1
|
Kadali KR, Nierobisch N, Maibach F, Heesen P, Alcaide-Leon P, Hüllner M, Weller M, Kulcsar Z, Hainc N. An effective MRI perfusion threshold based workflow to triage additional 18F-FET PET in posttreatment high grade glioma. Sci Rep 2025; 15:7749. [PMID: 40044711 PMCID: PMC11882894 DOI: 10.1038/s41598-025-90472-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
MRI is the preferred method for follow-up imaging of post-treatment WHO grade 3 or 4 gliomas. While positron emission tomography with O-(2-[18F]fluoroethyl)-L-tyrosine) (18F-FET PET) offers higher diagnostic accuracy, its use is limited due to low availability. We propose a sequential, threshold-based workflow to triage patients for additional 18F-FET PET scans based on MRI dynamic susceptibility contrast (DSC) perfusion-derived rCBV values, to optimize 18F-FET PET resource allocation. Patients with high-grade gliomas who had undergone standard-of-care treatment and developed new or enlarging contrast-enhancing post-treatment lesions on MRI were included, with a 18F-FET PET study performed within 4 months of the MRI. Patients were excluded if there were significant changes in lesion size or treatment between the MRI and 18F-FET PET scan. An rCBV threshold was determined and the performance of a threshold-based imaging workflow was evaluated compared to the gold standard defined here as surgical verification or long-term imaging follow-up without further intervention. Forty-one patients with a total of 49 lesions were included (tumor progression n = 40, treatment-related changes n = 9). Above the rCBV threshold of 2.4, MRI was 100% accurate (21/21 patients) in diagnosing tumor progression. Below the threshold, MRI identified 9 true negatives but produced 19 false negatives. 18F-FET PET reclassified 18/19 (95%) false negatives resulting in an overall accuracy of 48/49 (98%) for the workflow. Our MRI DSC perfusion rCBV-based threshold workflow for triaging patients for additional 18F-FET PET imaging in post-treatment high grade glioma has the potential to optimize 18F-FET PET resource allocation.
Collapse
Affiliation(s)
- Krishna Ranjith Kadali
- University of Zurich, Zurich, Switzerland
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nathalie Nierobisch
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Fabienne Maibach
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Philip Heesen
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Paula Alcaide-Leon
- Department of Medical Imaging, University of Toronto, Toronto, Canada
- Joint Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, University of Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nicolin Hainc
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
de Godoy LL, Mohan S, Wang S, Nasrallah MP, Sakai Y, O'Rourke DM, Bagley S, Desai A, Loevner LA, Poptani H, Chawla S. Validation of multiparametric MRI based prediction model in identification of pseudoprogression in glioblastomas. J Transl Med 2023; 21:287. [PMID: 37118754 PMCID: PMC10142504 DOI: 10.1186/s12967-023-03941-x] [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: 11/19/2022] [Accepted: 01/30/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Accurate differentiation of pseudoprogression (PsP) from tumor progression (TP) in glioblastomas (GBMs) is essential for appropriate clinical management and prognostication of these patients. In the present study, we sought to validate the findings of our previously developed multiparametric MRI model in a new cohort of GBM patients treated with standard therapy in identifying PsP cases. METHODS Fifty-six GBM patients demonstrating enhancing lesions within 6 months after completion of concurrent chemo-radiotherapy (CCRT) underwent anatomical imaging, diffusion and perfusion MRI on a 3 T magnet. Subsequently, patients were classified as TP + mixed tumor (n = 37) and PsP (n = 19). When tumor specimens were available from repeat surgery, histopathologic findings were used to identify TP + mixed tumor (> 25% malignant features; n = 34) or PsP (< 25% malignant features; n = 16). In case of non-availability of tumor specimens, ≥ 2 consecutive conventional MRIs using mRANO criteria were used to determine TP + mixed tumor (n = 3) or PsP (n = 3). The multiparametric MRI-based prediction model consisted of predictive probabilities (PP) of tumor progression computed from diffusion and perfusion MRI derived parameters from contrast enhancing regions. In the next step, PP values were used to characterize each lesion as PsP or TP+ mixed tumor. The lesions were considered as PsP if the PP value was < 50% and TP+ mixed tumor if the PP value was ≥ 50%. Pearson test was used to determine the concordance correlation coefficient between PP values and histopathology/mRANO criteria. The area under ROC curve (AUC) was used as a quantitative measure for assessing the discriminatory accuracy of the prediction model in identifying PsP and TP+ mixed tumor. RESULTS Multiparametric MRI model correctly predicted PsP in 95% (18/19) and TP+ mixed tumor in 57% of cases (21/37) with an overall concordance rate of 70% (39/56) with final diagnosis as determined by histopathology/mRANO criteria. There was a significant concordant correlation coefficient between PP values and histopathology/mRANO criteria (r = 0.56; p < 0.001). The ROC analyses revealed an accuracy of 75.7% in distinguishing PsP from TP+ mixed tumor. Leave-one-out cross-validation test revealed that 73.2% of cases were correctly classified as PsP and TP + mixed tumor. CONCLUSIONS Our multiparametric MRI based prediction model may be helpful in identifying PsP in GBM patients.
Collapse
Affiliation(s)
- Laiz Laura de Godoy
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sumei Wang
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Clinical Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yu Sakai
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Bagley
- Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Arati Desai
- Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Laurie A Loevner
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Harish Poptani
- Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Sanjeev Chawla
- Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Qi D, Li J, Quarles CC, Fonkem E, Wu E. Assessment and prediction of glioblastoma therapy response: challenges and opportunities. Brain 2023; 146:1281-1298. [PMID: 36445396 PMCID: PMC10319779 DOI: 10.1093/brain/awac450] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma is the most aggressive type of primary adult brain tumour. The median survival of patients with glioblastoma remains approximately 15 months, and the 5-year survival rate is <10%. Current treatment options are limited, and the standard of care has remained relatively constant since 2011. Over the last decade, a range of different treatment regimens have been investigated with very limited success. Tumour recurrence is almost inevitable with the current treatment strategies, as glioblastoma tumours are highly heterogeneous and invasive. Additionally, another challenging issue facing patients with glioblastoma is how to distinguish between tumour progression and treatment effects, especially when relying on routine diagnostic imaging techniques in the clinic. The specificity of routine imaging for identifying tumour progression early or in a timely manner is poor due to the appearance similarity of post-treatment effects. Here, we concisely describe the current status and challenges in the assessment and early prediction of therapy response and the early detection of tumour progression or recurrence. We also summarize and discuss studies of advanced approaches such as quantitative imaging, liquid biomarker discovery and machine intelligence that hold exceptional potential to aid in the therapy monitoring of this malignancy and early prediction of therapy response, which may decisively transform the conventional detection methods in the era of precision medicine.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Jing Li
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - C Chad Quarles
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ekokobe Fonkem
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Medical Education, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- Department of Oncology and LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
4
|
Bressler I, Ben Bashat D, Buchsweiler Y, Aizenstein O, Limon D, Bokestein F, Blumenthal TD, Nevo U, Artzi M. Model-free dynamic contrast-enhanced MRI analysis: differentiation between active tumor and necrotic tissue in patients with glioblastoma. MAGMA (NEW YORK, N.Y.) 2023; 36:33-42. [PMID: 36287282 DOI: 10.1007/s10334-022-01045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/09/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Treatment response assessment in patients with high-grade gliomas (HGG) is heavily dependent on changes in lesion size on MRI. However, in conventional MRI, treatment-related changes can appear as enhancing tissue, with similar presentation to that of active tumor tissue. We propose a model-free data-driven method for differentiation between these tissues, based on dynamic contrast-enhanced (DCE) MRI. MATERIALS AND METHODS The study included a total of 66 scans of patients with glioblastoma. Of these, 48 were acquired from 1 MRI vendor and 18 scans were acquired from a different MRI vendor and used as test data. Of the 48, 24 scans had biopsy results. Analysis included semi-automatic arterial input function (AIF) extraction, direct DCE pharmacokinetic-like feature extraction, and unsupervised clustering of the two tissue types. Validation was performed via (a) comparison to biopsy result (b) correlation to literature-based DCE curves for each tissue type, and (c) comparison to clinical outcome. RESULTS Consistency between the model prediction and biopsy results was found in 20/24 cases. An average correlation of 82% for active tumor and 90% for treatment-related changes was found between the predicted component and population-based templates. An agreement between the predicted results and radiologist's assessment, based on RANO criteria, was found in 11/12 cases. CONCLUSION The proposed method could serve as a non-invasive method for differentiation between lesion tissue and treatment-related changes.
Collapse
Affiliation(s)
- Idan Bressler
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Dafna Ben Bashat
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yuval Buchsweiler
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Orna Aizenstein
- Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel.,Division of Radiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Dror Limon
- Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel.,Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Felix Bokestein
- Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel.,Neuro-Oncology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - T Deborah Blumenthal
- Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel.,Neuro-Oncology Service, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Uri Nevo
- The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Moran Artzi
- Sagol Brain Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, 6 Weizmann St, 64239, Tel-Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
McCarthy L, Verma G, Hangel G, Neal A, Moffat BA, Stockmann JP, Andronesi OC, Balchandani P, Hadjipanayis CG. Application of 7T MRS to High-Grade Gliomas. AJNR Am J Neuroradiol 2022; 43:1378-1395. [PMID: 35618424 PMCID: PMC9575545 DOI: 10.3174/ajnr.a7502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/11/2022] [Indexed: 01/26/2023]
Abstract
MRS, including single-voxel spectroscopy and MR spectroscopic imaging, captures metabolites in high-grade gliomas. Emerging evidence indicates that 7T MRS may be more sensitive to aberrant metabolic activity than lower-field strength MRS. However, the literature on the use of 7T MRS to visualize high-grade gliomas has not been summarized. We aimed to identify metabolic information provided by 7T MRS, optimal spectroscopic sequences, and areas for improvement in and new applications for 7T MRS. Literature was found on PubMed using "high-grade glioma," "malignant glioma," "glioblastoma," "anaplastic astrocytoma," "7T," "MR spectroscopy," and "MR spectroscopic imaging." 7T MRS offers higher SNR, modestly improved spatial resolution, and better resolution of overlapping resonances. 7T MRS also yields reduced Cramér-Rao lower bound values. These features help to quantify D-2-hydroxyglutarate in isocitrate dehydrogenase 1 and 2 gliomas and to isolate variable glutamate, increased glutamine, and increased glycine with higher sensitivity and specificity. 7T MRS may better characterize tumor infiltration and treatment effect in high-grade gliomas, though further study is necessary. 7T MRS will benefit from increased sample size; reductions in field inhomogeneity, specific absorption rate, and acquisition time; and advanced editing techniques. These findings suggest that 7T MRS may advance understanding of high-grade glioma metabolism, with reduced Cramér-Rao lower bound values and better measurement of smaller metabolite signals. Nevertheless, 7T is not widely used clinically, and technical improvements are necessary. 7T MRS isolates metabolites that may be valuable therapeutic targets in high-grade gliomas, potentially resulting in wider ranging neuro-oncologic applications.
Collapse
Affiliation(s)
- L McCarthy
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| | - G Verma
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - G Hangel
- Department of Neurosurgery (G.H.)
