1
|
Gong Z, Zhou D, Wu D, Han Y, Yu H, Shen H, Feng W, Hou L, Chen Y, Xu T. Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 2025; 319:123180. [PMID: 39985979 DOI: 10.1016/j.biomaterials.2025.123180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Central nervous system (CNS) tumors, encompassing a diverse array of neoplasms in the brain and spinal cord, pose significant therapeutic challenges due to their intricate anatomy and the protective presence of the blood-brain barrier (BBB). The primary treatment obstacle is the effective delivery of therapeutics to the tumor site, which is hindered by multiple physiological, biological, and technical barriers, including the BBB. This comprehensive review highlights recent advancements in material science and nanotechnology aimed at surmounting these delivery challenges, with a focus on the development and application of nanomaterials. Nanomaterials emerge as potent tools in designing innovative drug delivery systems that demonstrate the potential to overcome the limitations posed by CNS tumors. The review delves into various strategies, including the use of lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, all of which are engineered to enhance drug stability, BBB penetration, and targeted tumor delivery. Additionally, this review highlights the burgeoning role of theranostic nanoparticles, integrating therapeutic and diagnostic functionalities to optimize treatment efficacy. The exploration extends to biocompatible materials like biodegradable polymers, liposomes, and advanced material-integrated delivery systems such as implantable drug-eluting devices and microfabricated devices. Despite promising preclinical results, the translation of these material-based strategies into clinical practice necessitates further research and optimization.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China; Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, 230601, PR China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, PR China
| | - Haotian Shen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lijun Hou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
2
|
Giuffrida AS, Sheriff S, Huang V, Weinberg BD, Cooper LAD, Liu Y, Soher BJ, Treadway M, Maudsley AA, Shim H. NNFit: A Self-Supervised Deep Learning Method for Accelerated Quantification of High-Resolution Short-Echo-Time MR Spectroscopy Datasets. Radiol Artif Intell 2025; 7:e230579. [PMID: 39812584 PMCID: PMC11950874 DOI: 10.1148/ryai.230579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Purpose To develop and evaluate the performance of NNFit, a self-supervised deep learning method for quantification of high-resolution short-echo-time (TE) echo-planar spectroscopic imaging (EPSI) datasets, with the goal of addressing the computational bottleneck of conventional spectral quantification methods in the clinical workflow. Materials and Methods This retrospective study included 89 short-TE whole-brain EPSI/generalized autocalibrating partial parallel acquisition scans from clinical trials for glioblastoma (trial 1, May 2014-October 2018) and major depressive disorder (trial 2, 2022-2023). The training dataset included 685 000 spectra from 20 participants (60 scans) in trial 1. The testing dataset included 115 000 spectra from five participants (13 scans) in trial 1 and 145 000 spectra from seven participants (16 scans) in trial 2. A comparative analysis was performed between NNFit and a widely used parametric-modeling spectral quantitation method (FITT). Metabolite maps generated by each method were compared using the structural similarity index measure (SSIM) and linear correlation coefficient (R2). Radiation treatment volumes for glioblastoma based on metabolite maps were compared using the Dice coefficient and a two-tailed t test. Results Mean SSIMs and R2 values for trial 1 test set data were 0.91 and 0.90 for choline, 0.93 and 0.93 for creatine, 0.93 and 0.93 for N-acetylaspartate, 0.80 and 0.72 for myo-inositol, and 0.59 and 0.47 for glutamate plus glutamine. Mean values for trial 2 test set data were 0.95 and 0.95, 0.98 and 0.97, 0.98 and 0.98, 0.92 and 0.92, and 0.79 and 0.81, respectively. The treatment volumes had a mean Dice coefficient of 0.92. The mean processing times were 90.1 seconds for NNFit and 52.9 minutes for FITT. Conclusion A deep learning approach to spectral quantitation offers performance similar to that of conventional quantification methods for EPSI data, but with faster processing at short TE. Keywords: MR Spectroscopy, Neural Networks, Brain/Brain Stem Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Alexander S. Giuffrida
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Sulaiman Sheriff
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Vicki Huang
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Brent D. Weinberg
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Lee A. D. Cooper
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Yuan Liu
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Brian J. Soher
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Michael Treadway
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Andrew A. Maudsley
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| | - Hyunsuk Shim
- From the Department of Radiation Oncology (A.S.G., V.H., H.S.) and
Department of Radiology and Imaging Sciences (B.D.W.), Emory University School
of Medicine, 1701 Uppergate Dr, C5008 Winship Cancer Institute, Atlanta, GA
30322; Department of Radiology, University of Miami School of Medicine, Miami,
Fla (S.S., A.A.M.); Department of Pathology, Northwestern University Feinberg
School of Medicine, Chicago, Ill (L.A.D.C.); Department of Biostatistics and
Bioinformatics, Emory University Rollins School of Public Health, Atlanta, Ga
(Y.L.); Department of Psychology, Emory University, Atlanta, Ga (M.T.); and
Department of Radiology, Duke University Medical Center, Durham, NC
(B.J.S.)
| |
Collapse
|
3
|
Giuffrida AS, Ramesh K, Sheriff S, Maudsley AA, Weinberg BD, Cooper LAD, Shim H. An Accelerated Spectroscopic MRI Metabolite Quantification Based on a Deep Learning Method for Radiation Therapy Planning in Brain Tumor Patients. Cancers (Basel) 2025; 17:423. [PMID: 39941791 PMCID: PMC11816355 DOI: 10.3390/cancers17030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Spectroscopic MRI (sMRI) is a quantitative imaging technique that maps infiltrated tumors in the brain without contrast injections. In a previous study (NCT03137888), sMRI-guided radiation treatment extended patient survival, showing promise for clinical translation. The spectral fitting of individual voxels in an sMRI dataset generate metabolite concentration maps that guide treatment. The established spectral analysis methods use iterative least-squares fitting (FITT) that are computationally demanding. This study compares the performance of NNFit, a neural network-based, accelerated spectral fitting model, to the established FITT for metabolite quantification and radiation treatment planning. METHODS NNFit is a self-supervised deep learning model trained on 50 ms echo-time (TE) sMRI data to estimate metabolite levels of choline (Cho), creatine (Cr), and NAA. We trained the model on 30 GBM patients (56 scans) and tested it on 17 GBM patients (29 scans). NNFit's performance was compared to the FITT using structural similarity indices (SSIM) and the Dice coefficient. RESULTS NNFit significantly improved processing speed while maintaining strong agreement with FITT. The radiation target volumes defined by Cho/NAA ≥ 2x were visually comparable, with fewer artifacts in NNFit. Structural similarity indices (SSIM) indicated minimal bias and high consistency across methods. CONCLUSIONS This study highlights NNFit's potential for rapid, accurate, and artifact-reduced metabolic imaging, enabling faster radiotherapy planning.
Collapse
Affiliation(s)
- Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
| | - Sulaiman Sheriff
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 45056, USA; (S.S.); (A.A.M.)
| | - Andrew A. Maudsley
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 45056, USA; (S.S.); (A.A.M.)
| | - Brent D. Weinberg
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lee A. D. Cooper
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60657, USA;
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.S.G.); (K.R.)
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
4
|
Vinogradskiy Y, Bahig H, Bucknell NW, Buchsbaum J, Shu HKG. Conference Report: Review of Clinical Implementation of Advanced Quantitative Imaging Techniques for Personalized Radiotherapy. Tomography 2024; 10:1798-1813. [PMID: 39590941 PMCID: PMC11598114 DOI: 10.3390/tomography10110132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The topic of quantitative imaging in radiation therapy was presented as a "Masterclass" at the 2023 annual meeting of the American Society of Radiation Oncology (ASTRO). Dual-energy computed tomography (CT) and single-positron computed tomography were reviewed in detail as the first portion of the meeting session, with data showing utility in many aspects of radiation oncology including treatment planning and dose response. Positron emission tomography/CT scans evaluating the functional volume of lung tissue so as to provide optimal avoidance of healthy lungs were presented second. Advanced brain imaging was then discussed in the context of different forms of magnetic resonance scanning methods as the third area noted with significant discussion of ongoing research programs. Quantitative image analysis was presented to provide clinical utility for the analysis of patients with head and neck cancer. Finally, quality assurance was reviewed for different forms of quantitative imaging given the critical nature of imaging when numerical valuation, not just relative contrast, plays a crucial role in clinical process and decision-making. Conclusions and thoughts are shared in the conclusion, noting strong data supporting the use of quantitative imaging in radiation therapy going forward and that more studies are needed to move the field forward.
Collapse
Affiliation(s)
- Yevgeniy Vinogradskiy
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Houda Bahig
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Centre Hospitalier de l’Universite de Montreal (CHUM), Montreal, QC H2X 3E4, Canada
| | | | | | - Hui-Kuo George Shu
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 19104, USA
| |
Collapse
|
5
|
Noble DJ, Ramaesh R, Brothwell M, Elumalai T, Barrett T, Stillie A, Paterson C, Ajithkumar T. The Evolving Role of Novel Imaging Techniques for Radiotherapy Planning. Clin Oncol (R Coll Radiol) 2024; 36:514-526. [PMID: 38937188 DOI: 10.1016/j.clon.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/29/2024]
Abstract
The ability to visualise cancer with imaging has been crucial to the evolution of modern radiotherapy (RT) planning and delivery. And as evolving RT technologies deliver increasingly precise treatment, the importance of accurate identification and delineation of disease assumes ever greater significance. However, innovation in imaging technology has matched that seen with RT delivery platforms, and novel imaging techniques are a focus of much research activity. How these imaging modalities may alter and improve the diagnosis and staging of cancer is an important question, but already well served by the literature. What is less clear is how novel imaging techniques may influence and improve practical and technical aspects of RT planning and delivery. In this review, current gold standard approaches to integration of imaging, and potential future applications of bleeding-edge imaging technology into RT planning pathways are explored.
Collapse
Affiliation(s)
- D J Noble
- Department of Clinical Oncology, Edinburgh Cancer Centre, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK.
| | - R Ramaesh
- Department of Radiology, Western General Hospital, Edinburgh, UK
| | - M Brothwell
- Department of Clinical Oncology, University College London Hospitals, London, UK
| | - T Elumalai
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - T Barrett
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - A Stillie
- Department of Clinical Oncology, Edinburgh Cancer Centre, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - C Paterson
- Beatson West of Scotland Cancer Centre, Great Western Road, Glasgow G12 0YN, UK
| | - T Ajithkumar
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
6
|
Peciu-Florianu I, Vannod-Michel Q, Vauleon E, Bonneterre ME, Reyns N. Long term follow-up of patients with newly diagnosed glioblastoma treated by intraoperative photodynamic therapy: an update from the INDYGO trial (NCT03048240). J Neurooncol 2024; 168:495-505. [PMID: 38753093 PMCID: PMC11186870 DOI: 10.1007/s11060-024-04693-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE Glioblastoma remains incurable despite optimal multimodal management. The interim analysis of open label, single arm INDYGO pilot trial showed actuarial 12-months progression-free survival (PFS) of 60% (median 17.1 months), actuarial 12-months overall survival (OS) of 80% (median 23.1 months). We report updated, exploratory analyses of OS, PFS, and health-related quality of life (HRQOL) for patients receiving intraoperative photodynamic therapy (PDT) with 5-aminolevulinic acid hydrochloride (5-ALA HCl). METHODS Ten patients were included (May 2017 - April 2021) for standardized therapeutic approach including 5-ALA HCl fluorescence-guided surgery (FGS), followed by intraoperative PDT with a single 200 J/cm2 dose of light. Postoperatively, patients received adjuvant therapy (Stupp protocol) then followed every 3 months (clinical and cerebral MRI) and until disease progression and/or death. Procedure safety and toxicity occurring during the first four weeks after PDT were assessed. Data concerning relapse, HRQOL and survival were prospectively collected and analyzed. RESULTS At the cut-off date (i.e., November 1st 2023), median follow-up was 23 months (9,7-71,4). No unacceptable or unexpected toxicities and no treatment-related deaths occurred during the study. Kaplan-Meier estimated 23.4 months median OS, actuarial 12-month PFS rate 60%, actuarial 12-month, 24-month, and 5-year OS rates 80%, 50% and 40%, respectively. Four patients were still alive (1 patient free of recurrence). CONCLUSION At 5 years-follow-up, intraoperative PDT with surgical maximal excision as initial therapy and standard adjuvant treatment suggests an increase of time to recurrence and overall survival in a high proportion of patients. Quality of life was maintained without any severe side effects. TRIAL REGISTRATION NCT NUMBER NCT03048240. EudraCT number: 2016-002706-39.
Collapse
Affiliation(s)
| | | | - Enora Vauleon
- Neuro-Oncology Department, CHU-Lille, F-59000, Lille, France
| | | | - Nicolas Reyns
- Neurosurgery Department, CHU-Lille, F-59000, Lille, France.
