1
|
Arevalo-Perez J, Yllera-Contreras E, Peck KK, Hatzoglou V, Yildirim O, Rosenblum MK, Holodny AI. Differentiating Low-Grade from High-Grade Intracranial Ependymomas: Comparison of Dynamic Contrast-Enhanced MRI and Diffusion-Weighted Imaging. AJNR Am J Neuroradiol 2024; 45:927-933. [PMID: 38782589 DOI: 10.3174/ajnr.a8226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/07/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND PURPOSE The aim of this study was to determine the diagnostic value of fractional plasma volume derived from dynamic contrast-enhanced perfusion MR imaging versus ADC, obtained from DWI in differentiating between grade 2 (low-grade) and grade 3 (high-grade) intracranial ependymomas. MATERIALS AND METHODS A hospital database was created for the period from January 2013 through June 2022, including patients with histologically-proved ependymoma diagnosis with available dynamic contrast-enhanced MR imaging. Both dynamic contrast-enhanced perfusion and DWI were performed on each patient using 1.5T and 3T scanners. Fractional plasma volume maps and ADC maps were calculated. ROIs were defined by a senior neuroradiologist manually by including the enhancing tumor on every section and conforming a VOI to obtain the maximum value of fractional plasma volume (Vpmax) and the minimum value of ADC (ADCmin). A Mann-Whitney U test at a significance level of corrected P = .01 was used to evaluate the differences. Additionally, receiver operating characteristic curve analysis was applied to assess the sensitivity and specificity of Vpmax and ADCmin values. RESULTS A total of 20 patients with ependymomas (10 grade 2 tumors and 10 grade 3 tumors) were included. Vpmax values for grade 3 ependymomas were significantly higher (P < .002) than those for grade 2. ADCmin values were overall lower in high-grade lesions. However, no statistically significant differences were found (P = .12114). CONCLUSIONS As a dynamic contrast-enhanced perfusion MR imaging metric, fractional plasma volume can be used as an indicator to differentiate grade 2 and grade 3 ependymomas. Dynamic contrast-enhanced perfusion MR imaging plays an important role with high diagnostic value in differentiating low- and high-grade ependymoma.
Collapse
Affiliation(s)
- Julio Arevalo-Perez
- From the Department of Radiology (J.A.-P., E.Y.-C., V.H., O.Y., A.I.H.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elena Yllera-Contreras
- From the Department of Radiology (J.A.-P., E.Y.-C., V.H., O.Y., A.I.H.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kyung K Peck
- Department of Medical Physics (K.K.P.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vaios Hatzoglou
- From the Department of Radiology (J.A.-P., E.Y.-C., V.H., O.Y., A.I.H.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Onur Yildirim
- From the Department of Radiology (J.A.-P., E.Y.-C., V.H., O.Y., A.I.H.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc K Rosenblum
- Department of Pathology (M.K.R.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrei I Holodny
- From the Department of Radiology (J.A.-P., E.Y.-C., V.H., O.Y., A.I.H.), Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
2
|
Prah MA, Schmainda KM. Practical guidance to identify and troubleshoot suboptimal DSC-MRI results. FRONTIERS IN RADIOLOGY 2024; 4:1307586. [PMID: 38445104 PMCID: PMC10913595 DOI: 10.3389/fradi.2024.1307586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
Relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) perfusion MR imaging (pMRI) has been shown to be a robust marker of neuroradiological tumor burden. Recent consensus recommendations in pMRI acquisition strategies have provided a pathway for pMRI inclusion in diverse patient care centers, regardless of size or experience. However, even with proper implementation and execution of the DSC-MRI protocol, issues will arise that many centers may not easily recognize or be aware of. Furthermore, missed pMRI issues are not always apparent in the resulting rCBV images, potentiating inaccurate or missed radiological diagnoses. Therefore, we gathered from our database of DSC-MRI datasets, true-to-life examples showcasing the breakdowns in acquisition, postprocessing, and interpretation, along with appropriate mitigation strategies when possible. The pMRI issues addressed include those related to image acquisition and postprocessing with a focus on contrast agent administration, timing, and rate, signal-to-noise quality, and susceptibility artifact. The goal of this work is to provide guidance to minimize and recognize pMRI issues to ensure that only quality data is interpreted.
Collapse
Affiliation(s)
- Melissa A. Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI,United States
| |
Collapse
|
3
|
Chan RW, Lam WW, Chen H, Murray L, Zhang B, Theriault A, Endre R, Moon S, Liebig P, Maralani PJ, Tseng CL, Myrehaug S, Detsky J, Lim-Fat MJ, Roberto K, Djayakarsana D, Lingamoorthy B, Mehrabian H, Khan BM, Sahgal A, Soliman H, Stanisz GJ. Is pulsed saturation transfer sufficient for differentiating radiation necrosis from tumor progression in brain metastases? Neurooncol Adv 2024; 6:vdae132. [PMID: 39220250 PMCID: PMC11364936 DOI: 10.1093/noajnl/vdae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Background Stereotactic radiosurgery (SRS) for the treatment of brain metastases delivers a high dose of radiation with excellent local control but comes with the risk of radiation necrosis (RN), which can be difficult to distinguish from tumor progression (TP). Magnetization transfer (MT) and chemical exchange saturation transfer (CEST) are promising techniques for distinguishing RN from TP in brain metastases. Previous studies used a 2D continuous-wave (ie, block radiofrequency [RF] saturation) MT/CEST approach. The purpose of this study is to investigate a 3D pulsed saturation MT/CEST approach with perfusion MRI for distinguishing RN from TP in brain metastases. Methods The study included 73 patients scanned with MT/CEST MRI previously treated with SRS or fractionated SRS who developed enhancing lesions with uncertain diagnoses of RN or TP. Perfusion MRI was acquired in 49 of 73 patients. Clinical outcomes were determined by at least 6 months of follow-up or via pathologic confirmation (in 20% of the lesions). Results Univariable logistic regression resulted in significant variables of the quantitative MT parameter 1/(RA·T2A), with 5.9 ± 2.7 for RN and 6.5 ± 2.9 for TP. The highest AUC of 75% was obtained using a multivariable logistic regression model for MT/CEST parameters, which included the CEST parameters of AREXAmide,0.625µT (P = .013), AREXNOE,0.625µT (P = .008), 1/(RA·T2A) (P = .004), and T1 (P = .004). The perfusion rCBV parameter did not reach significance. Conclusions Pulsed saturation transfer was sufficient for achieving a multivariable AUC of 75% for differentiating between RN and TP in brain metastases, but had lower AUCs compared to previous studies that used a block RF approach.
Collapse
Affiliation(s)
- Rachel W Chan
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Wilfred W Lam
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Hanbo Chen
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Leedan Murray
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Beibei Zhang
- Department of Medical Physics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Aimee Theriault
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Ruby Endre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Sangkyu Moon
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | - Pejman J Maralani
- Department of Medical Imaging, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Katrina Roberto
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Daniel Djayakarsana
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Hatef Mehrabian
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Benazir Mir Khan
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Hany Soliman
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre & University of Toronto, Toronto, Ontario, Canada
| | - Greg J Stanisz
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
4
|
Tseng CH, Jaspers J, Romero AM, Wielopolski P, Smits M, van Osch MJP, Vos F. Improved reliability of perfusion estimation in dynamic susceptibility contrast MRI by using the arterial input function from dynamic contrast enhanced MRI. NMR IN BIOMEDICINE 2024; 37:e5038. [PMID: 37712359 DOI: 10.1002/nbm.5038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/02/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
The arterial input function (AIF) plays a crucial role in estimating quantitative perfusion properties from dynamic susceptibility contrast (DSC) MRI. An important issue, however, is that measuring the AIF in absolute contrast-agent concentrations is challenging, due to uncertainty in relation to the measuredR 2 ∗ -weighted signal, signal depletion at high concentration, and partial-volume effects. A potential solution could be to derive the AIF from separately acquired dynamic contrast enhanced (DCE) MRI data. We aim to compare the AIF determined from DCE MRI with the AIF from DSC MRI, and estimated perfusion coefficients derived from DSC data using a DCE-driven AIF with perfusion coefficients determined using a DSC-based AIF. AIFs were manually selected in branches of the middle cerebral artery (MCA) in both DCE and DSC data in each patient. In addition, a semi-automatic AIF-selection algorithm was applied to the DSC data. The amplitude and full width at half-maximum of the AIFs were compared statistically using the Wilcoxon rank-sum test, applying a 0.05 significance level. Cerebral blood flow (CBF) was derived with different AIF approaches and compared further. The results showed that the AIFs extracted from DSC scans yielded highly variable peaks across arteries within the same patient. The semi-automatic DSC-AIF had significantly narrower width compared with the manual AIFs, and a significantly larger peak than the manual DSC-AIF. Additionally, the DCE-based AIF provided a more stable measurement of relative CBF and absolute CBF values estimated with DCE-AIFs that were compatible with previously reported values. In conclusion, DCE-based AIFs were reproduced significantly better across vessels, showed more realistic profiles, and delivered more stable and reasonable CBF measurements. The DCE-AIF can, therefore, be considered as an alternative AIF source for quantitative perfusion estimations in DSC MRI.
Collapse
Affiliation(s)
- Chih-Hsien Tseng
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
| | - Jaap Jaspers
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Alejandra Mendez Romero
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Piotr Wielopolski
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marion Smits
- Medical Delta, Delft, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Brain Tumour Center, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Matthias J P van Osch
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frans Vos
- Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
- Medical Delta, Delft, the Netherlands
- Holland Proton Therapy Center Consortium-Erasmus MC, Rotterdam, Holland Proton Therapy Centre, Delft, Leiden University Medical Center, Leiden and Delft University of Technology, Delft, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Senger KPS, Kesavadas C, Thomas B, Singh A, Multani GS, AN D, Label M, Suchandrima B, Shin D. Experimenting with ASL-based arterialized cerebral blood volume as a novel imaging biomarker in grading glial neoplasms. Neuroradiol J 2023; 36:728-735. [PMID: 37548164 PMCID: PMC10649543 DOI: 10.1177/19714009231193163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Perfusion imaging is one of the methods used to grade glial neoplasms, and in this study we evaluated the role of ASL perfusion in grading brain glioma. PURPOSE The aim is to evaluate the role of arterialized cerebral blood volume (aCBV) of multi-delay ASL perfusion for grading glial neoplasm. MATERIALS AND METHODS This study is a prospective observational study of 56 patients with glial neoplasms of the brain who underwent surgery, and only cases with positive diagnosis of glioma are included to evaluate the novel diagnostic parameter. RESULTS In the study, ASL-derived normalized aCBV (naCBV) and T2*DSC-derived normalized CBV (nCBV) are showing very high correlation (Pearson's correlation coefficient value of 0.94) in grading glial neoplasms. naCBV and nCBF are also showing very high correlation (Pearson's correlation coefficient value of 0.876). The study also provides cutoff values for differentiating LGG from HGG for normalized aCBV(naCBV) of ASL, normalized CBV (nCBV), and normalized nCBF derived from T2* DCS as 1.12, 1.254, and 1.31, respectively. ASL-derived aCBV also shows better diagnostic accuracy than ASL-derived CBF. CONCLUSION This study is one of its kind to the best of our knowledge where multi-delay ASL perfusion-derived aCBV is used as a novel imaging biomarker for grading glial neoplasms, and it has shown high statistical correlation with T2* DSC-derived perfusion parameters.
Collapse
Affiliation(s)
- Krishna Pratap Singh Senger
- 1Department of Imaging Sciences and Interventional Radiology, Sree Chita Institute of Medical Sciences and Technology, Trivandrum, Kerala, India
| | - C Kesavadas
- 1Department of Imaging Sciences and Interventional Radiology, Sree Chita Institute of Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Bejoy Thomas
- 1Department of Imaging Sciences and Interventional Radiology, Sree Chita Institute of Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Ankita Singh
- Department of Research, Army Hospital Research and Referral, New Delhi, India
| | - Gurpreet Singh Multani
- 1Department of Imaging Sciences and Interventional Radiology, Sree Chita Institute of Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Deepti AN
- 1Department of Imaging Sciences and Interventional Radiology, Sree Chita Institute of Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Marc Label
- Department of Research and Development, GEHealthcare, Calgary, AB, Canada
| | | | - David Shin
- Department of Research and Development, GEHealthcare, Calgary, AB, Canada
| |
Collapse
|
6
|
Wang F, Zhou X, Chen R, Kang J, Yang X, Lin J, Liu F, Cao D, Xing Z. Improved performance of non-preloaded and high flip-angle dynamic susceptibility contrast perfusion-weighted imaging sequences in the presurgical differentiation of brain lymphoma and glioblastoma. Eur Radiol 2023; 33:8800-8808. [PMID: 37439934 DOI: 10.1007/s00330-023-09917-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/26/2023] [Accepted: 05/08/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVE This study aimed to compare the accuracy of relative cerebral blood volume (rCBV) and percentage signal recovery (PSR) obtained from high flip-angle dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) sequences with and without contrast agent (CA) preload for presurgical discrimination of brain glioblastoma and lymphoma. METHODS Consecutive 336 patients (glioblastoma, 236; PCNSL, 100) were included. All the patients underwent DSC-PWI on 3.0-T magnetic resonance units before surgery. The rCBV and PSR with preloaded and non-preloaded CA were measured. The means of the continuous variables were compared using Welch's t-test. The diagnostic accuracies of the individual parameters were compared using the receiver operating characteristic curve analysis. RESULTS The rCBV was higher with preloaded CA than with non-preloaded CA (glioblastoma, 10.20 vs. 8.90, p = 0.020; PCNSL, 3.88 vs. 3.27, p = 0.020). The PSR was lower with preloaded CA than with non-preloaded CA (glioblastoma, 0.59 vs. 0.90; PCNSL, 0.70 vs. 1.63; all p < 0.001). Regarding the differentiation of glioblastoma and PCNSL, the AUC of rCBV with preloaded CA was indistinguishable from that of non-preloaded CA (0.940 vs. 0.949, p = 0.703), whereas the area under the curve of PSR with preloaded CA was lower than non-preloaded CA (0.529 vs. 0.884, p < 0.001). CONCLUSION With preloaded CA, diagnostic performance in differentiating glioblastoma and PCNSL did not improve for rCBV and it was decreased for PSR. Therefore, high flip-angle non-preload DSC-PWI sequences offer excellent accuracy and may be of choice sequence for presurgical discrimination of brain lymphoma and glioblastoma. CLINICAL RELEVANCE STATEMENT High flip-angle DSC-PWI using non-preloaded CA may be an excellent diagnostic method for distinguishing glioblastoma from PCNSL. KEY POINTS • Differentiating primary central nervous system lymphoma and glioblastoma accurately is critical for their management. • DSC-PWI sequences optimised for the most accurate CBV calculations may not be the optimal sequences for presurgical brain tumour diagnosis as they could be masquerading leakage phenomena that may provide interesting information in terms of differential diagnosis. • High flip-angle non-preloaded DSC-PWI sequences render the best accuracy in the presurgical differentiation of brain lymphoma and glioblastoma.
