1
|
Ghafarian M, Cao M, Kirby KM, Schneider CW, Deng J, Mellon EA, Kishan AU, Maziero D, Wu TC. Magnetic Resonance Imaging Sequences and Technologies in Adaptive Radiation Therapy. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00384-0. [PMID: 40298856 DOI: 10.1016/j.ijrobp.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/06/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
Radiation therapy is essential in both curative and palliative treatments for most cancers. However, traditional radiation therapy workflows using computed tomography (CT) simulation-based planning and cone beam CT image guidance face several technical challenges, including limited tumor visibility and daily fluctuations in tumor size and shape. Magnetic resonance imaging (MRI) guided linear accelerators (MR-Linacs) address these issues by enabling precise visualization of changes in tumor position and morphologic changes, as well as changes in surrounding organs-at-risk. The hybrid MR-Linac systems combine MRI with linear accelerator technology, offering enhanced soft tissue visualization and the potential for adaptive radiation therapy (ART). This narrative review provides a comprehensive introduction to MR guided ART technologies, covering protocol optimization with appropriate pulse sequence selection and parameter adjustment for clinical implementations on various disease sites.
Collapse
Affiliation(s)
- Melissa Ghafarian
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California.
| | - Minsong Cao
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Krystal M Kirby
- Department of Physics, Mary Bird Perkins Cancer Center, Baton Rouge, Louisiana
| | | | - Jie Deng
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Eric A Mellon
- Department of Radiation Oncology and Biomedical Engineering, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Coral Gables, Florida
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California
| | - Danilo Maziero
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California
| | - Trudy C Wu
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
2
|
Labbène E, Mahmoud M, Marrakchi-Kacem L, Ben Hamouda M. Prognostic value of preoperative diffusion restriction in glioblastoma. LA TUNISIE MEDICALE 2024; 102:94-99. [PMID: 38567475 PMCID: PMC11358806 DOI: 10.62438/tunismed.v102i2.4746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/13/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Although glioblastoma (GBM) has a very poor prognosis, overall survival (OS) in treated patients shows great difference varying from few days to several months. Identifying factors explaining this difference would improve management of patient treatment. AIM To determine the relevance of diffusion restriction in newly diagnosed treatment-naïve GBM patients. METHODS Preoperative magnetic resonance scans of 33 patients with GBM were reviewed. Regions of interest including all the T2 hyperintense lesion were drawn on diffusion weighted B0 images and transferred to the apparent diffusion coefficient (ADC) map. For each patient, a histogram displaying the ADC values within in the regions of interest was generated. Volumetric parameters including tumor regions with restricted diffusion, parameters derived from histogram and mean ADC value of the tumor were calculated. Their relationship with OS was analyzed. RESULTS Patients with mean ADC value < 1415x10-6 mm2/s had a significantly shorter OS (p=0.021). Among volumetric parameters, the percentage of volume within T2 lesion with a normalized ADC value <1.5 times that in white matter was significantly associated with OS (p=0.0045). Patients with a percentage>23.92% had a shorter OS. Among parameters derived from histogram, the 50th percentile showed a trend towards significance for OS (p=0.055) with patients living longer when having higher values of 50th percentile. A difference in OS was observed between patients according to ADC peak of histogram but this difference did not reach statistical significance (p=0.0959). CONCLUSION Diffusion magnetic resonance imaging may provide useful information for predicting GBM prognosis.
Collapse
Affiliation(s)
- Emna Labbène
- Department of radiology, MT Kassab institute of orthopaedics, Tunis, Tunisia
- Faculty of medicine of Tunis, Tunis-El Manar University, Tunis, Tunisia
| | - Maha Mahmoud
- Faculty of medicine of Tunis, Tunis-El Manar University, Tunis, Tunisia
- Department of neuroradiology, National institute of neurology Mongi-Ben Hamida, Tunis, Tunisia
| | - Linda Marrakchi-Kacem
- Higher institute of biotechnology of Sidi Thabet, Manouba University, Tunis, Tunisia
- National engineering school of Tunis (ENIT), L3S laboratory, Tunis-El Manar University, Tunis, Tunisia
| | - Mohamed Ben Hamouda
- Faculty of medicine of Tunis, Tunis-El Manar University, Tunis, Tunisia
- Department of neuroradiology, National institute of neurology Mongi-Ben Hamida, Tunis, Tunisia
| |
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
|
Laprie A, Noel G, Chaltiel L, Truc G, Sunyach MP, Charissoux M, Magne N, Auberdiac P, Biau J, Ken S, Tensaouti F, Khalifa J, Sidibe I, Roux FE, Vieillevigne L, Catalaa I, Boetto S, Uro-Coste E, Supiot S, Bernier V, Filleron T, Mounier M, Poublanc M, Olivier P, Delord JP, Cohen-Jonathan-Moyal E. Randomized phase III trial of metabolic imaging-guided dose escalation of radio-chemotherapy in patients with newly diagnosed glioblastoma (SPECTRO GLIO trial). Neuro Oncol 2024; 26:153-163. [PMID: 37417948 PMCID: PMC10768994 DOI: 10.1093/neuonc/noad119] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) systematically recurs after a standard 60 Gy radio-chemotherapy regimen. Since magnetic resonance spectroscopic imaging (MRSI) has been shown to predict the site of relapse, we analyzed the effect of MRSI-guided dose escalation on overall survival (OS) of patients with newly diagnosed GBM. METHODS In this multicentric prospective phase III trial, patients who had undergone biopsy or surgery for a GBM were randomly assigned to a standard dose (SD) of 60 Gy or a high dose (HD) of 60 Gy with an additional simultaneous integrated boost totaling 72 Gy to MRSI metabolic abnormalities, the tumor bed and residual contrast enhancements. Temozolomide was administered concomitantly and maintained for 6 months thereafter. RESULTS One hundred and eighty patients were included in the study between March 2011 and March 2018. After a median follow-up of 43.9 months (95% CI [42.5; 45.5]), median OS was 22.6 months (95% CI [18.9; 25.4]) versus 22.2 months (95% CI [18.3; 27.8]) for HD, and median progression-free survival was 8.6 (95% CI [6.8; 10.8]) versus 7.8 months (95% CI [6.3; 8.6]), in SD versus HD, respectively. No increase in toxicity rate was observed in the study arm. The pseudoprogression rate was similar across the SD (14.4%) and HD (16.7%) groups. For O(6)-methylguanine-DNA methyltransferase (MGMT) methylated patients, the median OS was 38 months (95% CI [23.2; NR]) for HD patients versus 28.5 months (95% CI [21.1; 35.7]) for SD patients. CONCLUSION The additional MRSI-guided irradiation dose totaling 72 Gy was well tolerated but did not improve OS in newly diagnosed GBM. TRIAL REGISTRATION NCT01507506; registration date: December 20, 2011. https://clinicaltrials.gov/ct2/show/NCT01507506?cond=NCT01507506&rank=1.
Collapse
Affiliation(s)
- Anne Laprie
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | | | - Leonor Chaltiel
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Gilles Truc
- Centre Georges-François Leclerc, Dijon, France
| | | | | | - Nicolas Magne
- Institut de Cancérologie de la Loire, Saint-Priest en Jarez, France
| | | | - Julian Biau
- Centre Jean-Perrin, Clermont-Ferrand, France
| | - Soléakhéna Ken
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, RadOpt-CRCT-INSERM, Toulouse, France
| | - Fatima Tensaouti
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole & ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Jonathan Khalifa
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | | - Franck-Emmanuel Roux
- Centre Hospitalier Universitaire de Toulouse, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Laure Vieillevigne
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | | - Sergio Boetto
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Emmanuelle Uro-Coste
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, RadOpt-CRCT-INSERM, Toulouse, France
| | - Stéphane Supiot
- Institut de Cancerologie de l’Ouest, Nantes st Herblain, France
| | - Valérie Bernier
- Institut de Cancérologie de Lorraine Centre Alexis Vautrin, Nancy, France
| | - Thomas Filleron
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Muriel Mounier
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Muriel Poublanc
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Pascale Olivier
- Service de Pharmacologie Médicale et Clinique, Centre Régional de Pharmacovigilance, de Pharmacoépidémiologie et d’Information sur le Médicament CHU de Toulouse, Toulouse, France
| | - Jean-Pierre Delord
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | |
Collapse
|
5
|
Lemarié A, Lubrano V, Delmas C, Lusque A, Cerapio JP, Perrier M, Siegfried A, Arnauduc F, Nicaise Y, Dahan P, Filleron T, Mounier M, Toulas C, Cohen-Jonathan Moyal E. The STEMRI trial: Magnetic resonance spectroscopy imaging can define tumor areas enriched in glioblastoma stem-like cells. SCIENCE ADVANCES 2023; 9:eadi0114. [PMID: 37922359 PMCID: PMC10624352 DOI: 10.1126/sciadv.adi0114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Despite maximally safe resection of the magnetic resonance imaging (MRI)-defined contrast-enhanced (CE) central tumor area and chemoradiotherapy, most patients with glioblastoma (GBM) relapse within a year in peritumoral FLAIR regions. Magnetic resonance spectroscopy imaging (MRSI) can discriminate metabolic tumor areas with higher recurrence potential as CNI+ regions (choline/N-acetyl-aspartate index >2) can predict relapse sites. As relapses are mainly imputed to glioblastoma stem-like cells (GSCs), CNI+ areas might be GSC enriched. In this prospective trial, 16 patients with GBM underwent MRSI/MRI before surgery/chemoradiotherapy to investigate GSC content in CNI-/+ biopsies from CE/FLAIR. Biopsy and derived-GSC characterization revealed a FLAIR/CNI+ sample enrichment in GSC and in gene signatures related to stemness, DNA repair, adhesion/migration, and mitochondrial bioenergetics. FLAIR/CNI+ samples generate GSC-enriched neurospheres faster than FLAIR/CNI-. Parameters assessing biopsy GSC content and time-to-neurosphere formation in FLAIR/CNI+ were associated with worse patient outcome. Preoperative MRI/MRSI would certainly allow better resection and targeting of FLAIR/CNI+ areas, as their GSC enrichment can predict worse outcomes.
Collapse
Affiliation(s)
- Anthony Lemarié
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Vincent Lubrano
- TONIC, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Toulouse Neuro Imaging Center, Toulouse, France
- CHU de Toulouse, Neurosurgery Department, Toulouse, France
| | - Caroline Delmas
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Interface Department, Toulouse, France
| | - Amélie Lusque
- Institut Claudius Regaud, IUCT-Oncopole, Biostatistics and Health Data Science Unit, Toulouse, France
| | - Juan-Pablo Cerapio
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Marion Perrier
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Aurore Siegfried
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- CHU de Toulouse, Anatomopathology Department, Toulouse, France
| | - Florent Arnauduc
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Yvan Nicaise
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Perrine Dahan
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Thomas Filleron
- Institut Claudius Regaud, IUCT-Oncopole, Biostatistics and Health Data Science Unit, Toulouse, France
| | - Muriel Mounier
- Institut Claudius Regaud, IUCT-Oncopole, Clinical Trials Office, Toulouse, France
| | - Christine Toulas
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Cancer Biology Department, Molecular Oncology Division, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Radiation Oncology Department, Toulouse, France
| |
Collapse
|
6
|
Sollmann N, Zhang H, Kloth C, Zimmer C, Wiestler B, Rosskopf J, Kreiser K, Schmitz B, Beer M, Krieg SM. Modern preoperative imaging and functional mapping in patients with intracranial glioma. ROFO-FORTSCHR RONTG 2023; 195:989-1000. [PMID: 37224867 DOI: 10.1055/a-2083-8717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Magnetic resonance imaging (MRI) in therapy-naïve intracranial glioma is paramount for neuro-oncological diagnostics, and it provides images that are helpful for surgery planning and intraoperative guidance during tumor resection, including assessment of the involvement of functionally eloquent brain structures. This study reviews emerging MRI techniques to depict structural information, diffusion characteristics, perfusion alterations, and metabolism changes for advanced neuro-oncological imaging. In addition, it reflects current methods to map brain function close to a tumor, including functional MRI and navigated transcranial magnetic stimulation with derived function-based tractography of subcortical white matter pathways. We conclude that modern preoperative MRI in neuro-oncology offers a multitude of possibilities tailored to clinical needs, and advancements in scanner technology (e. g., parallel imaging for acceleration of acquisitions) make multi-sequence protocols increasingly feasible. Specifically, advanced MRI using a multi-sequence protocol enables noninvasive, image-based tumor grading and phenotyping in patients with glioma. Furthermore, the add-on use of preoperatively acquired MRI data in combination with functional mapping and tractography facilitates risk stratification and helps to avoid perioperative functional decline by providing individual information about the spatial location of functionally eloquent tissue in relation to the tumor mass. KEY POINTS:: · Advanced preoperative MRI allows for image-based tumor grading and phenotyping in glioma.. · Multi-sequence MRI protocols nowadays make it possible to assess various tumor characteristics (incl. perfusion, diffusion, and metabolism).. · Presurgical MRI in glioma is increasingly combined with functional mapping to identify and enclose individual functional areas.. · Advancements in scanner technology (e. g., parallel imaging) facilitate increasing application of dedicated multi-sequence imaging protocols.. CITATION FORMAT: · Sollmann N, Zhang H, Kloth C et al. Modern preoperative imaging and functional mapping in patients with intracranial glioma. Fortschr Röntgenstr 2023; 195: 989 - 1000.
