Sahgal A, Perry J, Xu W, Tsao MN. External beam radiation dose escalation for high grade glioma. Cochrane Database Syst Rev 2020; 5:CD011475. [PMID: 32437039 PMCID: PMC7389526 DOI: 10.1002/14651858.cd011475.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND This is an updated version of the original Cochrane Review published in Issue 8, 2016. High grade glioma (HGG) is a rapidly growing brain tumour in the supporting cells of the nervous system, with several subtypes such as glioblastoma (grade IV astrocytoma), anaplastic (grade III) astrocytoma and anaplastic (grade III) oligodendroglioma. Studies have investigated the best strategy to give radiation to people with HGG. Conventional fractionated radiotherapy involves giving a daily radiation dose (called a fraction) of 180 cGy to 200 cGy. Hypofractionated radiotherapy uses higher daily doses, which reduces the overall number of fractions and treatment time. Hyperfractionated radiotherapy which uses a lower daily dose with a greater number of fractions and multiple fractions per day to deliver a total dose at least equivalent to external beam daily conventionally fractionated radiotherapy in the same time frame. The aim is to reduce the potential for late toxicity. Accelerated radiotherapy (dose escalation) refers to the delivery of multiple fractions per day using daily doses of radiation consistent with external beam daily conventionally fractionated radiotherapy doses. The aim is to reduce the overall treatment time; typically, two or three fractions per day may be delivered with a six to eight hour gap between fractions. OBJECTIVES To assess the effects of postoperative external beam radiation dose escalation in adults with HGG. SEARCH METHODS We searched CENTRAL, MEDLINE Ovid and Embase Ovid to August 2019 for relevant randomised phase III trials. SELECTION CRITERIA We included adults with a pathological diagnosis of HGG randomised to the following external beam radiation regimens: daily conventionally fractionated radiotherapy versus no radiotherapy; hypofractionated radiotherapy versus daily conventionally fractionated radiotherapy; hyperfractionated radiotherapy versus daily conventionally fractionated radiotherapy or accelerated radiotherapy versus daily conventionally fractionated radiotherapy. DATA COLLECTION AND ANALYSIS The primary outcomes were overall survival and adverse effects. The secondary outcomes were progression free survival and quality of life. We used the standard methodological procedures expected by Cochrane. We assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS Since the last version of this review, we identified no new relevant trials for inclusion. We included 11 randomised controlled trials (RCTs) with 2062 participants and 1537 in the relevant arms for this review. There was an overall survival benefit for people with HGG receiving postoperative radiotherapy compared to the participants receiving postoperative supportive care. For the four pooled RCTs (397 participants), the overall hazard ratio (HR) for survival was 2.01 favouring postoperative radiotherapy (95% confidence interval (CI) 1.58 to 2.55; P < 0.00001; moderate-certainty evidence). Although these trials may not have completely reported adverse effects, they did not note any significant toxicity attributable to radiation. Progression free survival and quality of life could not be pooled due to lack of data. Overall survival was similar between hypofractionated and conventional radiotherapy in five trials (943 participants), where the HR was 0.95 (95% CI 0.78 to 1.17; P = 0.63; very low-certainty evidence. The trials reported that hypofractionated and conventional radiotherapy were well tolerated with mild acute adverse effects. These trials only reported one participant in the hypofractionated arm developing symptomatic radiation necrosis that required surgery. Progression free survival and quality of life could not be pooled due to the lack of data. Overall survival was similar between hypofractionated and conventional radiotherapy in the subset of two trials (293 participants) which included participants aged 60 years and older with glioblastoma. For this category, the HR was 1.16 (95% CI 0.92 to 1.46; P = 0.21; high-certainty evidence). There were two trials which compared hyperfractionated radiotherapy versus conventional radiation and one trial which compared accelerated radiotherapy versus conventional radiation. However, the results could not be pooled. The conventionally fractionated radiotherapy regimens were 4500 cGy to 6000 cGy given in 180 cGy to 200 cGy daily fractions, over five to six weeks. All trials generally included participants with World Health Organization (WHO) performance status from 0 to 2 and Karnofsky performance status of 50 and higher. The risk of selection bias was generally low among these RCTs. The number of participants lost to follow-up for the outcome of overall survival was low. Attrition, performance, detection and reporting bias for the outcome of overall survival was low. There was unclear attrition, performance, detection and reporting bias relating to the outcomes of adverse effects, progression free survival and quality of life. AUTHORS' CONCLUSIONS Postoperative conventional daily radiotherapy probably improves survival for adults with good performance status and HGG compared to no postoperative radiotherapy. Hypofractionated radiotherapy has similar efficacy for survival compared to conventional radiotherapy, particularly for individuals aged 60 years and older with glioblastoma. There are insufficient data regarding hyperfractionation versus conventionally fractionated radiation (without chemotherapy) and for accelerated radiation versus conventionally fractionated radiation (without chemotherapy). There are HGG subsets who have poor prognosis even with treatment (e.g. glioblastoma histology, older age and poor performance status). These HGG individuals with poor prognosis have generally been excluded from randomised trials based on poor performance status. No randomised trial has compared comfort measures or best supportive care with an active intervention using radiotherapy or chemotherapy in these people with poor prognosis. Since the last version of this review, we found no new relevant studies. The search identified three new trials, but all were excluded as none had a conventionally fractionated radiotherapy arm.
