1
|
Kim AE, Lou KW, Giobbie-Hurder A, Chang K, Gidwani M, Hoebel K, Patel JB, Cleveland MC, Singh P, Bridge CP, Ahmed SR, Bearce BA, Liu W, Fuster-Garcia E, Lee EQ, Lin NU, Overmoyer B, Wen PY, Nayak L, Cohen JV, Dietrich J, Eichler A, Heist R, Krop I, Lawrence D, Ligibel J, Tolaney S, Mayer E, Winer E, Perrino CM, Summers EJ, Mahar M, Oh K, Shih HA, Cahill DP, Rosen BR, Yen YF, Kalpathy-Cramer J, Martinez-Lage M, Sullivan RJ, Brastianos PK, Emblem KE, Gerstner ER. Abnormal vascular structure and function within brain metastases is linked to pembrolizumab resistance. Neuro Oncol 2024; 26:965-974. [PMID: 38070147 PMCID: PMC11066943 DOI: 10.1093/neuonc/noad236] [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: 05/04/2024] Open
Abstract
BACKGROUND We recently conducted a phase 2 trial (NCT028865685) evaluating intracranial efficacy of pembrolizumab for brain metastases (BM) of diverse histologies. Our study met its primary efficacy endpoint and illustrates that pembrolizumab exerts promising activity in a select group of patients with BM. Given the importance of aberrant vasculature in mediating immunosuppression, we explored the relationship between immune checkpoint inhibitor (ICI) efficacy and vascular architecture in the hopes of identifying potential mechanisms of intracranial ICI response or resistance for BM. METHODS Using Vessel Architectural Imaging, a histologically validated quantitative metric for in vivo tumor vascular physiology, we analyzed dual-echo DSC/DCE MRI for 44 patients on trial. Tumor and peri-tumor cerebral blood volume/flow, vessel size, arterial and venous dominance, and vascular permeability were measured before and after treatment with pembrolizumab. RESULTS BM that progressed on ICI were characterized by a highly aberrant vasculature dominated by large-caliber vessels. In contrast, ICI-responsive BM possessed a more structurally balanced vasculature consisting of both small and large vessels, and there was a trend toward a decrease in under-perfused tissue, suggesting a reversal of the negative effects of hypoxia. In the peri-tumor region, the development of smaller blood vessels, consistent with neo-angiogenesis, was associated with tumor growth before radiographic evidence of contrast enhancement on anatomical MRI. CONCLUSIONS This study, one of the largest functional imaging studies for BM, suggests that vascular architecture is linked with ICI efficacy. Studies identifying modulators of vascular architecture, and effects on immune activity, are warranted and may inform future combination treatments.
Collapse
Affiliation(s)
- Albert E Kim
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kevin W Lou
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anita Giobbie-Hurder
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ken Chang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mishka Gidwani
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katharina Hoebel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jay B Patel
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mason C Cleveland
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Praveer Singh
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christopher P Bridge
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Syed Rakin Ahmed
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Benjamin A Bearce
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - William Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elies Fuster-Garcia
- Department of Physics and Computational Radiology, Division of Radiology & Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Instituto Universitario de Tecnologías de la Información y Comunicaciones, Universitat Politècnica de València, València, Spain
| | - Eudocia Q Lee
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Nancy U Lin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Beth Overmoyer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Lakshmi Nayak
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Justine V Cohen
- Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jorg Dietrich
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - April Eichler
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Heist
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ian Krop
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Donald Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Ligibel
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Tolaney
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Erica Mayer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Winer
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carmen M Perrino
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elizabeth J Summers
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Maura Mahar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin Oh
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Helen A Shih
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel P Cahill
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Maria Martinez-Lage
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Priscilla K Brastianos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Kyrre E Emblem
- Department of Physics and Computational Radiology, Division of Radiology & Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Elizabeth R Gerstner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Kim AE, Lou KW, Giobbie-Hurder A, Chang K, Gidwani M, Hoebel K, Patel JB, Cleveland MC, Singh P, Bridge CP, Ahmed SR, Bearce BA, Liu W, Fuster-Garcia E, Lee EQ, Lin NU, Overmoyer B, Wen PY, Nayak L, Cohen JV, Dietrich J, Eichler A, Heist R, Krop I, Lawrence D, Ligibel J, Tolaney S, Mayer E, Winer E, Perrino CM, Summers EJ, Mahar M, Oh K, Shih HA, Cahill DP, Rosen BR, Yen YF, Kalpathy-Cramer J, Martinez-Lage M, Sullivan RJ, Brastianos PK, Emblem KE, Gerstner ER. Structural and functional vascular dysfunction within brain metastases is linked to pembrolizumab inefficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554868. [PMID: 37693537 PMCID: PMC10491098 DOI: 10.1101/2023.08.25.554868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Structurally and functionally aberrant vasculature is a hallmark of tumor angiogenesis and treatment resistance. Given the synergistic link between aberrant tumor vasculature and immunosuppression, we analyzed perfusion MRI for 44 patients with brain metastases (BM) undergoing treatment with pembrolizumab. To date, vascular-immune communication, or the relationship between immune checkpoint inhibitor (ICI) efficacy and vascular architecture, has not been well-characterized in human imaging studies. We found that ICI-responsive BM possessed a structurally balanced vascular makeup, which was linked to improved vascular efficiency and an immune-stimulatory microenvironment. In contrast, ICI-resistant BM were characterized by a lack of immune cell infiltration and a highly aberrant vasculature dominated by large-caliber vessels. Peri-tumor region analysis revealed early functional changes predictive of ICI resistance before radiographic evidence on conventional MRI. This study was one of the largest functional imaging studies for BM and establishes a foundation for functional studies that illuminate the mechanisms linking patterns of vascular architecture with immunosuppression, as targeting these aspects of cancer biology may serve as the basis for future combination treatments.
Collapse
|
3
|
Velikina JV, Jung Y, Field AS, Samsonov AA. High-resolution dynamic susceptibility contrast perfusion imaging using higher-order temporal smoothness regularization. Magn Reson Med 2023; 89:112-127. [PMID: 36198002 PMCID: PMC9617779 DOI: 10.1002/mrm.29425] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE To improve image quality and resolution of dynamic susceptibility contrast perfusion weighted imaging (DSC-PWI) by developing acquisition and reconstruction methods exploiting the temporal regularity property of DSC-PWI signal. THEORY AND METHODS A novel regularized reconstruction is proposed that recovers DSC-PWI series from interleaved segmented spiral k-space acquisition using higher order temporal smoothness (HOTS) properties of the DSC-PWI signal. The HOTS regularization is designed to tackle representational insufficiency of the standard first-order temporal regularizations for supporting higher accelerations. The higher accelerations allow for k-space coverage with shorter spiral interleaves resulting in improved acquisition point spread function, and acquisition of images at multiple TEs for more accurate DSC-PWI analysis. RESULTS The methods were evaluated in simulated and in-vivo studies. HOTS regularization provided increasingly more accurate models for DSC-PWI than the standard first-order methods with either quadratic or robust norms at the expense of increased noise. HOTS DSC-PWI optimized for noise and accuracy demonstrated significant advantages over both spiral DSC-PWI without temporal regularization and traditional echo-planar DSC-PWI, improving resolution and mitigating image artifacts associated with long readout, including blurring and geometric distortions. In context of multi-echo DSC-PWI, the novel methods allowed ∼4.3× decrease of voxel volume, providing 2× number of TEs compared to the previously published results. CONCLUSIONS Proposed HOTS reconstruction combined with dynamic spiral sampling represents a valid mechanism for improving image quality and resolution of DSC-PWI significantly beyond those available with established fast imaging techniques.