- U1189-ONCO-THAI-Assisted Laser Therapy and Immunotherapy for Oncology, University of Lille, INSERM, CHU-Lille, F-59000, Lille, France.
| |
Collapse
|
7
|
Martinez Luque E, Liu Z, Sung D, Goldberg RM, Agarwal R, Bhattacharya A, Ahmed NS, Allen JW, Fleischer CC. An Update on MR Spectroscopy in Cancer Management: Advances in Instrumentation, Acquisition, and Analysis. Radiol Imaging Cancer 2024; 6:e230101. [PMID: 38578207 PMCID: PMC11148681 DOI: 10.1148/rycan.230101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 04/06/2024]
Abstract
MR spectroscopy (MRS) is a noninvasive imaging method enabling chemical and molecular profiling of tissues in a localized, multiplexed, and nonionizing manner. As metabolic reprogramming is a hallmark of cancer, MRS provides valuable metabolic and molecular information for cancer diagnosis, prognosis, treatment monitoring, and patient management. This review provides an update on the use of MRS for clinical cancer management. The first section includes an overview of the principles of MRS, current methods, and conventional metabolites of interest. The remainder of the review is focused on three key areas: advances in instrumentation, specifically ultrahigh-field-strength MRI scanners and hybrid systems; emerging methods for acquisition, including deuterium imaging, hyperpolarized carbon 13 MRI and MRS, chemical exchange saturation transfer, diffusion-weighted MRS, MR fingerprinting, and fast acquisition; and analysis aided by artificial intelligence. The review concludes with future recommendations to facilitate routine use of MRS in cancer management. Keywords: MR Spectroscopy, Spectroscopic Imaging, Molecular Imaging in Oncology, Metabolic Reprogramming, Clinical Cancer Management © RSNA, 2024.
Collapse
Affiliation(s)
- Eva Martinez Luque
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Zexuan Liu
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Dongsuk Sung
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Rachel M. Goldberg
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Rishab Agarwal
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Aditya Bhattacharya
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Nadine S. Ahmed
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Jason W. Allen
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| | - Candace C. Fleischer
- From the Departments of Radiology and Imaging Sciences (E.M.L., Z.L.,
D.S., J.W.A., C.C.F.) and Neurology (J.W.A.), Emory University School of
Medicine, Atlanta, Ga; Department of Biomedical Engineering (E.M.L., Z.L., D.S.,
J.W.A., C.C.F.), Georgia Institute of Technology and Emory University, Atlanta,
Ga; College of Arts and Sciences, Emory University, Atlanta, Ga (R.M.G.); and
College of Business (R.A.) and College of Sciences (A.B., N.S.A.), Georgia
Institute of Technology, Atlanta, Georgia
| |
Collapse
|
8
|
Harat M, Miechowicz I, Rakowska J, Zarębska I, Małkowski B. A Biopsy-Controlled Prospective Study of Contrast-Enhancing Diffuse Glioma Infiltration Based on FET-PET and FLAIR. Cancers (Basel) 2024; 16:1265. [PMID: 38610944 PMCID: PMC11010945 DOI: 10.3390/cancers16071265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 04/14/2024] Open
Abstract
Accurately defining glioma infiltration is crucial for optimizing radiotherapy and surgery, but glioma infiltration is heterogeneous and MRI imperfectly defines the tumor extent. Currently, it is impossible to determine the tumor infiltration gradient within a FLAIR signal. O-(2-[18F]fluoroethyl)-L-tyrosine (FET)-PET often reveals high-grade glioma infiltration beyond contrast-enhancing areas on MRI. Here, we studied FET uptake dynamics in tumor and normal brain structures by dual-timepoint (10 min and 40-60 min post-injection) acquisition to optimize analysis protocols for defining glioma infiltration. Over 300 serial stereotactic biopsies from 23 patients (mean age 47, 12 female/11 male) of diffuse contrast-enhancing gliomas were taken from areas inside and outside contrast enhancement or outside the FET hotspot but inside FLAIR. The final diagnosis was G4 in 11, grade 3 in 10, and grade 2 in 2 patients. The target-to-background (TBRs) ratios and standardized uptake values (SUVs) were calculated in areas used for biopsy planning and in background structures. The optimal method and threshold values were determined to find a preferred strategy for defining glioma infiltration. Standard thresholding (1.6× uptake in the contralateral brain) in standard acquisition PET images differentiated a tumor of any grade from astrogliosis, although the uptake in astrogliosis and grade 2 glioma was similar. Analyzing an optimal strategy for infiltration volume definition astrogliosis could be accurately differentiated from tumor samples using a choroid plexus as a background. Early acquisition improved the AUC in many cases, especially within FLAIR, from 56% to 90% sensitivity and 41% to 61% specificity (standard TBR 1.6 vs. early TBR plexus). The current FET-PET evaluation protocols for contrast-enhancing gliomas are limited, especially at the tumor border where grade 2 tumor and astrogliosis have similar uptake, but using choroid plexus uptake in early acquisitions as a background, we can precisely define a tumor within FLAIR that was outside of the scope of current FET-PET protocols.
Collapse
Affiliation(s)
- Maciej Harat
- Department of Neurooncology and Radiosurgery, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
- Department of Clinical Medicine, Faculty of Medicine, University of Science and Technology, 85-796 Bydgoszcz, Poland
| | - Izabela Miechowicz
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Józefina Rakowska
- Department of Neurosurgery, 10th Military Research Hospital, 85-681 Bydgoszcz, Poland;
| | - Izabela Zarębska
- Department of Radiotherapy, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland;
| | - Bogdan Małkowski
- Department of Nuclear Medicine, Franciszek Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
- Department of Diagnostic Imaging, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
9
|
Rejimon AC, Ramesh KK, Trivedi AG, Huang V, Schreibmann E, Weinberg BD, Kleinberg LR, Shu HKG, Shim H, Olson JJ. The Utility of Spectroscopic MRI in Stereotactic Biopsy and Radiotherapy Guidance in Newly Diagnosed Glioblastoma. Tomography 2024; 10:428-443. [PMID: 38535775 PMCID: PMC10975697 DOI: 10.3390/tomography10030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
Current diagnostic and therapeutic approaches for gliomas have limitations hindering survival outcomes. We propose spectroscopic magnetic resonance imaging as an adjunct to standard MRI to bridge these gaps. Spectroscopic MRI is a volumetric MRI technique capable of identifying tumor infiltration based on its elevated choline (Cho) and decreased N-acetylaspartate (NAA). We present the clinical translatability of spectroscopic imaging with a Cho/NAA ≥ 5x threshold for delineating a biopsy target in a patient diagnosed with non-enhancing glioma. Then, we describe the relationship between the undertreated tumor detected with metabolite imaging and overall survival (OS) from a pilot study of newly diagnosed GBM patients treated with belinostat and chemoradiation. Each cohort (control and belinostat) were split into subgroups using the median difference between pre-radiotherapy Cho/NAA ≥ 2x and the treated T1-weighted contrast-enhanced (T1w-CE) volume. We used the Kaplan-Meier estimator to calculate median OS for each subgroup. The median OS was 14.4 months when the difference between Cho/NAA ≥ 2x and T1w-CE volumes was higher than the median compared with 34.3 months when this difference was lower than the median. The T1w-CE volumes were similar in both subgroups. We find that patients who had lower volumes of undertreated tumors detected via spectroscopy had better survival outcomes.
Collapse
Affiliation(s)
- Abinand C. Rejimon
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA;
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (A.C.R.); (K.K.R.); (E.S.); (H.-K.G.S.); (H.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA;
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Jeffrey J. Olson
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Department of Neurosurgery, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Breen WG, Aryal MP, Cao Y, Kim MM. Integrating multi-modal imaging in radiation treatments for glioblastoma. Neuro Oncol 2024; 26:S17-S25. [PMID: 38437666 PMCID: PMC10911793 DOI: 10.1093/neuonc/noad187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Advances in diagnostic and treatment technology along with rapid developments in translational research may now allow the realization of precision radiotherapy. Integration of biologically informed multimodality imaging to address the spatial and temporal heterogeneity underlying treatment resistance in glioblastoma is now possible for patient care, with evidence of safety and potential benefit. Beyond their diagnostic utility, several candidate imaging biomarkers have emerged in recent early-phase clinical trials of biologically based radiotherapy, and their definitive assessment in multicenter prospective trials is already in development. In this review, the rationale for clinical implementation of candidate advanced magnetic resonance imaging and positron emission tomography imaging biomarkers to guide personalized radiotherapy, the current landscape, and future directions for integrating imaging biomarkers into radiotherapy for glioblastoma are summarized. Moving forward, response-adaptive radiotherapy using biologically informed imaging biomarkers to address emerging treatment resistance in rational combination with novel systemic therapies may ultimately permit improvements in glioblastoma outcomes and true individualization of patient care.
Collapse
Affiliation(s)
- William G Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Madhava P Aryal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Shu HKG, Shim H. SPECTRO GLIO trial aftermath: Where do we go from here? Neuro Oncol 2024; 26:164-165. [PMID: 37675932 PMCID: PMC10768972 DOI: 10.1093/neuonc/noad166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Indexed: 09/08/2023] Open
Affiliation(s)
- Hui-Kuo G Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Mauler J, Lohmann P, Maudsley AA, Sheriff S, Hoevels M, Meissner AK, Hamisch C, Brunn A, Deckert M, Filss CP, Stoffels G, Dammers J, Ruge MI, Galldiks N, Mottaghy FM, Langen KJ, Shah NJ. Diagnostic Accuracy of MR Spectroscopic Imaging and 18F-FET PET for Identifying Glioma: A Biopsy-Controlled Hybrid PET/MRI Study. J Nucl Med 2024; 65:16-21. [PMID: 37884332 DOI: 10.2967/jnumed.123.265868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/22/2023] [Indexed: 10/28/2023] Open
Abstract
Contrast-enhanced MRI is the method of choice for brain tumor diagnostics, despite its low specificity for tumor tissue. This study compared the contribution of MR spectroscopic imaging (MRSI) and amino acid PET to improve the detection of tumor tissue. Methods: In 30 untreated patients with suspected glioma, O-(2-[18F]fluoroethyl)-l-tyrosine (18F-FET) PET; 3-T MRSI with a short echo time; and fluid-attenuated inversion recovery, T2-weighted, and contrast-enhanced T1-weighted MRI were performed for stereotactic biopsy planning. Serial samples were taken along the needle trajectory, and their masks were projected to the preoperative imaging data. Each sample was individually evaluated neuropathologically. 18F-FET uptake and the MRSI signals choline (Cho), N-acetyl-aspartate (NAA), creatine, myoinositol, and derived ratios were evaluated for each sample and classified using logistic regression. The diagnostic accuracy was evaluated by receiver operating characteristic analysis. Results: On the basis of the neuropathologic evaluation of tissue from 88 stereotactic biopsies, supplemented with 18F-FET PET and MRSI metrics from 20 areas on the healthy-appearing contralateral hemisphere to balance the glioma/nonglioma groups, 18F-FET PET identified glioma with the highest accuracy (area under the receiver operating characteristic curve, 0.89; 95% CI, 0.81-0.93; threshold, 1.4 × background uptake). Among the MR spectroscopic metabolites, Cho/NAA normalized to normal brain tissue showed the highest diagnostic accuracy (area under the receiver operating characteristic curve, 0.81; 95% CI, 0.71-0.88; threshold, 2.2). The combination of 18F-FET PET and normalized Cho/NAA did not improve the diagnostic performance. Conclusion: MRI-based delineation of gliomas should preferably be supplemented by 18F-FET PET.
Collapse
Affiliation(s)
- Jörg Mauler
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany;
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andrew A Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Sulaiman Sheriff
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Moritz Hoevels
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anna-Katharina Meissner
- Department of General Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christina Hamisch
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anna Brunn
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuropathology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martina Deckert
- Institute of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute of Neuropathology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian P Filss
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
| | - Jürgen Dammers
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
| | - Maximillian I Ruge
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Center for Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-3/INM-4/INM-11), Forschungszentrum Juelich, Juelich, Germany
- Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany; and
- JARA-BRAIN-Translational Medicine, Aachen, Germany
| |
Collapse
|
13
|
Lemarié A, Lubrano V, Delmas C, Lusque A, Cerapio JP, Perrier M, Siegfried A, Arnauduc F, Nicaise Y, Dahan P, Filleron T, Mounier M, Toulas C, Cohen-Jonathan Moyal E. The STEMRI trial: Magnetic resonance spectroscopy imaging can define tumor areas enriched in glioblastoma stem-like cells. SCIENCE ADVANCES 2023; 9:eadi0114. [PMID: 37922359 PMCID: PMC10624352 DOI: 10.1126/sciadv.adi0114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Despite maximally safe resection of the magnetic resonance imaging (MRI)-defined contrast-enhanced (CE) central tumor area and chemoradiotherapy, most patients with glioblastoma (GBM) relapse within a year in peritumoral FLAIR regions. Magnetic resonance spectroscopy imaging (MRSI) can discriminate metabolic tumor areas with higher recurrence potential as CNI+ regions (choline/N-acetyl-aspartate index >2) can predict relapse sites. As relapses are mainly imputed to glioblastoma stem-like cells (GSCs), CNI+ areas might be GSC enriched. In this prospective trial, 16 patients with GBM underwent MRSI/MRI before surgery/chemoradiotherapy to investigate GSC content in CNI-/+ biopsies from CE/FLAIR. Biopsy and derived-GSC characterization revealed a FLAIR/CNI+ sample enrichment in GSC and in gene signatures related to stemness, DNA repair, adhesion/migration, and mitochondrial bioenergetics. FLAIR/CNI+ samples generate GSC-enriched neurospheres faster than FLAIR/CNI-. Parameters assessing biopsy GSC content and time-to-neurosphere formation in FLAIR/CNI+ were associated with worse patient outcome. Preoperative MRI/MRSI would certainly allow better resection and targeting of FLAIR/CNI+ areas, as their GSC enrichment can predict worse outcomes.