Collapse
Affiliation(s)
- Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, United States
| | - Haosu Zhang
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Christopher Kloth
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- TranslaTUM - Central Institute for Translational Cancer Research, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| | - Johannes Rosskopf
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Section of Neuroradiology, Bezirkskrankenhaus Günzburg, Günzburg, Germany
| | - Kornelia Kreiser
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Department of Radiology and Neuroradiology, Universitäts- und Rehabilitationskliniken Ulm, Ulm, Germany
| | - Bernd Schmitz
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
- Section of Neuroradiology, Bezirkskrankenhaus Günzburg, Günzburg, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Sandro M Krieg
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, München, Germany
- Department of Neurosurgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, München, Germany
| |
Collapse
|
7
|
Tensaouti F, Desmoulin F, Gilhodes J, Roques M, Ken S, Lotterie JA, Noël G, Truc G, Sunyach MP, Charissoux M, Magné N, Lubrano V, Péran P, Cohen-Jonathan Moyal E, Laprie A. Is pre-radiotherapy metabolic heterogeneity of glioblastoma predictive of progression-free survival? Radiother Oncol 2023; 183:109665. [PMID: 37024057 DOI: 10.1016/j.radonc.2023.109665] [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: 08/12/2022] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND AND PURPOSE All glioblastoma subtypes share the hallmark of aggressive invasion, meaning that it is crucial to identify their different components if we are to ensure effective treatment and improve survival. Proton MR spectroscopic imaging (MRSI) is a noninvasive technique that yields metabolic information and is able to identify pathological tissue with high accuracy. The aim of the present study was to identify clusters of metabolic heterogeneity, using a large MRSI dataset, and determine which of these clusters are predictive of progression-free survival (PFS). MATERIALS AND METHODS MRSI data of 180 patients acquired in a pre-radiotherapy examination were included in the prospective SPECTRO-GLIO trial. Eight features were extracted for each spectrum: Cho/NAA, NAA/Cr, Cho/Cr, Lac/NAA, and the ratio of each metabolite to the sum of all the metabolites. Clustering of data was performed using a mini-batch k-means algorithm. The Cox model and logrank test were used for PFS analysis. RESULTS Five clusters were identified as sharing similar metabolic information and being predictive of PFS. Two clusters revealed metabolic abnormalities. PFS was lower when Cluster 2 was the dominant cluster in patients' MRSI data. Among the metabolites, lactate (present in this cluster and in Cluster 5) was the most statistically significant predictor of poor outcome. CONCLUSION Results showed that pre-radiotherapy MRSI can be used to reveal tumor heterogeneity. Groups of spectra, which have the same metabolic information, reflect the different tissue components representative of tumor burden proliferation and hypoxia. Clusters with metabolic abnormalities and high lactate are predictive of PFS.
Collapse
Affiliation(s)
- Fatima Tensaouti
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.
| | - Franck Desmoulin
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Julia Gilhodes
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Biostatistics, Toulouse, France
| | - Margaux Roques
- CHU Toulouse, Neuroradiology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Soleakhena Ken
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Engineering and Medical Physics, Toulouse, France; Inserm U1037- Centre de Recherches contre le Cancer de Toulouse, Radiation oncology, Toulouse, France
| | - Jean-Albert Lotterie
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; CHU Toulouse, Nuclear Medicine, Toulouse, France
| | | | - Gilles Truc
- Centre Georges-François Leclerc, Radiation Oncology, Dijon, France
| | | | - Marie Charissoux
- Institut du Cancer de Montpellier, Radiation Oncology, Montpellier, France
| | - Nicolas Magné
- Institut de Cancérologie de la Loire Lucien Neuwirth, Radiation Oncology, Saint-Priest-en-Jarez, France
| | - Vincent Lubrano
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Elizabeth Cohen-Jonathan Moyal
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; Inserm U1037- Centre de Recherches contre le Cancer de Toulouse, Radiation oncology, Toulouse, France
| | - Anne Laprie
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| |
Collapse
|
8
|
Duval T, Lotterie JA, Lemarie A, Delmas C, Tensaouti F, Moyal ECJ, Lubrano V. Glioblastoma Stem-like Cell Detection Using Perfusion and Diffusion MRI. Cancers (Basel) 2022; 14:cancers14112803. [PMID: 35681782 PMCID: PMC9179449 DOI: 10.3390/cancers14112803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 01/25/2023] Open
Abstract
Simple Summary Glioblastoma stem-like cells (GSCs) are known to be aggressive and radio-resistant and proliferate heterogeneously in preferred environments. Additionally, quantitative diffusion and perfusion MRI biomarkers provide insight into the tissue micro-environment. This study assessed the sensitivity of these imaging biomarkers to GSCs in the hyperintensities-FLAIR region, where relapses may occur. A total of 16 patients underwent an MRI session and biopsies were extracted to study the GSCs. In vivo and in vitro biomarkers were compared and both Apparent Diffusion Coefficient (ADC) and relative Cerebral Blood Volume (rCBV) MRI metrics were found to be good predictors of GSCs presence and aggressiveness. Abstract Purpose: With current gold standard treatment, which associates maximum safe surgery and chemo-radiation, the large majority of glioblastoma patients relapse within a year in the peritumoral non contrast-enhanced region (NCE). A subpopulation of glioblastoma stem-like cells (GSC) are known to be particularly radio-resistant and aggressive, and are thus suspected to be the cause of these relapses. Previous studies have shown that their distribution is heterogeneous in the NCE compartment, but no study exists on the sensitivity of medical imaging for localizing these cells. In this work, we propose to study the magnetic resonance (MR) signature of these infiltrative cells. Methods: In the context of a clinical trial on 16 glioblastoma patients, relative Cerebral Blood Volume (rCBV) and Apparent Diffusion Coefficient (ADC) were measured in a preoperative diffusion and perfusion MRI examination. During surgery, two biopsies were extracted using image-guidance in the hyperintensities-FLAIR region. GSC subpopulation was quantified within the biopsies and then cultivated in selective conditions to determine their density and aggressiveness. Results: Low ADC was found to be a good predictor of the time to GSC neurospheres formation in vitro. In addition, GSCs were found in higher concentrations in areas with high rCBV. Conclusions: This study confirms that GSCs have a critical role for glioblastoma aggressiveness and supports the idea that peritumoral sites with low ADC or high rCBV should be preferably removed when possible during surgery and targeted by radiotherapy.
Collapse
Affiliation(s)
- Tanguy Duval
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, 31000 Toulouse, France; (J.-A.L.); (F.T.); (V.L.)
- Correspondence:
| | - Jean-Albert Lotterie
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, 31000 Toulouse, France; (J.-A.L.); (F.T.); (V.L.)
- Department of Nuclear Medicine, CHU Purpan, 31000 Toulouse, France
| | - Anthony Lemarie
- U1037 Toulouse Cancer Research Center CRCT, INSERM, 31000 Toulouse, France; (A.L.); (E.C.-J.M.)
- Université Paul Sabatier Toulouse III, 31000 Toulouse, France
| | - Caroline Delmas
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France;
| | - Fatima Tensaouti
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, 31000 Toulouse, France; (J.-A.L.); (F.T.); (V.L.)
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France;
| | - Elizabeth Cohen-Jonathan Moyal
- U1037 Toulouse Cancer Research Center CRCT, INSERM, 31000 Toulouse, France; (A.L.); (E.C.-J.M.)
- Université Paul Sabatier Toulouse III, 31000 Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, 31000 Toulouse, France;
| | - Vincent Lubrano
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, 31000 Toulouse, France; (J.-A.L.); (F.T.); (V.L.)
- Department of Nuclear Medicine, CHU Purpan, 31000 Toulouse, France
- Service de Neurochirurgie, Clinique de l’Union, 31240 Toulouse, France
| |
Collapse
|
9
|
Collet S, Guillamo JS, Berro DH, Chakhoyan A, Constans JM, Lechapt-Zalcman E, Derlon JM, Hatt M, Visvikis D, Guillouet S, Perrio C, Bernaudin M, Valable S. Simultaneous Mapping of Vasculature, Hypoxia, and Proliferation Using Dynamic Susceptibility Contrast MRI, 18F-FMISO PET, and 18F-FLT PET in Relation to Contrast Enhancement in Newly Diagnosed Glioblastoma. J Nucl Med 2021; 62:1349-1356. [PMID: 34016725 PMCID: PMC8724903 DOI: 10.2967/jnumed.120.249524] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/14/2021] [Indexed: 11/16/2022] Open
Abstract
Conventional MRI plays a key role in the management of patients with high-grade glioma, but multiparametric MRI and PET tracers could provide further information to better characterize tumor metabolism and heterogeneity by identifying regions having a high risk of recurrence. In this study, we focused on proliferation, hypervascularization, and hypoxia, all factors considered indicative of poor prognosis. They were assessed by measuring uptake of 18F-3'-deoxy-3'-18F-fluorothymidine (18F-FLT), relative cerebral blood volume (rCBV) maps, and uptake of 18F-fluoromisonidazole (18F-FMISO), respectively. For each modality, the volumes and high-uptake subvolumes (hot spots) were semiautomatically segmented and compared with the contrast enhancement (CE) volume on T1-weighted gadolinium-enhanced (T1w-Gd) images, commonly used in the management of patients with glioblastoma. Methods: Dynamic susceptibility contrast-enhanced MRI (31 patients), 18F-FLT PET (20 patients), or 18F-FMISO PET (20 patients), for a total of 31 patients, was performed on preoperative glioblastoma patients. Volumes and hot spots were segmented on SUV maps for 18F-FLT PET (using the fuzzy locally adaptive bayesian algorithm) and 18F-FMISO PET (using a mean contralateral image + 3.3 SDs) and on rCBV maps (using a mean contralateral image + 1.96 SDs) for dynamic susceptibility contrast-enhanced MRI and overlaid on T1w-Gd images. For each modality, the percentages of the peripheral volumes and the peripheral hot spots outside the CE volume were calculated. Results: All tumors showed highly proliferated, hypervascularized, and hypoxic regions. The images also showed pronounced heterogeneity of both tracers regarding their uptake and rCBV maps, within each individual patient. Overlaid volumes on T1w-Gd images showed that some proliferative, hypervascularized, and hypoxic regions extended beyond the CE volume but with marked differences between patients. The ranges of peripheral volume outside the CE volume were 1.6%-155.5%, 1.5%-89.5%, and 3.1%-78.0% for 18F-FLT, rCBV, and 18F-FMISO, respectively. All patients had hyperproliferative hot spots outside the CE volume, whereas hypervascularized and hypoxic hot spots were detected mainly within the enhancing region. Conclusion: Spatial analysis of multiparametric maps with segmented volumes and hot spots provides valuable information to optimize the management and treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Solène Collet
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Radiophysics Department, Centre François Baclesse, Caen, France
| | - Jean-Sébastien Guillamo
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neurology, CHU de Caen, Caen, France
- Department of Neurology, CHU de Nimes, Nimes, France
| | - David Hassanein Berro
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neurosurgery, CHU de Caen, Caen, France
| | - Ararat Chakhoyan
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Jean-Marc Constans
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Neuroradiology, CHU de Caen, Caen, France
| | - Emmanuèle Lechapt-Zalcman
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
- Department of Neuropathology, GHU Paris Psychiatry and Neuroscience, Paris, France
| | - Jean-Michel Derlon
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France; and
| | | | - Stéphane Guillouet
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Cécile Perrio
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Myriam Bernaudin
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie University, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France;
| |
Collapse
|
10
|
Maziero D, Straza MW, Ford JC, Bovi JA, Diwanji T, Stoyanova R, Paulson ES, Mellon EA. MR-Guided Radiotherapy for Brain and Spine Tumors. Front Oncol 2021; 11:626100. [PMID: 33763361 PMCID: PMC7982530 DOI: 10.3389/fonc.2021.626100] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
MRI is the standard modality to assess anatomy and response to treatment in brain and spine tumors given its superb anatomic soft tissue contrast (e.g., T1 and T2) and numerous additional intrinsic contrast mechanisms that can be used to investigate physiology (e.g., diffusion, perfusion, spectroscopy). As such, hybrid MRI and radiotherapy (RT) devices hold unique promise for Magnetic Resonance guided Radiation Therapy (MRgRT). In the brain, MRgRT provides daily visualizations of evolving tumors that are not seen with cone beam CT guidance and cannot be fully characterized with occasional standalone MRI scans. Significant evolving anatomic changes during radiotherapy can be observed in patients with glioblastoma during the 6-week fractionated MRIgRT course. In this review, a case of rapidly changing symptomatic tumor is demonstrated for possible therapy adaptation. For stereotactic body RT of the spine, MRgRT acquires clear isotropic images of tumor in relation to spinal cord, cerebral spinal fluid, and nearby moving organs at risk such as bowel. This visualization allows for setup reassurance and the possibility of adaptive radiotherapy based on anatomy in difficult cases. A review of the literature for MR relaxometry, diffusion, perfusion, and spectroscopy during RT is also presented. These techniques are known to correlate with physiologic changes in the tumor such as cellularity, necrosis, and metabolism, and serve as early biomarkers of chemotherapy and RT response correlating with patient survival. While physiologic tumor investigations during RT have been limited by the feasibility and cost of obtaining frequent standalone MRIs, MRIgRT systems have enabled daily and widespread physiologic measurements. We demonstrate an example case of a poorly responding tumor on the 0.35 T MRIgRT system with relaxometry and diffusion measured several times per week. Future studies must elucidate which changes in MR-based physiologic metrics and at which timepoints best predict patient outcomes. This will lead to early treatment intensification for tumors identified to have the worst physiologic responses during RT in efforts to improve glioblastoma survival.