Collapse
Affiliation(s)
- Julia V. Velikina
- Department of RadiologyUniversity of Wisconsin‐Madison
MadisonWisconsinUSA
| | - Youngkyoo Jung
- Department of RadiologyUniversity of California‐DavisDavisCaliforniaUSA
| | - Aaron S. Field
- Department of RadiologyUniversity of Wisconsin‐Madison
MadisonWisconsinUSA
| | - Alexey A. Samsonov
- Department of RadiologyUniversity of Wisconsin‐Madison
MadisonWisconsinUSA
| |
Collapse
|
4
|
Henriksen OM, del Mar Álvarez-Torres M, Figueiredo P, Hangel G, Keil VC, Nechifor RE, Riemer F, Schmainda KM, Warnert EAH, Wiegers EC, Booth TC. High-Grade Glioma Treatment Response Monitoring Biomarkers: A Position Statement on the Evidence Supporting the Use of Advanced MRI Techniques in the Clinic, and the Latest Bench-to-Bedside Developments. Part 1: Perfusion and Diffusion Techniques. Front Oncol 2022; 12:810263. [PMID: 35359414 PMCID: PMC8961422 DOI: 10.3389/fonc.2022.810263] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/05/2022] [Indexed: 01/16/2023] Open
Abstract
Objective Summarize evidence for use of advanced MRI techniques as monitoring biomarkers in the clinic, and highlight the latest bench-to-bedside developments. Methods Experts in advanced MRI techniques applied to high-grade glioma treatment response assessment convened through a European framework. Current evidence regarding the potential for monitoring biomarkers in adult high-grade glioma is reviewed, and individual modalities of perfusion, permeability, and microstructure imaging are discussed (in Part 1 of two). In Part 2, we discuss modalities related to metabolism and/or chemical composition, appraise the clinic readiness of the individual modalities, and consider post-processing methodologies involving the combination of MRI approaches (multiparametric imaging) or machine learning (radiomics). Results High-grade glioma vasculature exhibits increased perfusion, blood volume, and permeability compared with normal brain tissue. Measures of cerebral blood volume derived from dynamic susceptibility contrast-enhanced MRI have consistently provided information about brain tumor growth and response to treatment; it is the most clinically validated advanced technique. Clinical studies have proven the potential of dynamic contrast-enhanced MRI for distinguishing post-treatment related effects from recurrence, but the optimal acquisition protocol, mode of analysis, parameter of highest diagnostic value, and optimal cut-off points remain to be established. Arterial spin labeling techniques do not require the injection of a contrast agent, and repeated measurements of cerebral blood flow can be performed. The absence of potential gadolinium deposition effects allows widespread use in pediatric patients and those with impaired renal function. More data are necessary to establish clinical validity as monitoring biomarkers. Diffusion-weighted imaging, apparent diffusion coefficient analysis, diffusion tensor or kurtosis imaging, intravoxel incoherent motion, and other microstructural modeling approaches also allow treatment response assessment; more robust data are required to validate these alone or when applied to post-processing methodologies. Conclusion Considerable progress has been made in the development of these monitoring biomarkers. Many techniques are in their infancy, whereas others have generated a larger body of evidence for clinical application.
Collapse
Affiliation(s)
- Otto M. Henriksen
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Patricia Figueiredo
- Department of Bioengineering and Institute for Systems and Robotics-Lisboa, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Gilbert Hangel
- Department of Neurosurgery, Medical University, Vienna, Austria
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University, Vienna, Austria
| | - Vera C. Keil
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Ruben E. Nechifor
- International Institute for the Advanced Studies of Psychotherapy and Applied Mental Health, Department of Clinical Psychology and Psychotherapy, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Evita C. Wiegers
- Department of Radiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Thomas C. Booth
-
School of Biomedical Engineering and Imaging Sciences, St. Thomas’ Hospital, King’s College London, London, United Kingdom
- Department of Neuroradiology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
5
|
Gerstner ER, Emblem KE, Yen YF, Dietrich J, Jordan JT, Catana C, Wenchin KL, Hooker JM, Duda DG, Rosen BR, Kalpathy-Cramer J, Jain RK, Batchelor TT. Vascular dysfunction promotes regional hypoxia after bevacizumab therapy in recurrent glioblastoma patients. Neurooncol Adv 2020; 2:vdaa157. [PMID: 33392506 PMCID: PMC7764510 DOI: 10.1093/noajnl/vdaa157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Hypoxia is a driver of treatment resistance in glioblastoma. Antiangiogenic agents may transiently normalize blood vessels and decrease hypoxia before excessive pruning of vessels increases hypoxia. The time window of normalization is dose and time dependent. We sought to determine how VEGF blockade with bevacizumab modulates tumor vasculature and the impact that those vascular changes have on hypoxia in recurrent glioblastoma patients. Methods We measured tumor volume, vascular permeability (Ktrans), perfusion parameters (cerebral blood flow/volume, vessel caliber, and mean transit time), and regions of hypoxia in patients with recurrent glioblastoma before and after treatment with bevacizumab alone or with lomustine using [18F]FMISO PET-MRI. We also examined serial changes in plasma biomarkers of angiogenesis and inflammation. Results Eleven patients were studied. The magnitude of global tumor hypoxia was variable across these 11 patients prior to treatment and it did not significantly change after bevacizumab. The hypoxic regions had an inefficient vasculature characterized by elevated cerebral blood flow/volume and increased vessel caliber. In a subset of patients, there were tumor subregions with decreased mean transit times and a decrease in hypoxia, suggesting heterogeneous improvement in vascular efficiency. Bevacizumab significantly changed known pharmacodynamic biomarkers such as plasma VEGF and PlGF. Conclusions The vascular signature in hypoxic tumor regions indicates a disorganized vasculature which, in most tumors, does not significantly change after bevacizumab treatment. While some tumor regions showed improved vascular efficiency following treatment, bevacizumab did not globally alter hypoxia or normalize tumor vasculature in glioblastoma.
Collapse
Affiliation(s)
- Elizabeth R Gerstner
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Kyrre E Emblem
- Department of Diagnostic Physics, Oslo University, Oslo, Norway
| | - Yi-Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jorg Dietrich
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Justin T Jordan
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin Lou Wenchin
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Hormuth DA, Sorace AG, Virostko J, Abramson RG, Bhujwalla ZM, Enriquez-Navas P, Gillies R, Hazle JD, Mason RP, Quarles CC, Weis JA, Whisenant JG, Xu J, Yankeelov TE. Translating preclinical MRI methods to clinical oncology. J Magn Reson Imaging 2019; 50:1377-1392. [PMID: 30925001 PMCID: PMC6766430 DOI: 10.1002/jmri.26731] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
The complexity of modern in vivo magnetic resonance imaging (MRI) methods in oncology has dramatically changed in the last 10 years. The field has long since moved passed its (unparalleled) ability to form images with exquisite soft-tissue contrast and morphology, allowing for the enhanced identification of primary tumors and metastatic disease. Currently, it is not uncommon to acquire images related to blood flow, cellularity, and macromolecular content in the clinical setting. The acquisition of images related to metabolism, hypoxia, pH, and tissue stiffness are also becoming common. All of these techniques have had some component of their invention, development, refinement, validation, and initial applications in the preclinical setting using in vivo animal models of cancer. In this review, we discuss the genesis of quantitative MRI methods that have been successfully translated from preclinical research and developed into clinical applications. These include methods that interrogate perfusion, diffusion, pH, hypoxia, macromolecular content, and tissue mechanical properties for improving detection, staging, and response monitoring of cancer. For each of these techniques, we summarize the 1) underlying biological mechanism(s); 2) preclinical applications; 3) available repeatability and reproducibility data; 4) clinical applications; and 5) limitations of the technique. We conclude with a discussion of lessons learned from translating MRI methods from the preclinical to clinical setting, and a presentation of four fundamental problems in cancer imaging that, if solved, would result in a profound improvement in the lives of oncology patients. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;50:1377-1392.