Collapse
Affiliation(s)
- Anthony Lemarié
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Vincent Lubrano
- TONIC, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Toulouse Neuro Imaging Center, Toulouse, France
- CHU de Toulouse, Neurosurgery Department, Toulouse, France
| | - Caroline Delmas
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Interface Department, Toulouse, France
| | - Amélie Lusque
- Institut Claudius Regaud, IUCT-Oncopole, Biostatistics and Health Data Science Unit, Toulouse, France
| | - Juan-Pablo Cerapio
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Marion Perrier
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Aurore Siegfried
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- CHU de Toulouse, Anatomopathology Department, Toulouse, France
| | - Florent Arnauduc
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Yvan Nicaise
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
| | - Perrine Dahan
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Thomas Filleron
- Institut Claudius Regaud, IUCT-Oncopole, Biostatistics and Health Data Science Unit, Toulouse, France
| | - Muriel Mounier
- Institut Claudius Regaud, IUCT-Oncopole, Clinical Trials Office, Toulouse, France
| | - Christine Toulas
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Cancer Biology Department, Molecular Oncology Division, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III–Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- UFR Santé, Université de Toulouse III–Paul Sabatier, Toulouse, France
- Institut Claudius Regaud, IUCT-Oncopole, Radiation Oncology Department, Toulouse, France
| |
Collapse
|
14
|
Karschnia P, Smits M, Reifenberger G, Le Rhun E, Ellingson BM, Galldiks N, Kim MM, Huse JT, Schnell O, Harter PN, Mohme M, von Baumgarten L, Albert NL, Huang RY, Mehta MP, van den Bent M, Weller M, Vogelbaum MA, Chang SM, Berger MS, Tonn JC. A framework for standardised tissue sampling and processing during resection of diffuse intracranial glioma: joint recommendations from four RANO groups. Lancet Oncol 2023; 24:e438-e450. [PMID: 37922934 PMCID: PMC10849105 DOI: 10.1016/s1470-2045(23)00453-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/23/2023] [Accepted: 09/07/2023] [Indexed: 11/07/2023]
Abstract
Surgical resection represents the standard of care for people with newly diagnosed diffuse gliomas, and the neuropathological and molecular profile of the resected tissue guides clinical management and forms the basis for research. The Response Assessment in Neuro-Oncology (RANO) consortium is an international, multidisciplinary effort that aims to standardise research practice in neuro-oncology. These recommendations represent a multidisciplinary consensus from the four RANO groups: RANO resect, RANO recurrent glioblastoma, RANO radiotherapy, and RANO/PET for a standardised workflow to achieve a representative tumour evaluation in a disease characterised by intratumoural heterogeneity, including recommendations on which tumour regions should be surgically sampled, how to define those regions on the basis of preoperative imaging, and the optimal sample volume. Practical recommendations for tissue sampling are given for people with low-grade and high-grade gliomas, as well as for people with newly diagnosed and recurrent disease. Sampling of liquid biopsies is also addressed. A standardised workflow for subsequent handling of the resected tissue is proposed to avoid information loss due to decreasing tissue quality or insufficient clinical information. The recommendations offer a framework for prospective biobanking studies.
Collapse
Affiliation(s)
- Philipp Karschnia
- Department of Neurosurgery, Ludwig-Maximilians-University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Marion Smits
- Department of Neuroradiology and Nuclear Medicine, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Guido Reifenberger
- Institute of Neuropathology, Heinrich Heine University Medical Faculty and University Hospital Düsseldorf, Düsseldorf, Germany
| | - Emilie Le Rhun
- Department of Neurosurgery, University Hospital of Zurich and University of Zurich, Zurich, Switzerland; Department of Neurology, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; Research Center Juelich, Institute of Neuroscience and Medicine, Juelich, Germany
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan Hospital, Ann Arbor, MI, USA
| | - Jason T Huse
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oliver Schnell
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Patrick N Harter
- German Cancer Consortium, Partner Site Munich, Munich, Germany; Center for Neuropathology and Prion Research, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, Ludwig-Maximilians-University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Raymond Y Huang
- Division of Neuroradiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Martin van den Bent
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Michael Weller
- Department of Neurology, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| | | | - Susan M Chang
- Department of Neurosurgery and Division of Neuro-Oncology, University of California, San Francisco, CA, USA
| | - Mitchel S Berger
- Department of Neurosurgery and Division of Neuro-Oncology, University of California, San Francisco, CA, USA
| | - Joerg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Munich, Germany.
| |
Collapse
|
15
|
Trivedi AG, Ramesh KK, Huang V, Mellon EA, Barker PB, Kleinberg LR, Weinberg BD, Shu HKG, Shim H. Spectroscopic MRI-Based Biomarkers Predict Survival for Newly Diagnosed Glioblastoma in a Clinical Trial. Cancers (Basel) 2023; 15:3524. [PMID: 37444634 PMCID: PMC10340675 DOI: 10.3390/cancers15133524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Despite aggressive treatment, glioblastoma has a poor prognosis due to its infiltrative nature. Spectroscopic MRI-measured brain metabolites, particularly the choline to N-acetylaspartate ratio (Cho/NAA), better characterizes the extent of tumor infiltration. In a previous pilot trial (NCT03137888), brain regions with Cho/NAA ≥ 2x normal were treated with high-dose radiation for newly diagnosed glioblastoma patients. This report is a secondary analysis of that trial where spectroscopic MRI-based biomarkers are evaluated for how they correlate with progression-free and overall survival (PFS/OS). Subgroups were created within the cohort based on pre-radiation treatment (pre-RT) median cutoff volumes of residual enhancement (2.1 cc) and metabolically abnormal volumes used for treatment (19.2 cc). We generated Kaplan-Meier PFS/OS curves and compared these curves via the log-rank test between subgroups. For the subgroups stratified by metabolic abnormality, statistically significant differences were observed for PFS (p = 0.019) and OS (p = 0.020). Stratification by residual enhancement did not lead to observable differences in the OS (p = 0.373) or PFS (p = 0.286) curves. This retrospective analysis shows that patients with lower post-surgical Cho/NAA volumes had significantly superior survival outcomes, while residual enhancement, which guides high-dose radiation in standard treatment, had little significance in PFS/OS. This suggests that the infiltrating, non-enhancing component of glioblastoma is an important factor in patient outcomes and should be treated accordingly.
Collapse
Affiliation(s)
- Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eric A. Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 45056, USA
| | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
16
|
Wu T, Liu C, Thamizhchelvan AM, Fleischer C, Peng X, Liu G, Mao H. Label-Free Chemically and Molecularly Selective Magnetic Resonance Imaging. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:121-139. [PMID: 37235188 PMCID: PMC10207347 DOI: 10.1021/cbmi.3c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 05/28/2023]
Abstract
Biomedical imaging, especially molecular imaging, has been a driving force in scientific discovery, technological innovation, and precision medicine in the past two decades. While substantial advances and discoveries in chemical biology have been made to develop molecular imaging probes and tracers, translating these exogenous agents to clinical application in precision medicine is a major challenge. Among the clinically accepted imaging modalities, magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) exemplify the most effective and robust biomedical imaging tools. Both MRI and MRS enable a broad range of chemical, biological and clinical applications from determining molecular structures in biochemical analysis to imaging diagnosis and characterization of many diseases and image-guided interventions. Using chemical, biological, and nuclear magnetic resonance properties of specific endogenous metabolites and native MRI contrast-enhancing biomolecules, label-free molecular and cellular imaging with MRI can be achieved in biomedical research and clinical management of patients with various diseases. This review article outlines the chemical and biological bases of several label-free chemically and molecularly selective MRI and MRS methods that have been applied in imaging biomarker discovery, preclinical investigation, and image-guided clinical management. Examples are provided to demonstrate strategies for using endogenous probes to report the molecular, metabolic, physiological, and functional events and processes in living systems, including patients. Future perspectives on label-free molecular MRI and its challenges as well as potential solutions, including the use of rational design and engineered approaches to develop chemical and biological imaging probes to facilitate or combine with label-free molecular MRI, are discussed.
Collapse
Affiliation(s)
- Tianhe Wu
- Department
of Radiology and Imaging Sciences, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Claire Liu
- F.M.
Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| | - Anbu Mozhi Thamizhchelvan
- Department
of Radiology and Imaging Sciences, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Candace Fleischer
- Department
of Radiology and Imaging Sciences, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Xingui Peng
- Jiangsu
Key Laboratory of Molecular and Functional Imaging, Department of
Radiology, Zhongda Hospital, Medical School
of Southeast University, Nanjing, Jiangsu 210009, China
| | - Guanshu Liu
- F.M.
Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
- Russell
H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Hui Mao
- Department
of Radiology and Imaging Sciences, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
17
|
Trivedi AG, Kim SH, Ramesh KK, Giuffrida AS, Weinberg BD, Mellon EA, Kleinberg LR, Barker PB, Han H, Shu HKG, Shim H, Schreibmann E. Applying a Radiation Therapy Volume Analysis Pipeline to Determine the Utility of Spectroscopic MRI-Guided Adaptive Radiation Therapy for Glioblastoma. Tomography 2023; 9:1052-1061. [PMID: 37218946 PMCID: PMC10204497 DOI: 10.3390/tomography9030086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
Accurate radiation therapy (RT) targeting is crucial for glioblastoma treatment but may be challenging using clinical imaging alone due to the infiltrative nature of glioblastomas. Precise targeting by whole-brain spectroscopic MRI, which maps tumor metabolites including choline (Cho) and N-acetylaspartate (NAA), can quantify early treatment-induced molecular changes that other traditional modalities cannot measure. We developed a pipeline to determine how spectroscopic MRI changes during early RT are associated with patient outcomes to provide insight into the utility of adaptive RT planning. Data were obtained from a study (NCT03137888) where glioblastoma patients received high-dose RT guided by the pre-RT Cho/NAA twice normal (Cho/NAA ≥ 2x) volume, and received spectroscopic MRI scans pre- and mid-RT. Overlap statistics between pre- and mid-RT scans were used to quantify metabolic activity changes after two weeks of RT. Log-rank tests were used to quantify the relationship between imaging metrics and patient overall and progression-free survival (OS/PFS). Patients with lower Jaccard/Dice coefficients had longer PFS (p = 0.045 for both), and patients with lower Jaccard/Dice coefficients had higher OS trending towards significance (p = 0.060 for both). Cho/NAA ≥ 2x volumes changed significantly during early RT, putting healthy tissue at risk of irradiation, and warranting further study into using adaptive RT planning.
Collapse
Affiliation(s)
- Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Su Hyun Kim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eric A. Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 45056, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hui Han
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Chang CM, Ramesh KK, Huang V, Gurbani S, Kleinberg LR, Weinberg BD, Shim H, Shu HKG. Mutant Isocitrate Dehydrogenase 1 Expression Enhances Response of Gliomas to the Histone Deacetylase Inhibitor Belinostat. Tomography 2023; 9:942-954. [PMID: 37218937 PMCID: PMC10204413 DOI: 10.3390/tomography9030077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Histone deacetylase inhibitors (HDACis) are drugs that target the epigenetic state of cells by modifying the compaction of chromatin through effects on histone acetylation. Gliomas often harbor a mutation of isocitrate dehydrogenase (IDH) 1 or 2 that leads to changes in their epigenetic state presenting a hypermethylator phenotype. We postulated that glioma cells with IDH mutation, due to the presence of epigenetic changes, will show increased sensitivity to HDACis. This hypothesis was tested by expressing mutant IDH1 with a point alteration-converting arginine 132 to histidine-within glioma cell lines that contain wild-type IDH1. Glioma cells engineered to express mutant IDH1 produced D-2-hydroxyglutarate as expected. When assessed for response to the pan-HDACi drug belinostat, mutant IDH1-expressing glioma cells were subjected to more potent inhibition of growth than the corresponding control cells. Increased sensitivity to belinostat correlated with the increased induction of apoptosis. Finally, a phase I trial assessing the addition of belinostat to standard-of-care therapy for newly diagnosed glioblastoma patients included one patient with a mutant IDH1 tumor. This mutant IDH1 tumor appeared to display greater sensitivity to the addition of belinostat than the other cases with wild-type IDH tumors based on both standard magnetic resonance imaging (MRI) and advanced spectroscopic MRI criteria. These data together suggest that IDH mutation status within gliomas may serve as a biomarker of response to HDACis.