- High-field MR Center (G.H.), Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - A Neal
- Department of Medicine (A.N.), Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
- Department of Neurology (A.N.), Royal Melbourne Hospital, Melbourne, Australia
| | - B A Moffat
- The Melbourne Brain Centre Imaging Unit (B.A.M.), Department of Radiology, The University of Melbourne, Melbourne, Australia
| | - J P Stockmann
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - O C Andronesi
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - P Balchandani
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - C G Hadjipanayis
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| |
Collapse
|
6
|
Chawla S, Bukhari S, Afridi OM, Wang S, Yadav SK, Akbari H, Verma G, Nath K, Haris M, Bagley S, Davatzikos C, Loevner LA, Mohan S. Metabolic and physiologic magnetic resonance imaging in distinguishing true progression from pseudoprogression in patients with glioblastoma. NMR IN BIOMEDICINE 2022; 35:e4719. [PMID: 35233862 PMCID: PMC9203929 DOI: 10.1002/nbm.4719] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 05/15/2023]
Abstract
Pseudoprogression (PsP) refers to treatment-related clinico-radiologic changes mimicking true progression (TP) that occurs in patients with glioblastoma (GBM), predominantly within the first 6 months after the completion of surgery and concurrent chemoradiation therapy (CCRT) with temozolomide. Accurate differentiation of TP from PsP is essential for making informed decisions on appropriate therapeutic intervention as well as for prognostication of these patients. Conventional neuroimaging findings are often equivocal in distinguishing between TP and PsP and present a considerable diagnostic dilemma to oncologists and radiologists. These challenges have emphasized the need for developing alternative imaging techniques that may aid in the accurate diagnosis of TP and PsP. In this review, we encapsulate the current state of knowledge in the clinical applications of commonly used metabolic and physiologic magnetic resonance (MR) imaging techniques such as diffusion and perfusion imaging and proton spectroscopy in distinguishing TP from PsP. We also showcase the potential of promising imaging techniques, such as amide proton transfer and amino acid-based positron emission tomography, in providing useful information about the treatment response. Additionally, we highlight the role of "radiomics", which is an emerging field of radiology that has the potential to change the way in which advanced MR techniques are utilized in assessing treatment response in GBM patients. Finally, we present our institutional experiences and discuss future perspectives on the role of multiparametric MR imaging in identifying PsP in GBM patients treated with "standard-of-care" CCRT as well as novel/targeted therapies.
Collapse
Affiliation(s)
- Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sultan Bukhari
- Rowan School of Osteopathic Medicine at Rowan University, Voorhees, New Jersey, USA
| | - Omar M. Afridi
- Rowan School of Osteopathic Medicine at Rowan University, Voorhees, New Jersey, USA
| | - Sumei Wang
- Department of Cardiology, Lenox Hill Hospital, Northwell Health, New York, New York, USA
| | - Santosh K. Yadav
- Laboratory of Functional and Molecular Imaging, Sidra Medicine, Doha, Qatar
| | - Hamed Akbari
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gaurav Verma
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Kavindra Nath
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mohammad Haris
- Laboratory of Functional and Molecular Imaging, Sidra Medicine, Doha, Qatar
| | - Stephen Bagley
- Department of Hematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laurie A. Loevner
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Guglielmo P, Quartuccio N, Rossetti V, Celli M, Alongi P, Boero M, Arnone G, Baldari S, Matteucci F, Laudicella R. [ 18F] Fluorothymidine Positron Emission Tomography Imaging in Primary Brain Tumours: A Systematic Review. Curr Med Imaging 2022; 18:363-371. [PMID: 34533446 DOI: 10.2174/1573405617666210917123012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE This review aimed to summarize the available literature on the clinical application of [18F] FLT PET imaging in primary brain tumours. METHODS A comprehensive search strategy based on Pubmed/Medline, Scopus, Web of Science, Cochrane Library, Google Scholar, and the Embase databases was carried on using the following search string: ('3` Fluorothymidine'/exp OR 'FLT' OR '[81F]-FLT' OR '[18F] Fluorothymidine') AND ('pet'/exp OR 'pet' OR 'positron emission tomography') AND ('glioma'/exp OR 'glioma' OR 'brain tumour'/exp OR 'brain tumour'). The search was updated till March 2021 and only articles in English and studies investigating the clinical applications of [18F] FLT PET and PET/CT in primary brain tumours were considered eligible for inclusion. RESULTS The literature search ultimately yielded 52 studies included in the systematic review, with main results as follows: a) the uptake of [18F] FLT may guide stereotactic biopsy but does not discriminate between grade II and III glioma. b) [18F] FLT uptake and texture parameters correlate with overall survival (OS) in newly diagnosed gliomas. c) In patients with recurrent glioma, proliferative volume (PV) and tumour-to-normal brain (T/N) uptake ratio are independent predictors of survival. d) Patients demonstrating response to therapy at [18F] FLT PET scan show longer OS compared to non-responders. e) [18F] FLT PET demonstrated good performance in discriminating tumour recurrence from radionecrosis. However, controversial results exist in comparative literature examining the performance of [18F] FLT vs. other radiotracers in the assessment of recurrence. CONCLUSION [18F] FLT PET imaging has demonstrated potential benefits for grading, diagnostic and prognostic purposes, despite the small sample size studies due to the relatively low availability of the radiotracer.
Collapse
Affiliation(s)
| | - Natale Quartuccio
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Italy
| | - Virginia Rossetti
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Monica Celli
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Ct. da Pietra Pollastra-pisciotto, Cefalù. Italy
| | - Michele Boero
- Nuclear Medicine Unit, AO Brotzu, 09134 Cagliari, Italy
| | - Gaspare Arnone
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Italy
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Federica Matteucci
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
8
|
Qiu J, Tao ZC, Deng KX, Wang P, Chen CY, Xiao F, Luo Y, Yuan SY, Chen H, Huang H. Diagnostic accuracy of dynamic contrast-enhanced magnetic resonance imaging for distinguishing pseudoprogression from glioma recurrence: a meta-analysis. Chin Med J (Engl) 2021; 134:2535-2543. [PMID: 34748524 PMCID: PMC8577681 DOI: 10.1097/cm9.0000000000001445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND It is crucial to differentiate accurately glioma recurrence and pseudoprogression which have entirely different prognosis and require different treatment strategies. This study aimed to assess the diagnostic accuracy of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) as a tool for distinguishing glioma recurrence and pseudoprogression. METHODS According to particular criteria of inclusion and exclusion, related studies up to May 1, 2019, were thoroughly searched from several databases including PubMed, Embase, Cochrane Library, and Chinese biomedical databases. The quality assessment of diagnostic accuracy studies was applied to evaluate the quality of the included studies. By using the "mada" package in R, the heterogeneity, overall sensitivity, specificity, and diagnostic odds ratio were calculated. Moreover, funnel plots were used to visualize and estimate the publication bias in this study. The area under the summary receiver operating characteristic (SROC) curve was computed to display the diagnostic efficiency of DCE-MRI. RESULTS In the present meta-analysis, a total of 11 studies covering 616 patients were included. The results showed that the pooled sensitivity, specificity, and diagnostic odds ratio were 0.792 (95% confidence interval [CI] 0.707-0.857), 0.779 (95% CI 0.715-0.832), and 16.219 (97.5% CI 9.123-28.833), respectively. The value of the area under the SROC curve was 0.846. In addition, the SROC curve showed high sensitivities (>0.6) and low false positive rates (<0.5) from most of the included studies, which suggest that the results of our study were reliable. Furthermore, the funnel plot suggested the existence of publication bias. CONCLUSIONS While the DCE-MRI is not the perfect diagnostic tool for distinguishing glioma recurrence and pseudoprogression, it was capable of improving diagnostic accuracy. Hence, further investigations combining DCE-MRI with other imaging modalities are required to establish an efficient diagnostic method for glioma patients.
Collapse
Affiliation(s)
- Jun Qiu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Zhen-Chao Tao
- Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Ke-Xue Deng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Peng Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Chuan-Yu Chen
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Fang Xiao
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Yi Luo
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Shu-Ya Yuan
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Hao Chen
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Huan Huang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230036, China
| |
Collapse
|
9
|
Cao Y, Tang D, Xiang Y, Men L, Liu C, Zhou Q, Wu J, Huo L, Song T, Wang Y, Li Z, Wei R, Shen L, Yang Z, Hong J. Study on the Appropriate Timing of Postoperative Adaptive Radiotherapy for High-Grade Glioma. Cancer Manag Res 2021; 13:3561-3572. [PMID: 33953610 PMCID: PMC8089024 DOI: 10.2147/cmar.s300094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/02/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To investigate the appropriate timing of adaptive radiotherapy (ART) for high-grade glioma. METHODS Ten patients with high-grade gliomas were selected and underwent CT/MRI (CT1/MRI1, CT2/MRI2, CT3/MRI3, and CT4/MRI4) scans before RT and during 10-, 20- and 30-fraction RT, and the corresponding RT plans (plan1, plan2, plan3 and plan4) were made. The dose of the initial plan (plan1) was projected to CT2 and CT3 using the image registration technique to obtain the projection plans (plan1-2 and plan1-3) and by superimposing the doses to obtain the ART plans (plan10+20 and plan20+10), respectively. The dosimetric differences in the target volume and organs at risk (OARs) were compared between the projection and adaptive plans. The tumor control probability (TCP) for the planning target volume (PTV) and normal tissue complication probability (NTCP) for the OARs were compared between the two adaptive plans. RESULTS Compared with the projection plan, the D2 to the PTV of ART decreased, the conformity index (CI) to the PTV increased, and the D2/Dmean to the brainstem, optic chiasm and pituitary, as well as the V20, V30, V40 and V50 to the normal brain decreased. The D2 to the pituitary and optic chiasm as well as the V20, V30, V40 and V50 to the normal brain in plan10+20 were lower than those in plan20+10, while the CI to the PTV was higher than that in plan20+10. The TCP of the PTV in plan10+20 was higher than that in plan20+10. CONCLUSION ART can improve the precision of target volume irradiation and reduce the irradiation dose to the OARs in high-grade glioma. The time point after 10 fractions of RT is appropriate for ART.
Collapse
Affiliation(s)
- Ying Cao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Du Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yining Xiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Li Men
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chao Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Jun Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Lei Huo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Tao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Ying Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Rui Wei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhen Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Jidong Hong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
10
|
Nabors LB, Portnow J, Ahluwalia M, Baehring J, Brem H, Brem S, Butowski N, Campian JL, Clark SW, Fabiano AJ, Forsyth P, Hattangadi-Gluth J, Holdhoff M, Horbinski C, Junck L, Kaley T, Kumthekar P, Loeffler JS, Mrugala MM, Nagpal S, Pandey M, Parney I, Peters K, Puduvalli VK, Robins I, Rockhill J, Rusthoven C, Shonka N, Shrieve DC, Swinnen LJ, Weiss S, Wen PY, Willmarth NE, Bergman MA, Darlow SD. Central Nervous System Cancers, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 18:1537-1570. [PMID: 33152694 DOI: 10.6004/jnccn.2020.0052] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Central Nervous System (CNS) Cancers focus on management of adult CNS cancers ranging from noninvasive and surgically curable pilocytic astrocytomas to metastatic brain disease. The involvement of an interdisciplinary team, including neurosurgeons, radiation therapists, oncologists, neurologists, and neuroradiologists, is a key factor in the appropriate management of CNS cancers. Integrated histopathologic and molecular characterization of brain tumors such as gliomas should be standard practice. This article describes NCCN Guidelines recommendations for WHO grade I, II, III, and IV gliomas. Treatment of brain metastases, the most common intracranial tumors in adults, is also described.
Collapse
Affiliation(s)
| | | | - Manmeet Ahluwalia
- 3Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Henry Brem
- 5The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Steven Brem
- 6Abramson Cancer Center at the University of Pennsylvania
| | | | - Jian L Campian
- 8Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | - Craig Horbinski
- 13Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Larry Junck
- 14University of Michigan Rogel Cancer Center
| | | | - Priya Kumthekar
- 13Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Manjari Pandey
- 19St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | - Vinay K Puduvalli
- 21The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Ian Robins
- 22University of Wisconsin Carbone Cancer Center
| | - Jason Rockhill
- 23Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | | | - Lode J Swinnen
- 5The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | |
Collapse
|
11
|
Young RJ, Demétrio De Souza França P, Pirovano G, Piotrowski AF, Nicklin PJ, Riedl CC, Schwartz J, Bale TA, Donabedian PL, Kossatz S, Burnazi EM, Roberts S, Lyashchenko SK, Miller AM, Moss NS, Fiasconaro M, Zhang Z, Mauguen A, Reiner T, Dunphy MP. Preclinical and first-in-human-brain-cancer applications of [ 18F]poly (ADP-ribose) polymerase inhibitor PET/MR. Neurooncol Adv 2020; 2:vdaa119. [PMID: 33392502 PMCID: PMC7758909 DOI: 10.1093/noajnl/vdaa119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background We report preclinical and first-in-human-brain-cancer data using a targeted poly (ADP-ribose) polymerase 1 (PARP1) binding PET tracer, [18F]PARPi, as a diagnostic tool to differentiate between brain cancers and treatment-related changes. Methods We applied a glioma model in p53-deficient nestin/tv-a mice, which were injected with [18F]PARPi and then sacrificed 1 h post-injection for brain examination. We also prospectively enrolled patients with brain cancers to undergo dynamic [18F]PARPi acquisition on a dedicated positron emission tomography/magnetic resonance (PET/MR) scanner. Lesion diagnosis was established by pathology when available or by Response Assessment in Neuro-Oncology (RANO) or RANO-BM response criteria. Resected tissue also underwent PARPi-FL staining and PARP1 immunohistochemistry. Results In a preclinical mouse model, we illustrated that [18F]PARPi crossed the blood–brain barrier and specifically bound to PARP1 overexpressed in cancer cell nuclei. In humans, we demonstrated high [18F]PARPi uptake on PET/MR in active brain cancers and low uptake in treatment-related changes independent of blood–brain barrier disruption. Immunohistochemistry results confirmed higher PARP1 expression in cancerous than in noncancerous tissue. Specificity was also corroborated by blocking fluorescent tracer uptake with an excess unlabeled PARP inhibitor in patient cancer biospecimen. Conclusions Although larger studies are necessary to confirm and further explore this tracer, we describe the promising performance of [18F]PARPi as a diagnostic tool to evaluate patients with brain cancers and possible treatment-related changes.