Collapse
Affiliation(s)
- David A. Hormuth
- Institute for Computational Engineering and Sciences,Livestrong Cancer Institutes, The University of Texas at Austin
| | - Anna G. Sorace
- Department of Biomedical Engineering, The University of Texas at Austin,Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| | - John Virostko
- Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| | - Richard G. Abramson
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center
| | | | - Pedro Enriquez-Navas
- Departments of Cancer Imaging and Metabolism, Cancer Physiology, The Moffitt Cancer Center
| | - Robert Gillies
- Departments of Cancer Imaging and Metabolism, Cancer Physiology, The Moffitt Cancer Center
| | - John D. Hazle
- Imaging Physics, The University of Texas M.D. Anderson Cancer Center
| | - Ralph P. Mason
- Department of Radiology, The University of Texas Southwestern Medical Center
| | - C. Chad Quarles
- Department of NeuroImaging Research, The Barrow Neurological Institute
| | - Jared A. Weis
- Department of Biomedical Engineering Wake Forest School of Medicine
| | | | - Junzhong Xu
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center,Institute of Imaging Science, Vanderbilt University Medical Center
| | - Thomas E. Yankeelov
- Institute for Computational Engineering and Sciences,Department of Biomedical Engineering, The University of Texas at Austin,Department of Diagnostic Medicine, The University of Texas at Austin,Department of Oncology, The University of Texas at Austin,Livestrong Cancer Institutes, The University of Texas at Austin
| |
Collapse
|
7
|
Quarles CC, Bell LC, Stokes AM. Imaging vascular and hemodynamic features of the brain using dynamic susceptibility contrast and dynamic contrast enhanced MRI. Neuroimage 2018; 187:32-55. [PMID: 29729392 DOI: 10.1016/j.neuroimage.2018.04.069] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 12/22/2022] Open
Abstract
In the context of neurologic disorders, dynamic susceptibility contrast (DSC) and dynamic contrast enhanced (DCE) MRI provide valuable insights into cerebral vascular function, integrity, and architecture. Even after two decades of use, these modalities continue to evolve as their biophysical and kinetic basis is better understood, with improvements in pulse sequences and accelerated imaging techniques and through application of more robust and automated data analysis strategies. Here, we systematically review each of these elements, with a focus on how their integration improves kinetic parameter accuracy and the development of new hemodynamic biomarkers that provide sub-voxel sensitivity (e.g., capillary transit time and flow heterogeneity). Regarding contrast mechanisms, we discuss the dipole-dipole interactions and susceptibility effects that give rise to simultaneous T1, T2 and T2∗ relaxation effects, including their quantification, influence on pulse sequence parameter optimization, and use in methods such as vessel size and vessel architectural imaging. The application of technologic advancements, such as parallel imaging, simultaneous multi-slice, undersampled k-space acquisitions, and sliding window strategies, enables improved spatial and/or temporal resolution of DSC and DCE acquisitions. Such acceleration techniques have also enabled the implementation of, clinically feasible, simultaneous multi-echo spin- and gradient echo acquisitions, providing more comprehensive and quantitative interrogation of T1, T2 and T2∗ changes. Characterizing these relaxation rate changes through different post-processing options allows for the quantification of hemodynamics and vascular permeability. The application of different biophysical models provides insight into traditional hemodynamic parameters (e.g., cerebral blood volume) and more advanced parameters (e.g., capillary transit time heterogeneity). We provide insight into the appropriate selection of biophysical models and the necessary post-processing steps to ensure reliable measurements while minimizing potential sources of error. We show representative examples of advanced DSC- and DCE-MRI methods applied to pathologic conditions affecting the cerebral microcirculation, including brain tumors, stroke, aging, and multiple sclerosis. The maturation and standardization of conventional DSC- and DCE-MRI techniques has enabled their increased integration into clinical practice and use in clinical trials, which has, in turn, spurred renewed interest in their technological and biophysical development, paving the way towards a more comprehensive assessment of cerebral hemodynamics.
Collapse
Affiliation(s)
- C Chad Quarles
- Division of Neuro imaging Research, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, USA.
| | - Laura C Bell
- Division of Neuro imaging Research, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, USA
| | - Ashley M Stokes
- Division of Neuro imaging Research, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ, USA
| |
Collapse
|
8
|
Yan N, Zheng Y, Yang C. Functional and morphological effects of diazepam and midazolam on tumor vasculature in the 9L gliosarcoma brain tumor model using dynamic susceptibility contrast MRI: a comparative study. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2931-2936. [PMID: 29042753 PMCID: PMC5634367 DOI: 10.2147/dddt.s143838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antiangiogenic therapy attenuates tumor growth by reducing vascularization. Diazepam (DZP) and midazolam (MZL) have antiangiogenic properties in human umbilical vein endothelial cells. Thus, we investigated the antiangiogenic activity of DZP and MZL in the rat 9L gliosarcoma brain tumor model. The effect on tumor vasculature was evaluated using dynamic susceptibility contrast magnetic resonance imaging with gradient-echo (GE) and spin-echo (SE) to assess perfusion parameters, including cerebral blood volume (CBV), cerebral blood flow (CBF), mean transit time (MTT), and mean vessel diameter. The GE-normalized CBF (nCBF) in the tumors of untreated controls was significantly lower than that in normal brain tissue, whereas the CBV and MTT were higher. DZP- and MZL-treated rats had higher CBF and lower CBV and MTT values than did untreated controls. The tumor size decreased significantly to 33.5% in DZP-treated rats (P<0.001) and 22.5% in MZL-treated rats (P<0.01) relative to controls. The SE-normalized CBV was lower in DZP-treated (32.9%) and MZL-treated (10.6%) rats compared with controls. The mean vessel diameter decreased significantly by 32.5% in DPZ-treated and by 24.9% in MZL-treated rats compared with controls (P<0.01). The GE and SE nCBF values were higher in DZP-treated (49.9% and 40.1%, respectively) and MZL-treated (41.2% and 32.1%, respectively) rats than in controls. The GE- and SE-normalized MTTs were lower in DZP-treated (48.2% and 59.8%, respectively) and MZL-treated (40.5% and 51.2%, respectively) rats than in controls. Both DZP and MZL had antiangiogenic effects on tumor perfusion and vasculature; however, the antiangiogenic activity of DZP is more promising than that of MZL.
Collapse
Affiliation(s)
- Nuo Yan
- Second Department of Anesthesiology, The Affiliated Hospital to Logistics University of PAP, Tianjin
| | - Yuzhen Zheng
- Department of Anesthesiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Cheng Yang
- Second Department of Anesthesiology, The Affiliated Hospital to Logistics University of PAP, Tianjin
| |
Collapse
|
9
|
Iv M, Telischak N, Feng D, Holdsworth SJ, Yeom KW, Daldrup-Link HE. Clinical applications of iron oxide nanoparticles for magnetic resonance imaging of brain tumors. Nanomedicine (Lond) 2015; 10:993-1018. [PMID: 25867862 DOI: 10.2217/nnm.14.203] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Current neuroimaging provides detailed anatomic and functional evaluation of brain tumors, allowing for improved diagnostic and prognostic capabilities. Some challenges persist even with today's advanced imaging techniques, including accurate delineation of tumor margins and distinguishing treatment effects from residual or recurrent tumor. Ultrasmall superparamagnetic iron oxide nanoparticles are an emerging tool that can add clinically useful information due to their distinct physiochemical features and biodistribution, while having a good safety profile. Nanoparticles can be used as a platform for theranostic drugs, which have shown great promise for the treatment of CNS malignancies. This review will provide an overview of clinical ultrasmall superparamagnetic iron oxides and how they can be applied to the diagnostic and therapeutic neuro-oncologic setting.
Collapse
Affiliation(s)
- Michael Iv
- Department of Radiology, Stanford University & Stanford University Medical Center, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
10
|
Perfusion MRI derived indices of microvascular shunting and flow control correlate with tumor grade and outcome in patients with cerebral glioma. PLoS One 2015; 10:e0123044. [PMID: 25875182 PMCID: PMC4395250 DOI: 10.1371/journal.pone.0123044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/20/2015] [Indexed: 01/21/2023] Open
Abstract
Objectives Deficient microvascular blood flow control is thought to cause tumor hypoxia and increase resistance to therapy. In glioma patients, we tested whether perfusion-weighted MRI (PWI) based indices of microvascular flow control provide more information on tumor grade and patient outcome than does the established PWI angiogenesis marker, cerebral blood volume (CBV). Material and Methods Seventy-two glioma patients (sixty high-grade, twelve low-grade gliomas) were included. Capillary transit time heterogeneity (CTH) and the coefficient of variation (COV), its ratio to blood mean transit time, provide indices of microvascular flow control and the extent to which oxygen can be extracted by tumor tissue. The ability of these parameters and CBV to differentiate tumor grade were assessed by receiver operating characteristic curves and logistic regression. Their ability to predict time to progression and overall survival was examined by the Cox proportional-hazards regression model, and by survival curves using log-rank tests. Results The best prediction of grade (AUC = 0.876; p < 0.05) was achieved by combining knowledge of CBV and CTH in the enhancing tumor and peri-focal edema, and patients with glioblastoma multiforme were identified best by CTH (AUC = 0.763; p<0.001). CTH outperformed CBV and COV in predicting time to progression and survival in all gliomas and in a subgroup consisting of only high-grade gliomas. Conclusion Our study confirms the importance of microvascular flow control in tumor growth by demonstrating that determining CTH improves tumor grading and outcome prediction in glioma patients compared to CBV alone.