Collapse
Affiliation(s)
- Chi-Ming Chang
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Saumya Gurbani
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
| | | | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
19
|
Tensaouti F, Desmoulin F, Gilhodes J, Roques M, Ken S, Lotterie JA, Noël G, Truc G, Sunyach MP, Charissoux M, Magné N, Lubrano V, Péran P, Cohen-Jonathan Moyal E, Laprie A. Is pre-radiotherapy metabolic heterogeneity of glioblastoma predictive of progression-free survival? Radiother Oncol 2023; 183:109665. [PMID: 37024057 DOI: 10.1016/j.radonc.2023.109665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND AND PURPOSE All glioblastoma subtypes share the hallmark of aggressive invasion, meaning that it is crucial to identify their different components if we are to ensure effective treatment and improve survival. Proton MR spectroscopic imaging (MRSI) is a noninvasive technique that yields metabolic information and is able to identify pathological tissue with high accuracy. The aim of the present study was to identify clusters of metabolic heterogeneity, using a large MRSI dataset, and determine which of these clusters are predictive of progression-free survival (PFS). MATERIALS AND METHODS MRSI data of 180 patients acquired in a pre-radiotherapy examination were included in the prospective SPECTRO-GLIO trial. Eight features were extracted for each spectrum: Cho/NAA, NAA/Cr, Cho/Cr, Lac/NAA, and the ratio of each metabolite to the sum of all the metabolites. Clustering of data was performed using a mini-batch k-means algorithm. The Cox model and logrank test were used for PFS analysis. RESULTS Five clusters were identified as sharing similar metabolic information and being predictive of PFS. Two clusters revealed metabolic abnormalities. PFS was lower when Cluster 2 was the dominant cluster in patients' MRSI data. Among the metabolites, lactate (present in this cluster and in Cluster 5) was the most statistically significant predictor of poor outcome. CONCLUSION Results showed that pre-radiotherapy MRSI can be used to reveal tumor heterogeneity. Groups of spectra, which have the same metabolic information, reflect the different tissue components representative of tumor burden proliferation and hypoxia. Clusters with metabolic abnormalities and high lactate are predictive of PFS.
Collapse
Affiliation(s)
- Fatima Tensaouti
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.
| | - Franck Desmoulin
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Julia Gilhodes
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Biostatistics, Toulouse, France
| | - Margaux Roques
- CHU Toulouse, Neuroradiology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Soleakhena Ken
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Engineering and Medical Physics, Toulouse, France; Inserm U1037- Centre de Recherches contre le Cancer de Toulouse, Radiation oncology, Toulouse, France
| | - Jean-Albert Lotterie
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; CHU Toulouse, Nuclear Medicine, Toulouse, France
| | | | - Gilles Truc
- Centre Georges-François Leclerc, Radiation Oncology, Dijon, France
| | | | - Marie Charissoux
- Institut du Cancer de Montpellier, Radiation Oncology, Montpellier, France
| | - Nicolas Magné
- Institut de Cancérologie de la Loire Lucien Neuwirth, Radiation Oncology, Saint-Priest-en-Jarez, France
| | - Vincent Lubrano
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Elizabeth Cohen-Jonathan Moyal
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; Inserm U1037- Centre de Recherches contre le Cancer de Toulouse, Radiation oncology, Toulouse, France
| | - Anne Laprie
- Institut Claudius Regaud/Institut Universitaire du Cancer de Toulouse - Oncopôle, Radiation oncology, Toulouse, France; ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| |
Collapse
|
20
|
Goryawala M, Mellon EA, Shim H, Maudsley AA. Mapping early tumor response to radiotherapy using diffusion kurtosis imaging*. Neuroradiol J 2023; 36:198-205. [PMID: 36000488 PMCID: PMC10034702 DOI: 10.1177/19714009221122204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE In this pilot study, DKI measures of diffusivity and kurtosis were compared in active tumor regions and correlated to radiologic response to radiotherapy after completion of 2 weeks of treatment to derive potential early measures of tumor response. METHODS MRI and Magnetic Resonance Spectroscopic Imaging (MRSI) data were acquired before the beginning of RT (pre-RT) and 2 weeks after the initiation of treatment (during-RT) in 14 glioblastoma patients. The active tumor region was outlined as the union of the residual contrast-enhancing region and metabolically active tumor region. Average and standard deviation of mean, axial, and radial diffusivity (MD, AD, RD) and mean, axial, and radial kurtosis (MK, AK, RK) values were calculated for the active tumor VOI from images acquired pre-RT and during-RT and paired t-tests were executed to estimate pairwise differences. Receiver operating characteristic (ROC) curve analysis was conducted to evaluate the predictive capabilities of changes in diffusion metrics for progression-free survival (PFS). RESULTS Analysis showed significant pairwise differences for AD (p = 0.035; Cohen's d of 0.659) and AK (p = 0.019; Cohen's d of 0.753) in diffusion measures after 2 weeks of RT. ROC curve analysis showed that percentage change differences in AD and AK between pre-RT and during-RT scans provided an Area Under the Curve (AUC) of 0.524 and 0.762, respectively, in discriminating responders (PFS>180 days) and non-responders (PFS<180 days). CONCLUSION This pilot study, although preliminary in nature, showed significant changes in AD and AK maps, with kurtosis derived AK maps showing an increased sensitivity in mapping early changes in the active tumor regions.
Collapse
Affiliation(s)
| | - Eric A Mellon
- Department of Radiation Oncology, University of Miami, Miami, FL, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
21
|
Huang V, Rejimon A, Reddy K, Trivedi AG, Ramesh KK, Giuffrida AS, Muiruri R, Shim H, Eaton BR. Spectroscopic MRI-Guided Proton Therapy in Non-Enhancing Pediatric High-Grade Glioma. Tomography 2023; 9:633-646. [PMID: 36961010 PMCID: PMC10037577 DOI: 10.3390/tomography9020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Radiation therapy (RT) is a critical part of definitive therapy for pediatric high-grade glioma (pHGG). RT is designed to treat residual tumor defined on conventional MRI (cMRI), though pHGG lesions may be ill-characterized on standard imaging. Spectroscopic MRI (sMRI) measures endogenous metabolite concentrations in the brain, and Choline (Cho)/N-acetylaspartate (NAA) ratio is a highly sensitive biomarker for metabolically active tumor. We provide a preliminary report of our study introducing a novel treatment approach of whole brain sMRI-guided proton therapy for pHGG. An observational cohort (c1 = 10 patients) receives standard of care RT; a therapeutic cohort (c2 = 15 patients) receives sMRI-guided proton RT. All patients undergo cMRI and sMRI, a high-resolution 3D whole-brain echo-planar spectroscopic imaging (EPSI) sequence (interpolated resolution of 12 µL) prior to RT and at several follow-up timepoints integrated into diagnostic scans. Treatment volumes are defined by cMRI for c1 and by cMRI and Cho/NAA ≥ 2x for c2. A longitudinal imaging database is used to quantify changes in lesion and metabolite volumes. Four subjects have been enrolled (c1 = 1/c2 = 3) with sMRI imaging follow-up of 4-18 months. Preliminary data suggest sMRI improves identification of pHGG infiltration based on abnormal metabolic activity, and using proton therapy to target sMRI-defined high-risk regions is safe and feasible.
Collapse
Affiliation(s)
- Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Abinand Rejimon
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kartik Reddy
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Robert Muiruri
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bree R. Eaton
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
22
|
Bell JB, Jin W, Goryawala MZ, Azzam GA, Abramowitz MC, Diwanji T, Ivan ME, del Pilar Guillermo Prieto Eibl M, de la Fuente MI, Mellon EA. Delineation of recurrent glioblastoma by whole brain spectroscopic magnetic resonance imaging. Radiat Oncol 2023; 18:37. [PMID: 36814267 PMCID: PMC9948314 DOI: 10.1186/s13014-023-02219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/31/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) cellularity correlates with whole brain spectroscopic MRI (sMRI) generated relative choline to N-Acetyl-Aspartate ratio (rChoNAA) mapping. In recurrent GBM (rGBM), tumor volume (TV) delineation is challenging and rChoNAA maps may assist with re-RT targeting. METHODS Fourteen rGBM patients underwent sMRI in a prospective study. Whole brain sMRI was performed to generate rChoNAA maps. TVs were delineated by the union of rChoNAA ratio over 2 (rChoNAA > 2) on sMRI and T1PC. rChoNAA > 2 volumes were compared with multiparametric MRI sequences including T1PC, T2/FLAIR, diffusion-restriction on apparent diffusion coefficient (ADC) maps, and perfusion relative cerebral blood volume (rCBV). RESULTS rChoNAA > 2 (mean 27.6 cc, range 6.6-79.1 cc) was different from other imaging modalities (P ≤ 0.05). Mean T1PC volumes were 10.7 cc (range 1.2-31.4 cc). The mean non-overlapping volume of rChoNAA > 2 and T1PC was 29.2 cm3. rChoNAA > 2 was 287% larger (range 23% smaller-873% larger) than T1PC. T2/FLAIR volumes (mean 111.7 cc, range 19.0-232.7 cc) were much larger than other modalities. rCBV volumes (mean 6.2 cc, range 0.2-19.1 cc) and ADC volumes were tiny (mean 0.8 cc, range 0-3.7 cc). Eight in-field failures were observed. Three patients failed outside T1PC but within rChoNAA > 2. No grade 3 toxicities attributable to re-RT were observed. Median progression-free and overall survival for re-RT patients were 6.5 and 7.1 months, respectively. CONCLUSIONS Treatment of rGBM may be optimized by sMRI, and failure patterns suggest benefit for dose-escalation within sMRI-delineated volumes. Dose-escalation and radiologic-pathologic studies are underway to confirm the utility of sMRI in rGBM.
Collapse
Affiliation(s)
- Jonathan B. Bell
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| | - William Jin
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| | - Mohammed Z. Goryawala
- grid.26790.3a0000 0004 1936 8606Department of Radiology, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Gregory A. Azzam
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| | - Matthew C. Abramowitz
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| | - Tejan Diwanji
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| | - Michael E. Ivan
- grid.26790.3a0000 0004 1936 8606Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Maria del Pilar Guillermo Prieto Eibl
- grid.26790.3a0000 0004 1936 8606Department of Neurology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Macarena I. de la Fuente
- grid.26790.3a0000 0004 1936 8606Department of Neurology and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Eric A. Mellon
- grid.26790.3a0000 0004 1936 8606Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, 1475 NW 12th Ave, Miami, FL 33136 USA
| |
Collapse
|
23
|
Ramesh KK, Huang V, Rosenthal J, Mellon EA, Goryawala M, Barker PB, Gurbani SS, Trivedi AG, Giuffrida AS, Schreibmann E, Han H, de le Fuente M, Dunbar EM, Holdhoff M, Kleinberg LR, Shu HKG, Shim H, Weinberg BD. A Novel Approach to Determining Tumor Progression Using a Three-Site Pilot Clinical Trial of Spectroscopic MRI-Guided Radiation Dose Escalation in Glioblastoma. Tomography 2023; 9:362-374. [PMID: 36828381 PMCID: PMC9964256 DOI: 10.3390/tomography9010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Glioblastoma (GBM) is a fatal disease, with poor prognosis exacerbated by difficulty in assessing tumor extent with imaging. Spectroscopic MRI (sMRI) is a non-contrast imaging technique measuring endogenous metabolite levels of the brain that can serve as biomarkers for tumor extension. We completed a three-site study to assess survival benefits of GBM patients when treated with escalated radiation dose guided by metabolic abnormalities in sMRI. Escalated radiation led to complex post-treatment imaging, requiring unique approaches to discern tumor progression from radiation-related treatment effect through our quantitative imaging platform. The purpose of this study is to determine true tumor recurrence timepoints for patients in our dose-escalation multisite study using novel methodology and to report on median progression-free survival (PFS). Follow-up imaging for all 30 trial patients were collected, lesion volumes segmented and graphed, and imaging uploaded to our platform for visual interpretation. Eighteen months post-enrollment, the median PFS was 16.6 months with a median time to follow-up of 20.3 months. With this new treatment paradigm, incidence rate of tumor recurrence one year from treatment is 30% compared to 60-70% failure under standard care. Based on the delayed tumor progression and improved survival, a randomized phase II trial is under development (EAF211).
Collapse
Affiliation(s)
- Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Jeffrey Rosenthal
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eric A. Mellon
- Department of Radiation Oncology, University of Miami, Miami, FL 45056, USA
| | | | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Saumya S. Gurbani
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Han
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Erin M. Dunbar
- Department of Neuro-Oncology and Neurosurgery, Piedmont Atlanta Hospital, Atlanta, GA 30309, USA
| | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
24
|
Long H, Zhang P, Bi Y, Yang C, Wu M, He D, Huang S, Yang K, Qi S, Wang J. MRI radiomic features of peritumoral edema may predict the recurrence sites of glioblastoma multiforme. Front Oncol 2023; 12:1042498. [PMID: 36686829 PMCID: PMC9845721 DOI: 10.3389/fonc.2022.1042498] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/02/2022] [Indexed: 01/05/2023] Open
Abstract
Background and purpose As one of the most aggressive malignant tumor in the central nervous system, the main cause of poor outcome of glioblastoma (GBM) is recurrence, a non-invasive method which can predict the area of recurrence pre-operation is necessary.To investigate whether there is radiological heterogeneity within peritumoral edema and identify the reproducible radiomic features predictive of the sites of recurrence of glioblastoma(GBM), which may be of value to optimize patients' management. Materials and methods The clinical information and MR images (contrast-enhanced T1 weighted and FLAIR sequences) of 22 patients who have been histologically proven glioblastoma, were retrospectively evaluated. Kaplan-Meier methods was used for survival analysis. Oedematous regions were manually segmented by an expert into recurrence region, non-recurrence region. A set of 94 radiomic features were obtained from each region using the function of analyzing MR image of 3D slicer. Paired t test was performed to identify the features existing significant difference. Subsequently, the data of two patients from TCGA database was used to evaluate whether these features have clinical value. Results Ten features with significant differences between the recurrence and non-recurrence subregions were identified and verified on two individual patients from the TCGA database with pathologically confirmed diagnosis of GBM. Conclusions Our results suggested that heterogeneity does exist in peritumoral edema, indicating that the radiomic features of peritumoral edema from routine MR images can be utilized to predict the sites of GBM recurrence. Our findings may further guide the surgical treatment strategy for GBM.