Collapse
Affiliation(s)
- Luluel Khan
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Hany Soliman
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - James Perry
- Crolla Endowed Chair of Neuro-Oncology Research, Sunnybrook Health Sciences Centre and Odette Cancer Centre, Toronto, Canada
| | - Wei Xu
- Department of Biostatistics, University of Toronto, Toronto, Canada
| | - May N Tsao
- Department of Radiation Oncology, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
Schregel K, Nowicki MO, Palotai M, Nazari N, Zane R, Sinkus R, Lawler SE, Patz S. Magnetic Resonance Elastography reveals effects of anti-angiogenic glioblastoma treatment on tumor stiffness and captures progression in an orthotopic mouse model. Cancer Imaging 2020; 20:35. [PMID: 32398076 PMCID: PMC7218549 DOI: 10.1186/s40644-020-00314-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/27/2020] [Indexed: 01/09/2023] Open
Abstract
Background Anti-angiogenic treatment of glioblastoma (GBM) complicates radiologic monitoring. We evaluated magnetic resonance elastography (MRE) as an imaging tool for monitoring the efficacy of anti-VEGF treatment of GBM. Methods Longitudinal studies were performed in an orthotopic GBM xenograft mouse model. Animals treated with B20 anti-VEGF antibody were compared to untreated controls regarding survival (n = 13), classical MRI-contrasts and biomechanics as quantified via MRE (n = 15). Imaging was performed on a 7 T small animal horizontal bore MRI scanner. MRI and MRE parameters were compared to histopathology. Results Anti-VEGF-treated animals survived longer than untreated controls (p = 0.0011) with progressively increased tumor volume in controls (p = 0.0001). MRE parameters viscoelasticity |G*| and phase angle Y significantly decreased in controls (p = 0.02 for |G*| and p = 0.0071 for Y). This indicates that untreated tumors became softer and more elastic than viscous with progression. Tumor volume in treated animals increased more slowly than in controls, indicating efficacy of the therapy, reaching significance only at the last time point (p = 0.02). Viscoelasticity and phase angle Y tended to decrease throughout therapy, similar as for control animals. However, in treated animals, the decrease in phase angle Y was significantly attenuated and reached statistical significance at the last time point (p = 0.04). Histopathologically, control tumors were larger and more heterogeneous than treated tumors. Vasculature was normalized in treated tumors compared with controls, which showed abnormal vasculature and necrosis. In treated tumors, a higher amount of myelin was observed within the tumor area (p = 0.03), likely due to increased tumor invasion. Stiffness of the contralateral hemisphere was influenced by tumor mass effect and edema. Conclusions Anti-angiogenic GBM treatment prolonged animal survival, slowed tumor growth and softening, but did not prevent progression. MRE detected treatment effects on tumor stiffness; the decrease of viscoelasticity and phase angle in GBM was attenuated in treated animals, which might be explained by normalized vasculature and greater myelin preservation within treated tumors. Thus, further investigation of MRE is warranted to understand the potential for MRE in monitoring treatment in GBM patients by complementing existing MRI techniques.
Collapse
Affiliation(s)
- Katharina Schregel
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. .,Institute of Neuroradiology, University Medical Center Goettingen, Goettingen, Germany.
| | - Michal O Nowicki
- Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Miklos Palotai
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Navid Nazari
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rachel Zane
- Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Ralph Sinkus
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.,INSERM U1148, Laboratory for Vascular Translational Science, University Paris Diderot, University Paris 13, Paris, France
| | - Sean E Lawler
- Harvard Medical School, Boston, MA, USA. .,Harvey Cushing Neurooncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA.