Collapse
Affiliation(s)
- Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Paula Demétrio De Souza França
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, São Paulo, Brazil
| | - Giacomo Pirovano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anna F Piotrowski
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Philip J Nicklin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christopher C Riedl
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Tejus A Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Patrick L Donabedian
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Eva M Burnazi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexandra M Miller
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nelson S Moss
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Megan Fiasconaro
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zhigang Zhang
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Audrey Mauguen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Weill Cornell Medical College, New York, New York, USA.,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark P Dunphy
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
12
|
Hoxworth JM, Eschbacher JM, Gonzales AC, Singleton KW, Leon GD, Smith KA, Stokes AM, Zhou Y, Mazza GL, Porter AB, Mrugala MM, Zimmerman RS, Bendok BR, Patra DP, Krishna C, Boxerman JL, Baxter LC, Swanson KR, Quarles CC, Schmainda KM, Hu LS. Performance of Standardized Relative CBV for Quantifying Regional Histologic Tumor Burden in Recurrent High-Grade Glioma: Comparison against Normalized Relative CBV Using Image-Localized Stereotactic Biopsies. AJNR Am J Neuroradiol 2020; 41:408-415. [PMID: 32165359 DOI: 10.3174/ajnr.a6486] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/23/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Perfusion MR imaging measures of relative CBV can distinguish recurrent tumor from posttreatment radiation effects in high-grade gliomas. Currently, relative CBV measurement requires normalization based on user-defined reference tissues. A recently proposed method of relative CBV standardization eliminates the need for user input. This study compares the predictive performance of relative CBV standardization against relative CBV normalization for quantifying recurrent tumor burden in high-grade gliomas relative to posttreatment radiation effects. MATERIALS AND METHODS We recruited 38 previously treated patients with high-grade gliomas (World Health Organization grades III or IV) undergoing surgical re-resection for new contrast-enhancing lesions concerning for recurrent tumor versus posttreatment radiation effects. We recovered 112 image-localized biopsies and quantified the percentage of histologic tumor content versus posttreatment radiation effects for each sample. We measured spatially matched normalized and standardized relative CBV metrics (mean, median) and fractional tumor burden for each biopsy. We compared relative CBV performance to predict tumor content, including the Pearson correlation (r), against histologic tumor content (0%-100%) and the receiver operating characteristic area under the curve for predicting high-versus-low tumor content using binary histologic cutoffs (≥50%; ≥80% tumor). RESULTS Across relative CBV metrics, fractional tumor burden showed the highest correlations with tumor content (0%-100%) for normalized (r = 0.63, P < .001) and standardized (r = 0.66, P < .001) values. With binary cutoffs (ie, ≥50%; ≥80% tumor), predictive accuracies were similar for both standardized and normalized metrics and across relative CBV metrics. Median relative CBV achieved the highest area under the curve (normalized = 0.87, standardized = 0.86) for predicting ≥50% tumor, while fractional tumor burden achieved the highest area under the curve (normalized = 0.77, standardized = 0.80) for predicting ≥80% tumor. CONCLUSIONS Standardization of relative CBV achieves similar performance compared with normalized relative CBV and offers an important step toward workflow optimization and consensus methodology.
Collapse
Affiliation(s)
- J M Hoxworth
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| | | | | | - K W Singleton
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - G D Leon
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - K A Smith
- Keller Center for Imaging Innovation (A.M.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - A M Stokes
- Keller Center for Imaging Innovation (A.M.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - Y Zhou
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| | - G L Mazza
- Department of Health Sciences Research (G.L.M.), Division of Biomedical Statistics and Informatics, Mayo Clinic Scottsdale, Scottsdale, Arizona
| | | | | | | | - B R Bendok
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | - D P Patra
- Departments of Neurosurgery (D.P.P.)
| | | | - J L Boxerman
- Department of Diagnostic Imaging (J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - L C Baxter
- Neuropsychology (L.C.B.), Mayo Clinic Hospital, Phoenix, Arizona
| | - K R Swanson
- Precision Neurotherapeutics Lab (K.W.S., G.D.L., B.R.B., K.R.S.), Mayo Clinic in Arizona, Phoenix, Arizona
| | | | - K M Schmainda
- Department of Radiology (K.M.S.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - L S Hu
- From the Departments of Radiology (J.M.H., Y.Z., L.S.H.)
| |
Collapse
|
13
|
Hu LS, Hawkins-Daarud A, Wang L, Li J, Swanson KR. Imaging of intratumoral heterogeneity in high-grade glioma. Cancer Lett 2020; 477:97-106. [PMID: 32112907 DOI: 10.1016/j.canlet.2020.02.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
High-grade glioma (HGG), and particularly Glioblastoma (GBM), can exhibit pronounced intratumoral heterogeneity that confounds clinical diagnosis and management. While conventional contrast-enhanced MRI lacks the capability to resolve this heterogeneity, advanced MRI techniques and PET imaging offer a spectrum of physiologic and biophysical image features to improve the specificity of imaging diagnoses. Published studies have shown how integrating these advanced techniques can help better define histologically distinct targets for surgical and radiation treatment planning, and help evaluate the regional heterogeneity of tumor recurrence and response assessment following standard adjuvant therapy. Application of texture analysis and machine learning (ML) algorithms has also enabled the emerging field of radiogenomics, which can spatially resolve the regional and genetically distinct subpopulations that coexist within a single GBM tumor. This review focuses on the latest advances in neuro-oncologic imaging and their clinical applications for the assessment of intratumoral heterogeneity.
Collapse
Affiliation(s)
- Leland S Hu
- Department of Radiology, Mayo Clinic Arizona, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA.
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 East Mayo Blvd, Support, Services Building Suite 2-700, Phoenix, AZ, 85054, USA.
| | - Lujia Wang
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.
| | - Jing Li
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, 699 S Mill Ave, Tempe, AZ, 85281, USA.
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, 5777 East Mayo Blvd, Support, Services Building Suite 2-700, Phoenix, AZ, 85054, USA.
| |
Collapse
|
14
|
Henderson F, Brem S, O'Rourke DM, Nasrallah M, Buch VP, Young AJ, Doot RK, Pantel A, Desai A, Bagley SJ, Nabavizadeh SA. 18F-Fluciclovine PET to distinguish treatment-related effects from disease progression in recurrent glioblastoma: PET fusion with MRI guides neurosurgical sampling. Neurooncol Pract 2019; 7:152-157. [PMID: 32206320 PMCID: PMC7081387 DOI: 10.1093/nop/npz068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Differentiation of true tumor progression from treatment-related effects remains a major unmet need in caring for patients with glioblastoma. Here, we report how the intraoperative combination of MRI with18F-fluciclovine PET guided surgical sampling in 2 patients with recurrent glioblastoma.18F-Fluciclovine PET is FDA approved for use in prostate cancer and carries an orphan drug designation in glioma. To investigate its utility in recurrent glioblastoma, we fused PET and MRI images using 2 different surgical navigation systems and performed targeted stereotactic biopsies from the areas of high (“hot”) and low (“cold”) radiotracer uptake. Concordant histopathologic and imaging findings suggest that a combined18F-fluciclovine PET-MRI–guided approach can guide neurosurgical resection of viable recurrent glioblastoma in the background of treatment-related effects, which can otherwise look similar on MRI.
Collapse
Affiliation(s)
- Fraser Henderson
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia.,Department of Neurosurgery, Medical University of South Carolina, Charleston
| | - Steven Brem
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Donald M O'Rourke
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - MacLean Nasrallah
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivek P Buch
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
| | - Anthony J Young
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - Robert K Doot
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - Austin Pantel
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - Arati Desai
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Philadelphia.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Stephen J Bagley
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Philadelphia.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - S Ali Nabavizadeh
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| |
Collapse
|
15
|
Boxerman JL, Zhang Z, Safriel Y, Rogg JM, Wolf RL, Mohan S, Marques H, Sorensen AG, Gilbert MR, Barboriak DP. Prognostic value of contrast enhancement and FLAIR for survival in newly diagnosed glioblastoma treated with and without bevacizumab: results from ACRIN 6686. Neuro Oncol 2019; 20:1400-1410. [PMID: 29590461 DOI: 10.1093/neuonc/noy049] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background ACRIN 6686/RTOG 0825 was a phase III trial of conventional chemoradiation plus adjuvant temozolomide with bevacizumab or without (placebo) in newly diagnosed glioblastoma. This study investigated whether changes in contrast-enhancing and fluid attenuated inversion recovery (FLAIR)-hyperintense tumor assessed by central reading prognosticate overall survival (OS). Methods Two hundred eighty-four patients (171 men; median age 57 y, range 19-79; 159 on bevacizumab) had MRI at post-op (baseline) and pre-cycle 4 of adjuvant temozolomide (22 wk post chemoradiation initiation). Four central readers measured bidimensional lesion enhancement (2D-T1) and FLAIR hyperintensity at both time points. Changes from baseline to pre-cycle 4 for both markers were dichotomized (increasing vs non-increasing). Cox proportional hazards model and Kaplan-Meier survival estimates were used for inference. Results Adjusting for treatment, increasing 2D-T1 (n = 262, hazard ratio [HR] = 2.07, 95% CI: 1.48-2.91, P < 0.0001) and FLAIR (n = 273, HR = 1.75, 95% CI: 1.26-2.41, P = 0.0008) significantly predicted worse OS. Median OS (days) was significantly shorter for patients with increasing versus non-increasing 2D-T1 for both bevacizumab (443 vs 535, P = 0.004) and placebo (526 vs 887, P = 0.001). Median OS was significantly shorter for patients with increasing versus non-increasing FLAIR for placebo (595 vs 872, P = 0.001), and trended similarly for bevacizumab (499 vs 535, P = 0.0935). Adjusting for 2D-T1 and treatment, increasing FLAIR represented significantly higher risk for death (HR = 1.59 [1.11-2.26], P = 0.01). Conclusion Increased 2D-T1 significantly predicts worse OS in both treatment groups, implying absence of a substantial proportion of pseudoprogression 22 weeks after initiation of standard therapy. FLAIR adds value beyond 2D-T1 in predicting OS, potentially addressing the pseudoresponse effect by substratifying bevacizumab-treated patients with non-increasing 2D-T1.