Collapse
Affiliation(s)
- Feng Wang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Xiaofang Zhou
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ruiquan Chen
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jie Kang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Xinyi Yang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jinzhu Lin
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Fang Liu
- Department of Hyperbaric Oxygen, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Dairong Cao
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China.
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
- Department of Radiology, Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
| | - Zhen Xing
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, 20 Cha-Zhong Road, Fuzhou, Fujian, 350005, People's Republic of China.
- Department of Radiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
| |
Collapse
|
7
|
Shiroishi MS, Weinert D, Cen SY, Varghese B, Dondlinger T, Prah M, Mendoza J, Nazemi S, Ameli N, Amini N, Shohas S, Chen S, Bigjahan B, Zada G, Chen T, Neman-Ebrahim J, Chang EL, Chow FE, Fan Z, Yang W, Attenello FJ, Ye J, Kim PE, Patel VN, Lerner A, Acharya J, Hu LS, Quarles CC, Boxerman JL, Wu O, Schmainda KM. A cross-sectional study to test equivalence of low- versus intermediate-flip angle dynamic susceptibility contrast MRI measures of relative cerebral blood volume in patients with high-grade gliomas at 1.5 Tesla field strength. Front Oncol 2023; 13:1156843. [PMID: 37799462 PMCID: PMC10548232 DOI: 10.3389/fonc.2023.1156843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
Introduction 1.5 Tesla (1.5T) remain a significant field strength for brain imaging worldwide. Recent computer simulations and clinical studies at 3T MRI have suggested that dynamic susceptibility contrast (DSC) MRI using a 30° flip angle ("low-FA") with model-based leakage correction and no gadolinium-based contrast agent (GBCA) preload provides equivalent relative cerebral blood volume (rCBV) measurements to the reference-standard acquisition using a single-dose GBCA preload with a 60° flip angle ("intermediate-FA") and model-based leakage correction. However, it remains unclear whether this holds true at 1.5T. The purpose of this study was to test this at 1.5T in human high-grade glioma (HGG) patients. Methods This was a single-institution cross-sectional study of patients who had undergone 1.5T MRI for HGG. DSC-MRI consisted of gradient-echo echo-planar imaging (GRE-EPI) with a low-FA without preload (30°/P-); this then subsequently served as a preload for the standard intermediate-FA acquisition (60°/P+). Both normalized (nrCBV) and standardized relative cerebral blood volumes (srCBV) were calculated using model-based leakage correction (C+) with IBNeuro™ software. Whole-enhancing lesion mean and median nrCBV and srCBV from the low- and intermediate-FA methods were compared using the Pearson's, Spearman's and intraclass correlation coefficients (ICC). Results Twenty-three HGG patients composing a total of 31 scans were analyzed. The Pearson and Spearman correlations and ICCs between the 30°/P-/C+ and 60°/P+/C+ acquisitions demonstrated high correlations for both mean and median nrCBV and srCBV. Conclusion Our study provides preliminary evidence that for HGG patients at 1.5T MRI, a low FA, no preload DSC-MRI acquisition can be an appealing alternative to the reference standard higher FA acquisition that utilizes a preload.
Collapse
Affiliation(s)
- Mark S. Shiroishi
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Marina del Rey, CA, United States
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Dane Weinert
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Steven Y. Cen
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Bino Varghese
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | | | - Melissa Prah
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jesse Mendoza
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Sina Nazemi
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Nima Ameli
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Negin Amini
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Salman Shohas
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Shannon Chen
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Bavrina Bigjahan
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Gabriel Zada
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Thomas Chen
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Josh Neman-Ebrahim
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Eric L. Chang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Frances E. Chow
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Zhaoyang Fan
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Wensha Yang
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Frank J. Attenello
- Department of Neurological Surgery, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Jason Ye
- Department of Radiation Oncology, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Paul E. Kim
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Vishal N. Patel
- Department of Radiology, Mayo Clinic, Jacksonville, FL, United States
| | - Alexander Lerner
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Jay Acharya
- Department of Radiology, Keck School of Medicine of the University of Southern California (USC), Los Angeles, CA, United States
| | - Leland S. Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - C. Chad Quarles
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jerrold L. Boxerman
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Ona Wu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathleen M. Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
8
|
Haase R, Pinetz T, Kobler E, Paech D, Effland A, Radbruch A, Deike-Hofmann K. Artificial Contrast: Deep Learning for Reducing Gadolinium-Based Contrast Agents in Neuroradiology. Invest Radiol 2023; 58:539-547. [PMID: 36822654 DOI: 10.1097/rli.0000000000000963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
ABSTRACT Deep learning approaches are playing an ever-increasing role throughout diagnostic medicine, especially in neuroradiology, to solve a wide range of problems such as segmentation, synthesis of missing sequences, and image quality improvement. Of particular interest is their application in the reduction of gadolinium-based contrast agents, the administration of which has been under cautious reevaluation in recent years because of concerns about gadolinium deposition and its unclear long-term consequences. A growing number of studies are investigating the reduction (low-dose approach) or even complete substitution (zero-dose approach) of gadolinium-based contrast agents in diverse patient populations using a variety of deep learning methods. This work aims to highlight selected research and discusses the advantages and limitations of recent deep learning approaches, the challenges of assessing its output, and the progress toward clinical applicability distinguishing between the low-dose and zero-dose approach.
Collapse
Affiliation(s)
| | - Thomas Pinetz
- Institute of Applied Mathematics, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Erich Kobler
- From the Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn
| | | | - Alexander Effland
- Institute of Applied Mathematics, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | | |
Collapse
|
9
|
Haase R, Pinetz T, Bendella Z, Kobler E, Paech D, Block W, Effland A, Radbruch A, Deike-Hofmann K. Reduction of Gadolinium-Based Contrast Agents in MRI Using Convolutional Neural Networks and Different Input Protocols: Limited Interchangeability of Synthesized Sequences With Original Full-Dose Images Despite Excellent Quantitative Performance. Invest Radiol 2023; 58:420-430. [PMID: 36735399 DOI: 10.1097/rli.0000000000000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES The purpose of this study was to implement a state-of-the-art convolutional neural network used to synthesize artificial T1-weighted (T1w) full-dose images from corresponding noncontrast and low-dose images (using various settings of input sequences) and test its performance on a patient population acquired prospectively. MATERIALS AND METHODS In this monocentric, institutional review board-approved study, a total of 138 participants were included who received an adapted imaging protocol with acquisition of a T1w low dose after administration of 10% of the standard dose and acquisition of a T1w full dose after administration of the remaining 90% of the standard dose of a gadolinium-containing contrast agent. A total of 83 participants formed the training sample (51.7 ± 16.5 years, 36 women), 25 the validation sample (55.3 ± 16.4 years, 11 women), and 30 the test sample (55.0 ± 15.0 years, 9 women). Four input settings were differentiated: only the T1w noncontrast and T1w low-dose images (standard setting), only the T1w noncontrast and T1w low-dose images with a prolonged postinjection time of 5 minutes (5-minute setting), multiple noncontrast sequences (T1w, T2w, diffusion) and the T1w low-dose images (extended setting), and only noncontrast sequences (T1w, T2w, diffusion) were used (zero-dose setting). For each setting, a deep neural network was trained to synthesize artificial T1w full-dose images, which were assessed on the test sample using an objective evaluation based on quantitative metrics and a subjective evaluation through a reader-based study. Three readers scored the overall image quality, the interchangeability in regard to the clinical conclusion compared with the true T1w full-dose sequence, the contrast enhancement of lesions, and their conformity to the respective references in the true T1w full dose. RESULTS Quantitative analysis of the artificial T1w full-dose images of the standard setting provided a peak signal-to-noise ratio of 33.39 ± 0.62 (corresponding to an average improvement of the low-dose sequences of 5.2 dB) and a structural similarity index measure of 0.938 ± 0.005. In the 4-fold cross-validation, the extended setting yielded similar performance to the standard setting in terms of peak signal-to-noise ratio ( P = 0.20), but a slight improvement in structural similarity index measure ( P < 0.0001). For all settings, the reader study found comparable overall image quality between the original and artificial T1w full-dose images. The proportion of scans scored as fully or mostly interchangeable was 55%, 58%, 43%, and 3% and the average counts of false positives per case were 0.42 ± 0.83, 0.34 ± 0.71, 0.82 ± 1.15, and 2.00 ± 1.07 for the standard, 5-minute, extended, and zero-dose setting, respectively. Using a 5-point Likert scale (0 to 4, 0 being the worst), all settings of synthesized full-dose images showed significantly poorer contrast enhancement of lesions compared with the original full-dose sequence (difference of average degree of contrast enhancement-standard: -0.97 ± 0.83, P = <0.001; 5-minute: -0.93 ± 0.91, P = <0.001; extended: -0.96 ± 0.97, P = <0.001; zero-dose: -2.39 ± 1.14, P = <0.001). The average scores of conformity of the lesions compared with the original full-dose sequence were 2.25 ± 1.21, 2.22 ± 1.27, 2.24 ± 1.25, and 0.73 ± 0.93 for the standard, 5-minute, extended, and zero-dose setting, respectively. CONCLUSIONS The tested deep learning algorithm for synthesis of artificial T1w full-dose sequences based on images after administration of only 10% of the standard dose of a gadolinium-based contrast agent showed very good quantitative performance. Despite good image quality in all settings, both false-negative and false-positive signals resulted in significantly limited interchangeability of the synthesized sequences with the original full-dose sequences.
Collapse
Affiliation(s)
| | - Thomas Pinetz
- Institute of Applied Mathematics, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Zeynep Bendella
- From the Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn
| | - Erich Kobler
- From the Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn
| | | | - Wolfgang Block
- From the Department of Neuroradiology, University Medical Center Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn
| | - Alexander Effland
- Institute of Applied Mathematics, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | | |
Collapse
|
10
|
Venugopal K, Arzanforoosh F, van Dorth D, Smits M, van Osch MJP, Hernandez-Tamames JA, Warnert EAH, Poot DHJ. MR Vascular Fingerprinting with Hybrid Gradient–Spin Echo Dynamic Susceptibility Contrast MRI for Characterization of Microvasculature in Gliomas. Cancers (Basel) 2023; 15:cancers15072180. [PMID: 37046841 PMCID: PMC10093700 DOI: 10.3390/cancers15072180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Characterization of tumor microvasculature is important in tumor assessment and studying treatment response. This is possible by acquiring vascular biomarkers with magnetic resonance imaging (MRI) based on dynamic susceptibility contrast (DSC). We propose magnetic resonance vascular fingerprinting (MRVF) for hybrid echo planar imaging (HEPI) acquired during the first passage of the contrast agent (CA). The proposed approach was evaluated in patients with gliomas, and we simultaneously estimated vessel radius and relative cerebral blood volume. These parameters were also compared to the respective values estimated using the previously introduced vessel size imaging (VSI) technique. The results of both methods were found to be consistent. MRVF was also found to be robust to noise in the estimation of the parameters. DSC-HEPI-based MRVF provides characterization of microvasculature in gliomas with a short acquisition time and can be further improved in several ways to increase our understanding of tumor physiology.
Collapse
Affiliation(s)
- Krishnapriya Venugopal
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Fatemeh Arzanforoosh
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Daniëlle van Dorth
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Matthias J. P. van Osch
- C. J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Juan A. Hernandez-Tamames
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Medical Imaging, Faculty of Applied Physics, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Esther A. H. Warnert
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Dirk H. J. Poot
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
11
|
Powell SJ, Withey SB, Sun Y, Grist JT, Novak J, MacPherson L, Abernethy L, Pizer B, Grundy R, Morgan PS, Jaspan T, Bailey S, Mitra D, Auer DP, Avula S, Arvanitis TN, Peet A. Applying machine learning classifiers to automate quality assessment of paediatric dynamic susceptibility contrast (DSC-) MRI data. Br J Radiol 2023; 96:20201465. [PMID: 36802769 PMCID: PMC10161906 DOI: 10.1259/bjr.20201465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
OBJECTIVE Investigate the performance of qualitative review (QR) for assessing dynamic susceptibility contrast (DSC-) MRI data quality in paediatric normal brain and develop an automated alternative to QR. METHODS 1027 signal-time courses were assessed by Reviewer 1 using QR. 243 were additionally assessed by Reviewer 2 and % disagreements and Cohen's κ (κ) were calculated. The signal drop-to-noise ratio (SDNR), root mean square error (RMSE), full width half maximum (FWHM) and percentage signal recovery (PSR) were calculated for the 1027 signal-time courses. Data quality thresholds for each measure were determined using QR results. The measures and QR results trained machine learning classifiers. Sensitivity, specificity, precision, classification error and area under the curve from a receiver operating characteristic curve were calculated for each threshold and classifier. RESULTS Comparing reviewers gave 7% disagreements and κ = 0.83. Data quality thresholds of: 7.6 for SDNR; 0.019 for RMSE; 3 s and 19 s for FWHM; and 42.9 and 130.4% for PSR were produced. SDNR gave the best sensitivity, specificity, precision, classification error and area under the curve values of 0.86, 0.86, 0.93, 14.2% and 0.83. Random forest was the best machine learning classifier, giving sensitivity, specificity, precision, classification error and area under the curve of 0.94, 0.83, 0.93, 9.3% and 0.89. CONCLUSION The reviewers showed good agreement. Machine learning classifiers trained on signal-time course measures and QR can assess quality. Combining multiple measures reduces misclassification. ADVANCES IN KNOWLEDGE A new automated quality control method was developed, which trained machine learning classifiers using QR results.
Collapse
Affiliation(s)
- Stephen J Powell
- Physical Sciences for Health CDT, University of Birmingham, Birmingham, United Kingdom.,Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stephanie B Withey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.,Department of Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom.,RRPPS, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Yu Sun
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.,School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - James T Grist
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jan Novak
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.,Department of Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom.,Department of Psychology, Aston Brain Centre, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Lesley MacPherson
- Radiology, Birmingham Children's Hospital, Birmingham, United Kingdom
| | - Laurence Abernethy
- Radiology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Barry Pizer
- Oncology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Richard Grundy
- The Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Paul S Morgan
- The Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, United Kingdom.,Medical Physics, Nottingham University Hospitals, Nottingham, United Kingdom.,NIHR Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Tim Jaspan
- The Children's Brain Tumour Research Centre, University of Nottingham, Nottingham, United Kingdom.,Radiology, Nottingham University Hospitals, Nottingham, United Kingdom
| | - Simon Bailey
- Sir James Spence Institute of Child Health, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Dipayan Mitra
- Neuroradiology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Dorothee P Auer
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, United Kingdom
| | - Shivaram Avula
- Radiology, Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom
| | - Theodoros N Arvanitis
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.,Department of Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom.,Institute of Digital Healthcare, WMG, University of Warwick, Coventry, United Kingdom
| | - Andrew Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.,Department of Oncology, Birmingham Children's Hospital, Birmingham, United Kingdom
| |
Collapse
|
12
|
Bernstock JD, Gary SE, Klinger N, Valdes PA, Ibn Essayed W, Olsen HE, Chagoya G, Elsayed G, Yamashita D, Schuss P, Gessler FA, Peruzzi PP, Bag A, Friedman GK. Standard clinical approaches and emerging modalities for glioblastoma imaging. Neurooncol Adv 2022; 4:vdac080. [PMID: 35821676 PMCID: PMC9268747 DOI: 10.1093/noajnl/vdac080] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary adult intracranial malignancy and carries a dismal prognosis despite an aggressive multimodal treatment regimen that consists of surgical resection, radiation, and adjuvant chemotherapy. Radiographic evaluation, largely informed by magnetic resonance imaging (MRI), is a critical component of initial diagnosis, surgical planning, and post-treatment monitoring. However, conventional MRI does not provide information regarding tumor microvasculature, necrosis, or neoangiogenesis. In addition, traditional MRI imaging can be further confounded by treatment-related effects such as pseudoprogression, radiation necrosis, and/or pseudoresponse(s) that preclude clinicians from making fully informed decisions when structuring a therapeutic approach. A myriad of novel imaging modalities have been developed to address these deficits. Herein, we provide a clinically oriented review of standard techniques for imaging GBM and highlight emerging technologies utilized in disease characterization and therapeutic development.