Collapse
Affiliation(s)
- Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Ping Zhang
- Department of oncology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Yuewei Bi
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Chen Yang
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Manfeng Wu
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Dian He
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Shaozhuo Huang
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Kaijun Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China,The First Clinical Medicine College, Southern Medical University, Guangzhou, China,Neural Networks Surgery Team, Southern Medical University, Guangzhou, China,*Correspondence: Jun Wang,
| |
Collapse
|
25
|
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
|
26
|
Multiparametric Characterization of Intracranial Gliomas Using Dynamic [18F]FET-PET and Magnetic Resonance Spectroscopy. Diagnostics (Basel) 2022; 12:diagnostics12102331. [PMID: 36292019 PMCID: PMC9601276 DOI: 10.3390/diagnostics12102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Both static and dynamic O-(2-[18F]fluoroethyl)-l-tyrosine-(FET)-PET and 1H magnetic resonance spectroscopy (MRS) are useful tools for grading and prognostication in gliomas. However, little is known about the potential of multimodal imaging comprising both procedures. We therefore acquired NAA/Cr and Cho/Cr ratios in multi-voxel MRS as well as FET-PET parameters in 67 glioma patients and determined multiparametric parameter combinations. Using receiver operating characteristics, differentiation between low-grade and high-grade glioma was possible by static FET-PET (area under the curve (AUC) 0.86, p = 0.001), time-to-peak (TTP; AUC 0.79, p = 0.049), and using the Cho/Cr ratio (AUC 0.72, p = 0.039), while the multimodal analysis led to improved discrimination with an AUC of 0.97 (p = 0.001). In order to distinguish glioblastoma from non-glioblastoma, MRS (NAA/Cr ratio, AUC 0.66, p = 0.031), and dynamic FET-PET (AUC 0.88, p = 0.001) were superior to static FET imaging. The multimodal analysis increased the accuracy with an AUC of 0.97 (p < 0.001). In the survival analysis, PET parameters, but not spectroscopy, were significantly correlated with overall survival (OS, static PET p = 0.014, TTP p = 0.012), still, the multiparametric analysis, including MRS, was also useful for the prediction of OS (p = 0.002). In conclusion, FET-PET and MRS provide complementary information to better characterize gliomas before therapy, which is particularly interesting with respect to the increasing use of hybrid PET/MRI for brain tumors.
Collapse
|
27
|
Jain V, de Godoy LL, Mohan S, Chawla S, Learned K, Jain G, Wehrli FW, Alonso-Basanta M. Cerebral hemodynamic and metabolic dysregulation in the postradiation brain. J Neuroimaging 2022; 32:1027-1043. [PMID: 36156829 DOI: 10.1111/jon.13053] [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: 07/13/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
Technological advances in the delivery of radiation and other novel cancer therapies have significantly improved the 5-year survival rates over the last few decades. Although recent developments have helped to better manage the acute effects of radiation, the late effects such as impairment in cognition continue to remain of concern. Accruing data in the literature have implicated derangements in hemodynamic parameters and metabolic activity of the irradiated normal brain as predictive of cognitive impairment. Multiparametric imaging modalities have allowed us to precisely quantify functional and metabolic information, enhancing the anatomic and morphologic data provided by conventional MRI sequences, thereby contributing as noninvasive imaging-based biomarkers of radiation-induced brain injury. In this review, we have elaborated on the mechanisms of radiation-induced brain injury and discussed several novel imaging modalities, including MR spectroscopy, MR perfusion imaging, functional MR, SPECT, and PET that provide pathophysiological and functional insights into the postradiation brain, and its correlation with radiation dose as well as clinical neurocognitive outcomes. Additionally, we explored some innovative imaging modalities, such as quantitative blood oxygenation level-dependent imaging, susceptibility-based oxygenation measurement, and T2-based oxygenation measurement, that hold promise in delineating the potential mechanisms underlying deleterious neurocognitive changes seen in the postradiation setting. We aim that this comprehensive review of a range of imaging modalities will help elucidate the hemodynamic and metabolic injury mechanisms underlying cognitive impairment in the irradiated normal brain in order to optimize treatment regimens and improve the quality of life for these patients.
Collapse
Affiliation(s)
- Varsha Jain
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiation Oncology, Jefferson University Hospital, 111 South 11th Street, Philadelphia, PA, 19107, USA
| | - Laiz Laura de Godoy
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kim Learned
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gaurav Jain
- Department of Neurological Surgery, Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Felix W Wehrli
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle Alonso-Basanta
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Li ZC, Yan J, Zhang S, Liang C, Lv X, Zou Y, Zhang H, Liang D, Zhang Z, Chen Y. Glioma survival prediction from whole-brain MRI without tumor segmentation using deep attention network: a multicenter study. Eur Radiol 2022; 32:5719-5729. [PMID: 35278123 DOI: 10.1007/s00330-022-08640-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To develop and validate a deep learning model for predicting overall survival from whole-brain MRI without tumor segmentation in patients with diffuse gliomas. METHODS In this multicenter retrospective study, two deep learning models were built for survival prediction from MRI, including a DeepRisk model built from whole-brain MRI, and an original ResNet model built from expert-segmented tumor images. Both models were developed using a training dataset (n = 935) and an internal tuning dataset (n = 156) and tested on two external test datasets (n = 194 and 150) and a TCIA dataset (n = 121). C-index, integrated Brier score (IBS), prediction error curves, and calibration curves were used to assess the model performance. RESULTS In total, 1556 patients were enrolled (age, 49.0 ± 13.1 years; 830 male). The DeepRisk score was an independent predictor and can stratify patients in each test dataset into three risk subgroups. The IBS and C-index for DeepRisk were 0.14 and 0.83 in external test dataset 1, 0.15 and 0.80 in external dataset 2, and 0.16 and 0.77 in TCIA dataset, respectively, which were comparable with those for original ResNet. The AUCs at 6, 12, 24, 26, and 48 months for DeepRisk ranged between 0.77 and 0.94. Combining DeepRisk score with clinicomolecular factors resulted in a nomogram with a better calibration and classification accuracy (net reclassification improvement 0.69, p < 0.001) than the clinical nomogram. CONCLUSIONS DeepRisk that obviated the need of tumor segmentation can predict glioma survival from whole-brain MRI and offers incremental prognostic value. KEY POINTS • DeepRisk can predict overall survival directly from whole-brain MRI without tumor segmentation. • DeepRisk achieves comparable accuracy in survival prediction with deep learning model built using expert-segmented tumor images. • DeepRisk has independent and incremental prognostic value over existing clinical parameters and IDH mutation status.
Collapse
Affiliation(s)
- Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shenghai Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chaofeng Liang
- Department of Neurosurgery, The 3rd Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaofei Lv
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan Zou
- Department of Radiology, The 3rd Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huailing Zhang
- School of Information Engineering, Guangdong Medical University, Dongguan, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 1 Jian she Dong Road, Zhengzhou, 450052, Henan, China.
| | - Yinsheng Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
29
|
Xu J, Meng Y, Qiu K, Topatana W, Li S, Wei C, Chen T, Chen M, Ding Z, Niu G. Applications of Artificial Intelligence Based on Medical Imaging in Glioma: Current State and Future Challenges. Front Oncol 2022; 12:892056. [PMID: 35965542 PMCID: PMC9363668 DOI: 10.3389/fonc.2022.892056] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma is one of the most fatal primary brain tumors, and it is well-known for its difficulty in diagnosis and management. Medical imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and spectral imaging can efficiently aid physicians in diagnosing, treating, and evaluating patients with gliomas. With the increasing clinical records and digital images, the application of artificial intelligence (AI) based on medical imaging has reduced the burden on physicians treating gliomas even further. This review will classify AI technologies and procedures used in medical imaging analysis. Additionally, we will discuss the applications of AI in glioma, including tumor segmentation and classification, prediction of genetic markers, and prediction of treatment response and prognosis, using MRI, PET, and spectral imaging. Despite the benefits of AI in clinical applications, several issues such as data management, incomprehension, safety, clinical efficacy evaluation, and ethical or legal considerations, remain to be solved. In the future, doctors and researchers should collaborate to solve these issues, with a particular emphasis on interdisciplinary teamwork.
Collapse
Affiliation(s)
- Jiaona Xu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuting Meng
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefan Qiu
- Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Win Topatana
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shijie Li
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Wei
- Department of Neurology, Affiliated Ningbo First Hospital, Ningbo, China
| | - Tianwen Chen
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| | - Guozhong Niu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Mingyu Chen, ; Zhongxiang Ding, ; Guozhong Niu,
| |
Collapse
|
30
|
Ismail M, Prasanna P, Bera K, Statsevych V, Hill V, Singh G, Partovi S, Beig N, McGarry S, Laviolette P, Ahluwalia M, Madabhushi A, Tiwari P. Radiomic Deformation and Textural Heterogeneity (R-DepTH) Descriptor to Characterize Tumor Field Effect: Application to Survival Prediction in Glioblastoma. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:1764-1777. [PMID: 35108202 PMCID: PMC9575333 DOI: 10.1109/tmi.2022.3148780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The concept of tumor field effect implies that cancer is a systemic disease with its impact way beyond the visible tumor confines. For instance, in Glioblastoma (GBM), an aggressive brain tumor, the increase in intracranial pressure due to tumor burden often leads to brain herniation and poor outcomes. Our work is based on the rationale that highly aggressive tumors tend to grow uncontrollably, leading to pronounced biomechanical tissue deformations in the normal parenchyma, which when combined with local morphological differences in the tumor confines on MRI scans, will comprehensively capture tumor field effect. Specifically, we present an integrated MRI-based descriptor, radiomic-Deformation and Textural Heterogeneity (r-DepTH). This descriptor comprises measurements of the subtle perturbations in tissue deformations throughout the surrounding normal parenchyma due to mass effect. This involves non-rigidly aligning the patients' MRI scans to a healthy atlas via diffeomorphic registration. The resulting inverse mapping is used to obtain the deformation field magnitudes in the normal parenchyma. These measurements are then combined with a 3D texture descriptor, Co-occurrence of Local Anisotropic Gradient Orientations (COLLAGE), which captures the morphological heterogeneity and infiltration within the tumor confines, on MRI scans. In this work, we extensively evaluated r-DepTH for survival risk-stratification on a total of 207 GBM cases from 3 different cohorts (Cohort 1 ( n1 = 53 ), Cohort 2 ( n2 = 75 ), and Cohort 3 ( n3 = 79 )), where each of these three cohorts was used as a training set for our model separately, and the other two cohorts were used for testing, independently, for each training experiment. When employing Cohort 1 for training, r-DepTH yielded Concordance indices (C-indices) of 0.7 and 0.65, hazard ratios (HR) and Confidence Intervals (CI) of 10 (6 - 19) and 5 (3 - 8) on Cohorts 2 and 3, respectively. Similarly, training on Cohort 2 yielded C-indices of 0.6 and 0.7, HR and CI of 1 (0.7 - 2) and 3 (2 - 5) on Cohorts 1 and 3, respectively. Finally, training on Cohort 3 yielded C-indices of 0.75 and 0.63, HR and CI of 24 (10 - 57) and 12 (6 - 21) on Cohorts 1 and 2, respectively. Our results show that r-DepTH descriptor may serve as a comprehensive and a robust MRI-based prognostic marker of disease aggressiveness and survival in solid tumors.
Collapse
|
31
|
Lao Y, Ruan D, Vassantachart A, Fan Z, Ye JC, Chang EL, Chin R, Kaprealian T, Zada G, Shiroishi MS, Sheng K, Yang W. Voxelwise Prediction of Recurrent High-Grade Glioma via Proximity Estimation-Coupled Multidimensional Support Vector Machine. Int J Radiat Oncol Biol Phys 2022; 112:1279-1287. [PMID: 34963559 PMCID: PMC8923952 DOI: 10.1016/j.ijrobp.2021.12.153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/28/2023]
Abstract
PURPOSE To provide early and localized glioblastoma (GBM) recurrence prediction, we introduce a novel postsurgery multiparametric magnetic resonance-based support vector machine (SVM) method coupling with stem cell niche (SCN) proximity estimation. METHODS AND MATERIALS This study used postsurgery magnetic resonance imaging (MRI) scans from 50 patients with recurrent GBM, obtained approximately 2 months before clinically diagnosed recurrence. The main prediction pipeline consisted of a proximity-based estimator to identify regions with high risk of recurrence (HRRs) and an SVM classifier to provide voxelwise prediction in HRRs. The HRRs were estimated using the weighted sum of inverse distances to 2 possible origins of recurrence-the SCN and the tumor cavity. Subsequently, multiparametric voxels (from T1, T1 contrast-enhanced, fluid-attenuated inversion recovery, T2, and apparent diffusion coefficient) within the HRR were grouped into recurrent (warped from the clinical diagnosis) and nonrecurrent subregions and fed into the proximity estimation-coupled SVM classifier (SVMPE). The cohort was randomly divided into 40% and 60% for training and testing, respectively. The trained SVMPE was then extrapolated to an earlier time point for earlier recurrence prediction. As an exploratory analysis, the SVMPE predictive cluster sizes and the image intensities from the 5 magnetic resonance sequences were compared across time to assess the progressive subclinical traces. RESULTS On 2-month prerecurrence MRI scans from 30 test cohort patients, the SVMPE classifier achieved a recall of 0.80, a precision of 0.69, an F1-score of 0.73, and a mean boundary distance of 7.49 mm. Exploratory analysis at early time points showed spatially consistent but significantly smaller subclinical clusters and significantly increased T1 contrast-enhanced and apparent diffusion coefficient values over time. CONCLUSIONS We demonstrated a novel voxelwise early prediction method, SVMPE, for GBM recurrence based on clinical follow-up MR scans. The SVMPE is promising in localizing subclinical traces of recurrence 2 months ahead of clinical diagnosis and may be used to guide more effective personalized early salvage therapy.