Collapse
Affiliation(s)
- Jerrold L Boxerman
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Zheng Zhang
- Center for Statistical Sciences, Brown University, Providence, Rhode Island
| | - Yair Safriel
- Pharmascan Clinical Trials and Radiology Associates of Clearwater-University of South Florida, Clearwater, Florida
| | - Jeffrey M Rogg
- Department of Diagnostic Imaging, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island
| | - Ronald L Wolf
- Department of Radiology, Neuroradiology Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Suyash Mohan
- Department of Radiology, Neuroradiology Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Helga Marques
- Center for Statistical Sciences, Brown University, Providence, Rhode Island
| | - A Gregory Sorensen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts.,IMRIS, Deerfield Imaging, Inc, Minnetonka, Minnesota
| | - Mark R Gilbert
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Neuro-Oncology Branch of the National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel P Barboriak
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
16
|
Verma G, Chawla S, Mohan S, Wang S, Nasrallah M, Sheriff S, Desai A, Brem S, O'Rourke DM, Wolf RL, Maudsley AA, Poptani H. Three-dimensional echo planar spectroscopic imaging for differentiation of true progression from pseudoprogression in patients with glioblastoma. NMR IN BIOMEDICINE 2019; 32:e4042. [PMID: 30556932 PMCID: PMC6519064 DOI: 10.1002/nbm.4042] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 05/20/2023]
Abstract
Accurate differentiation of true progression (TP) from pseudoprogression (PsP) in patients with glioblastomas (GBMs) is essential for planning adequate treatment and for estimating clinical outcome measures and future prognosis. The purpose of this study was to investigate the utility of three-dimensional echo planar spectroscopic imaging (3D-EPSI) in distinguishing TP from PsP in GBM patients. For this institutional review board approved and HIPAA compliant retrospective study, 27 patients with GBM demonstrating enhancing lesions within six months of completion of concurrent chemo-radiation therapy were included. Of these, 18 were subsequently classified as TP and 9 as PsP based on histological features or follow-up MRI studies. Parametric maps of choline/creatine (Cho/Cr) and choline/N-acetylaspartate (Cho/NAA) were computed and co-registered with post-contrast T1 -weighted and FLAIR images. All lesions were segmented into contrast enhancing (CER), immediate peritumoral (IPR), and distal peritumoral (DPR) regions. For each region, Cho/Cr and Cho/NAA ratios were normalized to corresponding metabolite ratios from contralateral normal parenchyma and compared between TP and PsP groups. Logistic regression analyses were performed to obtain the best model to distinguish TP from PsP. Significantly higher Cho/NAA was observed from CER (2.69 ± 1.00 versus 1.56 ± 0.51, p = 0.003), IPR (2.31 ± 0.92 versus 1.53 ± 0.56, p = 0.030), and DPR (1.80 ± 0.68 versus 1.19 ± 0.28, p = 0.035) regions in TP patients compared with those with PsP. Additionally, significantly elevated Cho/Cr (1.74 ± 0.44 versus 1.34 ± 0.26, p = 0.023) from CER was observed in TP compared with PsP. When these parameters were incorporated in multivariate regression analyses, a discriminatory model with a sensitivity of 94% and a specificity of 87% was observed in distinguishing TP from PsP. These results indicate the utility of 3D-EPSI in differentiating TP from PsP with high sensitivity and specificity.
Collapse
Affiliation(s)
- Gaurav Verma
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Sanjeev Chawla
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Suyash Mohan
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Sumei Wang
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - MacLean Nasrallah
- Department of Pathology and Lab MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | | | - Arati Desai
- Department of Hematology‐OncologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Steven Brem
- Department of NeurosurgeryPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Donald M. O'Rourke
- Department of NeurosurgeryPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | - Ronald L. Wolf
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
| | | | - Harish Poptani
- Department of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPAUSA
- Department of Cellular and Molecular PhysiologyUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
17
|
Rowe LS, Butman JA, Mackey M, Shih JH, Cooley-Zgela T, Ning H, Gilbert MR, Smart DK, Camphausen K, Krauze AV. Differentiating pseudoprogression from true progression: analysis of radiographic, biologic, and clinical clues in GBM. J Neurooncol 2018; 139:145-152. [PMID: 29767308 PMCID: PMC7983158 DOI: 10.1007/s11060-018-2855-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/31/2018] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Pseudoprogression (PsP) is a diagnostic dilemma in glioblastoma (GBM) after chemoradiotherapy (CRT). Magnetic resonance imaging (MRI) features may fail to distinguish PsP from early true progression (eTP), however clinical findings may aid in their distinction. METHODS Sixty-seven patients received CRT for GBM between 2003 and 2016, and had pre- and post-treatment imaging suitable for retrospective evaluation using RANO criteria. Patients with signs of progression within the first 12-weeks post-radiation (P-12) were selected. Lesions that improved or stabilized were defined as PsP, and lesions that progressed were defined as eTP. RESULTS The median follow up for all patients was 17.6 months. Signs of progression developed in 35/67 (52.2%) patients within P-12. Of these, 20/35 (57.1%) were subsequently defined as eTP and 15/35 (42.9%) as PsP. MRI demonstrated increased contrast enhancement in 84.2% of eTP and 100% of PsP, and elevated CBV in 73.7% for eTP and 93.3% for PsP. A decrease in FLAIR was not seen in eTP patients, but was seen in 26.7% PsP patients. Patients with eTP were significantly more likely to require increased steroid doses or suffer clinical decline than PsP patients (OR 4.89, 95% CI 1.003-19.27; p = 0.046). KPS declined in 25% with eTP and none of the PsP patients. CONCLUSIONS MRI imaging did not differentiate eTP from PsP, however, KPS decline or need for increased steroids was significantly more common in eTP versus PsP. Investigation and standardization of clinical assessments in response criteria may help address the diagnostic dilemma of pseudoprogression after frontline treatment for GBM.
Collapse
Affiliation(s)
- Lindsay S Rowe
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA.
| | - John A Butman
- Radiology and Imaging Sciences, National Institutes of Health, 10 Center Drive Magnuson Clinical Center, MSC 1182, Bethesda, MD, 20892, USA
| | - Megan Mackey
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| | - Joanna H Shih
- Clinical Research Center, National Institutes of Health, 10 Center Drive Magnuson Clinical Center, Bethesda, MD, 20892, USA
| | - Theresa Cooley-Zgela
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| | - Holly Ning
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Building 82, Room 235A, Bethesda, MD, 20892, USA
| | - DeeDee K Smart
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| | - Andra V Krauze
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD, 20892, USA
| |
Collapse
|
18
|
Serial FLT PET imaging to discriminate between true progression and pseudoprogression in patients with newly diagnosed glioblastoma: a long-term follow-up study. Eur J Nucl Med Mol Imaging 2018; 45:2404-2412. [PMID: 30032322 PMCID: PMC6208814 DOI: 10.1007/s00259-018-4090-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/09/2018] [Indexed: 12/23/2022]
Abstract
Purpose Response evaluation in patients with glioblastoma after chemoradiotherapy is challenging due to progressive, contrast-enhancing lesions on MRI that do not reflect true tumour progression. In this study, we prospectively evaluated the ability of the PET tracer 18F-fluorothymidine (FLT), a tracer reflecting proliferative activity, to discriminate between true progression and pseudoprogression in newly diagnosed glioblastoma patients treated with chemoradiotherapy. Methods FLT PET and MRI scans were performed before and 4 weeks after chemoradiotherapy. MRI scans were also performed after three cycles of adjuvant temozolomide. Pseudoprogression was defined as progressive disease on MRI after chemoradiotherapy with stabilisation or reduction of contrast-enhanced lesions after three cycles of temozolomide, and was compared with the disease course during long-term follow-up. Changes in maximum standardized uptake value (SUVmax) and tumour-to-normal uptake ratios were calculated for FLT and are presented as the mean SUVmax for multiple lesions. Results Between 2009 and 2012, 30 patients were included. Of 24 evaluable patients, 7 showed pseudoprogression and 7 had true progression as defined by MRI response. FLT PET parameters did not significantly differ between patients with true progression and pseudoprogression defined by MRI. The correlation between change in SUVmax and survival (p = 0.059) almost reached the standard level of statistical significance. Lower baseline FLT PET uptake was significantly correlated with improved survival (p = 0.022). Conclusion Baseline FLT uptake appears to be predictive of overall survival. Furthermore, changes in SUVmax over time showed a tendency to be associated with improved survival. However, further studies are necessary to investigate the ability of FLT PET imaging to discriminate between true progression and pseudoprogression in patients with glioblastoma.
Collapse
|
19
|
Clinical, Radiographic, and Pathologic Findings in Patients Undergoing Reoperation Following Radiation Therapy and Temozolomide for Newly Diagnosed Glioblastoma. Am J Clin Oncol 2017; 40:219-222. [PMID: 26491903 DOI: 10.1097/coc.0000000000000136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Patients with glioblastoma (GBM) frequently deteriorate clinically and radiographically after chemoradiation and may require repeat surgical intervention. We attempted to correlate pathologic findings with preoperative clinical characteristics and survival in patients undergoing reoperation for GBM. MATERIALS AND METHODS Patients eligible for this retrospective analysis had pathologically confirmed GBM diagnosed between 2005 and 2010, received standard radiation and temozolomide, and underwent repeat resection within 18 months of diagnosis. RESULTS Thirty-eight patients were identified. Median age was 56 years (range, 30 to 80 y), 55% were male, and 66% had baseline performance status ≥90%. Median survival was 16.3 months (95% confidence interval [CI], 13.3-19.8) from initial surgery. At reoperation, 21% of patients had no pathologically evident tumor. Median time from initial diagnosis to second surgery was similar in patients with and without evident tumor (8.5 vs. 8.8 mo, respectively). Patients without evident tumor tended to have a worse performance status. Median overall survival from second surgery was 7 months (95% CI, 4.2-10.1) and 9.1 months (95% CI, 2.1-25.3) for patients with and without evident tumor, respectively. Multivariate proportional hazards analysis showed a hazard ratio for death of 0.61 (95% CI, 0.25-1.49) for patients without evident tumor after adjusting for Karnofsky performance status and second surgical procedure. CONCLUSIONS GBM patients with and without disease recurrence have similar clinical characteristics at the time of second surgical resection. Pathologic outcomes were not correlated with specific clinical or radiologic characteristics, including the time from diagnosis to reoperation. There was a trend toward improved overall survival among patients without evident tumor at reoperation.
Collapse
|
20
|
Brzezniak C, Oronsky B, Trepel J, Summers TA, Cabrales P, Lee MJ, Day R, Jha S, Caroen S, Zeman K, Ferry L, Harmer C, Oronsky N, Lybeck M, Lybeck HE, Brown JF, Reid TR, Carter CA. RRx-001 Priming of PD-1 Inhibition in the Treatment of Small Cell Carcinoma of the Vagina: A Rare Gynecological Tumor. Case Rep Oncol 2017; 10:276-280. [PMID: 28512410 PMCID: PMC5422722 DOI: 10.1159/000464101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/03/2022] Open
Abstract
Small cell carcinoma of the vagina is rare, so rare in fact that the total number reported in English-language journals is less than 30. Due to this extremely low incidence, no specific treatment guidelines have been established, and most of what is clinically known is derived from a handful of single case reports. However, as befitting its highly aggressive histologic features, which are reminiscent of small cell lung cancer (SCLC), first-line treatment is modeled after SCLC. Herein is reported the case of a 51-year-old African-American patient with metastatic biopsy-proven small cell carcinoma of the vagina that progressed through multiple therapies: first-line cisplatin and etoposide (making it platinum-resistant) and radiotherapy, followed by the tumor macrophage-stimulating agent RRx-001 in a clinical trial called QUADRUPLE THREAT, which per protocol preceded a mandated rechallenge with cisplatin and etoposide. RECIST v.1.1 tumor progression on both RRx-001 and cisplatin/etoposide was accompanied by central necrosis in several of the enlarged lymph nodes and hepatic metastases, which may have been evidence of pseudoprogression, accounting for her ongoing longer-than-expected survival, since the necrotic tissue may have primed the activity of the PD-1 inhibitor. The lack of response to RRx-001 is hypothesized to have correlated with sparse tumor macrophage infiltration, seen on pre- and post-treatment biopsies, since the mechanism of action of RRx-001 relates to stimulation of tumor-associated macrophages.