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Sam E Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Neil Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Pablo A Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Hannah E Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Gustavo Chagoya
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Galal Elsayed
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
| | - Patrick Schuss
- Department of Neurosurgery, Unfallkrankenhaus Berlin , Berlin, Germany
| | | | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School , Boston, Massachusetts, USA
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital , Memphis, TN USA
| | - Gregory K Friedman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham , AL, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham , Birmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham , AL, USA
| |
Collapse
|
13
|
Stadlbauer A, Marhold F, Oberndorfer S, Heinz G, Buchfelder M, Kinfe TM, Meyer-Bäse A. Radiophysiomics: Brain Tumors Classification by Machine Learning and Physiological MRI Data. Cancers (Basel) 2022; 14:2363. [PMID: 35625967 PMCID: PMC9139355 DOI: 10.3390/cancers14102363] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 01/06/2023] Open
Abstract
The precise initial characterization of contrast-enhancing brain tumors has significant consequences for clinical outcomes. Various novel neuroimaging methods have been developed to increase the specificity of conventional magnetic resonance imaging (cMRI) but also the increased complexity of data analysis. Artificial intelligence offers new options to manage this challenge in clinical settings. Here, we investigated whether multiclass machine learning (ML) algorithms applied to a high-dimensional panel of radiomic features from advanced MRI (advMRI) and physiological MRI (phyMRI; thus, radiophysiomics) could reliably classify contrast-enhancing brain tumors. The recently developed phyMRI technique enables the quantitative assessment of microvascular architecture, neovascularization, oxygen metabolism, and tissue hypoxia. A training cohort of 167 patients suffering from one of the five most common brain tumor entities (glioblastoma, anaplastic glioma, meningioma, primary CNS lymphoma, or brain metastasis), combined with nine common ML algorithms, was used to develop overall 135 classifiers. Multiclass classification performance was investigated using tenfold cross-validation and an independent test cohort. Adaptive boosting and random forest in combination with advMRI and phyMRI data were superior to human reading in accuracy (0.875 vs. 0.850), precision (0.862 vs. 0.798), F-score (0.774 vs. 0.740), AUROC (0.886 vs. 0.813), and classification error (5 vs. 6). The radiologists, however, showed a higher sensitivity (0.767 vs. 0.750) and specificity (0.925 vs. 0.902). We demonstrated that ML-based radiophysiomics could be helpful in the clinical routine diagnosis of contrast-enhancing brain tumors; however, a high expenditure of time and work for data preprocessing requires the inclusion of deep neural networks.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Institute of Medical Radiology, University Clinic St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
| | - Franz Marhold
- Department of Neurosurgery, University Clinic of St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic of St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Gertraud Heinz
- Institute of Medical Radiology, University Clinic St. Pölten, Karl Landsteiner University of Health Sciences, A-3100 St. Pölten, Austria;
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
| | - Thomas M. Kinfe
- Department of Neurosurgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany; (M.B.); (T.M.K.)
- Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Anke Meyer-Bäse
- Department of Scientific Computing, Florida State University, 400 Dirac Science Library, Tallahassee, FL 32306-4120, USA;
| |
Collapse
|
14
|
Malik DG, Rath TJ, Urcuyo Acevedo JC, Canoll PD, Swanson KR, Boxerman JL, Quarles CC, Schmainda KM, Burns TC, Hu LS. Advanced MRI Protocols to Discriminate Glioma From Treatment Effects: State of the Art and Future Directions. FRONTIERS IN RADIOLOGY 2022; 2:809373. [PMID: 37492687 PMCID: PMC10365126 DOI: 10.3389/fradi.2022.809373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/01/2022] [Indexed: 07/27/2023]
Abstract
In the follow-up treatment of high-grade gliomas (HGGs), differentiating true tumor progression from treatment-related effects, such as pseudoprogression and radiation necrosis, presents an ongoing clinical challenge. Conventional MRI with and without intravenous contrast serves as the clinical benchmark for the posttreatment surveillance imaging of HGG. However, many advanced imaging techniques have shown promise in helping better delineate the findings in indeterminate scenarios, as posttreatment effects can often mimic true tumor progression on conventional imaging. These challenges are further confounded by the histologic admixture that can commonly occur between tumor growth and treatment-related effects within the posttreatment bed. This review discusses the current practices in the surveillance imaging of HGG and the role of advanced imaging techniques, including perfusion MRI and metabolic MRI.
Collapse
Affiliation(s)
- Dania G. Malik
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Tanya J. Rath
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Javier C. Urcuyo Acevedo
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Peter D. Canoll
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Kristin R. Swanson
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Jerrold L. Boxerman
- Department of Diagnostic Imaging, Brown University, Providence, RI, United States
| | - C. Chad Quarles
- Department of Neuroimaging Research & Barrow Neuroimaging Innovation Center, Barrow Neurologic Institute, Phoenix, AZ, United States
| | - Kathleen M. Schmainda
- Department of Biophysics & Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Terry C. Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, Rochester, MN, United States
| | - Leland S. Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
15
|
Stadlbauer A, Kinfe TM, Zimmermann M, Eyüpoglu I, Brandner N, Buchfelder M, Zaiss M, Dörfler A, Brandner S. Association between tissue hypoxia, perfusion restrictions, and microvascular architecture alterations with lesion-induced impairment of neurovascular coupling. J Cereb Blood Flow Metab 2022; 42:526-539. [PMID: 32787542 PMCID: PMC8985434 DOI: 10.1177/0271678x20947546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Functional magnetic resonance imaging (fMRI) has been mainly utilized for the preoperative localization of eloquent cortical areas. However, lesion-induced impairment of neurovascular coupling (NVC) in the lesion border zone may lead to false-negative fMRI results. The purpose of this study was to determine physiological factors impacting the NVC. Twenty patients suffering from brain lesions were preoperatively examined using multimodal neuroimaging including fMRI, magnetoencephalography (MEG) during language or sensorimotor tasks (depending on lesion location), and a novel physiologic MRI approach for the combined quantification of oxygen metabolism, perfusion state, and microvascular architecture. Congruence of brain activity patterns between fMRI and MEG were found in 13 patients. In contrast, we observed missing fMRI activity in perilesional cortex that demonstrated MEG activity in seven patients, which was interpreted as lesion-induced impairment of NVC. In these brain regions with impaired NVC, physiologic MRI revealed significant brain tissue hypoxia, as well as significantly decreased macro- and microvascular perfusion and microvascular architecture. We demonstrated that perilesional hypoxia with reduced vascular perfusion and architecture is associated with lesion-induced impairment of NVC. Our physiologic MRI approach is a clinically applicable method for preoperative risk assessment for the presence of false-negative fMRI results and may prevent severe postoperative functional deficits.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany.,Institute of Medical Radiology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Thomas M Kinfe
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany.,Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Max Zimmermann
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany.,Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Ilker Eyüpoglu
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Nadja Brandner
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Moritz Zaiss
- Department of Neuroradiology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Dörfler
- Department of Neuroradiology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Brandner
- Department of Neurosurgery, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Cindil E, Sendur HN, Cerit MN, Erdogan N, Celebi F, Dag N, Celtikci E, Inan A, Oner Y, Tali T. Prediction of IDH Mutation Status in High-grade Gliomas Using DWI and High T1-weight DSC-MRI. Acad Radiol 2022; 29 Suppl 3:S52-S62. [PMID: 33685792 DOI: 10.1016/j.acra.2021.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/24/2021] [Accepted: 02/03/2021] [Indexed: 01/09/2023]
Abstract
RATIONALE AND OBJECTIVES We aimed to evaluate the diagnostic performance of diffusion-weighted imaging (DWI) and dynamic susceptibility contrast-enhanced (DSC) magnetic resonance imaging (MRI) parameters in the noninvasive prediction of the isocitrate dehydrogenase (IDH) mutation status in high-grade gliomas (HGGs). MATERIALS AND METHODS A total of 58 patients with histopathologically proved HGGs were included in this retrospective study. All patients underwent multiparametric MRI on 3-T, including DSC-MRI and DWI before surgery. The mean apparent diffusion coefficient (ADC), relative maximum cerebral blood volume (rCBV), and percentage signal recovery (PSR) of the tumor core were measured and compared depending on the IDH mutation status and tumor grade. The Mann-Whitney U test was used to detect statistically significant differences in parameters between IDH-mutant-type (IDH-m-type) and IDH-wild-type (IDH-w-type) HGGs. Receiver operating characteristic curve (ROC) analysis was performed to evaluate the diagnostic performance. RESULTS The rCBV was significantly higher, and the PSR value was significantly lower in IDH-w-type tumors than in the IDH-m group (p = 0.002 and <0.001, respectively).The ADC value in IDH-w-type tumors was significantly lower compared with the one in IDH-m types (p = 0.023), but remarkable overlaps were found between the groups. The PSR showed the best diagnostic performance with an AUC of 0.938 and with an accuracy rate of 0.87 at the optimal cutoff value of 86.85. The combination of the PSR and the rCBV for the identification of the IDH mutation status increased the discrimination ability at the AUC level of 0.955. In terms of each tumor grade, the PSR and rCBV showed significant differences between the IDH-m and IDH-w groups (p ≤0.001). CONCLUSION The rCBV and PSR from DSC-MRI may be feasible noninvasive imaging parameters for predicting the IDH mutation status in HGGs. The standardization of the imaging protocol is indispensable to the utility of DSC perfusion MRI in wider clinical usage.
Collapse
|
17
|
Stokes AM, Bergamino M, Alhilali L, Hu LS, Karis JP, Baxter LC, Bell LC, Quarles CC. Evaluation of single bolus, dual-echo dynamic susceptibility contrast MRI protocols in brain tumor patients. J Cereb Blood Flow Metab 2021; 41:3378-3390. [PMID: 34415211 PMCID: PMC8669280 DOI: 10.1177/0271678x211039597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Relative cerebral blood volume (rCBV) obtained from dynamic susceptibility contrast (DSC) MRI is adversely impacted by contrast agent leakage in brain tumors. Using simulations, we previously demonstrated that multi-echo DSC-MRI protocols provide improvements in contrast agent dosing, pulse sequence flexibility, and rCBV accuracy. The purpose of this study is to assess the in-vivo performance of dual-echo acquisitions in patients with brain tumors (n = 59). To verify pulse sequence flexibility, four single-dose dual-echo acquisitions were tested with variations in contrast agent dose, flip angle, and repetition time, and the resulting dual-echo rCBV was compared to standard single-echo rCBV obtained with preload (double-dose). Dual-echo rCBV was comparable to standard double-dose single-echo protocols (mean (standard deviation) tumor rCBV 2.17 (1.28) vs. 2.06 (1.20), respectively). High rCBV similarity was observed (CCC = 0.96), which was maintained across both flip angle (CCC = 0.98) and repetition time (CCC = 0.96) permutations, demonstrating that dual-echo acquisitions provide flexibility in acquisition parameters. Furthermore, a single dual-echo acquisition was shown to enable quantification of both perfusion and permeability metrics. In conclusion, single-dose dual-echo acquisitions provide similar rCBV to standard double-dose single-echo acquisitions, suggesting contrast agent dose can be reduced while providing significant pulse sequence flexibility and complementary tumor perfusion and permeability metrics.
Collapse
Affiliation(s)
- Ashley M Stokes
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Maurizio Bergamino
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Lea Alhilali
- Neuroradiology, Southwest Neuroimaging at Barrow Neurological Institute, Phoenix, AZ, USA
| | - Leland S Hu
- Department of Radiology, Division of Neuroradiology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - John P Karis
- Neuroradiology, Southwest Neuroimaging at Barrow Neurological Institute, Phoenix, AZ, USA
| | - Leslie C Baxter
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA.,Department of Radiology, Division of Neuroradiology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Laura C Bell
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
18
|
Jin S, Cho HJ. Model-free leakage index estimation of the blood-brain barrier using dual dynamic susceptibility contrast MRI acquisition. NMR IN BIOMEDICINE 2021; 34:e4570. [PMID: 34132432 DOI: 10.1002/nbm.4570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 06/12/2023]
Abstract
Pharmacokinetic K2 mapping from dynamic susceptibility contrast (DSC)-MRI can be a sensitive technique for evaluating the vascular permeability of the subtly damaged blood-brain barrier (BBB) in ischemic regions. However, the K2 values of ischemic lesions depend upon the selection of the intact BBB reference region. As previous observations suggest that the ΔR2* curve of pre-loaded DSC-MRI is not significantly affected by the extravasation of contrast agent, dual DSC-MRI acquisitions can be performed to derive the BBB leakage index from the voxel-wise reference input function for ischemic regions. This study aims to demonstrate the robustness of such model-free leakage index estimation in ischemic brains. By configuring the relationship between dual ΔR2* curves of the intact contralateral brain, the deviation of the measured ΔR2* curve from the unloaded DSC-MRI with respect to the non-deviated ΔR2* curve in the pre-loaded DSC-MRI can be quantified as the BBB leakage index. Such model-free leakage index values from rats with transient middle carotid artery occlusion (tMCAO) (n = 17) and normal controls (n = 3) were evaluated and compared with conventional K2 values with multiple reference regions. Inter-subject leakage index values were also compared with the corresponding ΔT1 map. Evans-blue-stained images were used to validate the leakage index. For the tMCAO group, leakage index values correlated well with ΔT1 (Pearson's r = 0.828). The hyperintense area on the leakage index map matched well with the corresponding Evans-blue-stained area (Dice correlation = 0.626). The slopes of the scatter-plot from the leakage index (0.97-1.00) were observed to be more robust against changes in the reference region than those from conventional K2 values (0.94-1.07). In a subtly damaged BBB tMCAO model, model-free evaluation of vascular permeability using dual DSC-MRIs would provide a consistent measure of inter-subject vascular permeability.