Collapse
|
11
|
Kellner E, Breyer T, Gall P, Müller K, Trippel M, Staszewski O, Stein F, Saborowski O, Dyakova O, Urbach H, Kiselev VG, Mader I. MR evaluation of vessel size imaging of human gliomas: Validation by histopathology. J Magn Reson Imaging 2015; 42:1117-25. [PMID: 25683112 DOI: 10.1002/jmri.24864] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 01/19/2015] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To compare the vessel size and the cerebral blood volume in human gliomas with histopathology. Vessel size imaging (VSI) is a dynamic susceptibility contrast method for the assessment of the vessel size in normal and pathological tissue. Previous publications in rodents showed a satisfactory conformity with the vessel size derived from histopathology. To assess the clinical value, further, the progression-free interval was determined and correlated. MATERIALS AND METHODS Twenty-five gliomas (WHO grade °II [n = 10], °III [n = 3], °IV [n = 12]) were prospectively included and received a stereotaxic biopsy after VSI. The vessel size and the cerebral blood volume (CBV) were calculated in regions of interest at the tumor edge and correlated with the vessel size measured by histopathology. RESULTS Both VSI and CBV showed a good correlation with the vessel size in histopathology (up to r = 0.84, P < 0.001, and r = 0.62, P < 0.001, respectively). Slope and offset of the linear regression (y = 0.77x + 0.36 μm) suggest that the size of normal capillaries is overestimated with VSI, while for grossly enlarged vessels an underestimation occurs. Both VSI and CBV were negatively correlated with the progression-free interval (r = -0.57, P = 0.008, and r = -0.50, P = 0.025, respectively). CONCLUSION The correlation between VSI and vessel size from histopathology is in good accordance with the animal studies. The overestimation of small capillary sizes is also known from the animal trials. Vessel size and CBV showed similar results, both for the correlation with the histopathological vessel size and the progression-free interval.
Collapse
Affiliation(s)
- Elias Kellner
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Germany
| | - Tobias Breyer
- Department of Neuroradiology, University Medical Center Freiburg, Germany
| | - Peter Gall
- Siemens AG, Healthcare Sector, Erlangen, Germany
| | - Klaus Müller
- Department of Neuropathology, University Medical Center Freiburg, Germany
| | - Michael Trippel
- Department of Stereotactic Neurosurgery, University Medical Center Freiburg, Germany
| | - Ori Staszewski
- Department of Neuropathology, University Medical Center Freiburg, Germany
| | - Florian Stein
- Department of Neuropathology, University Medical Center Freiburg, Germany
| | - Olaf Saborowski
- Department of Neuroradiology, University Medical Center Freiburg, Germany
| | - Olga Dyakova
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, University Medical Center Freiburg, Germany
| | - Valerij G Kiselev
- Department of Radiology, Medical Physics, University Medical Center Freiburg, Germany
| | - Irina Mader
- Department of Neuroradiology, University Medical Center Freiburg, Germany
| |
Collapse
|
12
|
Emblem KE, Farrar CT, Gerstner ER, Batchelor TT, Borra RJH, Rosen BR, Sorensen AG, Jain RK. Vessel caliber--a potential MRI biomarker of tumour response in clinical trials. Nat Rev Clin Oncol 2014; 11:566-84. [PMID: 25113840 PMCID: PMC4445139 DOI: 10.1038/nrclinonc.2014.126] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Our understanding of the importance of blood vessels and angiogenesis in cancer has increased considerably over the past decades, and the assessment of tumour vessel calibre and structure has become increasingly important for in vivo monitoring of therapeutic response. The preferred method for in vivo imaging of most solid cancers is MRI, and the concept of vessel-calibre MRI has evolved since its initial inception in the early 1990s. Almost a quarter of a century later, unlike traditional contrast-enhanced MRI techniques, vessel-calibre MRI remains widely inaccessible to the general clinical community. The narrow availability of the technique is, in part, attributable to limited awareness and a lack of imaging standardization. Thus, the role of vessel-calibre MRI in early phase clinical trials remains to be determined. By contrast, regulatory approvals of antiangiogenic agents that are not directly cytotoxic have created an urgent need for clinical trials incorporating advanced imaging analyses, going beyond traditional assessments of tumour volume. To this end, we review the field of vessel-calibre MRI and summarize the emerging evidence supporting the use of this technique to monitor response to anticancer therapy. We also discuss the potential use of this biomarker assessment in clinical imaging trials and highlight relevant avenues for future research.
Collapse
Affiliation(s)
- Kyrre E Emblem
- The Intervention Centre, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Christian T Farrar
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Tracy T Batchelor
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| | - Ronald J H Borra
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce R Rosen
- Department of Radiology and Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - A Gregory Sorensen
- Siemens Healthcare Health Services, 51 Valley Stream Parkway, Malvern, PA 19355, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratory of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Boston, MA 02114, USA
| |
Collapse
|
13
|
Troprès I, Pannetier N, Grand S, Lemasson B, Moisan A, Péoc'h M, Rémy C, Barbier EL. Imaging the microvessel caliber and density: Principles and applications of microvascular MRI. Magn Reson Med 2014; 73:325-41. [DOI: 10.1002/mrm.25396] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Irène Troprès
- IRMaGe; Université Grenoble Alpes; Grenoble France
- UMS 3552; CNRS; Grenoble France
- US 017; INSERM; Grenoble France
- IRMaGe, Hôpital Michallon; Centre Hospitalier Universitaire de Grenoble; Grenoble France
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France. INSERM; U836 Grenoble France
| | - Nicolas Pannetier
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Sylvie Grand
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
- CLUNI, Hôpital Michallon; Centre Hospitalier Universitaire de Grenoble; Grenoble France
| | - Benjamin Lemasson
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Anaïck Moisan
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Michel Péoc'h
- Service d'anatomo-pathologie; Centre Hospitalier Universitaire de Saint Etienne; Saint-Etienne France
- EA 2521; Université Jean Monnet; Saint-Etienne France
| | - Chantal Rémy
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| | - Emmanuel L. Barbier
- Université Joseph Fourier; Grenoble Institut des Neurosciences; Grenoble France
- INSERM; U836 Grenoble France
| |
Collapse
|
14
|
Martínez-Martínez A, Martínez-Bosch J. Perfusion magnetic resonance imaging for high grade astrocytomas: Can cerebral blood volume, peak height, and percentage of signal intensity recovery distinguish between progression and pseudoprogression? RADIOLOGIA 2014. [DOI: 10.1016/j.rxeng.2014.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Smith DS, Li X, Abramson RG, Chad Quarles C, Yankeelov TE, Brian Welch E. Potential of compressed sensing in quantitative MR imaging of cancer. Cancer Imaging 2013; 13:633-44. [PMID: 24434808 PMCID: PMC3893904 DOI: 10.1102/1470-7330.2013.0041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2013] [Indexed: 12/22/2022] Open
Abstract
Classic signal processing theory dictates that, in order to faithfully reconstruct a band-limited signal (e.g., an image), the sampling rate must be at least twice the maximum frequency contained within the signal, i.e., the Nyquist frequency. Recent developments in applied mathematics, however, have shown that it is often possible to reconstruct signals sampled below the Nyquist rate. This new method of compressed sensing (CS) requires that the signal have a concise and extremely dense representation in some mathematical basis. Magnetic resonance imaging (MRI) is particularly well suited for CS approaches, owing to the flexibility of data collection in the spatial frequency (Fourier) domain available in most MRI protocols. With custom CS acquisition and reconstruction strategies, one can quickly obtain a small subset of the full data and then iteratively reconstruct images that are consistent with the acquired data and sparse by some measure. Successful use of CS results in a substantial decrease in the time required to collect an individual image. This extra time can then be harnessed to increase spatial resolution, temporal resolution, signal-to-noise, or any combination of the three. In this article, we first review the salient features of CS theory and then discuss the specific barriers confronting CS before it can be readily incorporated into clinical quantitative MRI studies of cancer. We finally illustrate applications of the technique by describing examples of CS in dynamic contrast-enhanced MRI and dynamic susceptibility contrast MRI.