Collapse
Affiliation(s)
- Danilo Maziero
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Michael W Straza
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John C Ford
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Joseph A Bovi
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tejan Diwanji
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Radka Stoyanova
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Eric S Paulson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Eric A Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
11
|
Ruiz-Rodado V, Brender JR, Cherukuri MK, Gilbert MR, Larion M. Magnetic resonance spectroscopy for the study of cns malignancies. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2021; 122:23-41. [PMID: 33632416 PMCID: PMC7910526 DOI: 10.1016/j.pnmrs.2020.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 05/04/2023]
Abstract
Despite intensive research, brain tumors are amongst the malignancies with the worst prognosis; therefore, a prompt diagnosis and thoughtful assessment of the disease is required. The resistance of brain tumors to most forms of conventional therapy has led researchers to explore the underlying biology in search of new vulnerabilities and biomarkers. The unique metabolism of brain tumors represents one potential vulnerability and the basis for a system of classification. Profiling this aberrant metabolism requires a method to accurately measure and report differences in metabolite concentrations. Magnetic resonance-based techniques provide a framework for examining tumor tissue and the evolution of disease. Nuclear Magnetic Resonance (NMR) analysis of biofluids collected from patients suffering from brain cancer can provide biological information about disease status. In particular, urine and plasma can serve to monitor the evolution of disease through the changes observed in the metabolic profiles. Moreover, cerebrospinal fluid can be utilized as a direct reporter of cerebral activity since it carries the chemicals exchanged with the brain tissue and the tumor mass. Metabolic reprogramming has recently been included as one of the hallmarks of cancer. Accordingly, the metabolic rewiring experienced by these tumors to sustain rapid growth and proliferation can also serve as a potential therapeutic target. The combination of 13C tracing approaches with the utilization of different NMR spectral modalities has allowed investigations of the upregulation of glycolysis in the aggressive forms of brain tumors, including glioblastomas, and the discovery of the utilization of acetate as an alternative cellular fuel in brain metastasis and gliomas. One of the major contributions of magnetic resonance to the assessment of brain tumors has been the non-invasive determination of 2-hydroxyglutarate (2HG) in tumors harboring a mutation in isocitrate dehydrogenase 1 (IDH1). The mutational status of this enzyme already serves as a key feature in the clinical classification of brain neoplasia in routine clinical practice and pilot studies have established the use of in vivo magnetic resonance spectroscopy (MRS) for monitoring disease progression and treatment response in IDH mutant gliomas. However, the development of bespoke methods for 2HG detection by MRS has been required, and this has prevented the wider implementation of MRS methodology into the clinic. One of the main challenges for improving the management of the disease is to obtain an accurate insight into the response to treatment, so that the patient can be promptly diverted into a new therapy if resistant or maintained on the original therapy if responsive. The implementation of 13C hyperpolarized magnetic resonance spectroscopic imaging (MRSI) has allowed detection of changes in tumor metabolism associated with a treatment, and as such has been revealed as a remarkable tool for monitoring response to therapeutic strategies. In summary, the application of magnetic resonance-based methodologies to the diagnosis and management of brain tumor patients, in addition to its utilization in the investigation of its tumor-associated metabolic rewiring, is helping to unravel the biological basis of malignancies of the central nervous system.
Collapse
Affiliation(s)
- Victor Ruiz-Rodado
- Neuro-Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institute of Health, Bethesda, United States.
| | - Jeffery R Brender
- Radiation Biology Branch, Center for Cancer Research, National Institute of Health, Bethesda, United States
| | - Murali K Cherukuri
- Radiation Biology Branch, Center for Cancer Research, National Institute of Health, Bethesda, United States
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institute of Health, Bethesda, United States
| | - Mioara Larion
- Neuro-Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institute of Health, Bethesda, United States.
| |
Collapse
|
12
|
Evaluating survival in subjects with astrocytic brain tumors by dynamic susceptibility-weighted perfusion MR imaging. PLoS One 2021; 16:e0244275. [PMID: 33406116 PMCID: PMC7787526 DOI: 10.1371/journal.pone.0244275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/07/2020] [Indexed: 12/02/2022] Open
Abstract
Purpose Studies have evaluated the application of perfusion MR for predicting survival in patients with astrocytic brain tumors, but few of them statistically adjust their results to reflect the impact of the variability of treatment administered in the patients. Our aim was to analyze the association between the perfusion values and overall survival time, with adjustment for various clinical factors, including initial treatments and follow-up treatments. Materials and methods This study consisted of 51 patients with astrocytic brain tumors who underwent perfusion-weighted MRI with MultiHance® at a dose of 0.1 mmol/kg prior to initial surgery. We measured the mean rCBV, the 5% & 10% maximum rCBV, and the variation of rCBV in the tumors. Comparisons were made between patients with and without 2-year survival using two-sample t-test or Wilcoxon rank-sum test for the continuous data, or chi-square and Fisher exact tests for categorical data. The multivariate cox-proportional hazard regression was fit to evaluate the association between rCBV and overall survival time, with adjustment for clinical factors. Results Patients who survived less than 2 years after diagnosis had a higher mean and maximum rCBV and a larger variation of rCBV. After adjusting for clinical factors including therapeutic measures, we found no significant association of overall survival time within 2 years with any of these rCBV values. Conclusions Although patients who survived less than 2 years had a higher mean and maximum rCBV and a larger variation of rCBV, rCBV itself may not be used independently for predicting 2-year survival of patients with astrocytic brain tumors.
Collapse
|
13
|
Ahmed HAK, Mokhtar H. The diagnostic value of MR spectroscopy versus DWI-MRI in therapeutic planning of suspicious multi-centric cerebral lesions. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2020. [DOI: 10.1186/s43055-020-00154-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Background
A broad spectrum of non-neoplastic lesions can radiologically mimic cerebral neoplasms. Magnetic resonance imaging (MRI), magnetic resonance spectroscopy (MRS), and diffusion-weighted imaging (DWI) are the most extensively used for enabling lesional characterization of different brain disorders. We aimed to assess the diagnostic value of MRS versus DWI in the diagnosis and therapeutic planning of multicentric cerebral focal lesions and in our retrospective study, we enrolled 64 patients with 100 brain lesions who underwent pre- and post-contrast MRI, MRS, and DWI. Diagnoses supplied by the histopathology and follow up clinical results as a gold standard. Sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and diagnostic accuracy were calculated.
Results
Conventional MRI poorly differentiates multiple cerebral lesions with 89.33% sensitivity, 44.4% specificity, and 78% accuracy. MRS results revealed statistical significance for differentiating neoplastic from non-neoplastic lesions as regards Cho/Cr, Cho/NAA, and NAA/Cr ratios (M ± SD) with P < 0.001 (significant), and there is statistical significance for neoplastic lesion differentiation when Cho/NAA and Ch/Cr ratios measured in the pre-lesional areas outside the tumor margin. DWI showed mixed diffusion changes in most of the studied lesions and the measured ADC values ranges showed overlap in neoplastic and non-neoplastic lesions, P value = 0.236* (insignificant).
Conclusion
MRS was found to be a more accurate diagnostic tool than DWI with ADC measurements in the differentiation and therapeutic planning of multicentric cerebral focal lesions.
Collapse
|
14
|
Oberheim Bush NA, Hervey-Jumper SL, Berger MS. Management of Glioblastoma, Present and Future. World Neurosurg 2020; 131:328-338. [PMID: 31658576 DOI: 10.1016/j.wneu.2019.07.044] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 01/22/2023]
Abstract
Glioblastomas are the most common malignant brain tumor and despite extensive research have a dismal prognosis. This review focuses on the current treatment paradigms of glioblastoma and highlights current advances in surgical approaches, imaging techniques, molecular diagnostics, and translational efforts. Several promising clinical trials in immunotherapy and personalized medicine are discussed and the importance of quality of life in the patients and their caregivers both during active treatment and survivorship is also commented on.
Collapse
Affiliation(s)
- Nancy Ann Oberheim Bush
- Division of Neuro-Oncology, Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Shawn L Hervey-Jumper
- Division of Neuro-Oncology, Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA.
| |
Collapse
|
15
|
Diffusion- and perfusion-weighted MRI radiomics model may predict isocitrate dehydrogenase (IDH) mutation and tumor aggressiveness in diffuse lower grade glioma. Eur Radiol 2019; 30:2142-2151. [PMID: 31828414 DOI: 10.1007/s00330-019-06548-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/11/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To determine whether diffusion- and perfusion-weighted MRI-based radiomics features can improve prediction of isocitrate dehydrogenase (IDH) mutation and tumor aggressiveness in lower grade gliomas (LGGs) METHODS: Radiomics features (n = 6472) were extracted from multiparametric MRI including conventional MRI, apparent diffusion coefficient (ADC), and normalized cerebral blood volume, acquired on 127 LGG patients with determined IDH mutation status and grade (WHO II or III). Radiomics models were constructed using machine learning-based feature selection and generalized linear model classifiers. Segmentation stability was calculated between two readers using concordance correlation coefficients (CCCs). Diagnostic performance to predict IDH mutation and tumor grade was compared between the multiparametric and conventional MRI radiomics models using the area under the receiver operating characteristics curve (AUC). The models were tested using a temporally independent validation set (n = 28). RESULTS The multiparametric MRI radiomics model was optimized with a random forest feature selector, with segmentation stability of a CCC threshold of 0.8. For IDH mutation, multiparametric MR radiomics showed similar performance (AUC 0.795) to the conventional radiomics model (AUC 0.729). In tumor grading, multiparametric model with ADC features showed higher performance (AUC 0.932) than the conventional model (AUC 0.555). The independent validation set showed the same trend with AUCs of 0.747 for IDH prediction and 0.819 for tumor grading with multiparametric MRI radiomics model. CONCLUSION Multiparametric MRI radiomics model showed improved diagnostic performance in tumor grading and comparable diagnostic performance in IDH mutation status, with ADC features playing a significant role. KEY POINTS • The multiparametric MRI radiomics model was comparable with conventional MRI radiomics model in predicting IDH mutation. • The multiparametric MRI radiomics model outperformed conventional MRI in glioma grading. • Apparent diffusion coefficient played an important role in glioma grading and predicting IDH mutation status using radiomics.
Collapse
|
16
|
van Dijken BRJ, Jan van Laar P, Li C, Yan JL, Boonzaier NR, Price SJ, van der Hoorn A. Ventricle contact is associated with lower survival and increased peritumoral perfusion in glioblastoma. J Neurosurg 2019; 131:717-723. [PMID: 30485234 DOI: 10.3171/2018.5.jns18340] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/02/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The purpose of this study was to prospectively investigate outcome and differences in peritumoral MRI characteristics of glioblastomas (GBMs) that were in contact with the ventricles (ventricle-contacting tumors) and those that were not (noncontacting tumors). GBMs are heterogeneous tumors with variable survival. Lower survival is suggested for patients with ventricle-contacting tumors than for those with noncontacting tumors. This might be supported by aggressive peritumoral MRI features. However, differences in MRI characteristics of the peritumoral environment between ventricle-contacting and noncontacting GBMs have not yet been investigated. METHODS Patients with newly diagnosed GBM underwent preoperative MRI with contrast-enhanced T1-weighted, FLAIR, diffusion-weighted, and perfusion-weighted sequences. Tumors were categorized into ventricle-contacting or noncontacting based on contrast enhancement. Survival analysis was performed using log-rank for univariate analysis and Cox regression for multivariate analysis. Normalized perfusion (relative cerebral blood volume [rCBV]) and diffusion (apparent diffusion coefficient [ADC]) values were calculated in 2 regions: the peritumoral nonenhancing FLAIR region overlapping the subventricular zone and the remaining peritumoral nonenhancing FLAIR region. RESULTS Overall survival was significantly lower for patients with contacting tumors than for those with noncontacting tumors (434 vs 747 days, p < 0.001). Progression-free survival showed a comparable trend (260 vs 375 days, p = 0.094). Multivariate analysis confirmed a survival difference for both overall survival (HR 3.930, 95% CI 1.740-8.875, p = 0.001) and progression-free survival (HR 2.506, 95% CI 1.254-5.007, p = 0.009). Peritumoral perfusion was higher in contacting than in noncontacting tumors for both FLAIR regions (p = 0.04). There was no difference in peritumoral ADC values between the 2 groups. CONCLUSIONS Patients with ventricle-contacting tumors had poorer outcomes than patients with noncontacting tumors. This disadvantage of ventricle contact might be explained by higher peritumoral perfusion leading to more aggressive behavior.
Collapse
Affiliation(s)
- Bart Roelf Jan van Dijken
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter Jan van Laar
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chao Li
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom.,3Department of Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Jiun-Lin Yan
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom.,4Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Taiwan; and.,5Department of Neurosurgery, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Natalie Rosella Boonzaier
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| | | | -
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| | - Anouk van der Hoorn
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Gharzeddine K, Hatzoglou V, Holodny AI, Young RJ. MR Perfusion and MR Spectroscopy of Brain Neoplasms. Radiol Clin North Am 2019; 57:1177-1188. [PMID: 31582043 DOI: 10.1016/j.rcl.2019.07.008] [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: 12/30/2022]
Abstract
Advances in imaging techniques, such as MR perfusion and spectroscopy, are increasingly indispensable in the management and treatment plans of brain neoplasms: from diagnosing, molecular/genetic typing and grading neoplasms, augmenting biopsy results and improving accuracy, to ultimately directing and monitoring treatment and response. New developments in treatment methods have resulted in new diagnostic challenges for conventional MR imaging, such as pseudoprogression, where MR perfusion has the widest current application. MR spectroscopy is showing increasing promise in noninvasively determining genetic subtypes and, potentially, susceptibility to molecular targeted therapies.
Collapse
Affiliation(s)
- Karem Gharzeddine
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, 1275 York Avenue, New York, NY 10065, USA
| | - Andrei I Holodny
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, Weill Cornell Graduate School of Medical Sciences, 1275 York Avenue, New York, NY 10065, USA.
| | - Robert J Young
- Brain Imaging, Neuroradiology Research, Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
18
|
Zhang J, Jiang J, Zhao L, Zhang J, Shen N, Li S, Guo L, Su C, Jiang R, Zhu W. Survival prediction of high-grade glioma patients with diffusion kurtosis imaging. Am J Transl Res 2019; 11:3680-3688. [PMID: 31312379 PMCID: PMC6614625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/25/2019] [Indexed: 06/10/2023]
Abstract
PURPOSE To evaluate the prognostic value of diffusion kurtosis imaging (DKI) for survival prediction of patients with high-grade glioma (HGG). MATERIALS AND METHODS DKI was performed for fifty-eight patients with pathologically proven HGG by using a 3-T scanner. The mean kurtosis (MK), mean diffusivity (MD) and fractional anisotropy (FA) values in the solid part of the tumor were measured and normalized. Univariate Cox regression analysis was used to evaluate the association between overall survival (OS) and sex, age, Karnofsky performance status (KPS), tumor grade, Ki-67 labeling index (LI), extent of resection, use of chemoradiotherapy, MK, MD, and FA. Multivariate Cox regression analysis including sex, age, KPS, extent of resection, use of chemoradiotherapy, MK, MD, and FA was subsequently performed. Spearman's correlation coefficient for OS and the area under receiver operating characteristic curve (AUC) for predicting 2-year survival were calculated for each DKI parameter and further compared. RESULTS In univariate Cox regression analyses, OS was significantly associated with the tumor grade, Ki-67 LI, extent of resection, use of chemoradiotherapy, MK, and MD (P < 0.05 for all). Multivariate Cox regression analyses indicated that MK, MD (hazard ratio = 1.582 and 0.828, respectively, for each 0.1 increase in the normalized value), extent of resection and use of chemoradiotherapy were independent predictors of OS. The absolute value of the correlation coefficient for OS and AUC for predicting 2-year survival by MK (rho = -0.565, AUC = 0.841) were higher than those by MD (rho = 0.492, AUC = 0.772), but the difference was not significant. CONCLUSION DKI is a promising tool to predict the survival of HGG patients. MK and MD are independent predictors. MK is potentially better associated with OS than MD, which may lead to a more accurate evaluation of HGG patient survival.