Collapse
Affiliation(s)
- Yi Lao
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Dan Ruan
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - April Vassantachart
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Zhaoyang Fan
- Department of Radiology, Keck School of Medicine of USC, Los Angeles, USA
| | - Jason C. Ye
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Eric L. Chang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| | - Robert Chin
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Tania Kaprealian
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Gabriel Zada
- Department of Neurosurgery, Keck School of Medicine of USC, Los Angeles, USA
| | - Mark S Shiroishi
- Department of Radiology, Keck School of Medicine of USC, Los Angeles, USA
| | - Ke Sheng
- Department of Radiation Oncology, University of California - Los Angeles, USA
| | - Wensha Yang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, USA
| |
Collapse
|
32
|
Kumar M, Nanga RPR, Verma G, Wilson N, Brisset JC, Nath K, Chawla S. Emerging MR Imaging and Spectroscopic Methods to Study Brain Tumor Metabolism. Front Neurol 2022; 13:789355. [PMID: 35370872 PMCID: PMC8967433 DOI: 10.3389/fneur.2022.789355] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) provides a non-invasive biochemical profile of brain tumors. The conventional 1H-MRS methods present a few challenges mainly related to limited spatial coverage and low spatial and spectral resolutions. In the recent past, the advent and development of more sophisticated metabolic imaging and spectroscopic sequences have revolutionized the field of neuro-oncologic metabolomics. In this review article, we will briefly describe the scientific premises of three-dimensional echoplanar spectroscopic imaging (3D-EPSI), two-dimensional correlation spectroscopy (2D-COSY), and chemical exchange saturation technique (CEST) MRI techniques. Several published studies have shown how these emerging techniques can significantly impact the management of patients with glioma by determining histologic grades, molecular profiles, planning treatment strategies, and assessing the therapeutic responses. The purpose of this review article is to summarize the potential clinical applications of these techniques in studying brain tumor metabolism.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Ravi Prakash Reddy Nanga
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Gaurav Verma
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Neil Wilson
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | | | - Kavindra Nath
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Sanjeev Chawla
| |
Collapse
|
33
|
Xu K, Ramesh K, Huang V, Gurbani SS, Cordova JS, Schreibmann E, Weinberg BD, Sengupta S, Voloschin AD, Holdhoff M, Barker PB, Kleinberg LR, Olson JJ, Shu HKG, Shim H. Final Report on Clinical Outcomes and Tumor Recurrence Patterns of a Pilot Study Assessing Efficacy of Belinostat (PXD-101) with Chemoradiation for Newly Diagnosed Glioblastoma. Tomography 2022; 8:688-700. [PMID: 35314634 PMCID: PMC8938806 DOI: 10.3390/tomography8020057] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is highly aggressive and has a poor prognosis. Belinostat is a histone deacetylase inhibitor with blood-brain barrier permeability, anti-GBM activity, and the potential to enhance chemoradiation. The purpose of this clinical trial was to assess the efficacy of combining belinostat with standard-of-care therapy. Thirteen patients were enrolled in each of control and belinostat cohorts. The belinostat cohort was given a belinostat regimen (500-750 mg/m2 1×/day × 5 days) every three weeks (weeks 0, 3, and 6 of RT). All patients received temozolomide and radiation therapy (RT). RT margins of 5-10 mm were added to generate clinical tumor volumes and 3 mm added to create planning target volumes. Median overall survival (OS) was 15.8 months for the control cohort and 18.5 months for the belinostat cohort (p = 0.53). The recurrence volumes (rGTVs) for the control cohort occurred in areas that received higher radiation doses than that in the belinostat cohort. For those belinostat patients who experienced out-of-field recurrence, tumors were detectable by spectroscopic MRI before RT. Recurrence analysis suggests better in-field control with belinostat. This study highlights the potential of belinostat as a synergistic therapeutic agent for GBM. It may be particularly beneficial to combine this radio-sensitizing effect with spectroscopic MRI-guided RT.
Collapse
Affiliation(s)
- Karen Xu
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
| | - Karthik Ramesh
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Saumya S. Gurbani
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - James Scott Cordova
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA;
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Soma Sengupta
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; (S.S.); (A.D.V.)
| | - Alfredo D. Voloschin
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; (S.S.); (A.D.V.)
| | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Jeffrey J. Olson
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
- Department of Neurosurgery, Emory University, Atlanta, GA 30322, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, USA; (K.X.); (K.R.); (V.H.); (S.S.G.); (J.S.C.); (E.S.)
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA;
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| |
Collapse
|
34
|
Tang PLY, Méndez Romero A, Jaspers JPM, Warnert EAH. The potential of advanced MR techniques for precision radiotherapy of glioblastoma. MAGMA (NEW YORK, N.Y.) 2022; 35:127-143. [PMID: 35129718 PMCID: PMC8901515 DOI: 10.1007/s10334-021-00997-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 06/14/2023]
Abstract
As microscopic tumour infiltration of glioblastomas is not visible on conventional magnetic resonance (MR) imaging, an isotropic expansion of 1-2 cm around the visible tumour is applied to define the clinical target volume for radiotherapy. An opportunity to visualize microscopic infiltration arises with advanced MR imaging. In this review, various advanced MR biomarkers are explored that could improve target volume delineation for radiotherapy of glioblastomas. Various physiological processes in glioblastomas can be visualized with different advanced MR techniques. Combining maps of oxygen metabolism (CMRO2), relative cerebral blood volume (rCBV), vessel size imaging (VSI), and apparent diffusion coefficient (ADC) or amide proton transfer (APT) can provide early information on tumour infiltration and high-risk regions of future recurrence. Oxygen consumption is increased 6 months prior to tumour progression being visible on conventional MR imaging. However, presence of the Warburg effect, marking a switch from an infiltrative to a proliferative phenotype, could result in CMRO2 to appear unaltered in high-risk regions. Including information on biomarkers representing angiogenesis (rCBV and VSI) and hypercellularity (ADC) or protein concentration (APT) can omit misinterpretation due to the Warburg effect. Future research should evaluate these biomarkers in radiotherapy planning to explore the potential of advanced MR techniques to personalize target volume delineation with the aim to improve local tumour control and/or reduce radiation-induced toxicity.
Collapse
Affiliation(s)
- Patrick L Y Tang
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | - Alejandra Méndez Romero
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Jaap P M Jaspers
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Esther A H Warnert
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Ramesh K, Mellon EA, Gurbani SS, Weinberg BD, Schreibmann E, Sheriff SA, Goryawala M, de le Fuente M, Eaton BR, Zhong J, Voloschin AD, Sengupta S, Dunbar EM, Holdhoff M, Barker PB, Maudsley AA, Kleinberg LR, Shim H, Shu HKG. A multi-institutional pilot clinical trial of spectroscopic MRI-guided radiation dose escalation for newly diagnosed glioblastoma. Neurooncol Adv 2022; 4:vdac006. [PMID: 35382436 PMCID: PMC8976280 DOI: 10.1093/noajnl/vdac006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Glioblastomas (GBMs) are aggressive brain tumors despite radiation therapy (RT) to 60 Gy and temozolomide (TMZ). Spectroscopic magnetic resonance imaging (sMRI), which measures levels of specific brain metabolites, can delineate regions at high risk for GBM recurrence not visualized on contrast-enhanced (CE) MRI. We conducted a clinical trial to assess the feasibility, safety, and efficacy of sMRI-guided RT dose escalation to 75 Gy for newly diagnosed GBMs. Methods Our pilot trial (NCT03137888) enrolled patients at 3 institutions (Emory University, University of Miami, Johns Hopkins University) from September 2017 to June 2019. For RT, standard tumor volumes based on T2-FLAIR and T1w-CE MRIs with margins were treated in 30 fractions to 50.1 and 60 Gy, respectively. An additional high-risk volume based on residual CE tumor and Cho/NAA (on sMRI) ≥2× normal was treated to 75 Gy. Survival curves were generated by the Kaplan-Meier method. Toxicities were assessed according to CTCAE v4.0. Results Thirty patients were treated in the study. The median age was 59 years. 30% were MGMT promoter hypermethylated; 7% harbored IDH1 mutation. With a median follow-up of 21.4 months for censored patients, median overall survival (OS) and progression-free survival were 23.0 and 16.6 months, respectively. This regimen appeared well-tolerated with 70% of grade 3 or greater toxicity ascribed to TMZ and 23% occurring at least 1 year after RT. Conclusion Dose-escalated RT to 75 Gy guided by sMRI appears feasible and safe for patients with newly diagnosed GBMs. OS outcome is promising and warrants additional testing. Based on these results, a randomized phase II trial is in development.
Collapse
Affiliation(s)
- Karthik Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eric A Mellon
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Saumya S Gurbani
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | - Bree R Eaton
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jim Zhong
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alfredo D Voloschin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Soma Sengupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Present affiliation: Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter B Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Lawrence R Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA,Corresponding Authors: Hyunsuk Shim, PhD and Hui-Kuo G. Shu, MD, PhD, Department of Radiation Oncology, Winship Cancer Institute of Emory University, 1701 Uppergate Drive, Atlanta, GA 30322, USA (. )
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Ekici S, Nye J, Neill S, Allen J, Shu HK, Fleischer C. Glutamine Imaging: A New Avenue for Glioma Management. AJNR Am J Neuroradiol 2022; 43:11-18. [PMID: 34737183 PMCID: PMC8757564 DOI: 10.3174/ajnr.a7333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/04/2021] [Indexed: 01/03/2023]
Abstract
The glutamine pathway is emerging as an important marker of cancer prognosis and a target for new treatments. In gliomas, the most common type of brain tumors, metabolic reprogramming leads to abnormal consumption of glutamine as an energy source, and increased glutamine concentrations are associated with treatment resistance and proliferation. A key challenge in the development of glutamine-based biomarkers and therapies is the limited number of in vivo tools to noninvasively assess local glutamine metabolism and monitor its changes. In this review, we describe the importance of glutamine metabolism in gliomas and review the current landscape of translational and emerging imaging techniques to measure glutamine in the brain. These techniques include MRS, PET, SPECT, and preclinical methods such as fluorescence and mass spectrometry imaging. Finally, we discuss the roadblocks that must be overcome before incorporating glutamine into a personalized approach for glioma management.
Collapse
Affiliation(s)
- S. Ekici
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.)
| | - J.A. Nye
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.)
| | - S.G. Neill
- Pathology and Laboratory Medicine (S.G.N.), Emory University School of Medicine, Atlanta, Georgia
| | - J.W. Allen
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.),Neurology (J.W.A.), Emory University School of Medicine, Atlanta, Georgia
| | - H.-K. Shu
- Radiation Oncology (H.-K.S.), Emory University School of Medicine, Atlanta, Georgia
| | - C.C. Fleischer
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.),Wallace H. Coulter Department of Biomedical Engineering (C.C.F.), Geogria Institute of Technology and Emory University, Atlanta, Georgia
| |
Collapse
|
37
|
Advanced Imaging and Computational Techniques for the Diagnostic and Prognostic Assessment of Malignant Gliomas. Cancer J 2021; 27:344-352. [PMID: 34570448 DOI: 10.1097/ppo.0000000000000545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Advanced imaging techniques provide a powerful tool to assess the intratumoral and intertumoral heterogeneity of gliomas. Advances in the molecular understanding of glioma subgroups may allow improved diagnostic assessment combining imaging and molecular tumor features, with enhanced prognostic utility and implications for patient treatment. In this article, a comprehensive overview of the physiologic basis for conventional and advanced imaging techniques is presented, and clinical applications before and after treatment are discussed. An introduction to the principles of radiomics and the advanced integration of imaging, clinical outcomes, and genomic data highlights the future potential for this field of research to better stratify and select patients for standard as well as investigational therapies.