| | - Samuel Patz
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Gonçalves FG, Chawla S, Mohan S. Emerging MRI Techniques to Redefine Treatment Response in Patients With Glioblastoma. J Magn Reson Imaging 2020; 52:978-997. [PMID: 32190946 DOI: 10.1002/jmri.27105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and most malignant primary brain tumor. Despite aggressive multimodal treatment, its prognosis remains poor. Even with continuous developments in MRI, which has provided us with newer insights into the diagnosis and understanding of tumor biology, response assessment in the posttherapy setting remains challenging. We believe that the integration of additional information from advanced neuroimaging techniques can further improve the diagnostic accuracy of conventional MRI. In this article, we review the utility of advanced neuroimaging techniques such as diffusion-weighted imaging, diffusion tensor imaging, perfusion-weighted imaging, proton magnetic resonance spectroscopy, and chemical exchange saturation transfer in characterizing and evaluating treatment response in patients with glioblastoma. We will also discuss the existing challenges and limitations of using these techniques in clinical settings and possible solutions to avoiding pitfalls in study design, data acquisition, and analysis for future studies. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE: 3 J. Magn. Reson. Imaging 2020;52:978-997.
Collapse
Affiliation(s)
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
38
|
FASN Is a Biomarker Enriched in Malignant Glioma-Derived Extracellular Vesicles. Int J Mol Sci 2020; 21:ijms21061931. [PMID: 32178271 PMCID: PMC7139767 DOI: 10.3390/ijms21061931] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are known for their important role in cancer progression and hold considerable potential as a source for tumor biomarkers. However, purification of tumor-specific EVs from patient plasma is still an urgent unmet need due to contamination by normal host cell-derived EVs, that results in compromised analytical sensitivity. Here we identified fatty acid synthase (FASN), a key lipogenic enzyme which is highly expressed in malignant glioma cells, to be elevated in CD63- and CD81-positive EVs in glioma patient plasma samples, opening vital opportunities to sort brain tumor-specific EVs.
Collapse
|
39
|
Müller Bark J, Kulasinghe A, Chua B, Day BW, Punyadeera C. Circulating biomarkers in patients with glioblastoma. Br J Cancer 2020; 122:295-305. [PMID: 31666668 PMCID: PMC7000822 DOI: 10.1038/s41416-019-0603-6] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/23/2019] [Accepted: 09/23/2019] [Indexed: 12/28/2022] Open
Abstract
Gliomas are the most common tumours of the central nervous system and the most aggressive form is glioblastoma (GBM). Despite advances in treatment, patient survival remains low. GBM diagnosis typically relies on imaging techniques and postoperative pathological diagnosis; however, both procedures have their inherent limitations. Imaging modalities cannot differentiate tumour progression from treatment-related changes that mimic progression, known as pseudoprogression, which might lead to misinterpretation of therapy response and delay clinical interventions. In addition to imaging limitations, tissue biopsies are invasive and most of the time cannot be performed over the course of treatment to evaluate 'real-time' tumour dynamics. In an attempt to address these limitations, liquid biopsies have been proposed in the field. Blood sampling is a minimally invasive procedure for a patient to endure and could provide tumoural information to guide therapy. Tumours shed tumoural content, such as circulating tumour cells, cell-free nucleic acids, proteins and extracellular vesicles, into the circulation, and these biomarkers are reported to cross the blood-brain barrier. The use of liquid biopsies is emerging in the field of GBM. In this review, we aim to summarise the current literature on circulating biomarkers, namely circulating tumour cells, circulating tumour DNA and extracellular vesicles as potential non-invasively sampled biomarkers to manage the treatment of patients with GBM.
Collapse
Affiliation(s)
- Juliana Müller Bark
- Saliva and Liquid Biopsy Translational Research Team, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
- Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Arutha Kulasinghe
- Saliva and Liquid Biopsy Translational Research Team, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
- Translational Research Institute, Woolloongabba, QLD, 4102, Australia
| | - Benjamin Chua
- Faculty of Medicine, University of Queensland, 288 Herston Road, Herston, QLD, 4006, Australia
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4029, Australia
| | - Bryan W Day
- Faculty of Medicine, University of Queensland, 288 Herston Road, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Gardens Point, QLD, 4000, Australia
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer MRI, Brisbane, QLD, 4006, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Research Team, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia.