Collapse
Affiliation(s)
- Seokha Jin
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Hyung Joon Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| |
Collapse
|
19
|
Metabolic Tumor Microenvironment Characterization of Contrast Enhancing Brain Tumors Using Physiologic MRI. Metabolites 2021; 11:metabo11100668. [PMID: 34677383 PMCID: PMC8537028 DOI: 10.3390/metabo11100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The tumor microenvironment is a critical regulator of cancer development and progression as well as treatment response and resistance in brain neoplasms. The available techniques for investigation, however, are not well suited for noninvasive in vivo characterization in humans. A total of 120 patients (59 females; 61 males) with newly diagnosed contrast-enhancing brain tumors (64 glioblastoma, 20 brain metastases, 15 primary central nervous system (CNS) lymphomas (PCNSLs), and 21 meningiomas) were examined with a previously established physiological MRI protocol including quantitative blood-oxygen-level-dependent imaging and vascular architecture mapping. Six MRI biomarker maps for oxygen metabolism and neovascularization were fused for classification of five different tumor microenvironments: glycolysis, oxidative phosphorylation (OxPhos), hypoxia with/without neovascularization, and necrosis. Glioblastoma showed the highest metabolic heterogeneity followed by brain metastasis with a glycolysis-to-OxPhos ratio of approximately 2:1 in both tumor entities. In addition, glioblastoma revealed a significant higher percentage of hypoxia (24%) compared to all three other brain tumor entities: brain metastasis (7%; p < 0.001), PCNSL (8%; p = 0.001), and meningioma (8%; p = 0.003). A more aggressive biological brain tumor behavior was associated with a higher percentage of hypoxia and necrosis and a lower percentage of remaining vital tumor tissue and aerobic glycolysis. The proportion of oxidative phosphorylation, however, was rather similar (17–26%) for all four brain tumor entities. Tumor microenvironment (TME) mapping provides insights into neurobiological differences of contrast-enhancing brain tumors and deserves further clinical cancer research attention. Although there is a long roadmap ahead, TME mapping may become useful in order to develop new diagnostic and therapeutic approaches.
Collapse
|
20
|
Barajas RF, Politi LS, Anzalone N, Schöder H, Fox CP, Boxerman JL, Kaufmann TJ, Quarles CC, Ellingson BM, Auer D, Andronesi OC, Ferreri AJM, Mrugala MM, Grommes C, Neuwelt EA, Ambady P, Rubenstein JL, Illerhaus G, Nagane M, Batchelor TT, Hu LS. Consensus recommendations for MRI and PET imaging of primary central nervous system lymphoma: guideline statement from the International Primary CNS Lymphoma Collaborative Group (IPCG). Neuro Oncol 2021; 23:1056-1071. [PMID: 33560416 PMCID: PMC8248856 DOI: 10.1093/neuonc/noab020] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Advanced molecular and pathophysiologic characterization of primary central nervous system lymphoma (PCNSL) has revealed insights into promising targeted therapeutic approaches. Medical imaging plays a fundamental role in PCNSL diagnosis, staging, and response assessment. Institutional imaging variation and inconsistent clinical trial reporting diminishes the reliability and reproducibility of clinical response assessment. In this context, we aimed to: (1) critically review the use of advanced positron emission tomography (PET) and magnetic resonance imaging (MRI) in the setting of PCNSL; (2) provide results from an international survey of clinical sites describing the current practices for routine and advanced imaging, and (3) provide biologically based recommendations from the International PCNSL Collaborative Group (IPCG) on adaptation of standardized imaging practices. The IPCG provides PET and MRI consensus recommendations built upon previous recommendations for standardized brain tumor imaging protocols (BTIP) in primary and metastatic disease. A biologically integrated approach is provided to addresses the unique challenges associated with the imaging assessment of PCNSL. Detailed imaging parameters facilitate the adoption of these recommendations by researchers and clinicians. To enhance clinical feasibility, we have developed both “ideal” and “minimum standard” protocols at 3T and 1.5T MR systems that will facilitate widespread adoption.
Collapse
Affiliation(s)
- Ramon F Barajas
- Department of Radiology, Neuroradiology Section, Oregon Health & Science University, Portland Oregon, USA.,Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute Translational Oncology Program, Oregon Health & Science University, Portland, Oregon, USA
| | - Letterio S Politi
- Humanitas University and Humanitas Research and Clinical Center - IRCCS, Milan, Italy.,Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nicoletta Anzalone
- Neuroradiology Unit, IRCCS San Raffaele Hospital and Vita-Salute University, Milan, Italy
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christopher P Fox
- Department of Clinical Haematology, Nottingham University Hospitals NHS Trust, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jerrold L Boxerman
- Department of Diagnostic Imaging, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | | | - C Chad Quarles
- Department of Neuroimaging Research & Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, California, USA.,Departments of Radiological Sciences, Psychiatry, and Biobehavioral Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, California, USA
| | - Dorothee Auer
- Versus Arthritis Pain Centre, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ovidiu C Andronesi
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Andres J M Ferreri
- Lymphoma Unit, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maciej M Mrugala
- Department of Medicine, Division of Hematology and Oncology, Mayo Clinic Cancer Center, Phoenix, Arizona, USA.,Department of Neurology, Mayo Clinic, Phoenix, Arizona, USA
| | - Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Neurology, Weill Cornell Medical School, New York, New York, USA
| | - Edward A Neuwelt
- Blood-Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, USA.,Portland Veterans Affairs Medical Center, Portland, Oregon, USA
| | - Prakash Ambady
- Blood-Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - James L Rubenstein
- Division of Hematology/Oncology, University of California, San Francisco, California, USA.,Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Gerald Illerhaus
- Clinic of Hematology, Oncology and Palliative Care, Klinikum Stuttgart, Stuttgart, Germany
| | - Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leland S Hu
- Department of Radiology, Neuroradiology Division, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
21
|
Chen HSM, Jen ML, Hou P, Stafford RJ, Liu HL. A dynamic susceptibility contrast MRI digital reference object for testing software with leakage correction: Effect of background simulation. Med Phys 2021; 48:6051-6059. [PMID: 34293208 DOI: 10.1002/mp.15125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/22/2021] [Accepted: 07/17/2021] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Dynamic susceptibility contrast (DSC)-MRI is a perfusion imaging technique from which useful quantitative imaging biomarkers can be derived. Relative cerebral blood volume (rCBV) is such a biomarker commonly used for evaluating brain tumors. To account for the extravasation of contrast agents in tumors, post-processing leakage correction is often applied to improve rCBV accuracy. Digital reference objects (DRO) are ideal for testing the post-processing software, because the biophysical model used to generate the DRO can be matched to the one that the software uses. This study aims to develop DROs to validate the leakage correction of software using Weisskoff model and to examine the effect of background signal time curves that are required by the model. METHODS Three DROs were generated using the Weisskoff model, each composed of nine foreground lesion objects with combinations of different levels of rCBV and contrast leakage parameter (K2). Three types of background were implemented for these DROs: (1) a multi-compartment brain-like background, (2) a sphere background with a constant signal time curve, and (3) a sphere background with signal time curve identical to that of the brain-like DRO's white matter (WM). The DROs were then analyzed with an FDA-cleared software with and without leakage correction. Leakage correction was tested with and without brain segmentation. RESULTS Accuracy of leakage correction was able to be verified using the brain-like phantom and the sphere phantom with WM background. The sphere with constant background did not perform well with leakage correction with or without brain segmentation. The DROs were able to verify that for the particular software tested, leakage correction with brain segmentation achieved the lowest error. CONCLUSIONS DSC-MRI DROs with biophysical model matched to that of the post-processing software can be well used for the software's validation, provided that the background signals are also properly simulated for generating the reference time curve required by the model. Care needs to be taken to consider the interaction of the design of the DRO with the software's implementation of brain segmentation to extract the reference time curve.
Collapse
Affiliation(s)
- Henry Szu-Meng Chen
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mu-Lan Jen
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ping Hou
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Jason Stafford
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ho-Ling Liu
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Callewaert B, Jones EAV, Himmelreich U, Gsell W. Non-Invasive Evaluation of Cerebral Microvasculature Using Pre-Clinical MRI: Principles, Advantages and Limitations. Diagnostics (Basel) 2021; 11:diagnostics11060926. [PMID: 34064194 PMCID: PMC8224283 DOI: 10.3390/diagnostics11060926] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Alterations to the cerebral microcirculation have been recognized to play a crucial role in the development of neurodegenerative disorders. However, the exact role of the microvascular alterations in the pathophysiological mechanisms often remains poorly understood. The early detection of changes in microcirculation and cerebral blood flow (CBF) can be used to get a better understanding of underlying disease mechanisms. This could be an important step towards the development of new treatment approaches. Animal models allow for the study of the disease mechanism at several stages of development, before the onset of clinical symptoms, and the verification with invasive imaging techniques. Specifically, pre-clinical magnetic resonance imaging (MRI) is an important tool for the development and validation of MRI sequences under clinically relevant conditions. This article reviews MRI strategies providing indirect non-invasive measurements of microvascular changes in the rodent brain that can be used for early detection and characterization of neurodegenerative disorders. The perfusion MRI techniques: Dynamic Contrast Enhanced (DCE), Dynamic Susceptibility Contrast Enhanced (DSC) and Arterial Spin Labeling (ASL), will be discussed, followed by less established imaging strategies used to analyze the cerebral microcirculation: Intravoxel Incoherent Motion (IVIM), Vascular Space Occupancy (VASO), Steady-State Susceptibility Contrast (SSC), Vessel size imaging, SAGE-based DSC, Phase Contrast Flow (PC) Quantitative Susceptibility Mapping (QSM) and quantitative Blood-Oxygenation-Level-Dependent (qBOLD). We will emphasize the advantages and limitations of each strategy, in particular on applications for high-field MRI in the rodent's brain.
Collapse
Affiliation(s)
- Bram Callewaert
- Biomedical MRI Group, University of Leuven, Herestraat 49, bus 505, 3000 Leuven, Belgium; (B.C.); (W.G.)
- CMVB, Center for Molecular and Vascular Biology, University of Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium;
| | - Elizabeth A. V. Jones
- CMVB, Center for Molecular and Vascular Biology, University of Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium;
- CARIM, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Uwe Himmelreich
- Biomedical MRI Group, University of Leuven, Herestraat 49, bus 505, 3000 Leuven, Belgium; (B.C.); (W.G.)
- Correspondence:
| | - Willy Gsell
- Biomedical MRI Group, University of Leuven, Herestraat 49, bus 505, 3000 Leuven, Belgium; (B.C.); (W.G.)
| |
Collapse
|
23
|
Hypoxia and Microvascular Alterations Are Early Predictors of IDH-Mutated Anaplastic Glioma Recurrence. Cancers (Basel) 2021; 13:cancers13081797. [PMID: 33918764 PMCID: PMC8068871 DOI: 10.3390/cancers13081797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Anaplastic gliomas (AGs) are considered the most common and aggressive primary brain tumors of young adults with inevitable recurrence and treatment failure. The aim of this study was to investigate whether the imaging biomarkers of hypoxia, microvascular architecture and neovascularization activity can be of assistance to detect pathophysiological changes in the early developmental stages of isocitrate-dehydrogenase (IDH) mutated AG recurrence. We evaluated 142 physiological magnetic resonance imaging follow-up examinations as a part of the conventional magnetic resonance imaging (MRI) protocol in 60 AG patients after standard therapy. Physiological MRI biomarkers showed intensifying local tissue hypoxia 250 days prior to radiological recurrence with following upregulation of neovascularization activity 50 to 70 days later. Integration of physiological MRI in the monitoring of AG patients may be of clinical significance to make personalized decision of early tumor recurrence without an additional delay for multimodal therapy. Abstract Anaplastic gliomas (AG) represents aggressive brain tumors that often affect young adults. Although isocitrate-dehydrogenase (IDH) gene mutation has been identified as a more favorable prognostic factor, most IDH-mutated AG patients are confronted with tumor recurrence. Hence, increased knowledge about pathophysiological precursors of AG recurrence is urgently needed in order to develop precise diagnostic monitoring and tailored therapeutic approaches. In this study, 142 physiological magnetic resonance imaging (phyMRI) follow-up examinations in 60 AG patients after standard therapy were evaluated and magnetic resonance imaging (MRI) biomarker maps for microvascular architecture and perfusion, neovascularization activity, oxygen metabolism, and hypoxia calculated. From these 60 patients, 34 patients developed recurrence of the AG, and 26 patients showed no signs for AG recurrence during the study period. The time courses of MRI biomarker changes were analyzed regarding early pathophysiological alterations over a one-year period before radiological AG recurrence or a one-year period of stable disease for patients without recurrence, respectively. We detected intensifying local tissue hypoxia 250 days prior to radiological recurrence which initiated upregulation of neovascularization activity 50 to 70 days later. These changes were associated with a switch from an avascular infiltrative to a vascularized proliferative phenotype of the tumor cells another 30 days later. The dynamic changes of blood perfusion, microvessel density, neovascularization activity, and oxygen metabolism showed a close physiological interplay in the one-year period prior to radiological recurrence of IDH-mutated AG. These findings may path the wave for implementing both new MR-based imaging modalities for routine follow-up monitoring of AG patients after standard therapy and furthermore may support the development of novel, tailored therapy options in recurrent AG.
Collapse
|
24
|
Guo L, Li X, Cao H, Hua J, Mei Y, Pillai JJ, Wu Y. Inflow-based vascular-space-occupancy (iVASO) might potentially predict IDH mutation status and tumor grade in diffuse cerebral gliomas. J Neuroradiol 2021; 49:267-274. [PMID: 33482231 DOI: 10.1016/j.neurad.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/13/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE The aim of the study is to assess the diagnostic performance of inflow-based vascular-space-occupancy (iVASO) MR imaging for differentiating glioblastomas (grade IV, GBM) and lower-grade diffuse gliomas (grade II and III, LGG) and its potential to predict IDH mutation status. METHODS One hundred and two patients with diffuse cerebral glioma (56 males; median age, 43.5 years) underwent iVASO and dynamic susceptibility contrast (DSC) MR imaging. The iVASO-derived arteriolar cerebral blood volume (CBVa), relative CBVa (rCBVa), and the DSC-derived relative cerebral blood volume (rCBV) were obtained, and these measurements were compared between the GBM group (n = 43) and the LGG group (n = 59) and between the IDH-mutation group (n = 54) and the IDH-wild group (n = 48). RESULTS Significant correlation was observed between rCBV and CBVa (P < 0.001) or rCBVa (P < 0.001). Both CBVa (P < 0.001) and rCBVa (P < 0.001) were higher in the GBM group. Both CBVa (P < 0.001) and rCBVa (P < 0.001) were lower in the IDH-mutation group compared to the IDH-wild group. Receiver operating characteristic analyses showed the area under curve (AUC) of 0.95 with CBVa and 0.97 with rCBVa in differentiating GBM from LGG. The AUCs were 0.82 and 0.85 for CBVa and rCBVa in predicting IDH gene status, respectively, which were lower than that of rCBV (AUC = 0.90). Combined rCBV and rCBVa significantly improved the diagnostic performance (AUC = 0.95). CONCLUSIONS iVASO MR imaging has the potential to predict IDH mutation and grade in glioma.