Collapse
Affiliation(s)
- David S. Smith
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Xia Li
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Richard G. Abramson
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - C. Chad Quarles
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Thomas E. Yankeelov
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - E. Brian Welch
- Institute of Imaging Science, Departments of Radiology and Radiological Sciences, Biomedical Engineering, Physics and Astronomy, and Cancer Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
16
|
Martínez-Martínez A, Martínez-Bosch J. [Perfusion magnetic resonance imaging for high grade astrocytomas: Can cerebral blood volume, peak height, and percentage of signal intensity recovery distinguish between progression and pseudoprogression?]. RADIOLOGIA 2013; 56:35-43. [PMID: 23790618 DOI: 10.1016/j.rx.2013.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 01/22/2023]
Abstract
OBJECTIVES To study the usefulness of common MRI perfusion parameters for identifying pseudoprogression in high grade astrocytomas. MATERIAL AND METHODS This retrospective case-control study compared the relative cerebral blood volume (rCBV), the relative percentage of signal intensity recovery (rPSR), and the relative peak height (rPH) recorded in a sample of 17 cases of anaplastic astrocytomas and gliomas considered to be undergoing pseudoprogression by biopsy or follow-up with those recorded in a sample of histologically similar tumors that were treated and considered to be undergoing progression by histologic study or follow-up. We evaluated the accuracy of these parameters and the correlations among them. Statistical significance was set at P<.05. RESULTS The rCBV, rPSR, and rPH were significantly different between the two groups (P=.001). The cutoff values rPH=1.37, rCBV=0.9, and rPSR=99% yielded sensitivity (S)=88% and specificity (Sp)=82.2% for rPH, S=100% and Sp=100% for rCBV, and S=100% and Sp=70.6% for rPSR, respectively. We found negative correlations between rPRS and rPH (-0.76) and between rPRS and rCBV (-0.81) and a high positive correlation between rPH and rCBV (0.87). CONCLUSION The variables rPH and rCBV were useful for differentiating between pseudoprogression and true progression in our sample. The variable rPRS was also very sensitive, although the overlap in the values between samples make it less useful a priori.
Collapse
Affiliation(s)
- A Martínez-Martínez
- Sección de Neurorradiología, Unidad de Gestión Clínica de Radiodiagnóstico, Hospital Universitario Virgen de las Nieves, Granada, España.
| | - J Martínez-Bosch
- Sección de Neurorradiología, Unidad de Gestión Clínica de Radiodiagnóstico, Hospital Universitario Virgen de las Nieves, Granada, España
| |
Collapse
|
17
|
The Relationship between Tumor Blood Flow, Angiogenesis, Tumor Hypoxia, and Aerobic Glycolysis. Cancer Res 2013; 73:5618-24. [DOI: 10.1158/0008-5472.can-13-0964] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Reynaud O, Geffroy F, Ciobanu L. Quantification of microvascular cerebral blood flux and late-stage tumor compartmentalization in 9L gliosarcoma using flow enhanced MRI. NMR IN BIOMEDICINE 2013; 26:699-708. [PMID: 23335424 DOI: 10.1002/nbm.2915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 12/07/2012] [Accepted: 12/07/2012] [Indexed: 06/01/2023]
Abstract
Measurements of tumor microvasculature are important to obtain an understanding of tumor angiogenesis and for the evaluation of therapies. In this work, we characterize the evolution of the microvascular flux at different stages of tumor growth in the 9L rat brain tumor model. The absolute quantification of cerebral blood flux is achieved with MRI at 7 T using the flow enhanced signal intensity (FENSI) method. FENSI flux maps were obtained between 5 and 14 days after glioma cell inoculation. Based on cerebral blood flux maps, we highlighted two main stages of tumor growth, below and above 3 mm, presenting distinct flux patterns and vascular properties. No significant difference emerged from the group analysis performed on the data collected at an early developmental stage (tumor size < 3 mm) when compared with healthy tissue. At a late developmental stage (tumor size > 3 mm), we observed a significant decrease in the cerebral blood flux inside the gliosarcoma (-33%, p < 0.01) and compartmentalization of the tumor (p < 0.05). FENSI flux maps delineated a low-flux tumor core (58 ± 17 μL/min/cm(2) ) and higher vascularized regions around the tumor periphery (85 ± 21 μL/min/cm(2) ). Histology was performed on 11 animals to finely probe the intratumor heterogeneity and microvessel density, and the results were compared with the information derived from FENSI flux maps. The hyper- and hypoperfused tumor regions revealed with FENSI at the late tumor developmental stage correlated well with the ratios of high and low blood vessel density (R(2) = 0.41) and fractional vascular surface (R(2) = 0.67) observed with fluorescence microscopy [cluster of differentiation 31 (CD31) staining].
Collapse
Affiliation(s)
- Olivier Reynaud
- Commissariat a l'Energie Atomique/DSV, I2BM, NeuroSpin, LRMN, Gif sur Yvette, France
| | | | | |
Collapse
|
19
|
LaViolette PS, Cohen AD, Prah MA, Rand SD, Connelly J, Malkin MG, Mueller WM, Schmainda KM. Vascular change measured with independent component analysis of dynamic susceptibility contrast MRI predicts bevacizumab response in high-grade glioma. Neuro Oncol 2013; 15:442-50. [PMID: 23382287 DOI: 10.1093/neuonc/nos323] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Standard pre- and postcontrast (T1 + C) anatomical MR imaging is proving to be insufficient for accurately monitoring bevacizumab treatment response in recurrent glioblastoma (GBM). We present a novel imaging biomarker that detects abnormal tumor vasculature exhibiting both arterial and venous perfusion characteristics. We hypothesized that a decrease in the extent of this abnormal vasculature after bevacizumab treatment would predict treatment efficacy and overall survival. METHODS Dynamic susceptibility contrast perfusion MRI was gathered in 43 patients with high-grade glioma. Independent component analysis separated vasculature into arterial and venous components. Voxels with perfusion characteristics of both arteries and veins (ie, arterio-venous overlap [AVOL]) were measured in patients with de novo untreated GBM and patients with recurrent high-grade glioma before and after bevacizumab treatment. Treated patients were separated on the basis of an increase or decrease in AVOL volume (+/-ΔAVOL), and overall survival following bevacizumab onset was then compared between +/-ΔAVOL groups. RESULTS AVOL in untreated GBM was significantly higher than in normal vasculature (P < .001). Kaplan-Meier survival curves revealed a greater median survival (348 days) in patients with GBM with a negative ΔAVOL after bevacizumab treatment than in patients with a positive change (197 days; hazard ratio, 2.51; P < .05). Analysis of patients with combined grade III and IV glioma showed similar results, with median survivals of 399 days and 153 days, respectively (hazard ratio, 2.71; P < .01). Changes in T1+C volume and ΔrCBV after treatment were not significantly different across +/-ΔAVOL groups, and ΔAVOL was not significantly correlated with ΔT1+C or ΔrCBV. CONCLUSIONS The independent component analysis dynamic susceptibility contrast-derived biomarker AVOL adds additional information for determining bevacizumab treatment efficacy.
Collapse
Affiliation(s)
- Peter S LaViolette
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
This article presents an overview of advanced magnetic resonance (MR) imaging techniques using contrast media in neuroimaging, focusing on T2*-weighted dynamic susceptibility contrast MR imaging and T1-weighted dynamic contrast-enhanced MR imaging. Image acquisition and data processing methods and their clinical application in brain tumors, stroke, dementia, and multiple sclerosis are discussed.