Collapse
Affiliation(s)
- Ju Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Jingjing Jiang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Lingyun Zhao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Jiaxuan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Nanxi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Shihui Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Linying Guo
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Changliang Su
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| | - Rifeng Jiang
- Department of Radiology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, P. R. China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, P. R. China
| |
Collapse
|
19
|
Buemi F, Guzzardi G, Del Sette B, Sponghini AP, Matheoud R, Soligo E, Trisoglio A, Carriero A, Stecco A. Apparent diffusion coefficient and tumor volume measurements help stratify progression-free survival of bevacizumab-treated patients with recurrent glioblastoma multiforme. Neuroradiol J 2019; 32:241-249. [PMID: 31066622 DOI: 10.1177/1971400919847184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The aim of this study was to determine whether apparent diffusion coefficient (ADC) bi-component curve-fitting histogram analysis and volume percentage change (VPC) prior to bevacizumab treatment can stratify progression-free survival (PFS) and overall survival (OS) in patients with glioblastoma multiforme (GBM) on first recurrence. METHODS We retrospectively evaluated 17 patients with recurrent GBM who received bevacizumab and fotemustine (n = 13) or only bevacizumab (n = 4) on first recurrence at our institution between December 2009 and July 2015. Both T2/FLAIR abnormalities and enhancing tumor on T1 images were mapped to the ADC images. ADC-L and ADC-M values were obtained trough bi-Gaussian curve fitting histogram analysis. Furthermore, the study population was dichotomized into two subgroups: patients displaying a reduction in enhancing tumor volume of either >55% or <55% between the mean value calculated at baseline and first follow-up. Subsequently, a second dichotomization was performed according to a reduction in the T2 / FLAIR volume >41% or <41% at first check after treatment. OS and PFS were assessed using volume parameters in a Cox regression model adjusted for significant clinical parameters. RESULTS In univariate analysis, contrast-enhanced (CE)-ADC-L was significantly predictive of PFS (p = 0.01) and OS (p = 0.03). When we dichotomized our sample using the 55% cut-off for enhancing tumor volume, CE-VPC was able to predict PFS (p = 0.01) but not OS (p = 0.08). In multivariate analysis, only the CE-ADC-L was predictive of PFS (p = 0.01), albeit not predictive of OS (p = 0.14). CE-ADC-M, T2/FLAIR-ADC-L, T2/FLAIR-ADC, and T2/FLAIR VPC were not significantly predictive of PFS and OS (p > 0.05) in both univariate and multivariate analysis. CONCLUSIONS CE-ADC and CE-VPC can stratify PFS for patients with recurrent glioblastoma prior to bevacizumab treatment.
Collapse
Affiliation(s)
| | - Giuseppe Guzzardi
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Bruno Del Sette
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Andrea P Sponghini
- 3 Oncology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Roberta Matheoud
- 4 Medical Physics Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Eleonora Soligo
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Alessandra Trisoglio
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Alessandro Carriero
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| | - Alessandro Stecco
- 2 Radiology Department, University of Eastern Piedmont, "Maggiore della Carità" Hospital, Novara, Italy
| |
Collapse
|
20
|
Laprie A, Ken S, Filleron T, Lubrano V, Vieillevigne L, Tensaouti F, Catalaa I, Boetto S, Khalifa J, Attal J, Peyraga G, Gomez-Roca C, Uro-Coste E, Noel G, Truc G, Sunyach MP, Magné N, Charissoux M, Supiot S, Bernier V, Mounier M, Poublanc M, Fabre A, Delord JP, Cohen-Jonathan Moyal E. Dose-painting multicenter phase III trial in newly diagnosed glioblastoma: the SPECTRO-GLIO trial comparing arm A standard radiochemotherapy to arm B radiochemotherapy with simultaneous integrated boost guided by MR spectroscopic imaging. BMC Cancer 2019; 19:167. [PMID: 30791889 PMCID: PMC6385401 DOI: 10.1186/s12885-019-5317-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 01/24/2019] [Indexed: 02/05/2023] Open
Abstract
Background Glioblastoma, a high-grade glial infiltrating tumor, is the most frequent malignant brain tumor in adults and carries a dismal prognosis. External beam radiotherapy (EBRT) increases overall survival but this is still low due to local relapses, mostly occurring in the irradiation field. As the ratio of spectra of choline/N acetyl aspartate> 2 (CNR2) on MR spectroscopic imaging has been described as predictive for the site of local relapse, we hypothesized that dose escalation on these regions would increase local control and hence global survival. Methods/design In this multicenter prospective phase III trial for newly diagnosed glioblastoma, 220 patients having undergone biopsy or surgery are planned for randomization to two arms. Arm A is the Stupp protocol (EBRT 60 Gy on contrast enhancement + 2 cm margin with concomitant temozolomide (TMZ) and 6 months of TMZ maintenance); Arm B is the same treatment with an additional simultaneous integrated boost of intensity-modulated radiotherapy (IMRT) of 72Gy/2.4Gy delivered on the MR spectroscopic imaging metabolic volumes of CHO/NAA > 2 and contrast-enhancing lesions or resection cavity. Stratification is performed on surgical and MGMT status. Discussion This is a dose-painting trial, i.e. delivery of heterogeneous dose guided by metabolic imaging. The principal endpoint is overall survival. An online prospective quality control of volumes and dose is performed in the experimental arm. The study will yield a large amount of longitudinal multimodal MR imaging data including planning CT, radiotherapy dosimetry, MR spectroscopic, diffusion and perfusion imaging. Trial registration NCT01507506, registration date December 20, 2011.
Collapse
Affiliation(s)
- Anne Laprie
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France. .,ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, INSERM, UPS, Toulouse, France.
| | - Soléakhéna Ken
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, INSERM, UPS, Toulouse, France.,Department of Engineering and Medical Physics, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-OncopoleCancer de Toulouse-Oncopole, Toulouse, France
| | - Thomas Filleron
- Biostatistics Unit, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Vincent Lubrano
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, INSERM, UPS, Toulouse, France.,Neurosurgery Department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Laure Vieillevigne
- Department of Engineering and Medical Physics, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-OncopoleCancer de Toulouse-Oncopole, Toulouse, France
| | - Fatima Tensaouti
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Isabelle Catalaa
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, INSERM, UPS, Toulouse, France.,Neuroimaging Department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Sergio Boetto
- Neurosurgery Department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jonathan Khalifa
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Justine Attal
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Guillaume Peyraga
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Carlos Gomez-Roca
- Medical Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Emmanuelle Uro-Coste
- Pathology department, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Georges Noel
- Radiation Oncology Department, Centre Paul Strauss, Strasbourg, France
| | - Gilles Truc
- Radiation Oncology Department Centre Georges-François Leclerc, Dijon, France
| | | | - Nicolas Magné
- Radiation Oncology Department, Institut de Cancérologie de la Loire, Saint-Priest en Jarez, France
| | - Marie Charissoux
- Radiation Oncology Department - Centre Val d'aurelle, Montpellier, France
| | - Stéphane Supiot
- Radiation Oncology Department, Institut de Cancerologie de l'Ouest, Nantes st Herblain, France
| | - Valérie Bernier
- Radiation Oncology Department, Institut de cancérologie de Lorraine centre Alexis Vautrin, Nancy, France
| | - Muriel Mounier
- Clinical Research Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Muriel Poublanc
- Clinical Research Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Amandine Fabre
- Clinical Research Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jean-Pierre Delord
- Medical Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- Radiation Oncology Department, Institut Claudius Regaud- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.,INSERM UMR1037, Cancer Research Center of Toulouse, Oncopole, Toulouse, France
| |
Collapse
|
21
|
Peyraga G, Robaine N, Khalifa J, Cohen-Jonathan-Moyal E, Payoux P, Laprie A. Molecular PET imaging in adaptive radiotherapy: brain. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:337-348. [PMID: 30497232 DOI: 10.23736/s1824-4785.18.03116-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Owing to their heterogeneity and radioresistance, the prognosis of primitive brain tumors, which are mainly glial tumors, remains poor. Dose escalation in radioresistant areas is a potential issue for improving local control and overall survival. This review focuses on advances in biological and metabolic imaging of brain tumors that are proving to be essential for defining tumor target volumes in radiation therapy (RT) and for increasing the use of DPRT (dose painting RT) and ART (adaptative RT), to optimize dose in radio-resistant areas. EVIDENCE ACQUISITION Various biological imaging modalities such as PET (hypoxia, glucidic metabolism, protidic metabolism, cellular proliferation, inflammation, cellular membrane synthesis) and MRI (spectroscopy) may be used to identify these areas of radioresistance. The integration of these biological imaging modalities improves the diagnosis, prognosis and treatment of brain tumors. EVIDENCE SYNTHESIS Technological improvements (PET and MRI), the development of research, and intensive cooperation between different departments are necessary before using daily metabolic imaging (PET and MRI) to treat patients with brain tumors. CONCLUSIONS The adaptation of treatment volumes during RT (ART) seems promising, but its development requires improvements in several areas and an interdisciplinary approach involving radiology, nuclear medicine and radiotherapy. We review the literature on biological imaging to outline the perspectives for using DPRT and ART in brain tumors.
Collapse
Affiliation(s)
- Guillaume Peyraga
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Nesrine Robaine
- Department of Nuclear Medicine, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Jonathan Khalifa
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Paul Sabatier University, Toulouse III, Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,Paul Sabatier University, Toulouse III, Toulouse, France
| | - Pierre Payoux
- Department of Nuclear Medicine, Purpan University Hospital Center, Toulouse, France
| | - Anne Laprie
- Department of Radiation Therapy, Claudius Regaud Institute, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France - .,Paul Sabatier University, Toulouse III, Toulouse, France
| |
Collapse
|
22
|
Cordova JS, Gurbani SS, Holder CA, Olson JJ, Schreibmann E, Shi R, Guo Y, Shu HKG, Shim H, Hadjipanayis CG. Semi-Automated Volumetric and Morphological Assessment of Glioblastoma Resection with Fluorescence-Guided Surgery. Mol Imaging Biol 2017; 18:454-62. [PMID: 26463215 DOI: 10.1007/s11307-015-0900-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Glioblastoma (GBM) neurosurgical resection relies on contrast-enhanced MRI-based neuronavigation. However, it is well-known that infiltrating tumor extends beyond contrast enhancement. Fluorescence-guided surgery (FGS) using 5-aminolevulinic acid (5-ALA) was evaluated to improve extent of resection (EOR) of GBMs. Preoperative morphological tumor metrics were also assessed. PROCEDURES Thirty patients from a phase II trial evaluating 5-ALA FGS in newly diagnosed GBM were assessed. Tumors were segmented preoperatively to assess morphological features as well as postoperatively to evaluate EOR and residual tumor volume (RTV). RESULTS Median EOR and RTV were 94.3 % and 0.821 cm(3), respectively. Preoperative surface area to volume ratio and RTV were significantly associated with overall survival, even when controlling for the known survival confounders. CONCLUSIONS This study supports claims that 5-ALA FGS is helpful at decreasing tumor burden and prolonging survival in GBM. Moreover, morphological indices are shown to impact both resection and patient survival.
Collapse
Affiliation(s)
- J Scott Cordova
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Saumya S Gurbani
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Chad A Holder
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Ran Shi
- Department of Biostatistics, Emory University School of Public Health, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Ying Guo
- Department of Biostatistics, Emory University School of Public Health, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Hyunsuk Shim
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA. .,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA.
| | - Costas G Hadjipanayis
- Department of Neurosurgery, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA. .,Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA. .,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, 10 Union Square, 5th Floor, Suite 5E, New York, NY, 10003, USA.
| |
Collapse
|
23
|
Nelson SJ, Kadambi AK, Park I, Li Y, Crane J, Olson M, Molinaro A, Roy R, Butowski N, Cha S, Chang S. Association of early changes in 1H MRSI parameters with survival for patients with newly diagnosed glioblastoma receiving a multimodality treatment regimen. Neuro Oncol 2017; 19:430-439. [PMID: 27576874 DOI: 10.1093/neuonc/now159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
Background The heterogeneous biology of glioblastoma (GBM) emphasizes the need for imaging methods to assess tumor burden and assist in evaluating individual patients. The purpose of this study was to investigate early changes in metrics from 3D 1H magnetic resonance spectroscopic imaging (MRSI) data, compare them with anatomic lesion volumes, and determine whether they were associated with survival for patients with newly diagnosed GBM receiving a multimodality treatment regimen. Methods Serial MRI and MRSI scans provided estimates of anatomic lesion volumes and levels of choline, creatine, N-acetylaspartate, lactate, and lipid. The association of metrics derived from these data with survival was assessed using Cox proportional hazards models with adjustments for age, Karnofsky performance score, and extent of resection. Temporal changes in parameters were evaluated using a Wilcoxon signed rank test. Results Anatomic lesion volumes at the post-radiotherapy (RT) scan, metabolic lesion volume at mid-RT and post-RT scans, as well as metrics describing levels of choline, lactate, and lipid were associated with overall survival. There was a significant reduction in the enhancing lesion volume, increase in T2 lesion volume from mid-RT to post-RT, and decrease in parameters describing metabolite levels during these early time points. Conclusion The MRSI data provided metrics that described the effects of treatment on the metabolic lesion burden and were associated with overall survival. This suggests that adding these parameters to standard assessments of changes in anatomic lesion volumes could contribute to making early decisions about the efficacy of such combination therapies.