Collapse
Affiliation(s)
- Liuji Guo
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xiaodan Li
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Haimei Cao
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun Hua
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Yingjie Mei
- China International Center, Philips Healthcare, Guangzhou, PR China
| | - Jay J Pillai
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuankui Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
25
|
Boxerman JL, Quarles CC, Hu LS, Erickson BJ, Gerstner ER, Smits M, Kaufmann TJ, Barboriak DP, Huang RH, Wick W, Weller M, Galanis E, Kalpathy-Cramer J, Shankar L, Jacobs P, Chung C, van den Bent MJ, Chang S, Al Yung WK, Cloughesy TF, Wen PY, Gilbert MR, Rosen BR, Ellingson BM, Schmainda KM. Consensus recommendations for a dynamic susceptibility contrast MRI protocol for use in high-grade gliomas. Neuro Oncol 2020; 22:1262-1275. [PMID: 32516388 PMCID: PMC7523451 DOI: 10.1093/neuonc/noaa141] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite the widespread clinical use of dynamic susceptibility contrast (DSC) MRI, DSC-MRI methodology has not been standardized, hindering its utilization for response assessment in multicenter trials. Recently, the DSC-MRI Standardization Subcommittee of the Jumpstarting Brain Tumor Drug Development Coalition issued an updated consensus DSC-MRI protocol compatible with the standardized brain tumor imaging protocol (BTIP) for high-grade gliomas that is increasingly used in the clinical setting and is the default MRI protocol for the National Clinical Trials Network. After reviewing the basis for controversy over DSC-MRI protocols, this paper provides evidence-based best practices for clinical DSC-MRI as determined by the Committee, including pulse sequence (gradient echo vs spin echo), BTIP-compliant contrast agent dosing (preload and bolus), flip angle (FA), echo time (TE), and post-processing leakage correction. In summary, full-dose preload, full-dose bolus dosing using intermediate (60°) FA and field strength-dependent TE (40-50 ms at 1.5 T, 20-35 ms at 3 T) provides overall best accuracy and precision for cerebral blood volume estimates. When single-dose contrast agent usage is desired, no-preload, full-dose bolus dosing using low FA (30°) and field strength-dependent TE provides excellent performance, with reduced contrast agent usage and elimination of potential systematic errors introduced by variations in preload dose and incubation time.
Collapse
Affiliation(s)
- Jerrold L Boxerman
- Department of Diagnostic Imaging, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the American Society of Neuroradiology (ASNR)
- Representative of the American Society of Functional Neuroradiology (ASFNR)
| | - Chad C Quarles
- Department of Neuroimaging Research and Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Leland S Hu
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
- Representative of the Alliance for Clinical Trials in Oncology
- Representative of the American Society of Neuroradiology (ASNR)
| | - Bradley J Erickson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC–University Medical Center Rotterdam, Rotterdam, Netherlands
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Timothy J Kaufmann
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Daniel P Barboriak
- Department of Radiology, Duke University School of Medicine, Durham, North Carolina, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Raymond H Huang
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang Wick
- Department of Neurooncology, National Center of Tumor Disease, University Clinic Heidelberg, Heidelberg, Germany
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Evanthia Galanis
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Jayashree Kalpathy-Cramer
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lalitha Shankar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland, USA
| | - Paula Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), Bethesda, Maryland, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Representative of the Alliance for Clinical Trials in Oncology
| | - Martin J van den Bent
- Department of Neuro-Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Representative of the European Organisation for Research and Treatment of Cancer (EORTC)
| | - Susan Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - W K Al Yung
- Department of Neuro-Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program and UCLA Brain Tumor Imaging Laboratory (BTIL), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute (NCI), Bethesda, Maryland, USA
- Representative of the Radiation Therapy Oncology Group (RTOG)
| | - Bruce R Rosen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Ellingson
- UCLA Neuro-Oncology Program and UCLA Brain Tumor Imaging Laboratory (BTIL), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Departments of Radiological Sciences, Psychiatry, and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Representative of the Adult Brain Tumor Consortium (ABTC)
- Representative of the Ivy Consortium for Early Phase Clinical Trials
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
- Representative of the RSNA Quantitative Imaging Biomarker Alliance (QIBA)
- Representative of the American Society of Neuroradiology (ASNR)
| | - Kathleen M Schmainda
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Representative of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG-ACRIN) Cancer Research Group
| |
Collapse
|
26
|
Cindil E, Sendur HN, Cerit MN, Dag N, Erdogan N, Celebi FE, Oner Y, Tali T. Validation of combined use of DWI and percentage signal recovery-optimized protocol of DSC-MRI in differentiation of high-grade glioma, metastasis, and lymphoma. Neuroradiology 2020; 63:331-342. [PMID: 32821962 DOI: 10.1007/s00234-020-02522-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/13/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE With conventional MRI, it is often difficult to effectively differentiate between contrast-enhancing brain tumors, including primary central nervous system lymphoma (PCNSL), high-grade glioma (HGG), and metastasis. This study aimed to assess the discrimination ability of the parameters obtained from DWI and the percentage signal recovery- (PSR-) optimized protocol of DSC-MRI between these three tumor types at an initial step. METHODS DSC-MRI using a PSR-optimized protocol (TR/TE = 1500/30 ms, flip angle = 90°, no preload) and DWI of 99 solitary enhancing tumors (60 HGGs, 24 metastases, 15 PCNSLs) were retrospectively assessed before treatment. rCBV, PSR, ADC in the tumor core and rCBV, and ADC in peritumoral edema were measured. The differences were evaluated using one-way ANOVA, and the diagnostic performance was evaluated using ROC curve analysis. RESULTS PSR in the tumor core showed the best discriminating performance in differentiating these three tumor types with AUC values of 0.979 for PCNSL vs. others and 0.947 for HGG vs. metastasis. The ADC was only helpful in the tumor core and distinguishing PCNSLs from others (AUC = 0.897). CONCLUSION Different from CBV-optimized protocols (preload, intermediate FA), PSR derived from the PSR-optimized protocol seems to be the most important parameter in the differentiation of HGGs, metastases, and PCNSLs at initial diagnosis. This property makes PSR remarkable and carries the need for comprehensive DSC-MRI protocols, which provides PSR sensitivity and CBV accuracy together, such as the preload use of the PSR-optimized protocol before the CBV-optimized protocol.
Collapse
Affiliation(s)
- Emetullah Cindil
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey.
| | - Halit Nahit Sendur
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| | - Mahi Nur Cerit
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| | - Nurullah Dag
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| | - Nesrin Erdogan
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| | - Filiz Elbuken Celebi
- School of Medicine, Department of Radiology, Yeditepe University, Istanbul, Turkey
| | - Yusuf Oner
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| | - Turgut Tali
- School of Medicine, Department of Radiology, Gazi University, Ankara, Turkey
| |
Collapse
|
27
|
Predicting Glioblastoma Response to Bevacizumab Through MRI Biomarkers of the Tumor Microenvironment. Mol Imaging Biol 2020; 21:747-757. [PMID: 30361791 DOI: 10.1007/s11307-018-1289-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE Glioblastoma (GB) is one of the most vascularized of all solid tumors and, therefore, represents an attractive target for antiangiogenic therapies. Many lesions, however, quickly develop escape mechanisms associated with changes in the tumor microenvironment (TME) resulting in rapid treatment failure. To prevent patients from adverse effects of ineffective therapy, there is a strong need to better predict and monitor antiangiogenic treatment response. PROCEDURES We utilized a novel physiological magnetic resonance imaging (MRI) method combining the visualization of oxygen metabolism and neovascularization for classification of five different TME compartments: necrosis, hypoxia with/without neovascularization, oxidative phosphorylation, and aerobic glycolysis. This approach, termed TME mapping, was used to monitor changes in tumor biology and pathophysiology within the TME in response to bevacizumab treatment in 18 patients with recurrent GB. RESULTS We detected dramatic changes in the TME by rearrangement of its compartments after the onset of bevacizumab treatment. All patients showed a decrease in active tumor volume and neovascularization as well as an increase in hypoxia and necrosis in the first follow-up after 3 months. We found that recurrent GB with a high percentage of neovascularization and active tumor before bevacizumab onset showed a poor or no treatment response. CONCLUSIONS TME mapping might be useful to develop strategies for patient stratification and response prediction before bevacizumab onset.
Collapse
|
28
|
Jin S, Han S, Stoyanova R, Ackerstaff E, Cho H. Pattern recognition analysis of dynamic susceptibility contrast (DSC)‐MRI curves automatically segments tissue areas with intact blood–brain barrier in a rat stroke model: A feasibility and comparison study. J Magn Reson Imaging 2020; 51:1369-1381. [PMID: 31654463 PMCID: PMC8566029 DOI: 10.1002/jmri.26949] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/12/2019] [Indexed: 11/21/2023] Open
Abstract
BackgroundThe manual segmentation of intact blood–brain barrier (BBB) regions in the stroke brain is cumbersome, due to the coexistence of infarction, large blood vessels, ventricles, and intact BBB regions, specifically in areas with weak signal enhancement following contrast agent injection.HypothesisThat from dynamic susceptibility contrast (DSC)‐MRI alone, without user intervention, regions of weak BBB damage can be segmented based on the leakage‐related parameter K
2 and the extent of intact BBB regions, needed to estimate K
2 values, determined.Study TypeFeasibility.Animal ModelTen female Sprague–Dawley rats (SD, 200–250g) underwent 1‐hour middle carotid artery occlusion (MCAO) and 1‐day reperfusion. Two SD rats underwent 1‐hour MCAO with 3‐day and 5‐day reperfusion.Field Strength/Sequence7T; ADC and T1 maps using diffusion‐weighted echo planar imaging (EPI) and relaxation enhancement (RARE) with variable repetition time (TR), respectively. dynamic contrast‐enhanced (DCE)‐MRI using FLASH. DSC‐MRI using gradient‐echo EPI.AssessmentConstrained nonnegative matrix factorization (cNMF) was applied to the dynamic ‐curves of DSC‐MRI (<4 min) in a BBB‐disrupted rat model. Areas of voxels with intact BBB, classified by automated cNMF analyses, were then used in estimating K
1 and K
2 values, and compared with corresponding values from manually‐derived areas.Statistical TestsMean ± standard deviation of ΔT1‐differences between ischemic and healthy areas were displayed with unpaired Student's t‐tests. Scatterplots were displayed with slopes and intercepts and Pearson's r values were evaluated between K
2 maps obtained with automatic (cNMF)‐ and manually‐derived regions of interest (ROIs) of the intact BBB region.ResultsMildly BBB‐damaged areas (indistinguishable from DCE‐MRI (10 min) parameters) were automatically segmented. Areas of voxels with intact BBB, classified by automated cNMF, matched closely the corresponding, manually‐derived areas when respective areas were used in estimating K
2 maps (Pearson's r = 0.97, 12 slices).Data ConclusionAutomatic segmentation of short DSC‐MRI data alone successfully identified areas with intact and compromised BBB in the stroke brain and compared favorably with manual segmentation.Level of Evidence: 3Technical Efficacy: Stage 1J. Magn. Reson. Imaging 2020;51:1369–1381.
Collapse
Affiliation(s)
- Seokha Jin
- Department of Biomedical Engineering Ulsan National Institute of Science and Technology Ulsan South Korea
| | - SoHyun Han
- Center of Neuroscience Imaging Research Sungkyunkwan University Suwon South Korea
| | - Radka Stoyanova
- Department of Radiation Oncology Miller School of Medicine, University of Miami Miami Florida USA
| | - Ellen Ackerstaff
- Department of Medical Physics Memorial Sloan Kettering Cancer Center New York New York USA
| | - HyungJoon Cho
- Department of Biomedical Engineering Ulsan National Institute of Science and Technology Ulsan South Korea
| |
Collapse
|
29
|
Li X, Wang D, Liao S, Guo L, Xiao X, Liu X, Xu Y, Hua J, Pillai JJ, Wu Y. Discrimination between Glioblastoma and Solitary Brain Metastasis: Comparison of Inflow-Based Vascular-Space-Occupancy and Dynamic Susceptibility Contrast MR Imaging. AJNR Am J Neuroradiol 2020; 41:583-590. [PMID: 32139428 DOI: 10.3174/ajnr.a6466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/03/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Accurate differentiation between glioblastoma and solitary brain metastasis is of vital importance clinically. This study aimed to investigate the potential value of the inflow-based vascular-space-occupancy MR imaging technique, which has no need for an exogenous contrast agent, in differentiating glioblastoma and solitary brain metastasis and to compare it with DSC MR imaging. MATERIALS AND METHODS Twenty patients with glioblastoma and 22 patients with solitary brain metastasis underwent inflow-based vascular-space-occupancy and DSC MR imaging with a 3T clinical scanner. Two neuroradiologists independently measured the maximum inflow-based vascular-space-occupancy-derived arteriolar CBV and DSC-derived CBV values in intratumoral regions and peritumoral T2-hyperintense regions, which were normalized to the contralateral white matter (relative arteriolar CBV and relative CBV, inflow-based vascular-space-occupancy relative arteriolar CBV, and DSC-relative CBV). The intraclass correlation coefficient, Student t test, or Mann-Whitney U test and receiver operating characteristic analysis were performed. RESULTS All parameters of both regions had good or excellent interobserver reliability (0.74∼0.89). In peritumoral T2-hyperintese regions, DSC-relative CBV (P < .001), inflow-based vascular-space-occupancy arteriolar CBV (P = .001), and relative arteriolar CBV (P = .005) were significantly higher in glioblastoma than in solitary brain metastasis, with areas under the curve of 0.94, 0.83, and 0.72 for discrimination, respectively. In the intratumoral region, both inflow-based vascular-space-occupancy arteriolar CBV and relative arteriolar CBV were significantly higher in glioblastoma than in solitary brain metastasis (both P < .001), with areas under the curve of 0.91 and 0.90, respectively. Intratumoral DSC-relative CBV showed no significant difference (P = .616) between the 2 groups. CONCLUSIONS Inflow-based vascular-space-occupancy has the potential to discriminate glioblastoma from solitary brain metastasis, especially in the intratumoral region.
Collapse
Affiliation(s)
- X Li
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - D Wang
- School of Biomedical Engineering (D.W.), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - S Liao
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Division of CT and MR, Radiology Department (S.L.), First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - L Guo
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - X Xiao
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - X Liu
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Y Xu
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - J Hua
- Neurosection, Division of MR Research (J.H.)