Collapse
Affiliation(s)
- Jean-Christophe Ferré
- Department of Radiology, Keck Medical Center of University of Southern California, Los Angeles, CA 90033, USA.
| | | | | |
Collapse
|
21
|
Nielsen T, Bentzen L, Pedersen M, Tramm T, Rijken PFJW, Bussink J, Horsman MR, Østergaard L. Combretastatin A-4 phosphate affects tumor vessel volume and size distribution as assessed using MRI-based vessel size imaging. Clin Cancer Res 2012; 18:6469-77. [PMID: 23071260 DOI: 10.1158/1078-0432.ccr-12-2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Combretastatin A-4 disodium phosphate (CA4P) is a promising vascular disrupting agent (VDA) in clinical trials. As CA4P acts on dividing endothelial cells, we hypothesize that CA4P affects vessels of certain sizes. The aim of this study was to evaluate the effect of CA4P by the MRI-based vessel size imaging (VSI). EXPERIMENTAL DESIGN C3H mammary carcinomas were grown to 200 mm(3) in the right rear foot of female CDF(1) mice. A control group of mice received no treatment, and a treatment group had CA4P administered intraperitoneally at a dose of 250 mg/kg. VSI was conducted on a 3 Tesla MR scanner to estimate the tumor blood volume (ζ(0)) and mean vessel radius (R). Vascularization was also estimated histologically by endothelial and Hoechst 33342 staining. RESULTS ζ(0) and R showed different spatial heterogeneity. Tumor median and quartile values of ζ(0) were all significantly reduced by about 35% in the CA4P-treated group as compared with the control group, and the median and upper quartile of R were significantly increased. Histograms of ζ(0) and R showed a general decrease in ζ(0) following treatment, and values of R in a certain range (≈20-30 μm) were decreased in the treatment group. The drug-induced change in ζ(0) was in agreement with histology and our previous dynamic contrast enhanced MRI (DCE-MRI) data. CONCLUSIONS Tumor blood volume and mean vessel radius showed a clear response following treatment with CA4P. VSI may prove valuable in estimation of tumor angiogenesis and prediction of response to VDAs.
Collapse
Affiliation(s)
- Thomas Nielsen
- Department of Experimental Clinical Oncology, Danish National Research Foundations Center of Functionally Integrative Neuroscience, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
T(1)- and T(2)(*)-dominant extravasation correction in DSC-MRI: part II-predicting patient outcome after a single dose of cediranib in recurrent glioblastoma patients. J Cereb Blood Flow Metab 2011; 31:2054-64. [PMID: 21505476 PMCID: PMC3208147 DOI: 10.1038/jcbfm.2011.39] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A 'vascular normalization index' (VNI) based on the changes in the magnetic resonance imaging (MRI) parameters K(trans) and cerebral blood volume (CBV), combined with blood sampling, has been shown to correlate with patient outcome in recurrent glioblastoma after a single dose of antiangiogenic therapy. Here, by applying a novel contrast agent extravasation correction method insensitive to variations in tissue mean transit time, we show that a similar VNI parameter can be derived from a single dynamic susceptibility contrast MR acquisition rather than the three parameters shown previously. Our results show that this new VNI parameter, which combines changes in tumoral CBV and an apparent transfer constant from our leakage correction method, may provide prognostic information in an even simpler manner than prior efforts.
Collapse
|
23
|
Wang X, Duan X, Yang G, Zhang X, Deng L, Zheng H, Deng C, Wen J, Wang N, Peng C, Zhao X, Wei Y, Chen L. Honokiol crosses BBB and BCSFB, and inhibits brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. PLoS One 2011; 6:e18490. [PMID: 21559510 PMCID: PMC3084695 DOI: 10.1371/journal.pone.0018490] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 03/09/2011] [Indexed: 02/05/2023] Open
Abstract
Background Gliosarcoma is one of the most common malignant brain tumors, and anti-angiogenesis is a promising approach for the treatment of gliosarcoma. However, chemotherapy is obstructed by the physical obstacle formed by the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB). Honokiol has been known to possess potent activities in the central nervous system diseases, and anti-angiogenic and anti-tumor properties. Here, we hypothesized that honokiol could cross the BBB and BCSFB for the treatment of gliosarcoma. Methodologies We first evaluated the abilities of honokiol to cross the BBB and BCSFB by measuring the penetration of honokiol into brain and blood-cerebrospinal fluid, and compared the honokiol amount taken up by brain with that by other tissues. Then we investigated the effect of honokiol on the growth inhibition of rat 9L gliosarcoma cells and human U251 glioma cells in vitro. Finally we established rat 9L intracerebral gliosarcoma model in Fisher 344 rats and human U251 xenograft glioma model in nude mice to investigate the anti-tumor activity. Principal Findings We showed for the first time that honokiol could effectively cross BBB and BCSFB. The ratios of brain/plasma concentration were respectively 1.29, 2.54, 2.56 and 2.72 at 5, 30, 60 and 120 min. And about 10% of honokiol in plasma crossed BCSFB into cerebrospinal fluid (CSF). In vitro, honokiol produced dose-dependent inhibition of the growth of rat 9L gliosarcoma cells and human U251 glioma cells with IC50 of 15.61 µg/mL and 16.38 µg/mL, respectively. In vivo, treatment with 20 mg/kg body weight of honokiol (honokiol was given twice per week for 3 weeks by intravenous injection) resulted in significant reduction of tumor volume (112.70±10.16 mm3) compared with vehicle group (238.63±19.69 mm3, P = 0.000), with 52.77% inhibiting rate in rat 9L intracerebral gliosarcoma model, and (1450.83±348.36 mm3) compared with vehicle group (2914.17±780.52 mm3, P = 0.002), with 50.21% inhibiting rate in human U251 xenograft glioma model. Honokiol also significantly improved the survival over vehicle group in the two models (P<0.05). Conclusions/Significance This study provided the first evidence that honokiol could effectively cross BBB and BCSFB and inhibit brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. It suggested a significant strategy for offering a potential new therapy for the treatment of gliosarcoma.
Collapse
Affiliation(s)
- Xianhuo Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xingmei Duan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Guangli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xiaoyan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Linyu Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hao Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Chongyang Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jiaolin Wen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Xia Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
- * E-mail:
| |
Collapse
|
24
|
Haney CR, Pelizzari CA, Foxley S, Zamora MA, Mustafi D, Tretiakova M, Li S, Fan X, Karczmar GS. HiSStology: high spectral and spatial resolution magnetic resonance imaging detection of vasculature validated by histology and micro-computed tomography. Mol Imaging 2011; 10:187-96. [PMID: 21443840 DOI: 10.2310/7290.2010.00033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 03/31/2010] [Indexed: 11/18/2022] Open
Abstract
High spectral and spatial resolution (HiSS) data, acquired with echo-planar spectroscopic imaging (EPSI), can be used to acquire water spectra from each small image voxel. These images are sensitive to changes in local susceptibility caused by superparamagnetic iron oxide particles (SPIO); therefore, we hypothesized that images derived from HiSS data are very sensitive to tumor neovasculature following injection of SPIO. Accurate image registration was used to validate HiSS detection of neovasculature with histology and micro-computed tomographic (microCT) angiography. Athymic nude mice and Copenhagen rats were inoculated with Dunning AT6.1 prostate tumor cells in the right hind limb. The tumor region was imaged pre- and post-intravenous injection of SPIO. Three-dimensional assemblies of the CD31-stained histologic slices of the mouse legs and the microCT images of the rat vascular casts were registered with EPSI. The average distance between HiSS-predicted regions of high vascular density on magnetic resonance imaging and CD31-stained regions on histology was 200 μm. Similarly, vessels identified by HiSS in the rat images coincided with vasculature in the registered microCT image. The data demonstrate a strong correlation between tumor vasculature identified using HiSS and two gold standards: histology and microCT angiography.