Collapse
Affiliation(s)
- Sarah J Nelson
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Achuta K Kadambi
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Ilwoo Park
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Yan Li
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Jason Crane
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Marram Olson
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Annette Molinaro
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, California
| | - Ritu Roy
- Surbeck Laboratory of Advanced Imaging, University of California, San Francisco, California
| | - Nicholas Butowski
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, California
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Department of Neurological Surgery, University of California, San Francisco, California
| | - Susan Chang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| |
Collapse
|
24
|
Anwar M, Molinaro AM, Morin O, Chang SM, Haas-Kogan DA, Nelson SJ, Lupo JM. Identifying Voxels at Risk for Progression in Glioblastoma Based on Dosimetry, Physiologic and Metabolic MRI. Radiat Res 2017; 188:303-313. [PMID: 28723274 PMCID: PMC5628052 DOI: 10.1667/rr14662.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite the longstanding role of radiation in cancer treatment and the presence of advanced, high-resolution imaging techniques, delineation of voxels at-risk for progression remains purely a geometric expansion of anatomic images, missing subclinical disease at risk for recurrence while treating potentially uninvolved tissue and increasing toxicity. This remains despite the modern ability to precisely shape radiation fields. A striking example of this is the treatment of glioblastoma, a highly infiltrative tumor that may benefit from accurate identification of subclinical disease. In this study, we hypothesize that parameters from physiologic and metabolic magnetic resonance imaging (MRI) at diagnosis could predict the likelihood of voxel progression at radiographic recurrence in glioblastoma by identifying voxel characteristics that indicate subclinical disease. Integrating dosimetry can reveal its effect on voxel outcome, enabling risk-adapted voxel dosing. As a system example, 24 patients with glioblastoma treated with radiotherapy, temozolomide and an anti-angiogenic agent were analyzed. Pretreatment median apparent diffusion coefficient (ADC), fractional anisotropy (FA), relative cerebral blood volume (rCBV), vessel leakage (percentage recovery), choline-to-NAA index (CNI) and dose of voxels in the T2 nonenhancing lesion (NEL), T1 post-contrast enhancing lesion (CEL) or normal-appearing volume (NAV) of brain, were calculated for voxels that progressed [NAV→NEL, CEL (N = 8,765)] and compared against those that remained stable [NAV→NAV (N = 98,665)]. Voxels that progressed (NAV→NEL) had significantly different (P < 0.01) ADC (860), FA (0.36) and CNI (0.67) versus stable voxels (804, 0.43 and 0.05, respectively), indicating increased cell turnover, edema and decreased directionality, consistent with subclinical disease. NAV→CEL voxels were more abnormal (1,014, 0.28, 2.67, respectively) and leakier (percentage recovery = 70). A predictive model identified areas of recurrence, demonstrating that elevated CNI potentiates abnormal diffusion, even far (>2 cm) from the tumor and dose escalation >45 Gy has diminishing benefits. Integrating advanced MRI with dosimetry can identify at voxels at risk for progression and may allow voxel-level risk-adapted dose escalation to subclinical disease while sparing normal tissue. When combined with modern planning software, this technique may enable risk-adapted radiotherapy in any disease site with multimodal imaging.
Collapse
Affiliation(s)
- Mekhail Anwar
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Annette M. Molinaro
- Department of Neurosurgery, Division of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Olivier Morin
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Susan M. Chang
- Department of Neurosurgery, Division of Neuro-oncology, University of California, San Francisco, California
| | - Daphne A. Haas-Kogan
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Sarah J. Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| |
Collapse
|
25
|
Volumetric quantification of glioblastoma: experiences with different measurement techniques and impact on survival. J Neurooncol 2017; 135:391-402. [DOI: 10.1007/s11060-017-2587-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/25/2017] [Indexed: 11/27/2022]
|
26
|
Boonzaier NR, Larkin TJ, Matys T, van der Hoorn A, Yan JL, Price SJ. Multiparametric MR Imaging of Diffusion and Perfusion in Contrast-enhancing and Nonenhancing Components in Patients with Glioblastoma. Radiology 2017; 284:180-190. [DOI: 10.1148/radiol.2017160150] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Natalie R. Boonzaier
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| | - Timothy J. Larkin
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| | - Tomasz Matys
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| | - Anouk van der Hoorn
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| | - Jiun-Lin Yan
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| | - Stephen J. Price
- From the Cambridge Brain Tumour Imaging Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences (N.R.B., T.J.L., A.v.d.H., J.L.Y., S.J.P.), Wolfson Brain Imaging Centre, Department of Clinical Neurosciences (N.R.B., T.J.L., J.L.Y., S.J.P.), and Department of Radiology (T.M., A.v.d.H.), University of Cambridge, Cambridge, England; Cancer Trials Unit Department of Oncology, Addenbrooke’s Hospital, Cambridge, England (T.M.); Department of Radiology, University Medical Centre Groningen,
| |
Collapse
|
27
|
Brandão LA, Castillo M. Adult Brain Tumors: Clinical Applications of Magnetic Resonance Spectroscopy. Magn Reson Imaging Clin N Am 2017; 24:781-809. [PMID: 27742117 DOI: 10.1016/j.mric.2016.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Proton magnetic resonance spectroscopy (H-MRS) may be helpful in suggesting tumor histology and tumor grade and may better define tumor extension and the ideal site for biopsy compared with conventional magnetic resonance (MR) imaging. A multifunctional approach with diffusion-weighted imaging, perfusion-weighted imaging, and permeability maps, along with H-MRS, may enhance the accuracy of the diagnosis and characterization of brain tumors and estimation of therapeutic response. Integration of advanced imaging techniques with conventional MR imaging and the clinical history help to improve the accuracy, sensitivity, and specificity in differentiating tumors and nonneoplastic lesions.
Collapse
Affiliation(s)
- Lara A Brandão
- Clínica Felippe Mattoso, Av. Das Américas 700, sala 320, Barra da Tijuca, Rio de Janeiro 30112011, Brazil; Clínica IRM- Ressonância Magnética, Rua Capitão Salomão 44 Humaitá, Rio de Janeiro 22271040, Brazil.
| | - Mauricio Castillo
- Division of Neuroradiology, Department of Radiology, University of North Carolina School of Medicine, Room 3326, Old Infirmary Building, Manning Drive, Chapel Hill, NC 27599-7510, USA
| |
Collapse
|
28
|
Cata JP, Bhavsar S, Hagan KB, Arunkumar R, Grasu R, Dang A, Carlson R, Arnold B, Popat K, Rao G, Potylchansky Y, Lipski I, Ratty S, Nguyen AT, McHugh T, Feng L, Rahlfs TF. Intraoperative serum lactate is not a predictor of survival after glioblastoma surgery. J Clin Neurosci 2017; 43:224-228. [PMID: 28601568 DOI: 10.1016/j.jocn.2017.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/21/2017] [Accepted: 05/21/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND Cancer cells can produce lactate in high concentrations. Two previous studies examined the clinical relevance of serum lactate as a biomarker in patients with brain tumors. Patients with high-grade tumors have higher serum concentrations of lactate than those with low-grade tumors. We hypothesized that serum lactic could be used of biomarker to predictor of survival in patients with glioblastoma (GB). METHODS This was a retrospective study. Demographic, lactate concentrations and imaging data from 275 adult patients with primary GB was included in the analysis. The progression free survival (PFS) and overall survival (OS) rates were compared in patients who had above and below the median concentrations of lactate. We also investigated the correlation between lactate concentrations and tumor volume. Multivariate analyses were conducted to test the association lactate, tumor volume and demographic variables with PFS and OS. RESULTS The median serum concentration of lactate was 2.3mmol/L. A weak correlation was found between lactate concentrations and tumor volume. Kaplan-Meier curves demonstrated similar survival in patients with higher or lower than 2.3mmol/L of lactate. The multivariate analysis indicated that the intraoperative levels of lactate were not independently associated with changes in survival. On another hand, a preoperative T1 volume was an independent predictor PFS (HR 95%CI: 1.41, 1.02-1.82, p=0.006) and OS (HR 95%CI: 1.47, 1.11-1.96, p=0.006). CONCLUSION This retrospective study suggests that the serum concentrations of lactate cannot be used as a biomarker to predict survival after GB surgery. To date, there are no clinically available serum biomarkers to determine prognosis in patients with high-grade gliomas. These tumors may produce high levels of lactic acid. We hypothesized that serum lactic could be used of biomarker to predictor of survival in patients with glioblastoma (GB). In this study, we collected perioperative and survival data from 275 adult patients with primary high-grade gliomas to determine whether intraoperative serum acid lactic concentrations can serve as a marker of prognosis. The median serum concentration of lactate was 2.3mmol/L. Our analysis indicated the intraoperative levels of lactate were not independently associated with changes in survival. This retrospective study suggests that the serum concentrations of lactate cannot be used as a biomarker to predict survival after GB surgery.
Collapse
Affiliation(s)
- J P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.
| | - S Bhavsar
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - K B Hagan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Arunkumar
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Grasu
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - A Dang
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - R Carlson
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - B Arnold
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - K Popat
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Y Potylchansky
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - I Lipski
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Sally Ratty
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - A T Nguyen
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas McHugh
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - L Feng
- Department of Biostatistics, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - T F Rahlfs
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Krishnan AP, Karunamuni R, Leyden KM, Seibert TM, Delfanti RL, Kuperman JM, Bartsch H, Elbe P, Srikant A, Dale AM, Kesari S, Piccioni DE, Hattangadi-Gluth JA, Farid N, McDonald CR, White NS. Restriction Spectrum Imaging Improves Risk Stratification in Patients with Glioblastoma. AJNR Am J Neuroradiol 2017; 38:882-889. [PMID: 28279985 PMCID: PMC5507368 DOI: 10.3174/ajnr.a5099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE ADC as a marker of tumor cellularity has been promising for evaluating the response to therapy in patients with glioblastoma but does not successfully stratify patients according to outcomes, especially in the upfront setting. Here we investigate whether restriction spectrum imaging, an advanced diffusion imaging model, performed after an operation but before radiation therapy, could improve risk stratification in patients with newly diagnosed glioblastoma relative to ADC. MATERIALS AND METHODS Pre-radiation therapy diffusion-weighted and structural imaging of 40 patients with glioblastoma were examined retrospectively. Restriction spectrum imaging and ADC-based hypercellularity volume fraction (restriction spectrum imaging-FLAIR volume fraction, restriction spectrum imaging-contrast-enhanced volume fraction, ADC-FLAIR volume fraction, ADC-contrast-enhanced volume fraction) and intensities (restriction spectrum imaging-FLAIR 90th percentile, restriction spectrum imaging-contrast-enhanced 90th percentile, ADC-FLAIR 10th percentile, ADC-contrast-enhanced 10th percentile) within the contrast-enhanced and FLAIR hyperintensity VOIs were calculated. The association of diffusion imaging metrics, contrast-enhanced volume, and FLAIR hyperintensity volume with progression-free survival and overall survival was evaluated by using Cox proportional hazards models. RESULTS Among the diffusion metrics, restriction spectrum imaging-FLAIR volume fraction was the strongest prognostic metric of progression-free survival (P = .036) and overall survival (P = .007) in a multivariate Cox proportional hazards analysis, with higher values indicating earlier progression and shorter survival. Restriction spectrum imaging-FLAIR 90th percentile was also associated with overall survival (P = .043), with higher intensities, indicating shorter survival. None of the ADC metrics were associated with progression-free survival/overall survival. Contrast-enhanced volume exhibited a trend toward significance for overall survival (P = .063). CONCLUSIONS Restriction spectrum imaging-derived cellularity in FLAIR hyperintensity regions may be a more robust prognostic marker than ADC and conventional imaging for early progression and poorer survival in patients with glioblastoma. However, future studies with larger samples are needed to explore its predictive ability.
Collapse
Affiliation(s)
- A P Krishnan
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
| | - R Karunamuni
- Departments of Radiation Medicine (R.K., T.M.S., J.A.H.-G., C.R.M.)
| | - K M Leyden
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
| | - T M Seibert
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Departments of Radiation Medicine (R.K., T.M.S., J.A.H.-G., C.R.M.)
| | - R L Delfanti
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
| | - J M Kuperman
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
| | - H Bartsch
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
| | - P Elbe
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
| | - A Srikant
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
| | - A M Dale
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
- Neurosciences (A.M.D., D.E.P.)
| | - S Kesari
- Department of Translational Neuro-Oncology and Neurotherapeutics (S.K.), John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John's Health Center, Santa Monica, California
| | | | | | - N Farid
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
| | - C R McDonald
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Departments of Radiation Medicine (R.K., T.M.S., J.A.H.-G., C.R.M.)
- Psychiatry (C.R.M.), University of California, San Diego, La Jolla, California
| | - N S White
- From the Multimodal Imaging Laboratory (A.P.K., K.M.L., T.M.S., J.M.K., H.B., P.E., A.S., A.M.D., N.F., C.R.M., N.S.W.)
- Radiology (R.L.D., J.M.K., H.B., A.M.D., N.F., N.S.W.)
| |
Collapse
|
30
|
García-Figueiras R, Baleato-González S, Padhani AR, Oleaga L, Vilanova JC, Luna A, Cobas Gómez JC. Proton magnetic resonance spectroscopy in oncology: the fingerprints of cancer? Diagn Interv Radiol 2017; 22:75-89. [PMID: 26712681 DOI: 10.5152/dir.2015.15009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Abnormal metabolism is a key tumor hallmark. Proton magnetic resonance spectroscopy (1H-MRS) allows measurement of metabolite concentration that can be utilized to characterize tumor metabolic changes. 1H-MRS measurements of specific metabolites have been implemented in the clinic. This article performs a systematic review of image acquisition and interpretation of 1H-MRS for cancer evaluation, evaluates its strengths and limitations, and correlates metabolite peaks at 1H-MRS with diagnostic and prognostic parameters of cancer in different tumor types.