- Translational Research Institute, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
40
|
Daghighi S, Bahrami N, Tom WJ, Coley N, Seibert TM, Hattangadi-Gluth JA, Piccioni DE, Dale AM, Farid N, McDonald CR. Restriction Spectrum Imaging Differentiates True Tumor Progression From Immune-Mediated Pseudoprogression: Case Report of a Patient With Glioblastoma. Front Oncol 2020; 10:24. [PMID: 32047723 PMCID: PMC6997150 DOI: 10.3389/fonc.2020.00024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/08/2020] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy is increasingly used in the treatment of glioblastoma (GBM), with immune checkpoint therapy gaining in popularity given favorable outcomes achieved for other tumors. However, immune-mediated (IM)-pseudoprogression is common, remains poorly characterized, and renders conventional imaging of little utility when evaluating for treatment response. We present the case of a 64-year-old man with GBM who developed pathologically proven IM-pseudoprogression after initiation of a checkpoint inhibitor, and who subsequently developed true tumor progression at a distant location. Based on both qualitative and quantitative analysis, we demonstrate that an advanced diffusion-weighted imaging (DWI) technique called restriction spectrum imaging (RSI) can differentiate IM-pseudoprogression from true progression even when conventional imaging, including standard DWI/apparent diffusion coefficient (ADC), is not informative. These data complement existing literature supporting the ability of RSI to estimate tumor cellularity, which may help to resolve complex diagnostic challenges such as the identification of IM-pseudoprogression.
Collapse
Affiliation(s)
- Shadi Daghighi
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Naeim Bahrami
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
| | - William J. Tom
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Nicholas Coley
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Tyler M. Seibert
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jona A. Hattangadi-Gluth
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, United States
| | - David E. Piccioni
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Anders M. Dale
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Nikdokht Farid
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
| | - Carrie R. McDonald
- Center for Multimodal Imaging and Genetics, University of California, San Diego, La Jolla, CA, United States
- Department of Radiation Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
41
|
Monroe CL, Travers S, Woldu HG, Litofsky NS. Does Surveillance-Detected Disease Progression Yield Superior Patient Outcomes in High-Grade Glioma? World Neurosurg 2019; 135:e410-e417. [PMID: 31821913 DOI: 10.1016/j.wneu.2019.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Standard follow-up care for patients with high-grade glioma (HGG) involves routine surveillance imaging to detect disease progression, assess treatment response, and monitor clinical symptoms. Although logical in nature, evidence supporting this practice is limited. We hypothesize patients with tumor recurrence detected on routine surveillance imaging will experience superior outcomes relative to symptomatic detection, using measures of survival and postrecurrence neurologic function. METHODS Adult patients receiving treatment for HGG at our institution from 2004 to 2018 were identified, and data including tumor characteristics, imaging results, neurologic status, and survival were extracted from the medical records of patients meeting inclusion criteria. All participants were followed for a minimum of 12 months, or for survival duration. Survival and neurologic function differences were assessed using log rank and 2-sample t tests with 2-sided 0.05 alpha level of significance. RESULTS Of the 74 patients meeting inclusion criteria, 47 (63.5%) had recurrence detected via routine surveillance imaging, and 27 (36.5%) had symptomatic detection outside of the surveillance schedule. Neither median overall survival (14.8 months for surveillance and 15.7 months for symptomatic; P = 0.600) nor postrecurrence neurologic function (assessed by Karnofsky Performance Scale Index and Eastern Cooperative Oncology Group) differed between the surveillance and symptomatic detection groups (P = 0.699 and P = 0.908, respectively). CONCLUSIONS Recurrence detection occurring via routine surveillance imaging did not yield superior patient outcomes relative to symptomatic detection occurring outside of the standard surveillance schedule in patients with HGG. Further evaluation of surveillance imaging and alternative follow-up methods for this patient population may be warranted.
Collapse
Affiliation(s)
- Courtney L Monroe
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Sarah Travers
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Henok G Woldu
- Biostatistics and Research Design, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - N Scott Litofsky
- Division of Neurological Surgery, University of Missouri School of Medicine, Columbia, Missouri, USA.
| |
Collapse
|
42
|
Cuccarini V, Aquino D, Gioppo A, Anghileri E, Pellegatta S, Schettino C, Mazzi F, Finocchiaro G, Bruzzone MG, Eoli M. Advanced MRI Assessment during Dendritic Cell Immunotherapy Added to Standard Treatment Against Glioblastoma. J Clin Med 2019; 8:jcm8112007. [PMID: 31744235 PMCID: PMC6912338 DOI: 10.3390/jcm8112007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/18/2023] Open
Abstract
Evaluating changes induced by immunotherapies (IT) on conventional magnetic resonance imaging (MRI) is difficult because those treatments may produce inflammatory responses. To explore the potential contribution of advanced MRI to distinguish pseudoprogression (PsP) and true tumor progression (TTP), and to identify patients obtaining therapeutic benefit from IT, we examined aMRI findings in newly diagnosed glioblastoma treated with dendritic cell IT added to standard treatment. We analyzed longitudinal MRIs obtained in 22 patients enrolled in the EUDRACT N° 2008-005035-15 trial. According to RANO criteria, we observed 18 TTP and 8 PsP. Comparing MRI performed at the time of TTP/PsP with the previous exam performed two months before, a difference in cerebral blood volume ΔrCBVmax ≥ 0.47 distinguished TTP from PsP with a sensitivity of 67% and specificity of 75% (p = 0.004). A decrease in minimal apparent diffusion coefficient rADCmin (1.15 vs. 1.01, p = 0.003) was observed after four vaccinations only in patients with a persistent increase of natural killer cells (response effectors during IT) in peripheral blood. Basal rADCmin > 1 was independent predictor of longer progression free (16.1 vs. 9 months, p = 0.0001) and overall survival (32.8 vs. 17.5 months, p = 0.0005). In conclusion, rADC predicted response to immunotherapy and survival; Apparent Diffusion Coefficient (ADC) and Cerebral Blood Volume (CBV) modifications over time help differentiating PsP from TTP at onset.