Collapse
Affiliation(s)
- Chad R Haney
- Department of Radiology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Darpolor MM, Molthen RC, Schmainda KM. Multimodality imaging of abnormal vascular perfusion and morphology in preclinical 9L gliosarcoma model. PLoS One 2011; 6:e16621. [PMID: 21305001 PMCID: PMC3031600 DOI: 10.1371/journal.pone.0016621] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2010] [Accepted: 01/07/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND This study demonstrates that a dynamic susceptibility contrast-magnetic resonance imaging (DSC-MRI) perfusion parameter may indicate vascular abnormality in a brain tumor model and reflects an effect of dexamethasone treatment. In addition, X-ray computed tomography (CT) measurements of vascular tortuosity and tissue markers of vascular morphology were performed to investigate the underpinnings of tumor response to dexamethasone. METHODOLOGY/PRINCIPAL FINDINGS One cohort of Fisher 344 rats (N = 13), inoculated intracerebrally with 9L gliosarcoma cells, was treated with dexamethasone (i.p. 3 mg/kg/day) for five consecutive days, and another cohort (N = 11) was treated with equal volume of saline. Longitudinal DSC-MRI studies were performed at the first (baseline), third and fifth day of treatments. Relative cerebral blood volume (rCBV) was significantly reduced on the third day of dexamethasone treatment (0.65 ± .13) as compared to the fifth day during treatment (1.26 ±.19, p < 0.05). In saline treated rats, relative CBV gradually increased during treatment (0.89 ±.13, 1.00 ± .21, 1.13 ± .23) with no significant difference on the third day of treatment (p>0.05). In separate serial studies, microfocal X-ray CT of ex vivo brain specimens (N = 9) and immunohistochemistry for endothelial cell marker anti-CD31 (N = 8) were performed. Vascular morphology of ex vivo rat brains from micro-CT analysis showed hypervascular characteristics in tumors, and both vessel density (41.32 ± 2.34 branches/mm(3), p<0.001) and vessel tortuosity (p<0.05) were significantly reduced in tumors of rats treated with dexamethasone compared to saline (74.29 ± 3.51 branches/mm(3)). The vascular architecture of rat brain tissue was examined with anti-CD31 antibody, and dexamethasone treated tumor regions showed reduced vessel area (16.45 ± 1.36 µm(2)) as compared to saline treated tumor regions (30.83 ± 4.31 µm(2), p<0.001) and non-tumor regions (22.80 ± 1.11 µm(2), p<0.01). CONCLUSIONS/SIGNIFICANCE Increased vascular density and tortuosity are culprit to abnormal perfusion, which is transiently reduced during dexamethasone treatment.
Collapse
Affiliation(s)
- Moses M. Darpolor
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin, United States of America
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Robert C. Molthen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
26
|
Underhill HR, Yuan C, Hayes CE. A combined solenoid-surface RF coil for high-resolution whole-brain rat imaging on a 3.0 Tesla clinical MR scanner. Magn Reson Med 2011; 64:883-92. [PMID: 20535812 DOI: 10.1002/mrm.22466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Rat brain models effectively simulate a multitude of human neurological disorders. Improvements in coil design have facilitated the wider utilization of rat brain models by enabling the utilization of clinical MR scanners for image acquisition. In this study, a novel coil design, subsequently referred to as the rat brain coil, is described that exploits and combines the strengths of both solenoids and surface coils into a simple, multichannel, receive-only coil dedicated to whole-brain rat imaging on a 3.0 T clinical MR scanner. Compared with a multiturn solenoid mouse body coil, a 3-cm surface coil, a modified Helmholtz coil, and a phased-array surface coil, the rat brain coil improved signal-to-noise ratio by approximately 72, 61, 78, and 242%, respectively. Effects of the rat brain coil on amplitudes of static field and radiofrequency field uniformity were similar to each of the other coils. In vivo, whole-brain images of an adult male rat were acquired with a T(2)-weighted spin-echo sequence using an isotropic acquisition resolution of 0.25 x 0.25 x 0.25 mm(3) in 60.6 min. Multiplanar images of the in vivo rat brain with identification of anatomic structures are presented. Improvement in signal-to-noise ratio afforded by the rat brain coil may broaden experiments that utilize clinical MR scanners for in vivo image acquisition.
Collapse
Affiliation(s)
- Hunter R Underhill
- Department of Radiology, University of Washington, Seattle, Washington 98109, USA.
| | | | | |
Collapse
|
27
|
Shen Y, Ahearn T, Clemence M, Schwarzbauer C. Magnetic resonance imaging of the mean venous vessel size in the human brain using transient hyperoxia. Neuroimage 2011; 55:1063-7. [PMID: 21224003 DOI: 10.1016/j.neuroimage.2010.12.084] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 11/29/2010] [Accepted: 12/03/2010] [Indexed: 10/18/2022] Open
Abstract
Vessel size imaging is an emerging magnetic resonance imaging (MRI) technique which has been demonstrated to provide clinically relevant information about microvascular morphology. While previous studies of vessel size in humans relied on MRI contrast agents or hypercapnia-induced changes in blood oxygenation, the technique described here uses transient hyperoxia to alter the venous blood oxygenation. The experimental paradigm consisted of two 3-minute intervals of breathing 100% O(2) interleaved with three 2-minute intervals of breathing room air. Parametric maps of the mean venous vessel radius were calculated from changes in the blood oxygenation level dependent (BOLD) contrast which were measured using a combined spin-echo (SE) and gradient echo (GE) echo-planar imaging (EPI) sequence. The corresponding mean values in grey and white matter were r=6.5±0.3 μm and r=6.2±0.3 μm (n=6). While the hypercapnia technique requires a specialised gas mixture containing a low concentration of CO(2) (typically 5-6%), the hyperoxia technique presented here uses the inhalation of medical oxygen (100% O(2)) which is routinely available in a clinical environment. Furthermore, 100% O(2) is generally better tolerated than low doses of CO(2) which makes this technique particularly suitable for applications in critically ill patients.
Collapse
Affiliation(s)
- Yuji Shen
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Scotland, UK
| | | | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW To highlight the most recent advances in the imaging of gliomas with a focus on high-grade gliomas. In the last several years, MRI and PET imaging of the brain have moved beyond anatomic imaging and are now capable of probing different aspects of tumor biology and response to treatment. These new techniques are increasingly being incorporated into clinical trials and even into clinical practice. Therefore, it is important to be familiar with the available imaging techniques and their potential uses versus limitations. RECENT FINDINGS In 2010, updated response assessment criteria for high-grade gliomas were published. It was the first revision to imaging response criteria for gliomas since 1990 and represents an international effort to standardize criteria for clinical trials. Although this revision represents an important development, there are several promising imaging techniques on the horizon such as vascular permeability measurement, perfusion imaging, diffusion imaging, and new PET tracers that will continue to improve our understanding of tumor biology. SUMMARY Advances in neuroimaging of brain tumors are enabling investigators to noninvasively visualize tumor response to treatment over time. As these tools are validated, they will add substantially to our ability to understand glioma biology and response to therapy.
Collapse
Affiliation(s)
- Elizabeth R Gerstner
- Pappas Center for Neuro-Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA.
| | | |
Collapse
|
29
|
Just N. Histogram analysis of the microvasculature of intracerebral human and murine glioma xenografts. Magn Reson Med 2010; 65:778-89. [PMID: 21337410 DOI: 10.1002/mrm.22675] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 08/26/2010] [Accepted: 09/14/2010] [Indexed: 02/04/2023]
Abstract
The purpose of this study is to examine the usefulness of histogram analysis combined with vessel size index (VSI) magnetic resonance imaging for the specific characterization of brain tumor microvasculature in a panel of six volume-matched glioma xenografts. Using a simple descriptive histogram analysis, significant differences of the mean tumoral VSI (P=0.0035 for 9L, P=0.008 for glioma mix, P=0.05 for C6), the 75th VSI percentile (P=0.003-0.075) as well as the 25th and median blood volume (BV) percentiles were found in murine gliomas compared to their contralateral healthy brain. Using a segmented histogram analysis, dilatation of already existing vessels in murine gliomas and development of new small caliber vessels in human glioblastomas were suggested. Most gliomas showed a higher proportion of pixels with BV below 1% (glioma mix [21% vs 1%], Glioblastoma 2 (GBM2) [9% vs 3.7%]) and a smaller proportion of pixels with BV in the range 1.7-6.3% (65 vs 90% for glioma mix, 80 vs 85% in GBM2) relative to their contralateral part. In glioblastomas, VSI and BV distributions were similar to normal brain distributions and in agreement with immunohistochemical findings. The histogram analysis of VSI and BV heterogeneity in experimental brain tumors allowed detection of microregional differences in gliomas from different origins.