- F.M. Kirby Research Center for Functional Brain Imaging (J.H.), Kennedy Krieger Institute, Baltimore, Maryland
| | - J J Pillai
- Division of Neuroradiology (J.P.); Russell H. Morgan Department of Radiology and Radiological Science and
- Department of Neurosurgery (J.P.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Y Wu
- From the Department of Medical Imaging (X. Li, S.L., L.G., X.X., X. Liu, Y.X., Y.W.), Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
30
|
Stadlbauer A, Oberndorfer S, Zimmermann M, Renner B, Buchfelder M, Heinz G, Doerfler A, Kleindienst A, Roessler K. Physiologic MR imaging of the tumor microenvironment revealed switching of metabolic phenotype upon recurrence of glioblastoma in humans. J Cereb Blood Flow Metab 2020; 40:528-538. [PMID: 30732550 PMCID: PMC7026844 DOI: 10.1177/0271678x19827885] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Treating recurrent glioblastoma (GB) is one of the challenges in modern neurooncology. Hypoxia, neovascularization, and energy metabolism are of crucial importance for therapy failure and recurrence. Twenty-one patients with initially untreated GB who developed recurrence were examined with a novel MRI approach for noninvasive visualization of the tumor microenvironment (TME). Imaging biomarker information about oxygen metabolism (mitochondrial oxygen tension) and neovascularization (microvascular density and type) were fused for classification of five different TME compartments: necrosis, hypoxia with/without neovascularization, oxidative phosphorylation, and glycolysis. Volume percentages of these TME compartments were compared between untreated and recurrent GB. At initial diagnosis, all 21 GB showed either the features of a glycolytic dominant phenotype with a high percentage of functional neovasculature (N = 12) or those of a necrotic/hypoxic dominant phenotype with a high percentage of defective tumor neovasculature (N = 9). At recurrence, all 21 GB revealed switching of the initial metabolic phenotype: either from the glycolytic to the necrotic/hypoxic dominant phenotype or vice-versa. A necrotic/hypoxic phenotype at recurrence was associated with a higher rate of multifocality of the recurrent lesions. Our MRI approach may be helpful for a better understanding of treatment-induced metabolic phenotype switching and for future studies developing targeted therapeutic strategies for recurrent GB.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany.,Institute of Medical Radiology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Max Zimmermann
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Bertold Renner
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gertraud Heinz
- Institute of Medical Radiology, University Clinic of St. Pölten, St. Pölten, Austria
| | - Arnd Doerfler
- Department of Neuroradiology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Kleindienst
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Karl Roessler
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
31
|
Stadlbauer A, Eyüpoglu I, Buchfelder M, Dörfler A, Zimmermann M, Heinz G, Oberndorfer S. Vascular architecture mapping for early detection of glioblastoma recurrence. Neurosurg Focus 2019; 47:E14. [DOI: 10.3171/2019.9.focus19613] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/04/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVETreatment failure and inevitable tumor recurrence are the main reasons for the poor prognosis of glioblastoma (GB). Gross-total resection at repeat craniotomy for GB recurrence improves patient overall survival but requires early and reliable detection. It is known, however, that even advanced MRI approaches have limited diagnostic performance for distinguishing tumor progression from pseudoprogression. The novel MRI technique of vascular architectural mapping (VAM) provides deeper insight into tumor microvascularity and neovascularization. In this study the authors evaluated the usefulness of VAM for the monitoring of GB patients and quantitatively analyzed the features of neovascularization of early- and progressed-stage GB recurrence.METHODSIn total, a group of 115 GB patients who received overall 374 follow-up MRI examinations after standard treatment were retrospectively evaluated in this study. The clinical routine MRI (cMRI) protocol at 3 Tesla was extended with the authors’ experimental VAM approach, requiring 2 minutes of extra time for data acquisition. Custom-made MATLAB software was used for calculation of imaging biomarker maps of macrovascular perfusion from perfusion cMRI as well as of microvascular perfusion and architecture from VAM data. Additionally, cMRI data were analyzed by two board-certified radiologists in consensus. Statistical procedures included receiver operating characteristic (ROC) analysis to determine diagnostic performances for GB recurrence detection.RESULTSOverall, cMRI showed GB recurrence in 89 patients, and in 28 of these patients recurrence was detected earlier with VAM data, by 1 (20 patients) or 2 (8 patients) follow-up examinations, than with cMRI data. The mean time difference between recurrence detection with VAM and cMRI data was 147 days. During this time period the mean tumor volume increased significantly (p < 0.001) from 9.7 to 26.8 cm3. Quantitative analysis of imaging biomarkers demonstrated microvascular but no macrovascular hyperperfusion in early GB recurrence. Therefore, ROC analysis revealed superior diagnostic performance for VAM compared with cMRI.CONCLUSIONSThis study demonstrated that the targeted assessment of microvascular features using the VAM technique provided valuable information about early neovascularization activity in recurrent GB that is complementary to perfusion cMRI and may be helpful for earlier and more precise monitoring of patients suffering from GB. This VAM approach is compatible with existing cMRI protocols. Prospective clinical trials are necessary to investigate the clinical usefulness and potential benefit of increased overall survival with the use of VAM in patients with recurrent GB.
Collapse
Affiliation(s)
| | | | | | - Arnd Dörfler
- 3Neuroradiology, University of Erlangen-Nürnberg, Erlangen, Germany; and
| | | | | | - Stefan Oberndorfer
- 4Department of Neurology, University Clinic of St. Pölten, St. Pölten, Austria
| |
Collapse
|
32
|
Stadlbauer A, Zimmermann M, Doerfler A, Oberndorfer S, Buchfelder M, Coras R, Kitzwögerer M, Roessler K. Intratumoral heterogeneity of oxygen metabolism and neovascularization uncovers 2 survival-relevant subgroups of IDH1 wild-type glioblastoma. Neuro Oncol 2019; 20:1536-1546. [PMID: 29718366 DOI: 10.1093/neuonc/noy066] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The intratumoral heterogeneity of oxygen metabolism in combination with variable patterns of neovascularization (NV) as well as reprogramming of energy metabolism affects the landscape of tumor microenvironments (TMEs) in glioblastoma. Knowledge of the hypoxic and perivascular niches within the TME is essential for understanding treatment failure. Methods Fifty-two patients with untreated glioblastoma (isocitrate dehydrogenase 1 wild type [IDH1wt]) were examined with a physiological MRI protocol including a multiparametric quantitative blood oxygen level dependent (qBOLD) approach and vascular architecture mapping (VAM). Imaging biomarker information about oxygen metabolism (mitochondrial oxygen tension) and neovascularization (microvascular density and type) were fused for classification of 6 different TMEs: necrosis, hypoxia with/without neovascularization, oxidative phosphorylation (OxPhos), and glycolysis with/without neovascularization. Association of the different TME volume fractions with progression-free survival (PFS) was assessed using Kaplan-Meier analysis and Cox proportional hazards models. Results A common spatial structure of TMEs was detected: central necrosis surrounded by tumor hypoxia (with defective and functional neovasculature) and different TMEs with a predominance of OxPhos and glycolysis for energy production, respectively. The percentage of the different TMEs on the total tumor volume uncovered 2 clearly different subtypes of glioblastoma IDH1wt: a glycolytic dominated phenotype with predominantly functional neovasculature and a necrotic/hypoxic dominated phenotype with approximately 50% of defective neovasculature. Patients with a necrotic/hypoxic dominated phenotype showed significantly shorter PFS (P = 0.035). Conclusions Our non-invasive mapping approach allows for classification of the TME and detection of tumor-supportive niches in glioblastoma which may be helpful for both clinical patient management and research.
Collapse
Affiliation(s)
- Andreas Stadlbauer
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany.,Institute of Medical Radiology, University Clinic of St Pölten, St Pölten, Austria
| | - Max Zimmermann
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Doerfler
- Department of Neuroradiology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic of St Pölten, St Pölten, Austria
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Coras
- Department of Neuropathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Karl Roessler
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
33
|
Lee MD, Baird GL, Bell LC, Quarles CC, Boxerman JL. Utility of Percentage Signal Recovery and Baseline Signal in DSC-MRI Optimized for Relative CBV Measurement for Differentiating Glioblastoma, Lymphoma, Metastasis, and Meningioma. AJNR Am J Neuroradiol 2019; 40:1445-1450. [PMID: 31371360 DOI: 10.3174/ajnr.a6153] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE The percentage signal recovery in non-leakage-corrected (no preload, high flip angle, intermediate TE) DSC-MR imaging is known to differ significantly for glioblastoma, metastasis, and primary CNS lymphoma. Because the percentage signal recovery is influenced by preload and pulse sequence parameters, we investigated whether the percentage signal recovery can still differentiate these common contrast-enhancing neoplasms using a DSC-MR imaging protocol designed for relative CBV accuracy (preload, intermediate flip angle, low TE). MATERIALS AND METHODS We retrospectively analyzed DSC-MR imaging of treatment-naïve, pathology-proved glioblastomas (n = 14), primary central nervous system lymphomas (n = 7), metastases (n = 20), and meningiomas (n = 13) using a protocol designed for relative CBV accuracy (a one-quarter-dose preload and single-dose bolus of gadobutrol, TR/TE = 1290/40 ms, flip angle = 60° at 1.5T). Mean percentage signal recovery, relative CBV, and normalized baseline signal intensity were compared within contrast-enhancing lesion volumes. Classification accuracy was determined by receiver operating characteristic analysis. RESULTS Relative CBV best differentiated meningioma from glioblastoma and from metastasis with areas under the curve of 0.84 and 0.82, respectively. The percentage signal recovery best differentiated primary central nervous system lymphoma from metastasis with an area under the curve of 0.81. Relative CBV and percentage signal recovery were similar in differentiating primary central nervous system lymphoma from glioblastoma and from meningioma. Although neither relative CBV nor percentage signal recovery differentiated glioblastoma from metastasis, mean normalized baseline signal intensity achieved 86% sensitivity and 50% specificity. CONCLUSIONS Similar to results for non-preload-based DSC-MR imaging, percentage signal recovery for one-quarter-dose preload-based, intermediate flip angle DSC-MR imaging differentiates most pair-wise comparisons of glioblastoma, metastasis, primary central nervous system lymphoma, and meningioma, except for glioblastoma versus metastasis. Differences in normalized post-preload baseline signal for glioblastoma and metastasis, reflecting a snapshot of dynamic contrast enhancement, may motivate the use of single-dose multiecho protocols permitting simultaneous quantification of DSC-MR imaging and dynamic contrast-enhanced MR imaging parameters.
Collapse
Affiliation(s)
- M D Lee
- From the Warren Alpert Medical School of Brown University (M.D.L., J.L.B.), Providence, Rhode Island
| | - G L Baird
- Department of Diagnostic Imaging (G.L.B., J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| | - L C Bell
- Division of Neuroimaging Research (L.C.B., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - C C Quarles
- Division of Neuroimaging Research (L.C.B., C.C.Q.), Barrow Neurological Institute, Phoenix, Arizona
| | - J L Boxerman
- From the Warren Alpert Medical School of Brown University (M.D.L., J.L.B.), Providence, Rhode Island
- Department of Diagnostic Imaging (G.L.B., J.L.B.), Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
34
|
Stokes AM, Semmineh NB, Nespodzany A, Bell LC, Quarles CC. Systematic assessment of multi-echo dynamic susceptibility contrast MRI using a digital reference object. Magn Reson Med 2019; 83:109-123. [PMID: 31400035 DOI: 10.1002/mrm.27914] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 11/07/2022]
Abstract
PURPOSE Brain tumor dynamic susceptibility contrast (DSC) MRI is adversely impacted by T1 and T 2 ∗ contrast agent leakage effects that result in inaccurate hemodynamic metrics. While multi-echo acquisitions remove T1 leakage effects, there is no consensus on the optimal set of acquisition parameters. Using a computational approach, we systematically evaluated a wide range of acquisition strategies to determine the optimal multi-echo DSC-MRI perfusion protocol. METHODS Using a population-based DSC-MRI digital reference object (DRO), we assessed the influence of preload dosing (no preload and full dose preload), field strength (1.5 and 3T), pulse sequence parameters (echo time, repetition time, and flip angle), and leakage correction on relative cerebral blood volume (rCBV) and flow (rCBF) accuracy. We also compared multi-echo DSC-MRI protocols with standard single-echo protocols. RESULTS Multi-echo DSC-MRI is highly consistent across all protocols, and multi-echo rCBV (with or without use of a preload dose) had higher accuracy than single-echo rCBV. Regression analysis showed that choice of repetition time and flip angle had minimal impact on multi-echo rCBV and rCBV, indicating the potential for significant flexibility in acquisition parameters. The echo time combination had minimal impact on rCBV, though longer echo times should be avoided, particularly at higher field strengths. Leakage correction improved rCBV accuracy in all cases. Multi-echo rCBF was less biased than single-echo rCBF, although rCBF accuracy was reduced overall relative to rCBV. CONCLUSIONS Multi-echo acquisitions were more robust than single-echo, essentially decoupling both repetition time and flip angle from rCBV accuracy. Multi-echo acquisitions obviate the need for preload dosing, although leakage correction to remove residual T 2 ∗ leakage effects remains compulsory for high rCBV accuracy.
Collapse
Affiliation(s)
- Ashley M Stokes
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Natenael B Semmineh
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Ashley Nespodzany
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - Laura C Bell
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
35
|
Nabavizadeh SA, Akbari H, Ware JB, Nasrallah M, Guiry S, Bagley SJ, Desai A, Levy S, Sarchiapone W, Prior T, Detre J, Wolf RL, O'Rourke DM, Brem S, Davatzikos C. Arterial Spin Labeling and Dynamic Susceptibility Contrast-enhanced MR Imaging for evaluation of arteriovenous shunting and tumor hypoxia in glioblastoma. Sci Rep 2019; 9:8747. [PMID: 31217496 PMCID: PMC6584644 DOI: 10.1038/s41598-019-45312-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and carries a dismal prognosis. Significant challenges in the care of patients with GBM include marked vascular heterogeneity and arteriovenous (AV) shunting, which results in tumor hypoxia and inadequate delivery of systemic treatments to reach tumor cells. In this study, we investigated the utility of different MR perfusion techniques to detect and quantify arteriovenous (AV) shunting and tumor hypoxia in patients with GBM. Macrovascular shunting was present in 33% of subjects, with the degree of shunting ranging from (37-60%) using arterial spin labeling perfusion. Among the dynamic susceptibility contrast-enhanced perfusion curve features, there were a strong negative correlation between hypoxia score, DSC perfusion curve recovery slope (r = -0.72, P = 0.018) and angle (r = -0.73, P = 0.015). The results of this study support the possibility of using arterial spin labeling and pattern analysis of dynamic susceptibility contrast-enhanced MR Imaging for evaluation of arteriovenous shunting and tumor hypoxia in glioblastoma.
Collapse
Affiliation(s)
- S Ali Nabavizadeh
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Hamed Akbari
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey B Ware
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - MacLean Nasrallah
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samantha Guiry
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen J Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Arati Desai
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Scott Levy
- Department of Neurosurgery, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Whitney Sarchiapone
- Department of Neurosurgery, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy Prior
- Department of Neurosurgery, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Detre
- Department of Neurology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronald L Wolf
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald M O'Rourke
- Department of Neurosurgery, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Brem
- Department of Neurosurgery, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christos Davatzikos
- Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Ji B, Wang S, Liu Z, Weinberg BD, Yang X, Liu T, Wang L, Mao H. Revealing hemodynamic heterogeneity of gliomas based on signal profile features of dynamic susceptibility contrast-enhanced MRI. NEUROIMAGE-CLINICAL 2019; 23:101864. [PMID: 31176951 PMCID: PMC6558214 DOI: 10.1016/j.nicl.2019.101864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 04/30/2019] [Accepted: 05/19/2019] [Indexed: 01/25/2023]
Abstract
Dynamic susceptibility contrast enhanced magnetic resonance imaging (DSC MRI) is widely used for studying blood perfusion in brain tumors. While the time-dependent change of MRI signals related to the concentration of the tracer is used to derive the hemodynamic parameters such as regional blood volume and flow into tumors, the tissue-specific information associated with variations in profiles of signal time course is often overlooked. We report a new approach of combining model free independent component analysis (ICA) identification of specific signal profiles of DSC MRI time course data and extraction of the features from those time course profiles to interrogate time course data followed by calculating the region specific blood volume based on selected individual time courses. Based on the retrospective analysis of DSC MRI data from 38 patients with pathology confirmed low (n = 18) and high (n = 20) grade gliomas, the results reveal the spatially defined intra-tumoral hemodynamic heterogeneity of brain tumors based on features of time course profiles. The hemodynamic heterogeneity as measured by the number of independent components of time course data is associated with the tumor grade. Using 8 selected signal profile features, machine-learning trained algorithm, e.g., logistic regression, was able to differentiate pathology confirmed low intra-tumoral and high grade gliomas with an accuracy of 86.7%. Furthermore, the new method can potentially extract more tumor physiological information from DSC MRI comparing to the traditional model-based analysis and morphological analysis of tumor heterogeneity, thus may improve the characterizations of gliomas for better diagnosis and treatment decisions. Signal profiles of dynamic susceptibility contrast MRI data of brain tumors reflect hemodynamic properties of tumor tissue. Features in signal profiles extracted by machine learning methods revealed the hemodynamic heterogeneity of the gliomas. The reported approach is a new strategy to characterize the intra-tumor heterogeneity and physiological properties of gliomas.