Collapse
Affiliation(s)
| | | | - Jane Trepel
- cDevelopmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Pedro Cabrales
- dDepartment of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Min-Jung Lee
- cDevelopmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Regina Day
- eDepartment of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | | - Karen Zeman
- aWalter Reed National Medical Center, Bethesda, MD, USA
| | - Lindsey Ferry
- aWalter Reed National Medical Center, Bethesda, MD, USA
| | | | | | | | | | | | - Tony R Reid
- hMoores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
21
|
van Dijken BRJ, van Laar PJ, Holtman GA, van der Hoorn A. Diagnostic accuracy of magnetic resonance imaging techniques for treatment response evaluation in patients with high-grade glioma, a systematic review and meta-analysis. Eur Radiol 2017; 27:4129-4144. [PMID: 28332014 PMCID: PMC5579204 DOI: 10.1007/s00330-017-4789-9] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/01/2017] [Accepted: 02/23/2017] [Indexed: 01/04/2023]
Abstract
Objective Treatment response assessment in high-grade gliomas uses contrast enhanced T1-weighted MRI, but is unreliable. Novel advanced MRI techniques have been studied, but the accuracy is not well known. Therefore, we performed a systematic meta-analysis to assess the diagnostic accuracy of anatomical and advanced MRI for treatment response in high-grade gliomas. Methods Databases were searched systematically. Study selection and data extraction were done by two authors independently. Meta-analysis was performed using a bivariate random effects model when ≥5 studies were included. Results Anatomical MRI (five studies, 166 patients) showed a pooled sensitivity and specificity of 68% (95%CI 51–81) and 77% (45–93), respectively. Pooled apparent diffusion coefficients (seven studies, 204 patients) demonstrated a sensitivity of 71% (60–80) and specificity of 87% (77–93). DSC-perfusion (18 studies, 708 patients) sensitivity was 87% (82–91) with a specificity of 86% (77–91). DCE-perfusion (five studies, 207 patients) sensitivity was 92% (73–98) and specificity was 85% (76–92). The sensitivity of spectroscopy (nine studies, 203 patients) was 91% (79–97) and specificity was 95% (65–99). Conclusion Advanced techniques showed higher diagnostic accuracy than anatomical MRI, the highest for spectroscopy, supporting the use in treatment response assessment in high-grade gliomas. Key points • Treatment response assessment in high-grade gliomas with anatomical MRI is unreliable • Novel advanced MRI techniques have been studied, but diagnostic accuracy is unknown • Meta-analysis demonstrates that advanced MRI showed higher diagnostic accuracy than anatomical MRI • Highest diagnostic accuracy for spectroscopy and perfusion MRI • Supports the incorporation of advanced MRI in high-grade glioma treatment response assessment Electronic supplementary material The online version of this article (doi:10.1007/s00330-017-4789-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bart R J van Dijken
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Peter Jan van Laar
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands
- University Medical Center Groningen, Center for Medical Imaging-North East Netherlands, University of Groningen, Groningen, The Netherlands
| | - Gea A Holtman
- University Medical Center Groningen, Department of General Practice, University of Groningen, Groningen, The Netherlands
| | - Anouk van der Hoorn
- University Medical Center Groningen Department of Radiology, University of Groningen, Hanzeplein 1, P. O. Box 30.001, 9700 RB, Groningen, The Netherlands.
- University Medical Center Groningen, Center for Medical Imaging-North East Netherlands, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
22
|
Lin AL, White M, Miller-Thomas MM, Fulton RS, Tsien CI, Rich KM, Schmidt RE, Tran DD, Dahiya S. Molecular and histologic characteristics of pseudoprogression in diffuse gliomas. J Neurooncol 2016; 130:529-533. [PMID: 27704386 PMCID: PMC5518746 DOI: 10.1007/s11060-016-2247-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/21/2016] [Indexed: 10/20/2022]
Abstract
During the 6 month period following chemoradiotherapy, gliomas frequently develop new areas of contrast enhancement, which are due to treatment effect rather than tumor progression. We sought to characterize this phenomenon in oligodendrogliomas (OG) and mixed oligoastrocytomas (MOA). We reviewed the imaging findings from 143 patients with a WHO grade II or III OG or MOA for evidence of pseudoprogression (PsP) or early tumor progression. We characterized these cases for 1p/19q codeletions by FISH, IDH1 R132H mutation by immunohistochemistry, and TP53, ATRX, and EGFR mutations by next generation sequencing. We then reviewed the pathologic specimens of the patient cases in which a re-resection was performed. We found that OG and MOA that are 1p/19q intact developed PsP at a higher rate than tumors that are 1p/19q codeleted (27 vs. 8 %). Moreover, IDH1 wild-type (WT) tumors developed PsP at a higher rate than IDH1 R132H cases (27 vs. 11 %). Patients with ATRX or TP53 mutations developed PsP at an intermediate rate of 21 %. Ten patients in our cohort underwent a re-resection for early contrast enhancement; these tumors were predominantly 1p/19q intact (90 %) and had a low rate of IDH1 R132H mutation (50 %). 8 of 10 tumors demonstrated primarily treatment effects, while the remaining 2 of 10 demonstrated recurrent/residual tumor of the same grade. Early contrast enhancement that develops during the first 6 months after chemoradiotherapy is typically due to PsP and occurs primarily in OG and MOA that are 1p/19q intact and IDH WT.
Collapse
Affiliation(s)
- Andrew L Lin
- Department of Neurology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Michael White
- Department of Neurology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Michelle M Miller-Thomas
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Keith M Rich
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - David D Tran
- Neuro-Oncology Program, Department of Internal Medicine, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA.
- Division of Neuro-Oncology, Department of Neurosurgery, University of Florida, 1149 S Newell Drive, Suite L2-100, Gainesville, FL, 32611, USA.
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA.
| |
Collapse
|
23
|
Pseudo progression identification of glioblastoma with dictionary learning. Comput Biol Med 2016; 73:94-101. [PMID: 27100835 DOI: 10.1016/j.compbiomed.2016.03.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Although the use of temozolomide in chemoradiotherapy is effective, the challenging clinical problem of pseudo progression has been raised in brain tumor treatment. This study aims to distinguish pseudo progression from true progression. MATERIALS AND METHODS Between 2000 and 2012, a total of 161 patients with glioblastoma multiforme (GBM) were treated with chemoradiotherapy at our hospital. Among the patients, 79 had their diffusion tensor imaging (DTI) data acquired at the earliest diagnosed date of pseudo progression or true progression, and 23 had both DTI data and genomic data. Clinical records of all patients were kept in good condition. Volumetric fractional anisotropy (FA) images obtained from the DTI data were decomposed into a sequence of sparse representations. Then, a feature selection algorithm was applied to extract the critical features from the feature matrix to reduce the size of the feature matrix and to improve the classification accuracy. RESULTS The proposed approach was validated using the 79 samples with clinical DTI data. Satisfactory results were obtained under different experimental conditions. The area under the receiver operating characteristic (ROC) curve (AUC) was 0.87 for a given dictionary with 1024 atoms. For the subgroup of 23 samples, genomics data analysis was also performed. Results implied further perspective on pseudo progression classification. CONCLUSIONS The proposed method can determine pseudo progression and true progression with improved accuracy. Laboring segmentation is no longer necessary because this skillfully designed method is not sensitive to tumor location.
Collapse
|
24
|
Suchorska B, Albert NL, Tonn JC. Usefulness of PET Imaging to Guide Treatment Options in Gliomas. Curr Treat Options Neurol 2016; 18:4. [PMID: 26815310 DOI: 10.1007/s11940-015-0384-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OPINION STATEMENT Magnetic resonance imaging (MRI) is the gold standard guiding diagnostic and therapeutic management in glioma with its high resolution and possibility to depict blood-brain-barrier disruption when contrast medium is applied. In light of the shifting paradigms revealing distinct tumor subtypes based on the molecular and genetic characterization and increasing knowledge about the variability of glioma biology, additional imaging modalities such as positron emission tomography (PET) depicting metabolic processes gain further importance in the management of glioma.
Collapse
Affiliation(s)
- Bogdana Suchorska
- Department of Neurosurgery, University Hospital Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | | | - Jörg-Christian Tonn
- Department of Neurosurgery, University Hospital Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
25
|
Glucose-corrected standardized uptake value in the differentiation of high-grade glioma versus post-treatment changes. Nucl Med Commun 2016; 36:573-81. [PMID: 25714806 PMCID: PMC4422715 DOI: 10.1097/mnm.0000000000000288] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background Standardized uptake values (SUVs) of fluorine-18 fluorodeoxyglucose PET (18F-FDG PET) are used widely to differentiate residual or recurrent high-grade gliomas from post-treatment changes in patients with brain tumors. The aim of this study is to assess the accuracy of SUV corrected by blood glucose level (SUVgluc) compared with various quantitative methods in this role. Materials and methods In 55 patients with dynamic 18F-FDG PET scans, there were 97 glioma lesions: glioblastoma (n=60), grade III gliomas (n=22), grade III or IV gliomas (n=6), grade I/II (n=7), and prebiopsy lesions (n=2). The final actual diagnosis was made on the basis of pathology (n=33) and clinical outcome (n=64). Dynamic 18F-FDG PET scans were processed to generate parametric images of SUVgluc, SUVmax, and glucose metabolic rate (GMR). Lesion to cerebellum ratios (SUVRc) and contralateral white matter ratios (SUVRw) were also measured. The SUVgluc was calculated as SUVmax×blood glucose level/100. Results Using the thresholds of SUVmax>4.6, SUVRc>0.9, SUVRw>1.8, SUVgluc>4.3, and GMR>12.2 μmol/min/100 g to represent positivity for viable tumors, the accuracies were the same for the SUVgluc and SUVRw (80%) and were higher than the conventional SUVmax (72%). The area under the receiver operating characteristic curve for the SUVgluc (0.8933) was better than that for the SUVmax (0.8266) (P<0.01) and was similar to those of the GMR (0.8622), SUVRc (0.8606), and SUVRw (0.8981). Conclusion These results suggest that SUVgluc may aid in the differentiation of residual or recurrent high-grade tumor from post-treatment changes in patients with abnormal blood glucose levels. The simplicity of the SUVgluc avoids the complexity of kinetic analysis or the requirement of a reference tissue.
Collapse
|
26
|
Abstract
Glioblastoma is a refractory malignancy with limited treatment options at tumor recurrence. Only a small proportion of patients survive 2 years or longer with the current standard of care. Gene expression profiling can segregate newly diagnosed patients into groups with different prognoses, and these biomarkers are being incorporated into a new generation of personalized clinical trials. Using the experience from recently completed large scale, multi-faceted, randomized glioblastoma clinical trials, a new clinical trial paradigm is being established to move promising therapies forward into the newly diagnosed treatment setting. Upcoming trials using the immune check-point inhibitors are an example of this changing paradigm and these and other immunotherapies have potential as promising new treatment modalities for newly diagnosed GB patients.
Collapse
Affiliation(s)
- Brett J Theeler
- Department of Neurology and John P. Murtha Cancer Center, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Building 19, Bethesda, MD, 20889, USA.
| | - Mark R Gilbert
- National Institutes of Health, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
27
|
Ohue S, Kohno S, Inoue A, Yamashita D, Suehiro S, Seno T, Kumon Y, Kikuchi K, Ohnishi T. Evaluation of serial changes on computed tomography and magnetic resonance imaging after implantation of carmustine wafers in patients with malignant gliomas for differential diagnosis of tumor recurrence. J Neurooncol 2015; 126:119-126. [DOI: 10.1007/s11060-015-1941-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/12/2015] [Indexed: 10/23/2022]
|
28
|
Gzell C, Wheeler H, Huang D, Gaur P, Chen J, Kastelan M, Back M. Proliferation Index Predicts Survival after Second Craniotomy within 6 Months of Adjuvant Radiotherapy for High-grade Glioma. Clin Oncol (R Coll Radiol) 2015; 28:215-22. [PMID: 26382848 DOI: 10.1016/j.clon.2015.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/31/2015] [Accepted: 08/25/2015] [Indexed: 01/21/2023]
Abstract
AIMS To determine pathological features that predict survival in patients having repeat craniotomy within 6 months of radiotherapy for high-grade glioma (HGG). MATERIALS AND METHODS HGG patients (World Health Organization grade 3/4) managed with repeat craniotomy within 6 months of completing radiotherapy between 2008 and 2012 were included. Based on the presence of residual tumour cells, the pathology was reported as pathological progression or pathological pseudoprogression. The proliferation index (Ki67) was reported and compared with initial pathology as a percentage change. Tumour necrosis was estimated as a percentage of the specimen. Overall survival was calculated in months. RESULTS Of 327 patients managed with HGG, 27 patients underwent repeat craniotomy within 6 months of radiotherapy. The median survival after reoperation was 11 months (95% confidence interval 1-22). Ki67 at reoperation of 0%, 1-9% and >10% was associated with survival with a median survival of 13, 13 and 3 months, respectively (P = 0.007). Change in Ki67 was also associated with median survival, with <50% reduction median survival 3 months, 50-80% median survival 7 months and >80% reduction median survival 13 months, P = 0.02. Widespread treatment-related necrosis improved outcome, with >80% necrosis having a median survival of 13 months versus 3 months in those with <80% necrosis (P = 0.003). CONCLUSION The presence of residual tumour at repeat craniotomy within 6 months of radiotherapy is not an independent indicator of prognosis. Patients with residual tumour that had a low Ki67 had a similar median survival as those with only treatment necrosis. Reduced proliferation of residual tumour cells and widespread necrosis may be more important indicators for future outcome.