Collapse
Affiliation(s)
- Roberto García-Figueiras
- Department of Radiology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.
| | | | | | | | | | | | | |
Collapse
|
31
|
Zhang B, Chang K, Ramkissoon S, Tanguturi S, Bi WL, Reardon DA, Ligon KL, Alexander BM, Wen PY, Huang RY. Multimodal MRI features predict isocitrate dehydrogenase genotype in high-grade gliomas. Neuro Oncol 2017; 19:109-117. [PMID: 27353503 PMCID: PMC5193019 DOI: 10.1093/neuonc/now121] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND High-grade gliomas with mutations in the isocitrate dehydrogenase (IDH) gene family confer longer overall survival relative to their IDH-wild-type counterparts. Accurate determination of the IDH genotype preoperatively may have both prognostic and diagnostic value. The current study used a machine-learning algorithm to generate a model predictive of IDH genotype in high-grade gliomas based on clinical variables and multimodal features extracted from conventional MRI. METHODS Preoperative MRIs were obtained for 120 patients with primary grades III (n = 35) and IV (n = 85) glioma in this retrospective study. IDH genotype was confirmed for grade III (32/35, 91%) and IV (22/85, 26%) tumors by immunohistochemistry, spectrometry-based mutation genotyping (OncoMap), or multiplex exome sequencing (OncoPanel). IDH1 and IDH2 mutations were mutually exclusive, and all mutated tumors were collapsed into one IDH-mutated cohort. Cases were randomly assigned to either the training (n = 90) or validation cohort (n = 30). A total of 2970 imaging features were extracted from pre- and postcontrast T1-weighted, T2-weighted, and apparent diffusion coefficient map. Using a random forest algorithm, nonredundant features were integrated with clinical data to generate a model predictive of IDH genotype. RESULTS Our model achieved accuracies of 86% (area under the curve [AUC] = 0.8830) in the training cohort and 89% (AUC = 0.9231) in the validation cohort. Features with the highest predictive value included patient age as well as parametric intensity, texture, and shape features. CONCLUSION Using a machine-learning algorithm, we achieved accurate prediction of IDH genotype in high-grade gliomas with preoperative clinical and MRI features.
Collapse
Affiliation(s)
- Biqi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Ken Chang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shakti Ramkissoon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Shyam Tanguturi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Wenya Linda Bi
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - David A Reardon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Keith L Ligon
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Brian M Alexander
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Patrick Y Wen
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts (B.Z., K.C., R.Y.H.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Harvard Medical School, Boston, Massachusetts (S.R., K.L.L.); Department of Pathology, Boston Children's Hospital, Boston, Massachusetts (S.R., K.L.L.); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts(S.R., D.A.R., K.L.L., P.Y.W.); Harvard Radiation Oncology Program, Boston, Massachusetts (S.T.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (W.L.B.); Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (B.M.A.)
| |
Collapse
|
32
|
Nelson SJ, Li Y, Lupo JM, Olson M, Crane JC, Molinaro A, Roy R, Clarke J, Butowski N, Prados M, Cha S, Chang SM. Serial analysis of 3D H-1 MRSI for patients with newly diagnosed GBM treated with combination therapy that includes bevacizumab. J Neurooncol 2016; 130:171-179. [PMID: 27535746 PMCID: PMC5069332 DOI: 10.1007/s11060-016-2229-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/31/2016] [Indexed: 10/26/2022]
Abstract
Interpretation of changes in the T1- and T2-weighted MR images from patients with newly diagnosed glioblastoma (GBM) treated with standard of care in conjunction with anti-angiogenic agents is complicated by pseudoprogression and pseudoresponse. The hypothesis being tested in this study was that 3D H-1 magnetic resonance spectroscopic imaging (MRSI) provides estimates of levels of choline, creatine, N-acetylaspartate (NAA), lactate and lipid that change in response to treatment and that metrics describing these characteristics are associated with survival. Thirty-one patients with newly diagnosed GBM and being treated with radiation therapy (RT), temozolomide, erlotinib and bevacizumab were recruited to receive serial MR scans that included 3-D lactate edited MRSI at baseline, mid-RT, post-RT and at specific follow-up time points. The data were processed to provide estimates of metrics representing changes in metabolite levels relative to normal appearing brain. Cox proportional hazards analysis was applied to examine the relationship of these parameters with progression free survival (PFS) and overall survival (OS). There were significant reductions in parameters that describe relative levels of choline to NAA and creatine, indicating that the treatment caused a decrease in tumor cellularity. Changes in the levels of lactate and lipid relative to the NAA from contralateral brain were consistent with vascular normalization. Metabolic parameters from the first serial follow-up scan were associated with PFS and OS, when accounting for age and extent of resection. Integrating metabolic parameters into the assessment of patients with newly diagnosed GBM receiving therapies that include anti-angiogenic agents may be helpful for tracking changes in tumor burden, resolving ambiguities in anatomic images caused by non-specific treatment effects and for predicting outcome.
Collapse
Affiliation(s)
- Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Marram Olson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Jason C Crane
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Annette Molinaro
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
33
|
Huber T, Bette S, Wiestler B, Gempt J, Gerhardt J, Delbridge C, Barz M, Meyer B, Zimmer C, Kirschke JS. Fractional Anisotropy Correlates with Overall Survival in Glioblastoma. World Neurosurg 2016; 95:525-534.e1. [PMID: 27565465 DOI: 10.1016/j.wneu.2016.08.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 08/10/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Glioblastoma (GB) is an infiltrative disease that results in microstructural damage on a cellular level. Fractional anisotropy (FA) is an important estimate of diffusion tensor imaging (DTI) that can be used to assess microstructural integrity. The aim of this study was to examine the correlation between FA values and overall survival (OS) in patients with GB. METHODS This retrospective single-center study included 122 consecutive patients with GB (50 women; median age, 63 years) with preoperative MRI including fluid attenuated inversion recovery (FLAIR), contrast-enhanced T1-weighted sequences, and DTI. FA and apparent diffusion coefficient (ADC) values in contrast-enhancing lesions (FA-CEL, FA-ADC), nonenhancing lesions, and central tumor regions were correlated to histopathologic and clinical parameters. Univariate and multivariate survival analyses were performed. RESULTS Patients with low FA-CEL (median <0.31) showed significantly improved OS in univariate analysis (P = 0.028). FA-CEL also showed a positive correlation with Ki-67 proliferation index (P = 0.003). However, in a multivariate survival model, FA values could not be identified as independent prognostic parameters beside established factors such as age and Karnofsky performance scale score. FA values in nonenhancing lesions and central tumor regions and mean ADC values had no distinct influence on OS. CONCLUSIONS FA values can provide prognostic information regarding OS in patients with GB. There is a correlation between FA-CEL values and Ki-67 proliferation index, a marker for malignancy. Noninvasive identification of more aggressive GB growth patterns might be beneficial for preoperative risk evaluation and estimation of prognosis.
Collapse
Affiliation(s)
- Thomas Huber
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
| | - Stefanie Bette
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Julia Gerhardt
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claire Delbridge
- Department of Neuropathology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Melanie Barz
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jan S Kirschke
- Department of Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
34
|
Eidel O, Neumann JO, Burth S, Kieslich PJ, Jungk C, Sahm F, Kickingereder P, Kiening K, Unterberg A, Wick W, Schlemmer HP, Bendszus M, Radbruch A. Automatic Analysis of Cellularity in Glioblastoma and Correlation with ADC Using Trajectory Analysis and Automatic Nuclei Counting. PLoS One 2016; 11:e0160250. [PMID: 27467557 PMCID: PMC4965093 DOI: 10.1371/journal.pone.0160250] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 07/15/2016] [Indexed: 11/18/2022] Open
Abstract
Objective Several studies have analyzed a correlation between the apparent diffusion coefficient (ADC) derived from diffusion-weighted MRI and the tumor cellularity of corresponding histopathological specimens in brain tumors with inconclusive findings. Here, we compared a large dataset of ADC and cellularity values of stereotactic biopsies of glioblastoma patients using a new postprocessing approach including trajectory analysis and automatic nuclei counting. Materials and Methods Thirty-seven patients with newly diagnosed glioblastomas were enrolled in this study. ADC maps were acquired preoperatively at 3T and coregistered to the intraoperative MRI that contained the coordinates of the biopsy trajectory. 561 biopsy specimens were obtained; corresponding cellularity was calculated by semi-automatic nuclei counting and correlated to the respective preoperative ADC values along the stereotactic biopsy trajectory which included areas of T1-contrast-enhancement and necrosis. Results There was a weak to moderate inverse correlation between ADC and cellularity in glioblastomas that varied depending on the approach towards statistical analysis: for mean values per patient, Spearman’s ρ = -0.48 (p = 0.002), for all trajectory values in one joint analysis Spearman’s ρ = -0.32 (p < 0.001). The inverse correlation was additionally verified by a linear mixed model. Conclusions Our data confirms a previously reported inverse correlation between ADC and tumor cellularity. However, the correlation in the current article is weaker than the pooled correlation of comparable previous studies. Hence, besides cell density, other factors, such as necrosis and edema might influence ADC values in glioblastomas.
Collapse
Affiliation(s)
- Oliver Eidel
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Jan-Oliver Neumann
- Department of Neurosurgery, Division Stereotactic Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Sina Burth
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Pascal J. Kieslich
- Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Christine Jungk
- Department of Neurosurgery, Division Stereotactic Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Philipp Kickingereder
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Division Stereotactic Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Division Stereotactic Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic, University of Heidelberg Medical Center, Heidelberg, Germany
| | | | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- * E-mail:
| |
Collapse
|
35
|
Abstract
The revolution in cancer genomics has uncovered a variety of clinically relevant mutations in primary brain tumours, creating an urgent need to develop non-invasive imaging biomarkers to assess and integrate this genetic information into the clinical management of patients. Metabolic reprogramming is a central hallmark of cancer, including brain tumours; indeed, many of the molecular pathways implicated in the pathogenesis of brain tumours result in reprogramming of metabolism. This relationship provides the opportunity to devise in vivo metabolic imaging modalities to improve diagnosis, patient stratification, and monitoring of treatment response. Metabolic phenomena, such as the Warburg effect and altered mitochondrial metabolism, can be leveraged to image brain tumours using techniques including PET and MRI. Moreover, genetic alterations, such as mutations affecting isocitrate dehydrogenase, are associated with unique metabolic signatures that can be detected using magnetic resonance spectroscopy. The need to translate our understanding of the molecular features of brain tumours into imaging modalities with clinical utility is growing; metabolic imaging provides a unique platform to achieve this objective. In this Review, we examine the molecular basis for metabolic reprogramming in brain tumours, and examine current non-invasive metabolic imaging strategies that can be used to interrogate these molecular characteristics with the ultimate goal of guiding and improving patient care.
Collapse
|
36
|
Integrative analysis of diffusion-weighted MRI and genomic data to inform treatment of glioblastoma. J Neurooncol 2016; 129:289-300. [PMID: 27393347 DOI: 10.1007/s11060-016-2174-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/04/2016] [Indexed: 12/15/2022]
Abstract
Gene expression profiling from glioblastoma (GBM) patients enables characterization of cancer into subtypes that can be predictive of response to therapy. An integrative analysis of imaging and gene expression data can potentially be used to obtain novel biomarkers that are closely associated with the genetic subtype and gene signatures and thus provide a noninvasive approach to stratify GBM patients. In this retrospective study, we analyzed the expression of 12,042 genes for 558 patients from The Cancer Genome Atlas (TCGA). Among these patients, 50 patients had magnetic resonance imaging (MRI) studies including diffusion weighted (DW) MRI in The Cancer Imaging Archive (TCIA). We identified the contrast enhancing region of the tumors using the pre- and post-contrast T1-weighted MRI images and computed the apparent diffusion coefficient (ADC) histograms from the DW-MRI images. Using the gene expression data, we classified patients into four molecular subtypes, determined the number and composition of genes modules using the gap statistic, and computed gene signature scores. We used logistic regression to find significant predictors of GBM subtypes. We compared the predictors for different subtypes using Mann-Whitney U tests. We assessed detection power using area under the receiver operating characteristic (ROC) analysis. We computed Spearman correlations to determine the associations between ADC and each of the gene signatures. We performed gene enrichment analysis using Ingenuity Pathway Analysis (IPA). We adjusted all p values using the Benjamini and Hochberg method. The mean ADC was a significant predictor for the neural subtype. Neural tumors had a significantly lower mean ADC compared to non-neural tumors ([Formula: see text]), with mean ADC of [Formula: see text] and [Formula: see text] for neural and non-neural tumors, respectively. Mean ADC showed an area under the ROC of 0.75 for detecting neural tumors. We found eight gene modules in the GBM cohort. The mean ADC was significantly correlated with the gene signature related with dendritic cell maturation ([Formula: see text], [Formula: see text]). Mean ADC could be used as a biomarker of a gene signature associated with dendritic cell maturation and to assist in identifying patients with neural GBMs, known to be resistant to aggressive standard of care.
Collapse
|
37
|
Burth S, Kickingereder P, Eidel O, Tichy D, Bonekamp D, Weberling L, Wick A, Löw S, Hertenstein A, Nowosielski M, Schlemmer HP, Wick W, Bendszus M, Radbruch A. Clinical parameters outweigh diffusion- and perfusion-derived MRI parameters in predicting survival in newly diagnosed glioblastoma. Neuro Oncol 2016; 18:1673-1679. [PMID: 27298312 DOI: 10.1093/neuonc/now122] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/03/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The purpose of this study was to determine the relevance of clinical data, apparent diffusion coefficient (ADC), and relative cerebral blood volume (rCBV) from dynamic susceptibility contrast (DSC) perfusion and the volume transfer constant (ktrans) from dynamic contrast-enhanced (DCE) perfusion for predicting overall survival (OS) and progression-free survival (PFS) in newly diagnosed treatment-naïve glioblastoma patients. METHODS Preoperative MR scans including standardized contrast-enhanced T1 (cT1), T2 - fluid-attenuated inversion recovery (FLAIR), ADC, DSC, and DCE of 125 patients with subsequent histopathologically confirmed glioblastoma were performed on a 3 Tesla MRI scanner. ADC, DSC, and DCE parameters were analyzed in semiautomatically segmented tumor volumes on contrast-enhanced (CE) cT1 and hyperintense signal changes on T2 FLAIR (ED). Univariate and multivariable Cox regression analyses including age, sex, extent of resection (EOR), and KPS were performed to assess the influence of each parameter on OS and PFS. RESULTS Univariate Cox regression analysis demonstrated a significant association of age, KPS, and EOR with PFS and age, KPS, EOR, lower ADC, and higher rCBV with OS. Multivariable analysis showed independent significance of male sex, KPS, EOR, and increased rCBVCE for PFS, and age, sex, KPS, and EOR for OS. CONCLUSIONS MRI parameters help to predict OS in a univariate Cox regression analysis, and increased rCBVCE is associated with shorter PFS in the multivariable model. In summary, however, our findings suggest that the relevance of MRI parameters is outperformed by clinical parameters in a multivariable analysis, which limits their prognostic value for survival prediction at the time of initial diagnosis.