Collapse
Affiliation(s)
- Bing Ji
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Silun Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Zhou Liu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America; Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Xiaofeng Yang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Tianming Liu
- Department of Computer Sciences, University of Georgia, Athens, GA, United States of America
| | - Liya Wang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America; Medical College of Nanchang University, Nanchang, Jiangxi, China; Department of Radiology, The People's Hospital of Longhua, Shenzhen, Guangdong, China.
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
37
|
Vamvakas A, Williams S, Theodorou K, Kapsalaki E, Fountas K, Kappas C, Vassiou K, Tsougos I. Imaging biomarker analysis of advanced multiparametric MRI for glioma grading. Phys Med 2019; 60:188-198. [DOI: 10.1016/j.ejmp.2019.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/27/2019] [Accepted: 03/17/2019] [Indexed: 01/29/2023] Open
|
38
|
Schmainda KM, Prah MA, Hu LS, Quarles CC, Semmineh N, Rand SD, Connelly JM, Anderies B, Zhou Y, Liu Y, Logan B, Stokes A, Baird G, Boxerman JL. Moving Toward a Consensus DSC-MRI Protocol: Validation of a Low-Flip Angle Single-Dose Option as a Reference Standard for Brain Tumors. AJNR Am J Neuroradiol 2019; 40:626-633. [PMID: 30923088 DOI: 10.3174/ajnr.a6015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE DSC-MR imaging using preload, intermediate (60°) flip angle and postprocessing leakage correction has gained traction as a standard methodology. Simulations suggest that DSC-MR imaging with flip angle = 30° and no preload yields relative CBV practically equivalent to the reference standard. This study tested this hypothesis in vivo. MATERIALS AND METHODS Eighty-four patients with brain lesions were enrolled in this 3-institution study. Forty-three patients satisfied the inclusion criteria. DSC-MR imaging (3T, single-dose gadobutrol, gradient recalled-echo-EPI, TE = 20-35 ms, TR = 1.2-1.63 seconds) was performed twice for each patient, with flip angle = 30°-35° and no preload (P-), which provided preload (P+) for the subsequent intermediate flip angle = 60°. Normalized relative CBV and standardized relative CBV maps were generated, including postprocessing with contrast agent leakage correction (C+) and without (C-) contrast agent leakage correction. Contrast-enhancing lesion volume, mean relative CBV, and contrast-to-noise ratio obtained with 30°/P-/C-, 30°/P-/C+, and 60°/P+/C- were compared with 60°/P+/C+ using the Lin concordance correlation coefficient and Bland-Altman analysis. Equivalence between the 30°/P-/C+ and 60°/P+/C+ protocols and the temporal SNR for the 30°/P- and 60°/P+ DSC-MR imaging data was also determined. RESULTS Compared with 60°/P+/C+, 30°/P-/C+ had closest mean standardized relative CBV (P = .61), highest Lin concordance correlation coefficient (0.96), and lowest Bland-Altman bias (μ = 1.89), compared with 30°/P-/C- (P = .02, Lin concordance correlation coefficient = 0.59, μ = 14.6) and 60°/P+/C- (P = .03, Lin concordance correlation coefficient = 0.88, μ = -10.1) with no statistical difference in contrast-to-noise ratios across protocols. The normalized relative CBV and standardized relative CBV were statistically equivalent at the 10% level using either the 30°/P-/C+ or 60°/P+/C+ protocols. Temporal SNR was not significantly different for 30°/P- and 60°/P+ (P = .06). CONCLUSIONS Tumor relative CBV derived from low-flip angle, no-preload DSC-MR imaging with leakage correction is an attractive single-dose alternative to the higher dose reference standard.
Collapse
Affiliation(s)
- K M Schmainda
- From the Departments of Biophysics (K.M.S., M.A.P.) .,Radiology (K.M.S., S.D.R.)
| | - M A Prah
- From the Departments of Biophysics (K.M.S., M.A.P.)
| | - L S Hu
- Departments of Radiology (L.S.H., Y.Z.)
| | - C C Quarles
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - N Semmineh
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | | | | | - B Anderies
- Neurosurgery (B.A.), Mayo Clinic, Scottsdale, Arizona
| | - Y Zhou
- Departments of Radiology (L.S.H., Y.Z.)
| | - Y Liu
- Division of Biostatistics, Institute for Health and Society (Y.L., B.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - B Logan
- Division of Biostatistics, Institute for Health and Society (Y.L., B.L.), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A Stokes
- Division of Imaging Research (C.C.Q., N.S., A.S.), Barrow Neurological Institute, Phoenix, Arizona
| | - G Baird
- Department of Diagnostic Imaging (J.L.B., G.B.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - J L Boxerman
- Department of Diagnostic Imaging (J.L.B., G.B.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
39
|
Shah NJ, da Silva NA, Yun SD. Perfusion weighted imaging using combined gradient/spin echo EPIK: Brain tumour applications in hybrid MR-PET. Hum Brain Mapp 2019; 42:4144-4154. [PMID: 30761676 DOI: 10.1002/hbm.24537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 01/30/2023] Open
Abstract
Advanced perfusion-weighted imaging (PWI) methods that combine gradient echo (GE) and spin echo (SE) data are important tools for the study of brain tumours. In PWI, single-shot, EPI-based methods have been widely used due to their relatively high imaging speed. However, when used with increasing spatial resolution, single-shot EPI methods often show limitations in whole-brain coverage for multi-contrast applications. To overcome this limitation, this work employs a new version of EPI with keyhole (EPIK) to provide five echoes: two with GEs, two with mixed GESE and one with SE; the sequence is termed "GESE-EPIK." The performance of GESE-EPIK is evaluated against its nearest relative, EPI, in terms of the temporal signal-to-noise ratio (tSNR). Here, data from brain tumour patients were acquired using a hybrid 3T MR-BrainPET scanner. GESE-EPIK resulted in reduced susceptibility artefacts, shorter TEs for the five echoes and increased brain coverage when compared to EPI. Moreover, compared to EPI, EPIK achieved a comparable tSNR for the first and second echoes and significantly higher tSNR for other echoes. A new method to obtain multi-echo GE and SE data with shorter TEs and increased brain coverage is demonstrated. As proposed here, the workflow can be shortened and the integration of multimodal clinical MR-PET studies can be facilitated.
Collapse
Affiliation(s)
- N Jon Shah
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany.,Institute of Neuroscience-11, Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH, Jülich, Germany.,Department of Neurology, Faculty of Medicine, JARA, RWTH Aachen University, Aachen, Germany.,Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Nuno André da Silva
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Seong Dae Yun
- Institute of Neuroscience and Medicine - 4, Medical Imaging Physics, Forschungszentrum Jülich GmbH, Jülich, Germany
| |
Collapse
|
40
|
Novak J, Withey SB, Lateef S, MacPherson L, Pinkey B, Peet AC. A comparison of pseudo-continuous arterial spin labelling and dynamic susceptibility contrast MRI with and without contrast agent leakage correction in paediatric brain tumours. Br J Radiol 2019; 92:20170872. [PMID: 30358415 DOI: 10.1259/bjr.20170872] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE: To investigate correlations between MRI perfusion metrics measured by dynamic susceptibility contrast and arterial spin labelling in paediatric brain tumours. METHODS: 15 paediatric patients with brain tumours were scanned prospectively using pseudo-continuous arterial spin labelling (ASL) and dynamic susceptibility contrast (DSC-) MRI with a pre-bolus to minimise contrast agent leakage. Cerebral blood flow (CBF) maps were produced using ASL. Cerebral blood volume (CBV) maps with and without contrast agent leakage correction using the Boxerman technique and the leakage parameter, K2, were produced from the DSC data. Correlations between the metrics produced were investigated. RESULTS: Histology resulted in the following diagnoses: pilocytic astrocytoma (n = 7), glioblastoma (n = 1), medulloblastoma (n = 1), rosette-forming glioneuronal tumour of fourth ventricle (n = 1), atypical choroid plexus papilloma (n = 1) and pilomyxoid astrocytoma (n = 1). Three patients had a non-invasive diagnosis of low-grade glioma. DSC CBV maps of T1-enhancing tumours were difficult to interpret without the leakage correction. CBV values obtained with and without leakage correction were significantly different (p < 0.01). A significant positive correlation was observed between ASL CBF and DSC CBV (r = 0.516, p = 0.049) which became stronger when leakage correction was applied (r = 0.728, p = 0.002). K2 values were variable across the group (mean = 0.35, range = -0.49 to 0.64). CONCLUSION: CBV values from DSC obtained with and without leakage correction were significantly different. Large increases in CBV were observed following leakage correction in highly T1-enhancing tumours. DSC and ASL perfusion metrics were found to correlate significantly in a range of paediatric brain tumours. A stronger relationship between DSC and ASL was seen when leakage correction was applied to the DSC data. Leakage correction should be applied when analysing DSC data in enhancing paediatric brain tumours. ADVANCES IN KNOWLEDGE: We have shown that leakage correction should be applied when investigating enhancing paediatric brain tumours using DSC-MRI. A stronger correlation was found between CBF derived from ASL and CBV derived from DSC when a leakage correction was employed.
Collapse
Affiliation(s)
- Jan Novak
- 1 Birmingham Children's Hospital , Birmingham , UK.,2 Cancer Sciences, University of Birmingham , Birmingham , UK
| | - Stephanie Barbara Withey
- 1 Birmingham Children's Hospital , Birmingham , UK.,2 Cancer Sciences, University of Birmingham , Birmingham , UK.,3 RRPPS, University Hospitals Birmingham NHS Foundation Trust , Birmingham , UK
| | | | | | | | - Andrew C Peet
- 1 Birmingham Children's Hospital , Birmingham , UK.,2 Cancer Sciences, University of Birmingham , Birmingham , UK
| |
Collapse
|
41
|
Varallyay CG, Nesbit E, Horvath A, Varallyay P, Fu R, Gahramanov S, Muldoon LL, Li X, Rooney WD, Neuwelt EA. Cerebral blood volume mapping with ferumoxytol in dynamic susceptibility contrast perfusion MRI: Comparison to standard of care. J Magn Reson Imaging 2018; 48:441-448. [PMID: 29314418 PMCID: PMC6034979 DOI: 10.1002/jmri.25943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cerebral blood volume (CBV) mapping with a dynamic susceptibility contrast (DSC) perfusion technique has become a clinical tool in diagnosing and follow-up of brain tumors. Ferumoxytol, a long-circulating iron oxide nanoparticle, has been tested for CBV mapping, but the optimal dose has not been established. PURPOSE To compare ferumoxytol DSC of two different doses to standard of care gadoteridol by analyzing time-intensity curves and CBV maps in normal-appearing brain regions. STUDY TYPE Retrospective. SUBJECTS Fifty-four patients with various brain disorders. FIELD STRENGTH/SEQUENCE 3T MRI. DSC-MRI was performed with 0.1 mmol/kg gadoteridol and 1 day later with ferumoxytol in doses of 1 or 2 mg/kg. ASSESSMENT Signal changes during first pass, relative CBV (rCBV) in normal-appearing thalamus, putamen, and globus pallidus, and contrast-to-noise ratio (CNR) of the CBV maps were compared between gadoteridol and various doses of ferumoxytol using an automated method. To subjectively assess the quality of the CBV maps, two blinded readers also assessed visual conspicuity of the putamen. STATISTICAL TESTS Linear mixed effect model was used for statistical comparison. RESULTS Compared to gadoteridol, 1 mg/kg ferumoxytol showed no difference in CNR (P = 0.6505), peak ΔR2*, and rCBV in the putamen (P = 0.2669, 0.0871) or in the thalamus (P = 0.517, 0.9787); 2 mg/kg ferumoxytol increased peak ΔR2* as well as the CNR (P < 0.0001), but also mildly increased rCBV in putamen and globus pallidus (P = 0.0005, 0.0012). Signal intensities during first pass remained highly above the noise level, with overlapping of 95% confidence intervals with noise only in 3 out of 162 tested regions. Compared to gadoteridol, the visual image quality showed mild improvement with 1 mg/kg (P = 0.02) and marked improvement with 2 mg/kg ferumoxytol (P < 0.0001). DATA CONCLUSION 1 mg/kg ferumoxytol provides similar imaging results to standard gadoteridol for DSC-MRI, and 2 mg/kg has a benefit of increased CNR, but may also result in mildly increased rCBV values. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 1 J. MAGN. RESON. IMAGING 2018;48:441-448.
Collapse
Affiliation(s)
- Csanad G. Varallyay
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Eric Nesbit
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Andrea Horvath
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Peter Varallyay
- National Institute of Clinical Neurosciences, Budapest, Hungary
| | - Rongwei Fu
- School of Public Health, Oregon Health & Science University, Portland, OR
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University
| | - Seymur Gahramanov
- Department of Neurosurgery, University of New Mexico, Albuquerque, NM
| | - Leslie L. Muldoon
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | - Edward A. Neuwelt
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Neurosurgery, Oregon Health & Science University, Portland, OR
- Portland Veterans Affairs Medical Center, Portland, OR
| |
Collapse
|
42
|
Cerebral blood perfusion deficits using dynamic susceptibility contrast MRI with gadolinium chelates in rats with post-ischemic reperfusion without significant dynamic contrast-enhanced MRI-derived vessel permeabilities: A cautionary note. PLoS One 2018; 13:e0201076. [PMID: 30044884 PMCID: PMC6059480 DOI: 10.1371/journal.pone.0201076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/06/2018] [Indexed: 11/22/2022] Open
Abstract
In this study, we quantified perfusion deficits using dynamic susceptibility contrast magnetic resonance imaging (DSC-MRI) with an extravasating contrast agent (CA). We also investigated the efficacy of leakage compensation from CA pre-load in brains from post-ischemic rat models without significant dynamic contrast-enhanced MRI (DCE-MRI)-derived vessel wall permeability. DSC measurements were obtained using fast (0.3 s) echo-planar imaging in both normal rats and rats with transient middle carotid artery occlusion (MCAO) (1-h MCAO, 24-h reperfusion) after successive administrations of gadoterate meglumine (Dotarem) and intravascular superparamagnetic iron oxide nanoparticles (SPION). The relative cerebral blood volume (CBV) and cerebral blood flow (CBF) values acquired using Dotarem were significantly underestimated (~20%) when compared to those acquired using SPION in ipsilesional post-ischemic brain regions. A slight overestimation of relative mean transit time was observed. Areas with underestimated CBV and CBF values from the corresponding error maps encompassed the area of infarcted tissue (apparent diffusion coefficient < 500 μm2/s) and mostly coincided with the area wherein conspicuous longitudinal relaxation time differences were observed pre- vs. post-injection of Dotarem. The DSC measurements with significant pre-load (0.3 mmol·kg-1) of Dotarem displayed minimal perfusion deficits when compared to those determined using the reference intravascular SPION.