Collapse
Affiliation(s)
- C Gzell
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, Australia.
| | - H Wheeler
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, Australia
| | - D Huang
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - P Gaur
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - J Chen
- Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - M Kastelan
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - M Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
29
|
Kleinberg LR, Stieber V, Mikkelsen T, Judy K, Weingart J, Barnett G, Olson J, Desideri S, Ye X, Grossman S. Outcome of Adult Brain Tumor Consortium (ABTC) prospective dose-finding trials of I-125 balloon brachytherapy in high-grade gliomas: challenges in clinical trial design and technology development when MRI treatment effect and recurrence appear similar. ACTA ACUST UNITED AC 2015; 4:235-241. [PMID: 27695605 DOI: 10.1007/s13566-015-0210-y] [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] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The aim of this study is to define the maximal safe radiation dose to guide further study of the GliaSite balloon brachytherapy (GSBT) system in untreated newly diagnosed glioblastoma (NEW-GBM) and recurrent high-grade glioma (REC-HGG). GBST is a balloon placed in the resection cavity and later filled through a subcutaneous port with liquid I-125 Iotrex, providing radiation doses that diminish uniformly with distance from the balloon surface. METHODS The Adult Brain Tumor Consortium initiated prospective dose-finding studies to determine maximum tolerated dose in NEW-GBM treated before standard RT or after surgery for REC-HGG. Patients were inevaluable if there was progression before the 90-day posttreatment toxicity evaluation point. RESULTS Ten NEW-GBM patients had the balloon placed, and 2/10 reached the 90 day timepoint. Five REC-HGG enrolled and two were assessable at the 90-day evaluation endpoint. Imaging progression occurred before 90-day evaluation in 7/12 treated patients. The trials were closed as too few patients were assessable to allow dose escalation, although no dose-limiting toxicities (DLTs) were observed. Median survival from treatment was 15.3 months (95 % CI 7.1-23.6) for NEW-GBM and 12.8 months (95 % CI 4.2-20.9) for REC-HGG. CONCLUSION These trials failed to determine a maximum tolerated dose (MTD) for further testing as early imaging changes, presumed to be progression, were common and interfered with the assessment of treatment-related toxicity. The survival outcomes in these and other related studies, although based on small populations, suggest that GSBT may be worthy of further study using clinical and survival endpoints, rather than standard imaging results. The implications for local therapy development are discussed.
Collapse
Affiliation(s)
- L R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Cancer Center, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA
| | - V Stieber
- Piedmont Radiation Oncology, Winston-Salem, NC, USA
| | | | - K Judy
- Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - J Weingart
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Cancer Center, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA
| | - G Barnett
- Cleveland Clinic, Cleveland, OH, USA
| | - J Olson
- Emory University, Atlanta, USA
| | - S Desideri
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Cancer Center, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA
| | - X Ye
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Cancer Center, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA
| | - S Grossman
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Cancer Center, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA
| |
Collapse
|
30
|
Patient reported endpoints for measuring clinical benefit in (high grade glioma) primary brain tumor patients. Curr Treat Options Oncol 2015; 15:519-28. [PMID: 25173554 DOI: 10.1007/s11864-014-0302-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Symptom occurrence impacts primary brain tumor patients from the time of diagnosis and often heralds recurrence. In addition, the therapy can also result in symptoms that may compound tumor-associated symptoms, further impacting the patient's function and overall quality of life. There is increasing recognition that clinical studies evaluating tumor response using only measures of tumor size on imaging or survival are inadequate in brain tumor patients. Many symptoms can only be assessed from the patient, and patient reported outcome measures have been developed and have adequate reliability and validity. These measures are beginning to be incorporated into clinical trials. Guidelines on their use and meaning are needed to standardize assessment across trials and facilitate measurement of clinical benefit.
Collapse
|
31
|
Woernle CM, Péus D, Hofer S, Rushing EJ, Held U, Bozinov O, Krayenbühl N, Weller M, Regli L. Efficacy of Surgery and Further Treatment of Progressive Glioblastoma. World Neurosurg 2015; 84:301-7. [PMID: 25797075 DOI: 10.1016/j.wneu.2015.03.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/03/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Treatment options for patients with glioblastoma at progression have remained controversial, and selection criteria for the appropriate type of intervention remain poorly defined. The objectives were to determine which factors favor the decision for second surgery and which factors are associated with overall survival (OS) and to evaluate the National Institutes of Health (NIH) recurrent glioblastoma scale. The scale includes tumor involvement of eloquent brain regions, functional status, and tumor volume. METHODS A retrospective single-center analysis of patients with newly diagnosed glioblastoma undergoing initial surgery between January 2007 and December 2011 was performed. Patients were separated into two groups: those with versus those without second resection surgery at disease progression. OS was compared using the multiple logistic regression model, Cox proportional hazard regression, and Kaplan-Meier survival analysis. RESULTS The data of 98 patients were statistically analyzed. Among the patients, 58 had initial surgery only (age 61.27 years; median OS [mOS] 14.81 months) and 40 underwent second surgery at disease progression (age 55 years; mOS 18.86 months). Age was the only predictor for repeated surgery (P = 0.012; odds ratio 0.94). At the time of tumor progression, administration of alkylating chemotherapy (P = 0.004; hazard ratio [HR] 0.24) or bevacizumab (P = 0.001; HR 0.23) was associated with longer OS. Reoperation was associated with a lower HR (P = 0.134; HR 0.66). The NIH recurrent glioblastoma scale showed statistically significant improvement of prognosis prediction with the addition of age. CONCLUSIONS Surgery of progressive glioblastoma and postoperative treatment at the time of progression is associated with improved OS in some patients. The addition of age may improve survival prediction of the NIH recurrent glioblastoma scale.
Collapse
Affiliation(s)
| | - Dominik Péus
- Department of Neurosurgery, University Hospital Zurich, Switzerland.
| | - Silvia Hofer
- Department of Oncology, University Hospital Zurich, Switzerland
| | | | - Ulrike Held
- Horten Centre for Patient Oriented Research and Knowledge Transfer, University Hospital Zurich, Switzerland
| | - Oliver Bozinov
- Department of Neurosurgery, University Hospital Zurich, Switzerland
| | | | - Michael Weller
- Department of Neurology, University Hospital Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, Switzerland
| |
Collapse
|
32
|
Zach L, Guez D, Last D, Daniels D, Grober Y, Nissim O, Hoffmann C, Nass D, Talianski A, Spiegelmann R, Tsarfaty G, Salomon S, Hadani M, Kanner A, Blumenthal DT, Bukstein F, Yalon M, Zauberman J, Roth J, Shoshan Y, Fridman E, Wygoda M, Limon D, Tzuk T, Cohen ZR, Mardor Y. Delayed contrast extravasation MRI: a new paradigm in neuro-oncology. Neuro Oncol 2014; 17:457-65. [PMID: 25452395 DOI: 10.1093/neuonc/nou230] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/08/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Conventional magnetic resonance imaging (MRI) is unable to differentiate tumor/nontumor enhancing tissues. We have applied delayed-contrast MRI for calculating high resolution treatment response assessment maps (TRAMs) clearly differentiating tumor/nontumor tissues in brain tumor patients. METHODS One hundred and fifty patients with primary/metastatic tumors were recruited and scanned by delayed-contrast MRI and perfusion MRI. Of those, 47 patients underwent resection during their participation in the study. Region of interest/threshold analysis was performed on the TRAMs and on relative cerebral blood volume maps, and correlation with histology was studied. Relative cerebral blood volume was also assessed by the study neuroradiologist. RESULTS Histological validation confirmed that regions of contrast agent clearance in the TRAMs >1 h post contrast injection represent active tumor, while regions of contrast accumulation represent nontumor tissues with 100% sensitivity and 92% positive predictive value to active tumor. Significant correlation was found between tumor burden in the TRAMs and histology in a subgroup of lesions resected en bloc (r(2) = 0.90, P < .0001). Relative cerebral blood volume yielded sensitivity/positive predictive values of 51%/96% and there was no correlation with tumor burden. The feasibility of applying the TRAMs for differentiating progression from treatment effects, depicting tumor within hemorrhages, and detecting residual tumor postsurgery is demonstrated. CONCLUSIONS The TRAMs present a novel model-independent approach providing efficient separation between tumor/nontumor tissues by adding a short MRI scan >1 h post contrast injection. The methodology uses robust acquisition sequences, providing high resolution and easy to interpret maps with minimal sensitivity to susceptibility artifacts. The presented results provide histological validation of the TRAMs and demonstrate their potential contribution to the management of brain tumor patients.
Collapse
Affiliation(s)
- Leor Zach
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - David Guez
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - David Last
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Dianne Daniels
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Yuval Grober
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Ouzi Nissim
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Chen Hoffmann
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Dvora Nass
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Alisa Talianski
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Roberto Spiegelmann
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Galia Tsarfaty
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Sharona Salomon
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Moshe Hadani
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Andrew Kanner
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Deborah T Blumenthal
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Felix Bukstein
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Michal Yalon
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Jacob Zauberman
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Jonathan Roth
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Yigal Shoshan
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Evgeniya Fridman
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Marc Wygoda
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Dror Limon
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Tzahala Tzuk
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Zvi R Cohen
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| | - Yael Mardor
- Oncology Institute (L.Z., A.T.); Advanced Technology Center (D.G., D.L., D.D., S.S., Y.M.); Neurosurgery Department (Y.G., O.N., R.S., M.H., J.Z., Z.R.C.); Radiology Institute (C.H., G.T.); Pathology Institute (D.N.); Pediatric Hemato-Oncology Department, Sheba Medical Center, Ramat-Gan, Israel (M.Y.); Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel (L.Z., D.D., C.H., R.S., G.T., M.Y., Z.R.C., Y.M.); Neuro-Oncology Service (D.T.B., F.B.); Neurosurgery Department, Tel-Aviv Medical Center, Tel-Aviv, Israel (A.K., J.R.); Neuro-Oncology Service (E.F., M.W.); Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel (Y.S.); Oncology Institute, Davidoff Center, Rabin Medical Center, Petach Tikva, Israel (D.L.); Neuro-Oncology Service, Rambam Medical Center, Haifa, Israel (T.T.)
| |
Collapse
|
33
|
Paclitaxel poliglumex, temozolomide, and radiation for newly diagnosed high-grade glioma: a Brown University Oncology Group Study. Am J Clin Oncol 2014; 37:444-9. [PMID: 23388562 DOI: 10.1097/coc.0b013e31827de92b] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Paclitaxel poliglumex (PPX), a drug conjugate that links paclitaxel to poly-L-glutamic acid, is a potent radiation sensitizer. Prior studies in esophageal cancer have demonstrated that PPX (50 mg/m/wk) can be administered with concurrent radiation with acceptable toxicity. The primary objective of this study was to determine the safety of the combination of PPX with temozolomide and concurrent radiation for high-grade gliomas. METHODS Eligible patients were required to have WHO grade 3 or 4 gliomas. Patients received weekly PPX (50 mg/m/wk) combined with standard daily temozolomide (75 mg/m) for 6 weeks with concomitant radiation (2.0 Gy, 5 d/wk for a total dose of 60 Gy). RESULTS Twenty-five patients were enrolled, 17 with glioblastoma and 8 with grade 3 gliomas. Seven of 25 patients had grade 4 myelosuppression. Hematologic toxicity lasted up to 5 months suggesting a drug interaction between PPX and temozolomide. For patients with glioblastoma, the median progression-free survival was 11.5 months and the median overall survival was 18 months. CONCLUSIONS PPX could not be safely combined with temozolomide due to grade 4 hematologic toxicity. However, the favorable progression-free and overall survival suggest that PPX may enhance radiation for glioblastoma. A randomized study of single agent PPX/radiation versus temozolomide/radiation for glioblastoma without MGMT methylation is underway.