Collapse
Affiliation(s)
- Valeria Cuccarini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (V.C.); (D.A.); (A.G.); (F.M.)
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (V.C.); (D.A.); (A.G.); (F.M.)
| | - Andrea Gioppo
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (V.C.); (D.A.); (A.G.); (F.M.)
| | - Elena Anghileri
- Neuro-oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.A.); (S.P.); (C.S.); (G.F.); (M.E.)
| | - Serena Pellegatta
- Neuro-oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.A.); (S.P.); (C.S.); (G.F.); (M.E.)
| | - Carla Schettino
- Neuro-oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.A.); (S.P.); (C.S.); (G.F.); (M.E.)
| | - Federica Mazzi
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (V.C.); (D.A.); (A.G.); (F.M.)
| | - Gaetano Finocchiaro
- Neuro-oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.A.); (S.P.); (C.S.); (G.F.); (M.E.)
| | - Maria Grazia Bruzzone
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (V.C.); (D.A.); (A.G.); (F.M.)
- Correspondence: ; Tel.: +39-0223942775
| | - Marica Eoli
- Neuro-oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.A.); (S.P.); (C.S.); (G.F.); (M.E.)
| |
Collapse
|
43
|
Anthony C, Mladkova-Suchy N, Adamson DC. The evolving role of antiangiogenic therapies in glioblastoma multiforme: current clinical significance and future potential. Expert Opin Investig Drugs 2019; 28:787-797. [PMID: 31356114 DOI: 10.1080/13543784.2019.1650019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults, but its prognosis remains poor despite significant advances in our understanding of its molecular biology and investigation of numerous treatment modalities. Despite conventional treatment consisting of surgical resection, radiotherapy, and temozolomide marginally prolonging survival, most GBM patients die within 2 years of initial diagnosis. Bevacizumab (Bev) is the best-studied antiangiogenic agent for GBM and currently the only FDA-approved second-line treatment. Areas covered: Areas covered in this review include the molecular pathways of angiogenesis in glioblastoma, specifically the overexpression of vascular endothelial growth factor (VEGF) and robust formation of tumor neovasculature. In addition, this review covers pharmacological targeting of this process as a longstanding attractive clinical strategy, specifically by Bev. Expert opinion: This review attempts to discuss the history of early studies of antiangiogenic treatment for GBM that eventually failed in subsequent studies and the evolving modern role of Bev in the course of treatment for a variety of indications, including symptom control, reduced glucocorticoid use, and improved quality of life.
Collapse
Affiliation(s)
- Casey Anthony
- Department of Neurosurgery, Emory University , Atlanta , GA , USA
| | - Nikol Mladkova-Suchy
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London , UK
| | - David Cory Adamson
- Department of Neurosurgery, Emory University , Atlanta , GA , USA.,Neurosurgery section, Atlanta VA Medical Center , Decatur , GA , USA
| |
Collapse
|
44
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Radiol Clin North Am 2019; 57:1199-1216. [PMID: 31582045 DOI: 10.1016/j.rcl.2019.07.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
45
|
Pyrosequencing versus methylation-specific PCR for assessment of MGMT methylation in tumor and blood samples of glioblastoma patients. Sci Rep 2019; 9:11125. [PMID: 31366977 PMCID: PMC6668570 DOI: 10.1038/s41598-019-47642-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
Circulating biomarkers in blood may provide an interesting alternative to risky tissue biopsies in the diagnosis and follow-up of glioblastoma patients. We have assessed MGMT methylation status in blood and tissue samples from unresected glioblastoma patients who had been included in the randomized GENOM-009 trial. Paired blood and tissue samples were assessed by methylation-specific PCR (MSP) and pyrosequencing (PYR). After establishing the minimum PYR cut-off that could yield a significant difference in overall survival, we assessed the sensitivity, specificity, positive predictive value and negative predictive value (NPV) of the analyses. Methylation could be detected in cfDNA by both MSP and PYR but with low concordance with results in tissue. Sensitivity was low for both methods (31% and 38%, respectively), while specificity was higher for MSP in blood than for PYR in plasma (96% vs 76%) and NPV was similar (56 vs 57%). Concordance of results in tissue by MSP and PYR was 84.3% (P < 0.001) and correlated with outcome. We conclude that detection of cfDNA in the blood of glioblastoma patients can be an alternative when tumor tissue is not available but methods for the detection of cfDNA in blood must improve before it can replace analysis in tumor tissue.