Collapse
|
43
|
Hedderich D, Kluge A, Pyka T, Zimmer C, Kirschke JS, Wiestler B, Preibisch C. Consistency of normalized cerebral blood volume values in glioblastoma using different leakage correction algorithms on dynamic susceptibility contrast magnetic resonance imaging data without and with preload. J Neuroradiol 2018; 46:44-51. [PMID: 29753641 DOI: 10.1016/j.neurad.2018.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 02/27/2018] [Accepted: 04/21/2018] [Indexed: 10/16/2022]
Abstract
BACKGROUND AND PURPOSE Several leakage correction algorithms for dynamic susceptibility contrast (DSC) magnetic resonance imaging (MRI)-based cerebral blood volume (CBV) measurement have been proposed, and combination with a preload of contrast agent is generally recommended. A single bolus application scheme would largely simplify and facilitate standardized clinical applications, while reducing contrast agent (CA) dose. The aim of this study was, therefore, to investigate whether appropriate leakage correction redundantizes prebolus application by comparing normalized DSC-based CBV (nCBV) measures of two consecutive CA boli. MATERIALS AND METHODS Twenty-seven patients with suspected glioblastoma (WHO-grade-IV) underwent DSC-MRI during two consecutive boli of Gd-based CA. Four variants of two post-processing leakage correction techniques were compared with respect to nCBV in contrast enhancing tumor tissue. First, a reference curve approach with first pass and full integration of corrected ΔR2*(t), and second, a deconvolution-based approach using singular value decomposition (SVD) with a standard noise-dependent cutoff or Tikhonov regularization. RESULTS Compared to respective uncorrected values, all leakage correction techniques increased nCBV for data acquired without prebolus, while there was no consistent trend for data acquired with prebolus. The best agreement between corrected nCBV values in contrast enhancing tumor, obtained in the same patients without and with prebolus, respectively, was obtained for the reference curve-based correction approach with either first pass or full integration. CONCLUSION The reference curve-based leakage correction approach with integration-based nCBV calculation yielded a high accordance between nCBV values without and with prebolus, respectively. Thus, it appears possible to obtain valid nCBV in glioblastoma with a single CA injection.
Collapse
Affiliation(s)
- Dennis Hedderich
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Anne Kluge
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Thomas Pyka
- Clinic for Nuclear Medicine, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Jan S Kirschke
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany
| | - Christine Preibisch
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany; Clinic for Neurology, Technische Universität München, Ismaningerst. 22, 81675 Munich, Germany.
| |
Collapse
|
44
|
Monti L, Morbidelli L, Rossi A. Impaired Cerebral Perfusion in Multiple Sclerosis: Relevance of Endothelial Factors. Biomark Insights 2018; 13:1177271918774800. [PMID: 29795976 PMCID: PMC5960845 DOI: 10.1177/1177271918774800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/07/2018] [Indexed: 12/21/2022] Open
Abstract
Magnetic resonance imaging techniques measuring in vivo brain perfusion and integrity of the blood-brain barrier have developed rapidly in the past decade, resulting in a wide range of available methods. This review first discusses their principles, possible pitfalls, and potential for quantification and outlines clinical application in neurological disorders. Then, we focus on the endothelial cells of the blood-brain barrier, pointing out their contribution in regulating vascular tone by production of vasoactive substances. Finally, the role of these substances in brain hypoperfusion in multiple sclerosis is discussed.
Collapse
Affiliation(s)
- Lucia Monti
- Unit of Neuroimaging and Neurointervention, Department of Neurological and Neurosensory Sciences, "Santa Maria alle Scotte" General Hospital, University Hospital of Siena, Siena, Italy
| | | | - Alessandro Rossi
- Unit of Neurology and Clinical Neurophysiology, Department of Neurological and Neurosensory Sciences, University Hospital of Siena, Siena, Italy
| |
Collapse
|
45
|
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: 40] [Impact Index Per Article: 5.7] [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
|
46
|
Pyka T, Hiob D, Preibisch C, Gempt J, Wiestler B, Schlegel J, Straube C, Zimmer C. Diagnosis of glioma recurrence using multiparametric dynamic 18F-fluoroethyl-tyrosine PET-MRI. Eur J Radiol 2018; 103:32-37. [PMID: 29803382 DOI: 10.1016/j.ejrad.2018.04.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/27/2018] [Accepted: 04/02/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES To investigate the value of combined 18F-fluorethyltyrosine-(FET)-PET/MRI for differentiation between recurrence and treatment-related changes in glioma patients. METHODS 63 lesions suggestive of recurrence in 47 glioma patients were retrospectively identified. All patients had a dynamic FET scan, as well as morphologic MRI, PWI and DWI on a hybrid PET/MRI scanner. Lesions suggestive of recurrence were marked. ROC analysis was performed univariately and on parameter combination. RESULTS 50 lesions were classified as recurrence, 13 as radiation necrosis. Diagnosis was based on histology in 23 and follow-up imaging in 40 cases. Sensitivities and specificities for static PET were 80 and 85%, 66% and 77% for PWI, 62 and 77% for DWI and 64 and 79% for PET time-to-peak. AUC was 0.86 (p < 0.001) for static PET, 0.73 (p = 0.013) for PWI, 0.70 (p = 0.030) for DWI and 0.73 (p < 0.001) for dynamic PET. Multiparametric analysis resulted in an AUC of 0.89, notably yielding sensitivity of 76% vs. 56% for PET alone at 100% specificity. CONCLUSION Simultaneous dynamic FET-PET/MRI was reliably feasible for imaging of recurrent glioma. While all modalities were able to discriminate between recurrence and treatment-related changes, multiparametric analysis added value especially when high specificity was demanded.
Collapse
Affiliation(s)
- Thomas Pyka
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany; Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany.
| | - Daniela Hiob
- Department of Nuclear Medicine, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Christine Preibisch
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Jürgen Schlegel
- Department of Neuropathology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Christoph Straube
- Department of Radiation Oncology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, TU München, Ismaninger Str. 22, 81675 München, Germany
| |
Collapse
|
47
|
Soliman RK, Gamal SA, Essa AHA, Othman MH. Preoperative Grading of Glioma Using Dynamic Susceptibility Contrast MRI: Relative Cerebral Blood Volume Analysis of Intra-tumoural and Peri-tumoural Tissue. Clin Neurol Neurosurg 2018; 167:86-92. [DOI: 10.1016/j.clineuro.2018.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/27/2017] [Accepted: 01/07/2018] [Indexed: 11/28/2022]
|
48
|
Gong E, Pauly JM, Wintermark M, Zaharchuk G. Deep learning enables reduced gadolinium dose for contrast-enhanced brain MRI. J Magn Reson Imaging 2018; 48:330-340. [PMID: 29437269 DOI: 10.1002/jmri.25970] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/25/2018] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND There are concerns over gadolinium deposition from gadolinium-based contrast agents (GBCA) administration. PURPOSE To reduce gadolinium dose in contrast-enhanced brain MRI using a deep learning method. STUDY TYPE Retrospective, crossover. POPULATION Sixty patients receiving clinically indicated contrast-enhanced brain MRI. SEQUENCE 3D T1 -weighted inversion-recovery prepped fast-spoiled-gradient-echo (IR-FSPGR) imaging was acquired at both 1.5T and 3T. In 60 brain MRI exams, the IR-FSPGR sequence was obtained under three conditions: precontrast, postcontrast images with 10% low-dose (0.01mmol/kg) and 100% full-dose (0.1 mmol/kg) of gadobenate dimeglumine. We trained a deep learning model using the first 10 cases (with mixed indications) to approximate full-dose images from the precontrast and low-dose images. Synthesized full-dose images were created using the trained model in two test sets: 20 patients with mixed indications and 30 patients with glioma. ASSESSMENT For both test sets, low-dose, true full-dose, and the synthesized full-dose postcontrast image sets were compared quantitatively using peak-signal-to-noise-ratios (PSNR) and structural-similarity-index (SSIM). For the test set comprised of 20 patients with mixed indications, two neuroradiologists scored blindly and independently for the three postcontrast image sets, evaluating image quality, motion-artifact suppression, and contrast enhancement compared with precontrast images. STATISTICAL ANALYSIS Results were assessed using paired t-tests and noninferiority tests. RESULTS The proposed deep learning method yielded significant (n = 50, P < 0.001) improvements over the low-dose images (>5 dB PSNR gains and >11.0% SSIM). Ratings on image quality (n = 20, P = 0.003) and contrast enhancement (n = 20, P < 0.001) were significantly increased. Compared to true full-dose images, the synthesized full-dose images have a slight but not significant reduction in image quality (n = 20, P = 0.083) and contrast enhancement (n = 20, P = 0.068). Slightly better (n = 20, P = 0.039) motion-artifact suppression was noted in the synthesized images. The noninferiority test rejects the inferiority of the synthesized to true full-dose images for image quality (95% CI: -14-9%), artifacts suppression (95% CI: -5-20%), and contrast enhancement (95% CI: -13-6%). DATA CONCLUSION With the proposed deep learning method, gadolinium dose can be reduced 10-fold while preserving contrast information and avoiding significant image quality degradation. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 5 J. MAGN. RESON. IMAGING 2018;48:330-340.
Collapse
Affiliation(s)
- Enhao Gong
- Department of Electrical Engineering, Stanford University, Stanford, California, USA.,Department of Radiology, Stanford University, Stanford, California, USA
| | - John M Pauly
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Max Wintermark
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Greg Zaharchuk
- Department of Radiology, Stanford University, Stanford, California, USA
| |
Collapse
|
49
|
Li X, Varallyay CG, Gahramanov S, Fu R, Rooney WD, Neuwelt EA. Pseudo-extravasation rate constant of dynamic susceptibility contrast-MRI determined from pharmacokinetic first principles. NMR IN BIOMEDICINE 2017; 30:10.1002/nbm.3797. [PMID: 28885746 PMCID: PMC5870763 DOI: 10.1002/nbm.3797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 06/07/2023]
Abstract
Dynamic susceptibility contrast-magnetic resonance imaging (DSC-MRI) is widely used to obtain informative perfusion imaging biomarkers, such as the relative cerebral blood volume (rCBV). The related post-processing software packages for DSC-MRI are available from major MRI instrument manufacturers and third-party vendors. One unique aspect of DSC-MRI with low-molecular-weight gadolinium (Gd)-based contrast reagent (CR) is that CR molecules leak into the interstitium space and therefore confound the DSC signal detected. Several approaches to correct this leakage effect have been proposed throughout the years. Amongst the most popular is the Boxerman-Schmainda-Weisskoff (BSW) K2 leakage correction approach, in which the K2 pseudo-first-order rate constant quantifies the leakage. In this work, we propose a new method for the BSW leakage correction approach. Based on the pharmacokinetic interpretation of the data, the commonly adopted R2 * expression accounting for contributions from both intravascular and extravasating CR components is transformed using a method mathematically similar to Gjedde-Patlak linearization. Then, the leakage rate constant (KL ) can be determined as the slope of the linear portion of a plot of the transformed data. Using the DSC data of high-molecular-weight (~750 kDa), iron-based, intravascular Ferumoxytol (FeO), the pharmacokinetic interpretation of the new paradigm is empirically validated. The primary objective of this work is to empirically demonstrate that a linear portion often exists in the graph of the transformed data. This linear portion provides a clear definition of the Gd CR pseudo-leakage rate constant, which equals the slope derived from the linear segment. A secondary objective is to demonstrate that transformed points from the initial transient period during the CR wash-in often deviate from the linear trend of the linearized graph. The inclusion of these points will have a negative impact on the accuracy of the leakage rate constant, and even make it time dependent.
Collapse
Affiliation(s)
- Xin Li
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Csanad G. Varallyay
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Radiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Seymur Gahramanov
- Department of Neurosurgery, University of New Mexico, Albuquerque, New Mexico, USA
| | - Rongwei Fu
- School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| | - Edward A. Neuwelt
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
50
|
Prah MA, Al-Gizawiy MM, Mueller WM, Cochran EJ, Hoffmann RG, Connelly JM, Schmainda KM. Spatial discrimination of glioblastoma and treatment effect with histologically-validated perfusion and diffusion magnetic resonance imaging metrics. J Neurooncol 2017; 136:13-21. [PMID: 28900832 DOI: 10.1007/s11060-017-2617-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/04/2017] [Indexed: 11/30/2022]
Abstract
The goal of this study is to spatially discriminate tumor from treatment effect (TE), within the contrast-enhancing lesion, for brain tumor patients at all stages of treatment. To this end, the diagnostic accuracy of MRI-derived diffusion and perfusion parameters to distinguish pure TE from pure glioblastoma (GBM) was determined utilizing spatially-correlated biopsy samples. From July 2010 through June 2015, brain tumor patients who underwent pre-operative DWI and DSC-MRI and stereotactic image-guided biopsy were considered for inclusion in this IRB-approved study. MRI-derived parameter maps included apparent diffusion coefficient (ADC), normalized cerebral blood flow (nCBF), normalized and standardized relative cerebral blood volume (nRCBV, sRCBV), peak signal-height (PSR) and percent signal-recovery (PSR). These were co-registered to the Stealth MRI and median values extracted from the spatially-matched biopsy regions. A ROC analysis accounting for multiple subject samples was performed, and the optimal threshold for distinguishing TE from GBM determined for each parameter. Histopathologic diagnosis of pure TE (n = 10) or pure GBM (n = 34) was confirmed in tissue samples from 15 consecutive subjects with analyzable data. Perfusion thresholds of sRCBV (3575; SN/SP% = 79.4/90.0), nRCBV (1.13; SN/SP% = 82.1/90.0), and nCBF (1.05; SN/SP% = 79.4/80.0) distinguished TE from GBM (P < 0.05), whereas ADC, PSR, and PH could not (P > 0.05). The thresholds for CBF and CBV can be applied to lesions with any admixture of tumor or treatment effect, enabling the identification of true tumor burden within enhancing lesions. This approach overcomes current limitations of averaging values from both tumor and TE for quantitative assessments.
Collapse
Affiliation(s)
- Melissa A Prah
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Mona M Al-Gizawiy
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Wade M Mueller
- Department of Neurosurgery, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI, 53226, USA
| | - Elizabeth J Cochran
- Department of Pathology, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI, 53226, USA
| | - Raymond G Hoffmann
- Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Jennifer M Connelly
- Department of Neurology, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI, 53226, USA
| | - Kathleen M Schmainda
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA. .,Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| |
Collapse
|