Collapse
Affiliation(s)
- Nathalie Just
- Laboratory for functional and metabolic imaging (LIFMET), EPFL, UNIL, Centre d'Imagerie Biomédicale, Department of Radiology, Lausanne, Switzerland.
| |
Collapse
|
30
|
[Role of perfusion, vascular permeability and anatomic MR imaging in radiation therapy for gliomas]. Bull Cancer 2010; 97:753-8. [PMID: 20610378 DOI: 10.1684/bdc.2010.1146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is no clear consensus for tumour volume definition in radiotherapy of brain tumours, particularly for high-grade gliomas (HGG). They are infiltrative and heterogeneous, sub-populations of low and high grade can coexist inside one tumour volume, and peritumoral oedema is partly due to a vasogenic mechanism but also to a microscopic extension of sparse tumour cells. All these characteristics are not directly detectable using a conventional MR imaging (MRI). Complementary to the anatomical sequences (T1/T2), still always mandatory, functional maps using the dynamic MRI with a T2* weighted sequence reflect micro-vessel perfusion and permeability, more on a quantitative aspect and a qualitative one, respectively. These parameters better appreciate neo-vascularity of gliomas and areas associated with a higher value of perfusion are clearly correlated with a higher grade. Even a low-grade glioma but with detectable areas of high permeability presents a two-fold risk of recurrence versus another one with the same anatomical characteristics and treatment, but without any micro-vascular leakage. For high-grade gliomas, a high level of tissue perfusion seems to be better predictive for the risk of recurrence than histology itself. The exact co-registration of anatomic and vascular maps is currently available in clinical practice and can be incorporated during the dedicated brain MRI for radiotherapy. Its potential for better predicting the exact sites of recurrence after treatment has to be prospectively evaluated and a strong interest for a dose-escalating study is evident. Finally, T2* dynamic MRI has the ability to differentiate post-treatment modifications from recurrence better than conventional imaging.
Collapse
|
31
|
Thompson G, Mills SJ, Stivaros SM, Jackson A. Imaging of Brain Tumors: Perfusion/Permeability. Neuroimaging Clin N Am 2010; 20:337-53. [DOI: 10.1016/j.nic.2010.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Hokland SL, Nielsen T, Busk M, Horsman MR. Imaging tumour physiology and vasculature to predict and assess response to heat. Int J Hyperthermia 2010; 26:264-72. [PMID: 20388023 DOI: 10.3109/02656730903585982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The vascular supply of tumours and the tumour microenvironment both play an important role when tumours are treated with hyperthermia. Blood flow is one of the major vehicles by which heat is dissipated thus the vascular supply will influence the ability to heat the tumour. It also influences the type of microenvironment that exists within tumours, and it is now well-established that cells existing in areas of oxygen deficiency, nutrient deprivation and acidic conditions are more sensitive to the effect of hyperthermia. The vascular supply and microenvironment are also affected by hyperthermia. In general, mild heat temperatures transiently improve blood flow and oxygenation, while higher hyperthermia temperatures cause vascular collapse and so increase the adverse microenvironmental conditions. Being able to image these vascular and microenvironmental parameters both before and after heating will help in our ability to predict and assess response. Here we review the various techniques that can be applied to supply this information, especially using non-invasive imaging approaches.
Collapse
Affiliation(s)
- Steffen L Hokland
- Department of Experimental Clinical Oncology, Aarhus University Hospital NBG, Aarhus, Denmark
| | | | | | | |
Collapse
|
33
|
Jochimsen TH, Ivanov D, Ott DV, Heinke W, Turner R, Möller HE, Reichenbach JR. Whole-brain mapping of venous vessel size in humans using the hypercapnia-induced BOLD effect. Neuroimage 2010; 51:765-74. [DOI: 10.1016/j.neuroimage.2010.02.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 01/07/2010] [Accepted: 02/13/2010] [Indexed: 10/19/2022] Open
|
34
|
Garteiser P, Doblas S, Watanabe Y, Saunders D, Hoyle J, Lerner M, He T, Floyd RA, Towner RA. Multiparametric assessment of the anti-glioma properties of OKN007 by magnetic resonance imaging. J Magn Reson Imaging 2010; 31:796-806. [DOI: 10.1002/jmri.22106] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
35
|
Advanced Imaging of Adult Brain Tumors with MRI and PET. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/b978-0-7506-7516-1.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
36
|
Lacerda S, Law M. Magnetic Resonance Perfusion and Permeability Imaging in Brain Tumors. Neuroimaging Clin N Am 2009; 19:527-57. [DOI: 10.1016/j.nic.2009.08.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
37
|
Pike MM, Stoops CN, Langford CP, Akella NS, Nabors LB, Gillespie GY. High-resolution longitudinal assessment of flow and permeability in mouse glioma vasculature: Sequential small molecule and SPIO dynamic contrast agent MRI. Magn Reson Med 2009; 61:615-25. [PMID: 19235262 DOI: 10.1002/mrm.21931] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The poor prognosis associated with malignant glioma is largely attributable to its invasiveness and robust angiogenesis. Angiogenesis involves host-tumor interaction and requires in vivo evaluation. Despite their versatility, few studies have used mouse glioma models with perfusion MRI approaches, and generally lack longitudinal study design. Using a micro-MRI system (8.5 Tesla), a novel dual bolus-tracking perfusion MRI strategy was implemented. Using the small molecule contrast agent Magnevist, dynamic contrast enhanced MRI was implemented in the intracranial 4C8 mouse glioma model to determine K(trans) and v(e), indices of tumor vascular permeability and cellularity, respectively. Dynamic susceptibility contrast MRI was subsequently implemented to assess both cerebral blood flow and volume, using the macromolecular superparamagnetic iron oxide, Feridex, which circumvented tumor bolus susceptibility curve distortions from first-pass extravasation. The high-resolution parametric maps obtained over 4 weeks, indicated a progression of tumor vascularization, permeability, and decreased cellularity with tumor growth. In conclusion, a comprehensive array of key parameters were reliably quantified in a longitudinal mouse glioma study. The syngeneic 4C8 intracerebral mouse tumor model has excellent characteristics for studies of glioma angiogenesis. This approach provides a useful platform for noninvasive and highly diagnostic longitudinal investigations of anti-angiogenesis strategies in a relevant orthotopic animal model.
Collapse
Affiliation(s)
- M M Pike
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Valable S, Eddi D, Constans JM, Guillamo JS, Bernaudin M, Roussel S, Petit E. MRI assessment of hemodynamic effects of angiopoietin-2 overexpression in a brain tumor model. Neuro Oncol 2009; 11:488-502. [PMID: 19168695 DOI: 10.1215/15228517-2008-117] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Despite treatment efforts, the median survival in patients with glioblastoma multiforme, the most aggressive form of glioma, does not extend beyond 12-15 months. One of the major pathophysiological characteristics of these tumors is their ability to induce active angiogenesis. Thus, based on the lack of efficient therapies, agents that inhibit angiogenesis are particularly attractive as a therapeutic option. However, it has been recently proposed that although specifically targeting vascular endothelial growth factor, the main angiogenic factor, certainly leads to significant tumor regression, it could also be followed by tumor relapses. In this case, angiogenesis is driven by alternate pathways that include other angiogenic factors. One possible strategy to overcome this therapeutic obstacle is to overexpress antivascular factors such as angiopoietin-2 (Ang2). Here, by using MRI and histological analysis, we studied the vascular events involved in glioma growth impairment induced by Ang2 overexpression. Our results show that an increase in Ang2 expression, during the tumor growth, leads to a significant decrease in tumor growth ( approximately 86%) along with an increase in the survival median ( approximately 70%) but does not modify the tumor vascular area or cerebral blood volume. However, tumor Ang2-derived blood vessels display an abnormal, enlarged morphology. We show that the presence of Ang2 leads to an enhancement of tumor perfusion and a decrease in tumor vessel permeability. Based on our MR image evaluations of hemodynamic tumor vessel changes, we propose that Ang2-derived tumor vessels lead to an inadequate oxygenation of the tumor tissue, leading to impairment in tumor growth.
Collapse
|
39
|
Advances in neuroimaging techniques for the evaluation of tumor growth, vascular permeability, and angiogenesis in gliomas. Curr Opin Neurol 2008; 21:728-35. [DOI: 10.1097/wco.0b013e328318402a] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Pathak AP. Magnetic resonance susceptibility based perfusion imaging of tumors using iron oxide nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2008; 1:84-97. [DOI: 10.1002/wnan.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Arvind P. Pathak
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University In Vivo Cellular Molecular Imaging Center (JHU ICMIC), Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Consider Hidden IT Costs When Purchasing an MRI or CT Scanner. J Am Coll Radiol 2008; 5:935-6. [DOI: 10.1016/j.jacr.2008.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Indexed: 11/22/2022]
|