Collapse
Affiliation(s)
- Sina Burth
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Philipp Kickingereder
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Oliver Eidel
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Diana Tichy
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - David Bonekamp
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Lukas Weberling
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Antje Wick
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Sarah Löw
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Anne Hertenstein
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Martha Nowosielski
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Heinz-Peter Schlemmer
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Wolfgang Wick
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| | - Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany (S.B., P.K., O.E., D.B., L.W., M.B., A.R.); Division of Bioststatistics, German Cancer Research Center, Heidelberg, Germany (D.T.); Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany (A.W., S.L., A.H., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.); Department of Radiology, German Cancer Research Center, Heidelberg, Germany (H.S.)
| |
Collapse
|
38
|
Zarifi M, Tzika AA. Proton MRS imaging in pediatric brain tumors. Pediatr Radiol 2016; 46:952-62. [PMID: 27233788 DOI: 10.1007/s00247-016-3547-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 11/30/2015] [Accepted: 01/13/2016] [Indexed: 12/14/2022]
Abstract
Magnetic resonance (MR) techniques offer a noninvasive, non-irradiating yet sensitive approach to diagnosing and monitoring pediatric brain tumors. Proton MR spectroscopy (MRS), as an adjunct to MRI, is being more widely applied to monitor the metabolic aspects of brain cancer. In vivo MRS biomarkers represent a promising advance and may influence treatment choice at both initial diagnosis and follow-up, given the inherent difficulties of sequential biopsies to monitor therapeutic response. When combined with anatomical or other types of imaging, MRS provides unique information regarding biochemistry in inoperable brain tumors and can complement neuropathological data, guide biopsies and enhance insight into therapeutic options. The combination of noninvasively acquired prognostic information and the high-resolution anatomical imaging provided by conventional MRI is expected to surpass molecular analysis and DNA microarray gene profiling, both of which, although promising, depend on invasive biopsy. This review focuses on recent data in the field of MRS in children with brain tumors.
Collapse
Affiliation(s)
- Maria Zarifi
- Department of Radiology, Aghia Sophia Children's Hospital, Athens, Greece
| | - A Aria Tzika
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Shriners Burn Hospital, 51 Blossom St., Room #261, Boston, MA, 02114, USA.
| |
Collapse
|
39
|
Basic Principles and Clinical Applications of Magnetic Resonance Spectroscopy in Neuroradiology. J Comput Assist Tomogr 2016; 40:1-13. [PMID: 26484954 DOI: 10.1097/rct.0000000000000322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Magnetic resonance spectroscopy is a powerful tool to assist daily clinical diagnostics. This review is intended to give an overview on basic principles of the technology, discuss some of its technical aspects, and present typical applications in daily clinical routine in neuroradiology.
Collapse
|
40
|
Current and future strategies for treatment of glioma. Neurosurg Rev 2016; 40:1-14. [PMID: 27085859 DOI: 10.1007/s10143-016-0709-8] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/25/2016] [Indexed: 01/12/2023]
Abstract
Gliomas are one of the most common types of primary brain tumors and have remained particularly challenging to treat. This review illustrates a multidisciplinary approach to the treatment of glioma and glioblastoma. We will review current advances in surgical approaches, novel imaging techniques, advanced molecular characterization of tumors and translational efforts for treatment. We will focus on current clinical trials as well as the pursuit of personalized or precision therapy. We will also comment on the importance of both quality of life of our patients and their care givers.
Collapse
|
41
|
Mörén L, Wibom C, Bergström P, Johansson M, Antti H, Bergenheim AT. Characterization of the serum metabolome following radiation treatment in patients with high-grade gliomas. Radiat Oncol 2016; 11:51. [PMID: 27039175 PMCID: PMC4818859 DOI: 10.1186/s13014-016-0626-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/22/2016] [Indexed: 11/26/2022] Open
Abstract
Background Glioblastomas progress rapidly making response evaluation using MRI insufficient since treatment effects are not detectable until months after initiation of treatment. Thus, there is a strong need for supplementary biomarkers that could provide reliable and early assessment of treatment efficacy. Analysis of alterations in the metabolome may be a source for identification of new biomarker patterns harboring predictive information. Ideally, the biomarkers should be found within an easily accessible compartment such as the blood. Method Using gas-chromatographic- time-of-flight-mass spectroscopy we have analyzed serum samples from 11 patients with glioblastoma during the initial phase of radiotherapy. Fasting serum samples were collected at admittance, on the same day as, but before first treatment and in the morning after the second and fifth dose of radiation. The acquired data was analyzed and evaluated by chemometrics based bioinformatics methods. Our findings were compared and discussed in relation to previous data from microdialysis in tumor tissue, i.e. the extracellular compartment, from the same patients. Results We found a significant change in metabolite pattern in serum comparing samples taken before radiotherapy to samples taken during early radiotherapy. In all, 68 metabolites were lowered in concentration following treatment while 16 metabolites were elevated in concentration. All detected and identified amino acids and fatty acids together with myo-inositol, creatinine, and urea were among the metabolites that decreased in concentration during treatment, while citric acid was among the metabolites that increased in concentration. Furthermore, when comparing results from the serum analysis with findings in tumor extracellular fluid we found a common change in metabolite patterns in both compartments on an individual patient level. On an individual metabolite level similar changes in ornithine, tyrosine and urea were detected. However, in serum, glutamine and glutamate were lowered after treatment while being elevated in the tumor extracellular fluid. Conclusion Cross-validated multivariate statistical models verified that the serum metabolome was significantly changed in relation to radiation in a similar pattern to earlier findings in tumor tissue. However, all individual changes in tissue did not translate into changes in serum. Our study indicates that serum metabolomics could be of value to investigate as a potential marker for assessing early response to radiotherapy in malignant glioma. Electronic supplementary material The online version of this article (doi:10.1186/s13014-016-0626-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lina Mörén
- Department of Chemistry, Computational Life Science Cluster, Umeå University, SE 901 87, Umeå, Sweden. .,Department of Chemistry, Umeå University, SE 90187, Umeå, Sweden.
| | - Carl Wibom
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Per Bergström
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Henrik Antti
- Department of Chemistry, Computational Life Science Cluster, Umeå University, SE 901 87, Umeå, Sweden
| | - A Tommy Bergenheim
- Department of Clinical Neuroscience, Neurosurgery, Umeå University, SE 901 85, Umeå, Sweden
| |
Collapse
|
42
|
Henker C, Kriesen T, Fürst K, Goody D, Glass Ä, Pützer BM, Piek J. Effect of 10 different polymorphisms on preoperative volumetric characteristics of glioblastoma multiforme. J Neurooncol 2015; 126:585-92. [PMID: 26603163 DOI: 10.1007/s11060-015-2005-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/19/2015] [Indexed: 01/09/2023]
Abstract
There is a distinct diversity between the appearance of every glioblastoma multiforme (GBM) on pretreatment magnetic resonance imaging (MRI) with a potential impact on clinical outcome and survival of the patients. The object of this study was to determine the impact of 10 different single nucleotide polymorphisms (SNPs) on various volumetric parameters in patients harboring a GBM. We prospectively analyzed 20 steroid-naïve adult patients who had been treated for newly diagnosed GBM. The volumetry was performed using MRI with the help of a semiautomated quantitative software measuring contrast enhancing tumor volume including necrosis, central necrosis alone and peritumoral edema (PTE). We calculated ratios between the tumor volume and edema (ETR), respectively necrosis (NTR). SNP analysis was done using genomic DNA extracted from peripheral blood genotyped via PCR and sequencing. There was a strong correlation between tumor volume and PTE (p < 0.001), necrosis (p < 0.001) and NTR (p = 0.003). Age and sex had no influence on volumetric data. The Aquaporin 4-31G > A SNP had a significant influence on the ETR (p = 0.042) by decreasing the measured edema compared with the tumor volume. The Interleukin 8-251A > T SNP was significantly correlated with an increased tumor (p = 0.048), PTE (p = 0.033) and necrosis volume (p = 0.028). We found two SNPs with a distinct impact on pretreatment tumor characteristics, presenting a potential explanation for the individual diversity of GBM appearance on MRI and influence on survival.
Collapse
Affiliation(s)
- Christian Henker
- Department of Neurosurgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany.
| | - Thomas Kriesen
- Department of Neurosurgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany
| | - Katharina Fürst
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| | - Deborah Goody
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| | - Änne Glass
- Institute for Biostatistics and Informatics in Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 8, 18057, Rostock, Germany
| | - Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| | - Jürgen Piek
- Department of Neurosurgery, University Hospital Rostock, Schillingallee 35, 18057, Rostock, Germany
| |
Collapse
|
43
|
Chaumeil MM, Lupo JM, Ronen SM. Magnetic Resonance (MR) Metabolic Imaging in Glioma. Brain Pathol 2015; 25:769-80. [PMID: 26526945 PMCID: PMC8029127 DOI: 10.1111/bpa.12310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/25/2022] Open
Abstract
This review is focused on describing the use of magnetic resonance (MR) spectroscopy for metabolic imaging of brain tumors. We will first review the MR metabolic imaging findings generated from preclinical models, focusing primarily on in vivo studies, and will then describe the use of metabolic imaging in the clinical setting. We will address relatively well-established (1) H MRS approaches, as well as (31) P MRS, (13) C MRS and emerging hyperpolarized (13) C MRS methodologies, and will describe the use of metabolic imaging for understanding the basic biology of glioma as well as for improving the characterization and monitoring of brain tumors in the clinic.
Collapse
Affiliation(s)
| | - Janine M. Lupo
- Department of Radiology and Biomedical ImagingMission Bay Campus
| | - Sabrina M. Ronen
- Department of Radiology and Biomedical ImagingMission Bay Campus
- Brain Tumor Research CenterUniversity of CaliforniaSan FranciscoCA
| |
Collapse
|
44
|
Ken S, Deviers A, Filleron T, Catalaa I, Lotterie JA, Khalifa J, Lubrano V, Berry I, Péran P, Celsis P, Moyal ECJ, Laprie A. Voxel-based evidence of perfusion normalization in glioblastoma patients included in a phase I-II trial of radiotherapy/tipifarnib combination. J Neurooncol 2015; 124:465-73. [PMID: 26189058 DOI: 10.1007/s11060-015-1860-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/14/2015] [Indexed: 01/24/2023]
Abstract
We previously showed that the farnesyl transferase inihibitor, Tipifarnib induced vascularization normalization, oxygenation and radiosensitization in a pre-clinical glioblastoma (GBM) model. The aim of this study was to assess by dynamic-susceptibility-contrast MRI (DSC-MRI) the effect of radiotherapy (RT) and Tipifarnib combination on tumor perfusion in GBM patients. Eighteen patients with newly diagnosed GBM, enrolled in a phase I-II clinical trial associating RT with Tipifarnib, underwent anatomical MR imaging and DSC-MRI before (M0) and two months after treatment (M2). Anatomic volumes of interest (VOIs) were delineated according to contrast-enhanced and hyper-intense signal areas on T1-Gd and T2 images, respectively. Perfusion variations between M0 and M2 were assessed with median relative cerebral blood volume (rCBV) inside these VOIs. Another voxel by voxel analysis of CBV values classified 405,117 tumor voxels into High_, Normal_ and Low_CBVTUMOR according to the distribution of CBV in the contralateral normal tissue. These three categories of CBVTUMOR voxels were color-coded over anatomical MRI. Variations of median rCBV were significantly different for two groups of patients (P < 0.013): rCBV decreased when initial rCBV was ≥ 1.0 (Group_rCBV_M0 > 1) and rCBV increased when initial rCBV was < 1.0 (Group_rCBV_M0 < 1). Mapping of color-coded voxels provided additional spatial and quantitative information about tumor perfusion: Group_rCBV_M0 > 1 presented a significant decrease of High_CBVTUMOR volume (P = 0.015) simultaneously with a significant increase of Normal_CBVTUMOR volume (P = 0.009) after treatment. Group_rCBV_M0 < 1 presented a decrease of Low_CBVTUMOR volume with an increase of Normal_ and High_CBV TUMOR volume after treatment. Pre and post-treatment CBV measurements with DSC-MRI characterized tumor perfusion evolution in GBM patients treated with RT combined to Tipifarnib; showing variations in favour of tumor perfusion normalization in agreement with our pre-clinical results of vascular normalization.
Collapse
Affiliation(s)
- Soléakhéna Ken
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France. .,Department of Medical Physics, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France. .,INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.
| | - Alexandra Deviers
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France.,INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.,Université Paul Sabatier, Toulouse III, 118, route de Narbonne, 31062, Toulouse, France
| | - Thomas Filleron
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, Bureau des Essais Cliniques, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France
| | - Isabelle Catalaa
- Centre Hospitalier Universitaire de Purpan, 31059, Toulouse, France
| | - Jean-Albert Lotterie
- INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.,Centre Hospitalier Universitaire de Rangueil, 31059, Toulouse, France
| | - Jonathan Khalifa
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France
| | - Vincent Lubrano
- INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.,Centre Hospitalier Universitaire de Rangueil, 31059, Toulouse, France
| | - Isabelle Berry
- INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.,Centre Hospitalier Universitaire de Rangueil, 31059, Toulouse, France
| | - Patrice Péran
- INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France
| | - Pierre Celsis
- INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France.,Université Paul Sabatier, Toulouse III, 118, route de Narbonne, 31062, Toulouse, France.,INSERM, UMR 1037, CRCT, 1, avenue Irene Joliot-Curie, 31000, Toulouse, France
| | - Anne Laprie
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse - Oncopôle, 1, avenue Irene Joliot-Curie, 31059, Toulouse, France.,INSERM UMR 825 Imagerie Cérébrale et Handicaps Neurologiques, CHU Purpan - Pavillon Baudot, 31024, Toulouse, France.,Université Paul Sabatier, Toulouse III, 118, route de Narbonne, 31062, Toulouse, France
| |
Collapse
|
45
|
Macyszyn L, Akbari H, Pisapia JM, Da X, Attiah M, Pigrish V, Bi Y, Pal S, Davuluri RV, Roccograndi L, Dahmane N, Martinez-Lage M, Biros G, Wolf RL, Bilello M, O'Rourke DM, Davatzikos C. Imaging patterns predict patient survival and molecular subtype in glioblastoma via machine learning techniques. Neuro Oncol 2015; 18:417-25. [PMID: 26188015 DOI: 10.1093/neuonc/nov127] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND MRI characteristics of brain gliomas have been used to predict clinical outcome and molecular tumor characteristics. However, previously reported imaging biomarkers have not been sufficiently accurate or reproducible to enter routine clinical practice and often rely on relatively simple MRI measures. The current study leverages advanced image analysis and machine learning algorithms to identify complex and reproducible imaging patterns predictive of overall survival and molecular subtype in glioblastoma (GB). METHODS One hundred five patients with GB were first used to extract approximately 60 diverse features from preoperative multiparametric MRIs. These imaging features were used by a machine learning algorithm to derive imaging predictors of patient survival and molecular subtype. Cross-validation ensured generalizability of these predictors to new patients. Subsequently, the predictors were evaluated in a prospective cohort of 29 new patients. RESULTS Survival curves yielded a hazard ratio of 10.64 for predicted long versus short survivors. The overall, 3-way (long/medium/short survival) accuracy in the prospective cohort approached 80%. Classification of patients into the 4 molecular subtypes of GB achieved 76% accuracy. CONCLUSIONS By employing machine learning techniques, we were able to demonstrate that imaging patterns are highly predictive of patient survival. Additionally, we found that GB subtypes have distinctive imaging phenotypes. These results reveal that when imaging markers related to infiltration, cell density, microvascularity, and blood-brain barrier compromise are integrated via advanced pattern analysis methods, they form very accurate predictive biomarkers. These predictive markers used solely preoperative images, hence they can significantly augment diagnosis and treatment of GB patients.