Collapse
|
46
|
Multicenter study demonstrates radiomic features derived from magnetic resonance perfusion images identify pseudoprogression in glioblastoma. Nat Commun 2019; 10:3170. [PMID: 31320621 PMCID: PMC6639324 DOI: 10.1038/s41467-019-11007-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/07/2019] [Indexed: 01/04/2023] Open
Abstract
Pseudoprogression (PsP) is a diagnostic clinical dilemma in cancer. In this study, we retrospectively analyse glioblastoma patients, and using their dynamic susceptibility contrast and dynamic contrast-enhanced perfusion MRI images we build a classifier using radiomic features obtained from both Ktrans and rCBV maps coupled with support vector machines. We achieve an accuracy of 90.82% (area under the curve (AUC) = 89.10%, sensitivity = 91.36%, 67 specificity = 88.24%, p = 0.017) in differentiating between pseudoprogression (PsP) and progressive disease (PD). The diagnostic performances of the models built using radiomic features from Ktrans and rCBV separately were equally high (Ktrans: AUC = 94%, 69 p = 0.012; rCBV: AUC = 89.8%, p = 0.004). Thus, this MR perfusion-based radiomic model demonstrates high accuracy, sensitivity and specificity in discriminating PsP from PD, thus provides a reliable alternative for noninvasive identification of PsP versus PD at the time of clinical/radiologic question. This study also illustrates the successful application of radiomic analysis as an advanced processing step on different MR perfusion maps. MRI scans of glioblastoma patients can be misleading and some patients appear to show features of progressive disease although they respond to treatment. Here, the authors use MRI images of progressive disease or pseudoprogression and build a classifier using machine learning to distinguish the two.
Collapse
|
47
|
Hafeez U, Cher LM. Biomarkers and smart intracranial devices for the diagnosis, treatment, and monitoring of high-grade gliomas: a review of the literature and future prospects. Neurooncol Adv 2019; 1:vdz013. [PMID: 32642651 PMCID: PMC7212884 DOI: 10.1093/noajnl/vdz013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain neoplasm with median overall survival (OS) around 15 months. There is a dearth of effective monitoring strategies for patients with high-grade gliomas. Relying on magnetic resonance images of brain has its challenges, and repeated brain biopsies add significant morbidity. Hence, it is imperative to establish a less invasive way to diagnose, monitor, and guide management of patients with high-grade gliomas. Currently, multiple biomarkers are in various phases of development and include tissue, serum, cerebrospinal fluid (CSF), and imaging biomarkers. Here we review and summarize the potential biomarkers found in blood and CSF, including extracellular macromolecules, extracellular vesicles, circulating tumor cells, immune cells, endothelial cells, and endothelial progenitor cells. The ability to detect tumor-specific biomarkers in blood and CSF will potentially not only reduce the need for repeated brain biopsies but also provide valuable information about the heterogeneity of tumor, response to current treatment, and identify disease resistance. This review also details the status and potential scope of brain tumor-related cranial devices and implants including Ommaya reservoir, microelectromechanical systems-based depot device, Alzet mini-osmotic pump, Metronomic Biofeedback Pump (MBP), ipsum G1 implant, ultra-thin needle implant, and putative devices. An ideal smart cranial implant will overcome the blood-brain barrier, deliver various drugs, provide access to brain tissue, and potentially measure and monitor levels of various biomarkers.