Collapse
|
34
|
Radbruch A, Fladt J, Kickingereder P, Wiestler B, Nowosielski M, Bäumer P, Schlemmer HP, Wick A, Heiland S, Wick W, Bendszus M. Pseudoprogression in patients with glioblastoma: clinical relevance despite low incidence. Neuro Oncol 2014; 17:151-9. [PMID: 25038253 DOI: 10.1093/neuonc/nou129] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND According to the Response Assessment in Neuro-Oncology criteria, new enhancement within the radiation field on contrast enhanced T1-weighted images within 12 weeks after completion of radiotherapy should not qualify for progressive disease, since up to 50% of these cases may be pseudoprogression (PsP). To validate this concept, we assessed incidence and overall survival (OS) of patients with suspected and confirmed PsP dependent on different time intervals and definitions of PsP. METHODS Patients with newly diagnosed glioblastoma and an enhancement increase of at least 25% after completion of standard radiochemotherapy at month 1, 4, 7, or 10 were eligible. Based on the development of the enhancement in follow-up examinations, patients were categorized as either PsP (subgrouped as complete resolution/decrease >50% and decrease <50%/stable) or true progression. RESULTS Out of 548 patients, 79 fulfilled the inclusion criteria. Of these 79 patients, 9 (11.4%) showed PsP (6/45 patients at 1 month, 2/17 at 4 months, 1/9 at 7 months, and 0/8 at 10 months). Complete resolution of the enhancement was found in 1, decrease >50% in 3, decrease <50% in 2, and stable enhancement in 3 patients with PsP. Patients with PsP showed a significantly longer OS (P < .012). No difference in OS was found among PsP subgroups. CONCLUSIONS This series challenges the current concept of PsP. Even though we could confirm a prolonged OS of patients with PsP, the incidence of PsP was lower than reported previously and extended beyond 12 weeks.
Collapse
Affiliation(s)
- Alexander Radbruch
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Joachim Fladt
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Philipp Kickingereder
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Benedikt Wiestler
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Martha Nowosielski
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Philipp Bäumer
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Heinz-Peter Schlemmer
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Antje Wick
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Sabine Heiland
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Wolfgang Wick
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| | - Martin Bendszus
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany (A.R., J.F., P.K., P.B., S.H., M.B.); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (A.R., H.-P.S.); Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany (B.W., A.W., W.W.); German Cancer Consortium (DKTK) Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany (B.W., A.W., W.W.); Department of Neurology, Innsbruck Medical University, Innsbruck, Austria (M.N.)
| |
Collapse
|
35
|
3-Dimensional magnetic resonance spectroscopic imaging at 3 Tesla for early response assessment of glioblastoma patients during external beam radiation therapy. Int J Radiat Oncol Biol Phys 2014; 90:181-9. [PMID: 24986746 DOI: 10.1016/j.ijrobp.2014.05.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/09/2014] [Accepted: 05/13/2014] [Indexed: 11/20/2022]
Abstract
PURPOSE To evaluate the utility of 3-dimensional magnetic resonance (3D-MR) proton spectroscopic imaging for treatment planning and its implications for early response assessment in glioblastoma multiforme. METHODS AND MATERIALS Eighteen patients with newly diagnosed, histologically confirmed glioblastoma had 3D-MR proton spectroscopic imaging (MRSI) along with T2 and T1 gadolinium-enhanced MR images at simulation and at boost treatment planning after 17 to 20 fractions of radiation therapy. All patients received standard radiation therapy (RT) with concurrent temozolomide followed by adjuvant temozolomide. Imaging for response assessment consisted of MR scans every 2 months. Progression-free survival was defined by the criteria of MacDonald et al. MRSI images obtained at initial simulation were analyzed for choline/N-acetylaspartate ratios (Cho/NAA) on a voxel-by-voxel basis with abnormal activity defined as Cho/NAA ≥2. These images were compared on anatomically matched MRSI data collected after 3 weeks of RT. Changes in Cho/NAA between pretherapy and third-week RT scans were tested using Wilcoxon matched-pairs signed rank tests and correlated with progression-free survival, radiation dose and location of recurrence using Cox proportional hazards regression. RESULTS After a median follow-up time of 8.6 months, 50% of patients had experienced progression based on imaging. Patients with a decreased or stable mean or median Cho/NAA values had less risk of progression (P<.01). Patients with an increase in mean or median Cho/NAA values at the third-week RT scan had a significantly greater chance of early progression (P<.01). An increased Cho/NAA at the third-week MRSI scan carried a hazard ratio of 2.72 (95% confidence interval, 1.10-6.71; P=.03). Most patients received the prescription dose of RT to the Cho/NAA ≥2 volume, where recurrence most often occurred. CONCLUSION Change in mean and median Cho/NAA detected at 3 weeks was a significant predictor of early progression. The potential impact for risk-adaptive therapy based on early spectroscopic findings is suggested.
Collapse
|
36
|
Abstract
Radiotherapy is a primary mode of treatment of many of the disease entities seen by the neurologist. Therefore knowledge of how ionizing radiation works and when it is indicated is a crucial part of the field of Neurology. The neurologist may also be confronted with some of the side effects and complications or radiotherapy treatment. This chapter attempts to serve as a review of the current day process of radiotherapy, a brief review of biology and physics of radiation, and how it is used in the treatment diseases which are common to the Neurologist. In addition we review the more commonly seen side effects and complications of treatment which may be seen by the neurologist.
Collapse
|
37
|
Nabors LB, Ammirati M, Bierman PJ, Brem H, Butowski N, Chamberlain MC, DeAngelis LM, Fenstermaker RA, Friedman A, Gilbert MR, Hesser D, Holdhoff M, Junck L, Lawson R, Loeffler JS, Maor MH, Moots PL, Morrison T, Mrugala MM, Newton HB, Portnow J, Raizer JJ, Recht L, Shrieve DC, Sills AK, Tran D, Tran N, Vrionis FD, Wen PY, McMillian N, Ho M. Central nervous system cancers. J Natl Compr Canc Netw 2014; 11:1114-51. [PMID: 24029126 DOI: 10.6004/jnccn.2013.0132] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Primary and metastatic tumors of the central nervous system are a heterogeneous group of neoplasms with varied outcomes and management strategies. Recently, improved survival observed in 2 randomized clinical trials established combined chemotherapy and radiation as the new standard for treating patients with pure or mixed anaplastic oligodendroglioma harboring the 1p/19q codeletion. For metastatic disease, increasing evidence supports the efficacy of stereotactic radiosurgery in treating patients with multiple metastatic lesions but low overall tumor volume. These guidelines provide recommendations on the diagnosis and management of this group of diseases based on clinical evidence and panel consensus. This version includes expert advice on the management of low-grade infiltrative astrocytomas, oligodendrogliomas, anaplastic gliomas, glioblastomas, medulloblastomas, supratentorial primitive neuroectodermal tumors, and brain metastases. The full online version, available at NCCN. org, contains recommendations on additional subtypes.
Collapse
|
38
|
In vivo chemical exchange saturation transfer imaging allows early detection of a therapeutic response in glioblastoma. Proc Natl Acad Sci U S A 2014; 111:4542-7. [PMID: 24616497 DOI: 10.1073/pnas.1323855111] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM), which account for more than 50% of all gliomas, is among the deadliest of all human cancers. Given the dismal prognosis of GBM, it would be advantageous to identify early biomarkers of a response to therapy to avoid continuing ineffective treatments and to initiate other therapeutic strategies. The present in vivo longitudinal study in an orthotopic mouse model demonstrates quantitative assessment of early treatment response during short-term chemotherapy with temozolomide (TMZ) by amide proton transfer (APT) imaging. In a GBM line, only one course of TMZ (3 d exposure and 4 d rest) at a dose of 80 mg/kg resulted in substantial reduction in APT signal compared with untreated control animals, in which the APT signal continued to increase. Although there were no detectable differences in tumor volume, cell density, or apoptosis rate between groups, levels of Ki67 (index of cell proliferation) were substantially reduced in treated tumors. In another TMZ-resistant GBM line, the APT signal and levels of Ki67 increased despite the same course of TMZ treatment. As metabolite changes are known to occur early in the time course of chemotherapy and precede morphologic changes, these results suggest that the APT signal in glioma may be a useful functional biomarker of treatment response or degree of tumor progression. Thus, APT imaging may serve as a sensitive biomarker of early treatment response and could potentially replace invasive biopsies to provide a definitive diagnosis. This would have a major impact on the clinical management of patients with glioma.
Collapse
|
39
|
Martínez-Martínez A, Martínez-Bosch J. Perfusion magnetic resonance imaging for high grade astrocytomas: Can cerebral blood volume, peak height, and percentage of signal intensity recovery distinguish between progression and pseudoprogression? RADIOLOGIA 2014. [DOI: 10.1016/j.rxeng.2014.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
40
|
Pinho MC, Polaskova P, Kalpathy-Cramer J, Jennings D, Emblem KE, Jain RK, Rosen BR, Wen PY, Sorensen AG, Batchelor TT, Gerstner ER. Low incidence of pseudoprogression by imaging in newly diagnosed glioblastoma patients treated with cediranib in combination with chemoradiation. Oncologist 2013; 19:75-81. [PMID: 24309981 DOI: 10.1634/theoncologist.2013-0101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chemoradiation (CRT) can significantly modify the radiographic appearance of malignant gliomas, especially within the immediate post-CRT period. Pseudoprogression (PsP) is an increasingly recognized phenomenon in this setting, and is thought to be secondary to increased permeability as a byproduct of the complex process of radiation-induced tissue injury, possibly enhanced by temozolomide. We sought to determine whether the addition of a vascular endothelial growth factor (VEGF) signaling inhibitor (cediranib) to conventional CRT had an impact on the frequency of PsP, by comparing two groups of patients with newly diagnosed glioblastoma before, during, and after CRT. METHODS All patients underwent serial magnetic resonance imaging as part of institutional review board-approved clinical studies. Eleven patients in the control group received only chemoradiation, whereas 29 patients in the study group received chemoradiation and cediranib until disease progression or toxicity. Response assessment was defined according to Response Assessment in Neuro-Oncology criteria, and patients with enlarging lesions were classified into true tumor progressions (TTP) or PsP, based on serial radiographic follow-up. RESULTS Two patients in the study group (7%) showed signs of apparent early tumor progression, and both were subsequently classified as TTP. Six patients in the control group (54%) showed signs of apparent early tumor progression, and three were subsequently classified as TTP and three as PsP. The frequency of PsP was significantly higher in the control group. CONCLUSION Administration of a VEGF inhibitor during and after CRT modifies the expression of PsP by imaging.
Collapse
Affiliation(s)
- Marco C Pinho
- Departments of Radiology, Neurology, and Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA; Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA; The Intervention Centre, Oslo University Hospital, Oslo, Norway; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ricard C, Stanchi F, Rodriguez T, Amoureux MC, Rougon G, Debarbieux F. Dynamic quantitative intravital imaging of glioblastoma progression reveals a lack of correlation between tumor growth and blood vessel density. PLoS One 2013; 8:e72655. [PMID: 24069154 PMCID: PMC3771993 DOI: 10.1371/journal.pone.0072655] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/12/2013] [Indexed: 11/19/2022] Open
Abstract
The spatiotemporal and longitudinal monitoring of cellular processes occurring in tumors is critical for oncological research. We focused on glioblastoma multiforme (GBM), an untreatable highly vascularized brain tumor whose progression is thought to critically depend on the oxygen and metabolites supplied by blood vessels. We optimized protocols for orthotopic GBM grafting in mice that were able to recapitulate the biophysical constraints normally governing tumor progression and were suitable for intravital multiphoton microscopy. We repeatedly imaged tumor cells and blood vessels during GBM development. We established methods for quantitative correlative analyses of dynamic imaging data over wide fields in order to cover the entire tumor. We searched whether correlations existed between blood vessel density, tumor cell density and proliferation in control tumors. Extensive vascular remodeling and the formation of new vessels accompanied U87 tumor cell growth, but no strong correlation was found between local cell density and the extent of local blood vessel density irrespective of the tumor area or time points. The technique moreover proves useful for comparative analysis of mice subjected either to Bevacizumab anti-angiogenic treatment that targets VEGF or to AMD3100, an antagonist of CXCR4 receptor. Bevacizumab treatment massively reduced tumoral vessel densities but only transiently reduced U87 tumor growth rate. Again, there was no correlation between local blood vessel density and local cell density. Moreover, Bev applied only prior to tumor implantation inhibited tumor growth to the same extent as post-grafting treatment. AMD3100 achieved a potent inhibition of tumor growth without significant reduction in blood vessel density. These results indicate that in the brain, in this model, tumor growth can be sustained without an increase in blood vessel density and suggest that GBM growth is rather governed by stromal properties.