Collapse
Affiliation(s)
- Luke Macyszyn
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Hamed Akbari
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Jared M Pisapia
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Xiao Da
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Mark Attiah
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Vadim Pigrish
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Yingtao Bi
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Sharmistha Pal
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Ramana V Davuluri
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Laura Roccograndi
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Nadia Dahmane
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Maria Martinez-Lage
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - George Biros
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Ronald L Wolf
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Michel Bilello
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Donald M O'Rourke
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| | - Christos Davatzikos
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania (L.M., J.M.P., M.A., L.R., N.D., D.M.O.); Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., R.L.W., M.B., C.D.); Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, Pennsylvania (H.A., X.D., V.P., M.B., C.D.); Department of Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Y.B., S.P., R.V.D.); Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas (G.B.); Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (M.M.-L., D.M.O.)
| |
Collapse
|
46
|
Mabray MC, Barajas RF, Cha S. Modern brain tumor imaging. Brain Tumor Res Treat 2015; 3:8-23. [PMID: 25977902 PMCID: PMC4426283 DOI: 10.14791/btrt.2015.3.1.8] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 12/16/2022] Open
Abstract
The imaging and clinical management of patients with brain tumor continue to evolve over time and now heavily rely on physiologic imaging in addition to high-resolution structural imaging. Imaging remains a powerful noninvasive tool to positively impact the management of patients with brain tumor. This article provides an overview of the current state-of-the art clinical brain tumor imaging. In this review, we discuss general magnetic resonance (MR) imaging methods and their application to the diagnosis of, treatment planning and navigation, and disease monitoring in patients with brain tumor. We review the strengths, limitations, and pitfalls of structural imaging, diffusion-weighted imaging techniques, MR spectroscopy, perfusion imaging, positron emission tomography/MR, and functional imaging. Overall this review provides a basis for understudying the role of modern imaging in the care of brain tumor patients.
Collapse
Affiliation(s)
- Marc C Mabray
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Ramon F Barajas
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
47
|
Paech D, Burth S, Windschuh J, Meissner JE, Zaiss M, Eidel O, Kickingereder P, Nowosielski M, Wiestler B, Sahm F, Floca RO, Neumann JO, Wick W, Heiland S, Bendszus M, Schlemmer HP, Ladd ME, Bachert P, Radbruch A. Nuclear Overhauser Enhancement imaging of glioblastoma at 7 Tesla: region specific correlation with apparent diffusion coefficient and histology. PLoS One 2015; 10:e0121220. [PMID: 25789657 PMCID: PMC4366097 DOI: 10.1371/journal.pone.0121220] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/30/2015] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE To explore the correlation between Nuclear Overhauser Enhancement (NOE)-mediated signals and tumor cellularity in glioblastoma utilizing the apparent diffusion coefficient (ADC) and cell density from histologic specimens. NOE is one type of chemical exchange saturation transfer (CEST) that originates from mobile macromolecules such as proteins and might be associated with tumor cellularity via altered protein synthesis in proliferating cells. PATIENTS AND METHODS For 15 patients with newly diagnosed glioblastoma, NOE-mediated CEST-contrast was acquired at 7 Tesla (asymmetric magnetization transfer ratio (MTRasym) at 3.3ppm, B1 = 0.7 μT). Contrast enhanced T1 (CE-T1), T2 and diffusion-weighted MRI (DWI) were acquired at 3 Tesla and coregistered. The T2 edema and the CE-T1 tumor were segmented. ADC and MTRasym values within both regions of interest were correlated voxelwise yielding the correlation coefficient rSpearman (rSp). In three patients who underwent stereotactic biopsy, cell density of 12 specimens per patient was correlated with corresponding MTRasym and ADC values of the biopsy site. RESULTS Eight of 15 patients showed a weak or moderate positive correlation of MTRasym and ADC within the T2 edema (0.16≤rSp≤0.53, p<0.05). Seven correlations were statistically insignificant (p>0.05, n = 4) or yielded rSp≈0 (p<0.05, n = 3). No trend towards a correlation between MTRasym and ADC was found in CE-T1 tumor (-0.31<rSp<0.28, p<0.05, n = 9; p>0.05, n = 6). The biopsy-analysis within CE-T1 tumor revealed a strong positive correlation between tumor cellularity and MTRasym values in two of the three patients (rSppatient3 = 0.69 and rSppatient15 = 0.87, p<0.05), while the correlation of ADC and cellularity was heterogeneous (rSppatient3 = 0.545 (p = 0.067), rSppatient4 = -0.021 (p = 0.948), rSppatient15 = -0.755 (p = 0.005)). DISCUSSION NOE-imaging is a new contrast promising insight into pathophysiologic processes in glioblastoma regarding cell density and protein content, setting itself apart from DWI. Future studies might be based on the assumption that NOE-mediated CEST visualizes cellularity more accurately than ADC, especially in the CE-T1 tumor region.
Collapse
Affiliation(s)
- Daniel Paech
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
- Neurooncologic Imaging, Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Sina Burth
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
- Neurooncologic Imaging, Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Johannes Windschuh
- Department of Medical Physics in Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Jan-Eric Meissner
- Neurooncologic Imaging, Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Department of Medical Physics in Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Moritz Zaiss
- Department of Medical Physics in Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Oliver Eidel
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Philipp Kickingereder
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Martha Nowosielski
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Benedikt Wiestler
- Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Ralf Omar Floca
- Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Jan-Oliver Neumann
- Department of Neurosurgery, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Heinz-Peter Schlemmer
- Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Mark Edward Ladd
- Department of Medical Physics in Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Peter Bachert
- Department of Medical Physics in Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg Medical Center, Heidelberg, Germany
- Neurooncologic Imaging, Department of Radiology, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- * E-mail:
| |
Collapse
|
48
|
Elson A, Bovi J, Siker M, Schultz C, Paulson E. Evaluation of absolute and normalized apparent diffusion coefficient (ADC) values within the post-operative T2/FLAIR volume as adverse prognostic indicators in glioblastoma. J Neurooncol 2015; 122:549-58. [PMID: 25700835 DOI: 10.1007/s11060-015-1743-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/14/2015] [Indexed: 02/03/2023]
Abstract
To evaluate the association of normalized and absolute ADC metrics with progression free survival (PFS) and overall survival (OS) in patients treated for glioblastoma multiforme (GBM). Fifty-two patients with preradiotherapy diffusion weighted imaging treated with post-operative chemoradiation for GBM were evaluated. Region of interest analysis for ADC metrics including mean and minimum ADC value (ADCmean) and (ADCmin) was performed within the T2/FLAIR volume. Normalized (N)ADC values were generated relative to contralateral white matter. PFS and OS were analyzed relative to ADC parameters using a regression model. Kaplan-Meier and Cox proportional hazards analysis with respect to (N)ADCmean, and (N)ADCmin was performed. A (N)ADC threshold <1.3 within the T2/FLAIR volume was analyzed with respect to PFS and OS. Regression analysis indicated that normalized ADC values provide the strongest association with PFS and OS. Kaplan-Meier analysis revealed a non-significant trend toward inferior PFS and OS associated with (N)ADCmean <1.7, and a significant decrement to PFS and OS associated with (N)ADCmin <0.3. (N)ADCmin was a significant prognostic factor when taking into account age, performance status, and extent of resection. ADC thresholding analysis revealed that a retained volume of >0.45 cc per mL FLAIR volume was associated with a trend toward inferior PFS and OS. In the post-operative, pre-radiotherapy setting, the (N)ADCmin is the strongest predictor of outcomes in patients treated for GBM. ADC thresholding analysis indicates that a large volume of normalized ADC value <1.3 may be associated with adverse outcomes.
Collapse
Affiliation(s)
- Andrew Elson
- Department of Radiation Oncology, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Froedtert Hospital East Clinics 3rd Floor, Milwaukee, WI, 53226, USA,
| | | | | | | | | |
Collapse
|
49
|
Schmainda KM, Zhang Z, Prah M, Snyder BS, Gilbert MR, Sorensen AG, Barboriak DP, Boxerman JL. Dynamic susceptibility contrast MRI measures of relative cerebral blood volume as a prognostic marker for overall survival in recurrent glioblastoma: results from the ACRIN 6677/RTOG 0625 multicenter trial. Neuro Oncol 2015; 17:1148-56. [PMID: 25646027 DOI: 10.1093/neuonc/nou364] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/24/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The study goal was to determine whether changes in relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) MRI are predictive of overall survival (OS) in patients with recurrent glioblastoma multiforme (GBM) when measured 2, 8, and 16 weeks after treatment initiation. METHODS Patients with recurrent GBM (37/123) enrolled in ACRIN 6677/RTOG 0625, a multicenter, randomized, phase II trial of bevacizumab with irinotecan or temozolomide, consented to DSC-MRI plus conventional MRI, 21 with DSC-MRI at baseline and at least 1 postbaseline scan. Contrast-enhancing regions of interest were determined semi-automatically using pre- and postcontrast T1-weighted images. Mean tumor rCBV normalized to white matter (nRCBV) and standardized rCBV (sRCBV) were determined for these regions of interest. The OS rates for patients with positive versus negative changes from baseline in nRCBV and sRCBV were compared using Wilcoxon rank-sum and Kaplan-Meier survival estimates with log-rank tests. RESULTS Patients surviving at least 1 year (OS-1) had significantly larger decreases in nRCBV at week 2 (P = .0451) and sRCBV at week 16 (P = .014). Receiver operating characteristic analysis found the percent changes of nRCBV and sRCBV at week 2 and sRCBV at week 16, but not rCBV data at week 8, to be good prognostic markers for OS-1. Patients with positive change from baseline rCBV had significantly shorter OS than those with negative change at both week 2 and week 16 (P = .0015 and P = .0067 for nRCBV and P = .0251 and P = .0004 for sRCBV, respectively). CONCLUSIONS Early decreases in rCBV are predictive of improved survival in patients with recurrent GBM treated with bevacizumab.
Collapse
Affiliation(s)
- Kathleen M Schmainda
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Zheng Zhang
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Melissa Prah
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Bradley S Snyder
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Mark R Gilbert
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - A Gregory Sorensen
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Daniel P Barboriak
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Jerrold L Boxerman
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| |
Collapse
|
50
|
Bonekamp D, Deike K, Wiestler B, Wick W, Bendszus M, Radbruch A, Heiland S. Association of overall survival in patients with newly diagnosed glioblastoma with contrast-enhanced perfusion MRI: Comparison of intraindividually matched T1 - and T2 (*) -based bolus techniques. J Magn Reson Imaging 2014; 42:87-96. [PMID: 25244574 DOI: 10.1002/jmri.24756] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/27/2014] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND To compare intraindividual dynamic susceptibility contrast (DSC) and dynamic contrast enhanced (DCE) MR perfusion parameters and determine the association of DCE parameters with overall survival (OS) with the established predictive DSC parameter cerebral blood volume (CBV) in patients with newly diagnosed glioblastoma. METHODS Perfusion data were analyzed retrospectively, and included scans performed preoperatively at 3.0 Tesla in 37 patients (25 males, 12 females, 39-83 years, median 65) later diagnosed with glioblastoma. All patients received standard treatment consisting of surgery and radiochemotherapy. Images were spatially coregistered and maximum region of interest-based DCE and DSC parameter measurements compared and thresholds identified using multivariate linear regression, Pearson's correlation coefficients and using receiver operating characteristic analysis. Survival analysis was performed using Kaplan-Meier curves. RESULTS While both, elevated volume transfer constant (K(trans) ) (>0.29 min(-1) ; P = 0.041) and CBV (>23.7 mL/100 mL; P < 0.001) were significantly associated with OS, elevated CBV was associated with worse OS compared with elevated K(trans) . K(trans) was significantly correlated with the leakage correction factor K2 but not with CBV. CONCLUSION The combined use of DSC and DCE MR perfusion may provide additional information of prognostic value for glioblastoma patient survival prediction. As K(trans) was not tightly coupled to CBV, both parameters may reflect different stages in the pathogenetic sequence of glioblastoma growth.
Collapse
Affiliation(s)
- David Bonekamp
- Department of Neuroradiology, University Hospital Heidelberg, Germany.,Division of Experimental Radiology, Department of Neuroradiology, University Hospital Heidelberg, Germany
| | - Katerina Deike
- Department of Neuroradiology, University Hospital Heidelberg, Germany
| | - Benedikt Wiestler
- Department of Neurooncology, University Hospital Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurooncology, University Hospital Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Germany
| | - Alexander Radbruch
- Department of Neuroradiology, University Hospital Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Heiland
- Division of Experimental Radiology, Department of Neuroradiology, University Hospital Heidelberg, Germany
| |
Collapse
|