Collapse
Affiliation(s)
- Umbreen Hafeez
- Olivia Newton-John Cancer Research Institute, Austin Hospital, Melbourne, Australia
- Latrobe University School of Cancer Medicine, Melbourne, Australia
- Department of Medical Oncology, Austin Hospital, Melbourne, Australia
| | - Lawrence M Cher
- Olivia Newton-John Cancer Research Institute, Austin Hospital, Melbourne, Australia
- Department of Medical Oncology, Austin Hospital, Melbourne, Australia
- Corresponding Author: Lawrence M. Cher, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, VIC 3084, Australia ()
| |
Collapse
|
48
|
Werner JM, Stoffels G, Lichtenstein T, Borggrefe J, Lohmann P, Ceccon G, Shah NJ, Fink GR, Langen KJ, Kabbasch C, Galldiks N. Differentiation of treatment-related changes from tumour progression: a direct comparison between dynamic FET PET and ADC values obtained from DWI MRI. Eur J Nucl Med Mol Imaging 2019; 46:1889-1901. [PMID: 31203420 DOI: 10.1007/s00259-019-04384-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Following brain cancer treatment, the capacity of anatomical MRI to differentiate neoplastic tissue from treatment-related changes (e.g., pseudoprogression) is limited. This study compared apparent diffusion coefficients (ADC) obtained by diffusion-weighted MRI (DWI) with static and dynamic parameters of O-(2-[18F]fluoroethyl)-L-tyrosine (FET) PET for the differentiation of treatment-related changes from tumour progression. PATIENTS AND METHODS Forty-eight pretreated high-grade glioma patients with anatomical MRI findings suspicious for progression (median time elapsed since last treatment was 16 weeks) were investigated using DWI and dynamic FET PET. Maximum and mean tumour-to-brain ratios (TBRmax, TBRmean) as well as dynamic parameters (time-to-peak and slope values) of FET uptake were calculated. For mean ADC calculation, regions-of-interest analyses were performed on ADC maps calculated from DWI coregistered with the contrast-enhanced MR image. Diagnoses were confirmed neuropathologically (21%) or clinicoradiologically. Diagnostic performance was evaluated using receiver-operating-characteristic analyses or Fisher's exact test for a combinational approach. RESULTS Ten of 48 patients had treatment-related changes (21%). The diagnostic performance of FET PET was significantly higher (threshold for both TBRmax and TBRmean, 1.95; accuracy, 83%; AUC, 0.89 ± 0.05; P < 0.001) than that of ADC values (threshold ADC, 1.09 × 10-3 mm2/s; accuracy, 69%; AUC, 0.73 ± 0.09; P = 0.13). The addition of static FET PET parameters to ADC values increased the latter's accuracy to 89%. The highest accuracy was achieved by combining static and dynamic FET PET parameters (93%). Moreover, in contrast to ADC values, TBRs <1.95 at suspected progression predicted a significantly longer survival (P = 0.01). CONCLUSIONS Data suggest that static and dynamic FET PET provide valuable information concerning the differentiation of early treatment-related changes from tumour progression and outperform ADC measurement for this highly relevant clinical question.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Thorsten Lichtenstein
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan Borggrefe
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Garry Ceccon
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Department of Neurology, University Hospital Aachen, Aachen, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Christoph Kabbasch
- Department of Neuroradiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Cologne, Germany.
- Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Leo-Brandt-St. 5, 52425, Juelich, Germany.
- Department of Neurology, University Hospital Cologne, Kerpener St. 62, 50937, Cologne, Germany.
| |
Collapse
|
49
|
Durand-Muñoz C, Flores-Alvarez E, Moreno-Jimenez S, Roldan-Valadez E. Pre-operative apparent diffusion coefficient values and tumour region volumes as prognostic biomarkers in glioblastoma: correlation and progression-free survival analyses. Insights Imaging 2019; 10:36. [PMID: 30887267 PMCID: PMC6423260 DOI: 10.1186/s13244-019-0724-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/20/2019] [Indexed: 02/08/2023] Open
Abstract
Objectives Glioblastoma (GB) contains diverse histologic regions. Apparent diffusion coefficient (ADC) values are surrogates for the degree of number of cells within the tumour regions. Because an assessment of ADC values and volumes within tumour sub-compartments of GB is missing in the literature, we aimed to evaluate these associations. Methods A retrospective cohort of 48 patients with GB underwent segmentation to calculate tumour region volumes (in cubic centimetre) and ADC values in tumour regions: normal tissue, enhancing tumour, proximal oedema, distal oedema, and necrosis. Correlation, Kaplan-Meier, and Cox hazard regression analyses were performed. Results We found a statistically significant difference among ADC values for tumour regions: F (4, 220) = 166.71 and p ≤ .001 and tumour region volumes (necrosis, enhancing tumour, peritumoural oedema): F (2, 141) = 136.3 and p ≤ .001. Post hoc comparisons indicated that the only significantly different mean score was the peritumoural volume in oedema region (p < .001). We observed a positive significant correlation between ADC of distal oedema and peritumoural volume, r = .418, df = 34, and p = .011. Cox proportional hazards regression analysis considering only tumour region volumes provided an almost significant model: − 2 log-likelihood = 146.066, χ2 (4) = 9.303, and p = .054 with a trend towards significance of the hazard function: p = .067 and HR = 1.077 for the non-enhancing tumour volume. Conclusions ADC values together with volumes of oedema region might have a role as predictors of progression-free survival (PFS) in patients with GB; we recommend a routine MRI assessment with the calculation of these biomarkers in GB.