Collapse
|
38
|
Hampton DG, Goldman-Yassen AE, Sun PZ, Hu R. Metabolic Magnetic Resonance Imaging in Neuroimaging: Magnetic Resonance Spectroscopy, Sodium Magnetic Resonance Imaging and Chemical Exchange Saturation Transfer. Semin Ultrasound CT MR 2021; 42:452-462. [PMID: 34537114 DOI: 10.1053/j.sult.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Magnetic resonance (MR) is a powerful and versatile technique that offers much more beyond conventional anatomic imaging and has the potential of probing in vivo metabolism. Although MR spectroscopy (MRS) predates clinical MR imaging (MRI), its clinical application has been limited by technical and practical challenges. Other MR techniques actively being developed for in vivo metabolic imaging include sodium concentration imaging and chemical exchange saturation transfer. This article will review some of the practical aspects of MRS in neuroimaging, introduce sodium MRI and chemical exchange saturation transfer MRI, and highlight some of their emerging clinical applications.
Collapse
Affiliation(s)
- Daniel G Hampton
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA.
| | - Adam E Goldman-Yassen
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Phillip Zhe Sun
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA; Yerkes Imaging Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA
| | - Ranliang Hu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
39
|
Li M, Zhang Q, Yang K. Role of MRI-Based Functional Imaging in Improving the Therapeutic Index of Radiotherapy in Cancer Treatment. Front Oncol 2021; 11:645177. [PMID: 34513659 PMCID: PMC8429950 DOI: 10.3389/fonc.2021.645177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 07/30/2021] [Indexed: 02/05/2023] Open
Abstract
Advances in radiation technology, such as intensity-modulated radiation therapy (IMRT), have largely enabled a biological dose escalation of the target volume (TV) and reduce the dose to adjacent tissues or organs at risk (OARs). However, the risk of radiation-induced injury increases as more radiation dose utilized during radiation therapy (RT), which predominantly limits further increases in TV dose distribution and reduces the local control rate. Thus, the accurate target delineation is crucial. Recently, technological improvements for precise target delineation have obtained more attention in the field of RT. The addition of functional imaging to RT can provide a more accurate anatomy of the tumor and normal tissues (such as location and size), along with biological information that aids to optimize the therapeutic index (TI) of RT. In this review, we discuss the application of some common MRI-based functional imaging techniques in clinical practice. In addition, we summarize the main challenges and prospects of these imaging technologies, expecting more inspiring developments and more productive research paths in the near future.
Collapse
Affiliation(s)
- Mei Li
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Zhang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Kaixuan Yang
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Wang C, Padgett KR, Su MY, Mellon EA, Maziero D, Chang Z. Multi-parametric MRI (mpMRI) for treatment response assessment of radiation therapy. Med Phys 2021; 49:2794-2819. [PMID: 34374098 DOI: 10.1002/mp.15130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 11/11/2022] Open
Abstract
Magnetic resonance imaging (MRI) plays an important role in the modern radiation therapy (RT) workflow. In comparison with computed tomography (CT) imaging, which is the dominant imaging modality in RT, MRI possesses excellent soft-tissue contrast for radiographic evaluation. Based on quantitative models, MRI can be used to assess tissue functional and physiological information. With the developments of scanner design, acquisition strategy, advanced data analysis, and modeling, multiparametric MRI (mpMRI), a combination of morphologic and functional imaging modalities, has been increasingly adopted for disease detection, localization, and characterization. Integration of mpMRI techniques into RT enriches the opportunities to individualize RT. In particular, RT response assessment using mpMRI allows for accurate characterization of both tissue anatomical and biochemical changes to support decision-making in monotherapy of radiation treatment and/or systematic cancer management. In recent years, accumulating evidence have, indeed, demonstrated the potentials of mpMRI in RT response assessment regarding patient stratification, trial benchmarking, early treatment intervention, and outcome modeling. Clinical application of mpMRI for treatment response assessment in routine radiation oncology workflow, however, is more complex than implementing an additional imaging protocol; mpMRI requires additional focus on optimal study design, practice standardization, and unified statistical reporting strategy to realize its full potential in the context of RT. In this article, the mpMRI theories, including image mechanism, protocol design, and data analysis, will be reviewed with a focus on the radiation oncology field. Representative works will be discussed to demonstrate how mpMRI can be used for RT response assessment. Additionally, issues and limits of current works, as well as challenges and potential future research directions, will also be discussed.
Collapse
Affiliation(s)
- Chunhao Wang
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Kyle R Padgett
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA.,Department of Radiology, University of Miami, Miami, Florida, USA
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, California, USA.,Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Eric A Mellon
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Danilo Maziero
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Zheng Chang
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
41
|
Kim MM, Sun Y, Aryal MP, Parmar HA, Piert M, Rosen B, Mayo CS, Balter JM, Schipper M, Gabel N, Briceño EM, You D, Heth J, Al-Holou W, Umemura Y, Leung D, Junck L, Wahl DR, Lawrence TS, Cao Y. A Phase 2 Study of Dose-intensified Chemoradiation Using Biologically Based Target Volume Definition in Patients With Newly Diagnosed Glioblastoma. Int J Radiat Oncol Biol Phys 2021; 110:792-803. [PMID: 33524546 PMCID: PMC8920120 DOI: 10.1016/j.ijrobp.2021.01.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE We hypothesized that dose-intensified chemoradiation therapy targeting adversely prognostic hypercellular (TVHCV) and hyperperfused (TVCBV) tumor volumes would improve outcomes in patients with glioblastoma. METHODS AND MATERIALS This single-arm, phase 2 trial enrolled adult patients with newly diagnosed glioblastoma. Patients with a TVHCV/TVCBV >1 cm3, identified using high b-value diffusion-weighted magnetic resonance imaging (MRI) and dynamic contrast-enhanced perfusion MRI, were treated over 30 fractions to 75 Gy to the TVHCV/TVCBV with temozolomide. The primary objective was to estimate improvement in 12-month overall survival (OS) versus historical control. Secondary objectives included evaluating the effect of 3-month TVHCV/TVCBV reduction on OS using Cox proportional-hazard regression and characterizing coverage (95% isodose line) of metabolic tumor volumes identified using correlative 11C-methionine positron emission tomography. Clinically meaningful change was assessed for quality of life by the European Organisation for the Research and Treatment of Cancer Quality of Life Questionnaire C30, for symptom burden by the MD Anderson Symptom Inventory for brain tumor, and for neurocognitive function (NCF) by the Controlled Oral Word Association Test, the Trail Making Test, parts A and B, and the Hopkins Verbal Learning Test-Revised. RESULTS Between 2016 and 2018, 26 patients were enrolled. Initial patients were boosted to TVHCV alone, and 13 patients were boosted to both TVHCV/TVCBV. Gross or subtotal resection was performed in 87% of patients; 22% were O6-methylguanine-DNA methyltransferase (MGMT) methylated. With 26-month follow-up (95% CI, 19-not reached), the 12-month OS rate among patients boosted to the combined TVHCV/TVCBV was 92% (95% CI, 78%-100%; P = .03) and the median OS was 20 months (95% CI, 18-not reached); the median OS for the whole study cohort was 20 months (95% CI, 14-29 months). Patients whose 3-month TVHCV/TVCBV decreased to less than the median volume (3 cm3) had superior OS (29 vs 12 months; P = .02). Only 5 patients had central or in-field failures, and 93% (interquartile range, 59%-100%) of the 11C-methionine metabolic tumor volumes received high-dose coverage. Late grade 3 neurologic toxicity occurred in 2 patients. Among non-progressing patients, 1-month and 7-month deterioration in quality of life, symptoms, and NCF were similar in incidence to standard therapy. CONCLUSIONS Dose intensification against hypercellular/hyperperfused tumor regions in glioblastoma yields promising OS with favorable outcomes for NCF, symptom burden, and quality of life, particularly among patients with greater tumor reduction 3 months after radiation therapy.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Madhava P Aryal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Hemant A Parmar
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Morand Piert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin Rosen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Charles S Mayo
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - James M Balter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Nicolette Gabel
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, Michigan
| | - Emily M Briceño
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, Michigan
| | - Daekeun You
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Jason Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Wajd Al-Holou
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Denise Leung
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Department of Radiology, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
42
|
Zhong J, Huang V, Gurbani SS, Ramesh K, Scott Cordova J, Schreibmann E, Shu HKG, Olson J, Han H, Giuffrida A, Shim H, Weinberg BD. 3D whole-brain metabolite imaging to improve characterization of low-to-intermediate grade gliomas. J Neurooncol 2021; 153:303-311. [PMID: 33983570 PMCID: PMC8237861 DOI: 10.1007/s11060-021-03770-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE MRI is the standard imaging modality used for diagnosis, treatment planning, and post-treatment management of gliomas. Contrast-enhanced T1-weighted (CE-T1w) MRI is used to plan biopsy and radiation for grade IV gliomas but is less effective for grade II and III gliomas (i.e., low-to-intermediate grade gliomas) which may have minimal or no enhancement. Magnetic resonance spectroscopic imaging (MRSI) is an advanced MRI technique that has been shown, to improve diagnostic yield of biopsy and target delineation for grade IV glioma. The purpose of this study is to determine if MRSI can improve characterization and tissue sampling of low-to-intermediate grade gliomas. METHODS Prospective grade II and grade III glioma patients were enrolled to undergo whole brain high-resolution MRSI prior to tissue sampling. Choline/N-acetyl-aspartate (Cho/NAA) maps were overlaid on anatomic imaging and imported into stereotactic biopsy software. Patients were treated with standard-of-care surgery and radiation. Volumes of spectroscopically abnormal tissue were generated and compared with anatomic imaging and areas of enhancing recurrence on follow-up imaging. RESULTS Ten patients had pathologic diagnosis of grade II (n = 4) or grade III (n = 6) with a median follow-up of 27.3 months. Five patients had recurrence, and regions of recurrence were found to overlap with metabolically abnormal regions on MRSI at the time of diagnosis. CONCLUSION MRSI in low-to-intermediate grade glioma patients is predictive of areas of subsequent recurrence. Larger studies are needed to determine if MRSI can be used to guide surgical and radiation treatment planning in these patients.
Collapse
Affiliation(s)
- Jim Zhong
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Vicki Huang
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Saumya S Gurbani
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Karthik Ramesh
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - J Scott Cordova
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA
| | - Jeffrey Olson
- Department of Neurosurgery, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hui Han
- Biomedical Sciences and Biomedical Imaging Research Institute, Cedars Sinai, Los Angeles, CA, 90048, USA
| | - Alexander Giuffrida
- Department of Biomedical Engineering, Winship Cancer Institute of Emory University, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Winship Cancer Institute of Emory University, Emory University School of Medicine, 1701 Uppergate Drive, C5018, Atlanta, GA, 30322, USA.
| |
Collapse
|
43
|
Weinberg BD, Kuruva M, Shim H, Mullins ME. Clinical Applications of Magnetic Resonance Spectroscopy in Brain Tumors: From Diagnosis to Treatment. Radiol Clin North Am 2021; 59:349-362. [PMID: 33926682 PMCID: PMC8272438 DOI: 10.1016/j.rcl.2021.01.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Magnetic resonance spectroscopy (MRS) is a valuable tool for imaging brain tumors, primarily as an adjunct to conventional imaging and clinical presentation. MRS is useful in initial diagnosis of brain tumors, helping differentiate tumors from possible mimics such as metastatic disease, lymphoma, demyelination, and infection, as well as in the subsequent follow-up of patients after resection and chemoradiation. Unfortunately, the spectroscopic appearance of many pathologies can overlap, and ultimately follow-up or biopsy may be required to make a definitive diagnosis. Future developments may continue to increase the value of MRS for initial diagnosis, treatment planning, and early detection of recurrence.
Collapse
Affiliation(s)
- Brent D Weinberg
- Radiology and Imaging Sciences, Emory University, 1364 Clifton Road Northeast BG20, Atlanta, GA 30322, USA.
| | - Manohar Kuruva
- Radiology and Imaging Sciences, Emory University, 1364 Clifton Road Northeast BG20, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Radiation Oncology, Emory University, 1365 Clifton Road Northeast, Atlanta, GA 30322, USA
| | - Mark E Mullins
- Radiology and Imaging Sciences, Emory University, 1364 Clifton Road Northeast BG20, Atlanta, GA 30322, USA
| |
Collapse
|
44
|
Maudsley AA, Andronesi OC, Barker PB, Bizzi A, Bogner W, Henning A, Nelson SJ, Posse S, Shungu DC, Soher BJ. Advanced magnetic resonance spectroscopic neuroimaging: Experts' consensus recommendations. NMR IN BIOMEDICINE 2021; 34:e4309. [PMID: 32350978 PMCID: PMC7606742 DOI: 10.1002/nbm.4309] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 02/01/2020] [Accepted: 03/10/2020] [Indexed: 05/04/2023]
Abstract
Magnetic resonance spectroscopic imaging (MRSI) offers considerable promise for monitoring metabolic alterations associated with disease or injury; however, to date, these methods have not had a significant impact on clinical care, and their use remains largely confined to the research community and a limited number of clinical sites. The MRSI methods currently implemented on clinical MRI instruments have remained essentially unchanged for two decades, with only incremental improvements in sequence implementation. During this time, a number of technological developments have taken place that have already greatly benefited the quality of MRSI measurements within the research community and which promise to bring advanced MRSI studies to the point where the technique becomes a true imaging modality, while making the traditional review of individual spectra a secondary requirement. Furthermore, the increasing use of biomedical MR spectroscopy studies has indicated clinical areas where advanced MRSI methods can provide valuable information for clinical care. In light of this rapidly changing technological environment and growing understanding of the value of MRSI studies for biomedical studies, this article presents a consensus from a group of experts in the field that reviews the state-of-the-art for clinical proton MRSI studies of the human brain, recommends minimal standards for further development of vendor-provided MRSI implementations, and identifies areas which need further technical development.