Collapse
Affiliation(s)
- Clément Ricard
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
- European Center for Medical Imaging (CERIMED), Marseille, France
| | - Fabio Stanchi
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
- VIB3 Vesalius Onderzoekscentrum, KU, Leuven, Belgium
| | - Thieric Rodriguez
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
| | - Marie-Claude Amoureux
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
- European Center for Medical Imaging (CERIMED), Marseille, France
| | - Geneviève Rougon
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
- European Center for Medical Imaging (CERIMED), Marseille, France
- * E-mail: (GR); (FD)
| | - Franck Debarbieux
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University-CNRS 7288, Marseille, France
- European Center for Medical Imaging (CERIMED), Marseille, France
- * E-mail: (GR); (FD)
| |
Collapse
|
42
|
Bevacizumab for radiation necrosis following treatment of high grade glioma: a systematic review of the literature. J Neurooncol 2013; 115:317-22. [PMID: 24005770 DOI: 10.1007/s11060-013-1233-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
Abstract
This review identifies the current literature on the use of bevacizumab for cerebral radiation necrosis in patients with high-grade gliomas, summarizes the clinical course and complications following bevacizumab, and discusses the relative costs and benefits of this therapeutic option. A Medline search was conducted of all clinical studies before September 2012 investigating outcomes following use of bevacizumab therapy for radiation necrosis in patients with high-grade gliomas. Clinical and radiographic outcomes are reviewed. Seven studies reported a total of 30 patients with high-grade gliomas treated with bevacizumab for radiation necrosis. All patients demonstrated decreased radiographic volume of edema on T1 and T2 MRI sequences. Clinical outcomes were reported for 23 patients: 16 (70 %) had improvement in neurologic signs or symptoms, 5 (22 %) had mixed results, and 2 (9 %) remained neurologically unchanged. Complications were documented in 5 of 7 studies (18 of 29 patients, 62 %) and included deep vein thrombosis, pulmonary embolism, visual field worsening, worsening hemiplegia, pneumonia, seizure, and fatigue. Only one study evaluated quality of life measures and none evaluated cost or cost effectiveness. Data regarding the use of bevacizumab to treat radiation necrosis in patients with high-grade gliomas is limited and primarily class III evidence. While bevacizumab improves neurological symptoms and reduces radiographic volume of necrosis-associated cerebral edema, it comes at the expense of a high rate of potentially serious complications. Definitive evidence for the utility, cost-effectiveness, and overall efficacy of this management strategy is currently lacking and additional investigation is warranted.
Collapse
|
43
|
Martínez-Martínez A, Martínez-Bosch J. [Perfusion magnetic resonance imaging for high grade astrocytomas: Can cerebral blood volume, peak height, and percentage of signal intensity recovery distinguish between progression and pseudoprogression?]. RADIOLOGIA 2013; 56:35-43. [PMID: 23790618 DOI: 10.1016/j.rx.2013.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 01/22/2023]
Abstract
OBJECTIVES To study the usefulness of common MRI perfusion parameters for identifying pseudoprogression in high grade astrocytomas. MATERIAL AND METHODS This retrospective case-control study compared the relative cerebral blood volume (rCBV), the relative percentage of signal intensity recovery (rPSR), and the relative peak height (rPH) recorded in a sample of 17 cases of anaplastic astrocytomas and gliomas considered to be undergoing pseudoprogression by biopsy or follow-up with those recorded in a sample of histologically similar tumors that were treated and considered to be undergoing progression by histologic study or follow-up. We evaluated the accuracy of these parameters and the correlations among them. Statistical significance was set at P<.05. RESULTS The rCBV, rPSR, and rPH were significantly different between the two groups (P=.001). The cutoff values rPH=1.37, rCBV=0.9, and rPSR=99% yielded sensitivity (S)=88% and specificity (Sp)=82.2% for rPH, S=100% and Sp=100% for rCBV, and S=100% and Sp=70.6% for rPSR, respectively. We found negative correlations between rPRS and rPH (-0.76) and between rPRS and rCBV (-0.81) and a high positive correlation between rPH and rCBV (0.87). CONCLUSION The variables rPH and rCBV were useful for differentiating between pseudoprogression and true progression in our sample. The variable rPRS was also very sensitive, although the overlap in the values between samples make it less useful a priori.
Collapse
Affiliation(s)
- A Martínez-Martínez
- Sección de Neurorradiología, Unidad de Gestión Clínica de Radiodiagnóstico, Hospital Universitario Virgen de las Nieves, Granada, España.
| | - J Martínez-Bosch
- Sección de Neurorradiología, Unidad de Gestión Clínica de Radiodiagnóstico, Hospital Universitario Virgen de las Nieves, Granada, España
| |
Collapse
|
44
|
Boxerman JL, Zhang Z, Safriel Y, Larvie M, Snyder BS, Jain R, Chi TL, Sorensen AG, Gilbert MR, Barboriak DP. Early post-bevacizumab progression on contrast-enhanced MRI as a prognostic marker for overall survival in recurrent glioblastoma: results from the ACRIN 6677/RTOG 0625 Central Reader Study. Neuro Oncol 2013; 15:945-54. [PMID: 23788270 DOI: 10.1093/neuonc/not049] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND RTOG 0625/ACRIN 6677 is a multicenter, randomized, phase II trial of bevacizumab with irinotecan or temozolomide in recurrent glioblastoma (GBM). This study investigated whether early posttreatment progression on FLAIR or postcontrast MRI assessed by central reading predicts overall survival (OS). METHODS Of 123 enrolled patients, 107 had baseline and at least 1 posttreatment MRI. Two central neuroradiologists serially measured bidimensional (2D) and volumetric (3D) enhancement on postcontrast T1-weighted images and volume of FLAIR hyperintensity. Progression status on all posttreatment MRIs was determined using Macdonald and RANO imaging threshold criteria, with a third neuroradiologist adjudicating discrepancies of both progression occurrence and timing. For each MRI pulse sequence, Kaplan-Meier survival estimates and log-rank test were used to compare OS between cases with or without radiologic progression. RESULTS Radiologic progression occurred after 2 chemotherapy cycles (8 weeks) in 9 of 97 (9%), 9 of 73 (12%), and 11 of 98 (11%) 2D-T1, 3D-T1, and FLAIR cases, respectively, and 34 of 80 (43%), 21 of 58 (36%), and 37 of 79 (47%) corresponding cases after 4 cycles (16 weeks). Median OS among patients progressing at 8 or 16 weeks was significantly less than that among nonprogressors, as determined on 2D-T1 (114 vs 278 days and 214 vs 426 days, respectively; P < .0001 for both) and 3D-T1 (117 vs 306 days [P < .0001] and 223 vs 448 days [P = .0003], respectively) but not on FLAIR (201 vs 276 days [P = .38] and 303 vs 321 days [P = .13], respectively). CONCLUSION Early progression on 2D-T1 and 3D-T1, but not FLAIR MRI, after 8 and 16 weeks of anti-vascular endothelial growth factor therapy has highly significant prognostic value for OS in recurrent GBM.
Collapse
Affiliation(s)
- Jerrold L Boxerman
- Rhode Island Hospital, Department of Diagnostic Imaging, 593 Eddy St., Providence, RI 02903, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nozawa A, Rivandi AH, Kesari S, Hoh CK. Glucose corrected standardized uptake value (SUVgluc) in the evaluation of brain lesions with 18F-FDG PET. Eur J Nucl Med Mol Imaging 2013; 40:997-1004. [DOI: 10.1007/s00259-013-2396-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 03/11/2013] [Indexed: 11/29/2022]
|
46
|
|
47
|
Shiroishi MS, Booker MT, Agarwal M, Jain N, Naghi I, Lerner A, Law M. Posttreatment evaluation of central nervous system gliomas. Magn Reson Imaging Clin N Am 2013; 21:241-68. [PMID: 23642552 DOI: 10.1016/j.mric.2013.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although conventional contrast-enhanced MR imaging remains the standard-of-care imaging method in the posttreatment evaluation of gliomas, recent developments in therapeutic options such as chemoradiation and antiangiogenic agents have caused the neuro-oncology community to rethink traditional imaging criteria. This article highlights the latest recommendations. These recommendations should be viewed as works in progress. As more is learned about the pathophysiology of glioma treatment response, quantitative imaging biomarkers will be validated within this context. There will likely be further refinements to glioma response criteria, although the lack of technical standardization in image acquisition, postprocessing, and interpretation also need to be addressed.
Collapse
Affiliation(s)
- Mark S Shiroishi
- Division of Neuroradiology, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
van Akkooi A, Nijsten T. A costly revolution for a subgroup of patients with metastatic melanoma. Br J Dermatol 2013; 168:467-70; discussion 470-1. [DOI: 10.1111/bjd.12238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
49
|
Neal ML, Trister AD, Ahn S, Baldock A, Bridge CA, Guyman L, Lange J, Sodt R, Cloke T, Lai A, Cloughesy TF, Mrugala MM, Rockhill JK, Rockne RC, Swanson KR. Response classification based on a minimal model of glioblastoma growth is prognostic for clinical outcomes and distinguishes progression from pseudoprogression. Cancer Res 2013; 73:2976-86. [PMID: 23400596 DOI: 10.1158/0008-5472.can-12-3588] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Glioblastoma multiforme is the most aggressive type of primary brain tumor. Glioblastoma growth dynamics vary widely across patients, making it difficult to accurately gauge their response to treatment. We developed a model-based metric of therapy response called Days Gained that accounts for this heterogeneity. Here, we show in 63 newly diagnosed patients with glioblastoma that Days Gained scores from a simple glioblastoma growth model computed at the time of the first postradiotherapy MRI scan are prognostic for time to tumor recurrence and overall patient survival. After radiation treatment, Days Gained also distinguished patients with pseudoprogression from those with true progression. Because Days Gained scores can be easily computed with routinely available clinical imaging devices, this model offers immediate potential to be used in ongoing prospective studies.
Collapse
Affiliation(s)
- Maxwell Lewis Neal
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
de Groot J, Reardon DA, Batchelor TT. Antiangiogenic therapy for glioblastoma: the challenge of translating response rate into efficacy. Am Soc Clin Oncol Educ Book 2013:00113000e71. [PMID: 23714460 DOI: 10.14694/edbook_am.2013.33.e71] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Glioblastoma are one of the mostly vascularized tumors and are histologically characterized by abundant endothelial cell proliferation. Vascular endothelial growth factor (VEGF) is responsible for a degree of vascular proliferation and vessel permeability leading to symptomatic cerebral edema. Initial excitement generated from the impressive radiographic response rates has waned due to concerns of limited long-term efficacy and the promotion of a treatment-resistant phenotype. Reasons for the discrepancy between high radiographic response rates and lack of survival benefit have led to a focus on identifying potential mechanisms of resistance to antiangiogenic therapy. However, equally important is the need to focus on identification of basic mechanisms of action of this class of drugs, determining the optimal biologic dose for each agent and identify the effect of antiangiogenic therapy on oxygen and drug delivery to tumor to optimize drug combinations. Finally, alternatives to overall survival (OS) need to be pursued using the application of validated parameters to reliably assess neurologic function and quality of life.
Collapse
Affiliation(s)
- John de Groot
- From the Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA; Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital Cancer Center, Boston, MA
| | | | | |
Collapse
|