Collapse
Affiliation(s)
- Coral Durand-Muñoz
- Department of Internal Medicine, Medica Sur Clinic and Foundation, Mexico City, Mexico
| | - Eduardo Flores-Alvarez
- Department of Neurosurgery, Secretariat of Health, General Hospital of Mexico, Mexico City, Mexico
| | - Sergio Moreno-Jimenez
- Radioneurosurgery Unit, The National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Ernesto Roldan-Valadez
- Directorate of Research, Secretariat of Health, General Hospital of Mexico, Mexico City, Mexico. .,Department of Radiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str., 8, b. 2, Moscow, Russia, 119992.
| |
Collapse
|
50
|
Rodriguez D, Chambers T, Warmuth-Metz M, Aliaga ES, Warren D, Calmon R, Hargrave D, Garcia J, Vassal G, Grill J, Zahlmann G, Morgan PS, Jaspan T. Evaluation of the Implementation of the Response Assessment in Neuro-Oncology Criteria in the HERBY Trial of Pediatric Patients with Newly Diagnosed High-Grade Gliomas. AJNR Am J Neuroradiol 2019; 40:568-575. [PMID: 30819765 DOI: 10.3174/ajnr.a5982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/31/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE HERBY was a Phase II multicenter trial setup to establish the efficacy and safety of adding bevacizumab to radiation therapy and temozolomide in pediatric patients with newly diagnosed non-brain stem high-grade gliomas. This study evaluates the implementation of the radiologic aspects of HERBY. MATERIALS AND METHODS We analyzed multimodal imaging compliance rates and scan quality for participating sites, adjudication rates and reading times for the central review process, the influence of different Response Assessment in Neuro-Oncology criteria in the final response, the incidence of pseudoprogression, and the benefit of incorporating multimodal imaging into the decision process. RESULTS Multimodal imaging compliance rates were the following: diffusion, 82%; perfusion, 60%; and spectroscopy, 48%. Neuroradiologists' responses differed for 50% of scans, requiring adjudication, with a total average reading time per patient of approximately 3 hours. Pseudoprogression occurred in 10/116 (9%) cases, 8 in the radiation therapy/temozolomide arm and 2 in the bevacizumab arm (P < .01). Increased target enhancing lesion diameter was a reason for progression in 8/86 cases (9.3%) but never the only radiologic or clinical reason. Event-free survival was predicted earlier in 5/86 (5.8%) patients by multimodal imaging (diffusion, n = 4; perfusion, n = 1). CONCLUSIONS The addition of multimodal imaging to the response criteria modified the assessment in a small number of cases, determining progression earlier than structural imaging alone. Increased target lesion diameter, accounting for a large proportion of reading time, was never the only reason to designate disease progression.
Collapse
Affiliation(s)
- D Rodriguez
- Medical Physics and Clinical Engineering (D.R., P.S.M.)
| | - T Chambers
- Cardiff University, School of Medicine (T.C.), Cardiff, UK
| | - M Warmuth-Metz
- Würzburg University, Institute for Diagnostic and Interventional Neuroradiology (M.W.-M.), Würzburg, Germany
| | - E Sanchez Aliaga
- VU University Medical Center, Department of Radiology & Nuclear Medicine (E.S.A.), Amsterdam, the Netherlands
| | - D Warren
- Leeds Teaching Hospital, Department of Radiology (D.W.), Leeds, UK
| | - R Calmon
- Assistance Publique-Hôpitaux de Paris, Pediatric Radiology (R.C.), Paris, France
| | - D Hargrave
- Great Ormond Street Hospital, Haematology and Oncology Department (D.H.), London, UK
| | - J Garcia
- F. Hoffmann-La Roche (J.Garcia, G.Z.), Basel, Switzerland
| | - G Vassal
- Gustave Roussy and Paris-Sud University, Pediatric and Adolescent Oncology and Unite Mixte de Recherche (G.V., J.Grill), Villejuif, France
| | - J Grill
- Gustave Roussy and Paris-Sud University, Pediatric and Adolescent Oncology and Unite Mixte de Recherche (G.V., J.Grill), Villejuif, France
| | - G Zahlmann
- F. Hoffmann-La Roche (J.Garcia, G.Z.), Basel, Switzerland
| | - P S Morgan
- Medical Physics and Clinical Engineering (D.R., P.S.M.).,Nottingham Biomedical Research Centre of the UK National Institute of Health Research (P.S.M.), Nottingham, UK
| | - T Jaspan
- From Nottingham University Hospitals, Department of Radiology (T.J.)
| |
Collapse
|