Collapse
Affiliation(s)
- Andrew A Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ovidiu C Andronesi
- Department of Radiology, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, Massachusetts
| | - Peter B Barker
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, and the Kennedy Krieger Institute, F.M. Kirby Center for Functional Brain Imaging, Baltimore, Maryland
| | - Alberto Bizzi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University Vienna, Vienna, Austria
| | - Anke Henning
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Stefan Posse
- Department of Neurology, University of New Mexico, Albuquerque, New Mexico
| | - Dikoma C Shungu
- Department of Neuroradiology, Weill Cornell Medical College, New York, New York
| | - Brian J Soher
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
45
|
Wala K, Szlasa W, Saczko J, Rudno-Rudzińska J, Kulbacka J. Modulation of Blood-Brain Barrier Permeability by Activating Adenosine A2 Receptors in Oncological Treatment. Biomolecules 2021; 11:biom11050633. [PMID: 33923147 PMCID: PMC8146369 DOI: 10.3390/biom11050633] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
The blood–brain barrier (BBB) plays an important protective role in the central nervous system and maintains its homeostasis. It regulates transport into brain tissue and protects neurons against the toxic effects of substances circulating in the blood. However, in the case of neurological diseases or primary brain tumors, i.e., gliomas, the higher permeability of the blood-derived substances in the brain tissue is necessary. Currently applied methods of treatment for the primary brain neoplasms include surgical removal of the tumor, radiation therapy, and chemotherapy. Despite the abovementioned treatment methods, the prognosis of primary brain tumors remains bad. Moreover, chemotherapy options seem to be limited due to low drug penetration into the cancerous tissue. Modulation of the blood–brain barrier permeability may contribute to an increase in the concentration of the drug in the CNS and thus increase the effectiveness of therapy. Interestingly, endothelial cells in cerebral vessels are characterized by the presence of adenosine 2A receptors (A2AR). It has been shown that substances affecting these receptors regulate the permeability of the BBB. The mechanism of increasing the BBB permeability by A2AR agonists is the actin-cytoskeletal reorganization and acting on the tight junctions. In this case, the A2AR seems to be a promising therapy target. This article aims to assess the possibility of increasing the BBB permeability through A2AR agonists to increase the effectiveness of chemotherapy and to improve the results of cancer therapy.
Collapse
Affiliation(s)
- Kamila Wala
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julia Rudno-Rudzińska
- Department of General and Oncological Surgery, Medical University Hospital, Borowska 213, 50-556 Wrocław, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-784-06-92
| |
Collapse
|
46
|
Zhvansky E, Sorokin A, Shurkhay V, Zavorotnyuk D, Bormotov D, Pekov S, Potapov A, Nikolaev E, Popov I. Comparison of Dimensionality Reduction Methods in Mass Spectra of Astrocytoma and Glioblastoma Tissues. Mass Spectrom (Tokyo) 2021; 10:A0094. [PMID: 33747696 PMCID: PMC7953827 DOI: 10.5702/massspectrometry.a0094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/21/2021] [Indexed: 11/24/2022] Open
Abstract
Recently developed methods of ambient ionization allow the collection of mass spectrometric datasets for biological and medical applications at an unprecedented pace. One of the areas that could employ such analysis is neurosurgery. The fast in situ identification of dissected tissues could assist the neurosurgery procedure. In this paper tumor tissues of astrocytoma and glioblastoma are compared. The vast majority of the data representation methods are hard to use, as the number of features is high and the amount of samples is limited. Furthermore, the ratio of features and samples number restricts the use of many machine learning methods. The number of features could be reduced through feature selection algorithms or dimensionality reduction methods. Different algorithms of dimensionality reduction are considered along with the traditional noise thresholding for the mass spectra. From our analysis, the Isomap algorithm appears to be the most effective dimensionality reduction algorithm for negative mode, whereas the positive mode could be processed with a simple noise reduction by a threshold. Also, negative and positive mode correspond to different sample properties: negative mode is responsible for the inner variability and the details of the sample, whereas positive mode describes measurement in general.
Collapse
Affiliation(s)
- Evgeny Zhvansky
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
| | - Anatoly Sorokin
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
- Institute of Cell Biophysics RAS, Pushchino, Russian
Federation
- Institute of Systems, Molecular and Integrative Biology,
University of Liverpool, Liverpool, UK
| | - Vsevolod Shurkhay
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
- Federal State Autonomous Institution «N.N. Burdenko
National Scientific and Practical Center for Neurosurgery» of the Ministry of
Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Denis Zavorotnyuk
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
| | - Denis Bormotov
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
| | - Stanislav Pekov
- N.N. Semenov Federal Research Center of Chemical Physics
Russian Academy of Sciences, Moscow, Russian Federation
| | - Alexander Potapov
- Federal State Autonomous Institution «N.N. Burdenko
National Scientific and Practical Center for Neurosurgery» of the Ministry of
Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Evgeny Nikolaev
- Skolkovo Institute of Science and Technology, Moscow,
Russian Federation
| | - Igor Popov
- Moscow Institute of Physics and Technology, Dolgoprudny,
Moscow Region, Russian Federation
| |
Collapse
|
47
|
Maziero D, Straza MW, Ford JC, Bovi JA, Diwanji T, Stoyanova R, Paulson ES, Mellon EA. MR-Guided Radiotherapy for Brain and Spine Tumors. Front Oncol 2021; 11:626100. [PMID: 33763361 PMCID: PMC7982530 DOI: 10.3389/fonc.2021.626100] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
MRI is the standard modality to assess anatomy and response to treatment in brain and spine tumors given its superb anatomic soft tissue contrast (e.g., T1 and T2) and numerous additional intrinsic contrast mechanisms that can be used to investigate physiology (e.g., diffusion, perfusion, spectroscopy). As such, hybrid MRI and radiotherapy (RT) devices hold unique promise for Magnetic Resonance guided Radiation Therapy (MRgRT). In the brain, MRgRT provides daily visualizations of evolving tumors that are not seen with cone beam CT guidance and cannot be fully characterized with occasional standalone MRI scans. Significant evolving anatomic changes during radiotherapy can be observed in patients with glioblastoma during the 6-week fractionated MRIgRT course. In this review, a case of rapidly changing symptomatic tumor is demonstrated for possible therapy adaptation. For stereotactic body RT of the spine, MRgRT acquires clear isotropic images of tumor in relation to spinal cord, cerebral spinal fluid, and nearby moving organs at risk such as bowel. This visualization allows for setup reassurance and the possibility of adaptive radiotherapy based on anatomy in difficult cases. A review of the literature for MR relaxometry, diffusion, perfusion, and spectroscopy during RT is also presented. These techniques are known to correlate with physiologic changes in the tumor such as cellularity, necrosis, and metabolism, and serve as early biomarkers of chemotherapy and RT response correlating with patient survival. While physiologic tumor investigations during RT have been limited by the feasibility and cost of obtaining frequent standalone MRIs, MRIgRT systems have enabled daily and widespread physiologic measurements. We demonstrate an example case of a poorly responding tumor on the 0.35 T MRIgRT system with relaxometry and diffusion measured several times per week. Future studies must elucidate which changes in MR-based physiologic metrics and at which timepoints best predict patient outcomes. This will lead to early treatment intensification for tumors identified to have the worst physiologic responses during RT in efforts to improve glioblastoma survival.
Collapse
Affiliation(s)
- Danilo Maziero
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Michael W Straza
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John C Ford
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Joseph A Bovi
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tejan Diwanji
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Radka Stoyanova
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Eric S Paulson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Eric A Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
48
|
Castellano A, Bailo M, Cicone F, Carideo L, Quartuccio N, Mortini P, Falini A, Cascini GL, Minniti G. Advanced Imaging Techniques for Radiotherapy Planning of Gliomas. Cancers (Basel) 2021; 13:1063. [PMID: 33802292 PMCID: PMC7959155 DOI: 10.3390/cancers13051063] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
The accuracy of target delineation in radiation treatment (RT) planning of cerebral gliomas is crucial to achieve high tumor control, while minimizing treatment-related toxicity. Conventional magnetic resonance imaging (MRI), including contrast-enhanced T1-weighted and fluid-attenuated inversion recovery (FLAIR) sequences, represents the current standard imaging modality for target volume delineation of gliomas. However, conventional sequences have limited capability to discriminate treatment-related changes from viable tumors, owing to the low specificity of increased blood-brain barrier permeability and peritumoral edema. Advanced physiology-based MRI techniques, such as MR spectroscopy, diffusion MRI and perfusion MRI, have been developed for the biological characterization of gliomas and may circumvent these limitations, providing additional metabolic, structural, and hemodynamic information for treatment planning and monitoring. Radionuclide imaging techniques, such as positron emission tomography (PET) with amino acid radiopharmaceuticals, are also increasingly used in the workup of primary brain tumors, and their integration in RT planning is being evaluated in specialized centers. This review focuses on the basic principles and clinical results of advanced MRI and PET imaging techniques that have promise as a complement to RT planning of gliomas.
Collapse
Affiliation(s)
- Antonella Castellano
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Michele Bailo
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
| | - Luciano Carideo
- National Cancer Institute, G. Pascale Foundation, 80131 Naples, Italy;
| | - Natale Quartuccio
- A.R.N.A.S. Ospedale Civico Di Cristina Benfratelli, 90144 Palermo, Italy;
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Andrea Falini
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Giuseppe Lucio Cascini
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, 53100 Siena, Italy;
- IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| |
Collapse
|
49
|
Evaluation of interim MRI changes during limited-field radiation therapy for glioblastoma and implications for treatment planning. Radiother Oncol 2021; 158:237-243. [PMID: 33587967 DOI: 10.1016/j.radonc.2021.01.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/10/2021] [Accepted: 01/29/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND PURPOSE Consensus for defining gross tumor volume (GTV) and clinical target volume (CTV) for limited-field radiation therapy (LFRT) of GBM are not well established. We leveraged a department MRI simulator to image patients before and during LFRT to address these questions. MATERIALS AND METHODS Supratentorial GBM patients receiving LFRT (46 Gy + boost to 60 Gy) underwent baseline MRI (MRI1) and interim MRI during RT (MRI2). GTV1 was defined as T1 enhancement + surgical cavity on MRI1 without routine inclusion of T2 abnormality (unless tumor did not enhance). The initial CTV margin was 15 mm from GTV1, and the boost CTV margin was 5-7 mm. The GTV1 characteristics were categorized into three groups: identical T1 and T2 abnormality (Group A), T1 only with larger T2 abnormality not included (Group B), and T2 abnormality when tumor lacked enhancement (Group C). GTV2 was contoured on MRI2 and compared with GTV1 plus 5-15 mm expansions. RESULTS Among 120 patients treated from 2014-2019, 29 patients (24%) underwent replanning based on MRI2. On MRI2, 84% of GTV2 were covered by GTV1 + 5 mm, 93% by GTV1 + 7 mm, and 98% by GTV1 + 15 mm. On MRI1, 43% of GTV1 could be categorized into Group A, 39% Group B, and 18% Group C. Group B's patterns of failure, local control, or progression-free survival were similar to Group A/C. CONCLUSIONS Initial CTV margin of 15 mm followed by a boost CTV margin of 7 mm is a reasonable approach for LFRT of GBM. Omitting routine inclusion of T2 abnormality from GTV delineation may not jeopardize disease control.
Collapse
|
50
|
Reuter G, Moïse M, Roll W, Martin D, Lombard A, Scholtes F, Stummer W, Suero Molina E. Conventional and advanced imaging throughout the cycle of care of gliomas. Neurosurg Rev 2021; 44:2493-2509. [PMID: 33411093 DOI: 10.1007/s10143-020-01448-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
Although imaging of gliomas has evolved tremendously over the last decades, published techniques and protocols are not always implemented into clinical practice. Furthermore, most of the published literature focuses on specific timepoints in glioma management. This article reviews the current literature on conventional and advanced imaging techniques and chronologically outlines their practical relevance for the clinical management of gliomas throughout the cycle of care. Relevant articles were located through the Pubmed/Medline database and included in this review. Interpretation of conventional and advanced imaging techniques is crucial along the entire process of glioma care, from diagnosis to follow-up. In addition to the described currently existing techniques, we expect deep learning or machine learning approaches to assist each step of glioma management through tumor segmentation, radiogenomics, prognostication, and characterization of pseudoprogression. Thorough knowledge of the specific performance, possibilities, and limitations of each imaging modality is key for their adequate use in glioma management.
Collapse
Affiliation(s)
- Gilles Reuter
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium. .,GIGA-CRC In-vivo Imaging Center, ULiege, Liège, Belgium.
| | - Martin Moïse
- Department of Radiology, University Hospital of Liège, Liège, Belgium
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Didier Martin
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Arnaud Lombard
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium
| | - Félix Scholtes
- Department of Neurosurgery, University Hospital of Liège, Liège, Belgium.,Department of Neuroanatomy, University of Liège, Liège, Belgium
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, Münster, Germany
| |
Collapse
|