RSNA 2016

Abstract Archives of the RSNA, 2016


RC305

Neuroradiology Series: Brain Tumors

Tuesday, Nov. 29 8:30AM - 12:00PM Room: S102AB

NR

AMA PRA Category 1 Credits ™: 3.25
ARRT Category A+ Credits: 4.00

FDA Discussions may include off-label uses.

Yvonne W. Lui, MD, New York, NY, (yvonne.lui@nyumc.org) (Moderator) Nothing to Disclose
Whitney B. Pope, MD, PhD, Los Angeles, CA (Moderator) Research Consultant, F. Hoffmann-La Roche Ltd; Research Consultant, Amgen Inc; Research Consultant, Tocagen Inc; ;
LEARNING OBJECTIVES

This session will highlight cutting-edge advances in brain tumor imaging including classification, radiogenomics, and quantitative pitfalls from leaders in the field.

ABSTRACT

Sub-Events
RC305-01
Soonmee Cha, MD, San Francisco, CA (Presenter) Nothing to Disclose
LEARNING OBJECTIVES

1) Present salient features of the new 2016 WHO Classification of Tumors or the Central Nervous System. 2) Discuss rationale and implication of the new classification in clinical practice of neuro-oncology and neuroradiology. 3) Apply tissue-based data outlined in the new classification to understand complex and heterogeneous brain tumor biology and to improve diagnostic imaging interpretation of brain tumors before, during, and after treatment.

ABSTRACT

RC305-02
Liya Wang, MD, Atlanta, GA (Abstract Co-Author) Nothing to Disclose
Jeffrey Olson, PHD, Atlanta, GA (Abstract Co-Author) Nothing to Disclose
James Ross, Atlanta, GA (Abstract Co-Author) Nothing to Disclose
Erwin G. Van Meir, Atlanta, GA (Abstract Co-Author) Nothing to Disclose
Zhou Liu, MD, Shenzhen, China (Abstract Co-Author) Nothing to Disclose
Hui Mao, PhD, Atlanta, GA (Presenter) Nothing to Disclose
PURPOSE

Mutations in isocitrate dehydrogenase 1 or 2 (IDH1/2) occur in 80% of low grade gliomas. Onco-metabolite, R(-)-2-hydroxyglutarate (2HG) is a biomarker for IDH mutation (mIDH) for molecular classification of tumors and can be detected with magnetic resonance spectroscopy (MRS) for predicting prognosis[1,2]. To determine whether MRS measurement of 2HG can provide more prognostic and therapeutic information, we investigated the correlations of 2HG level with chromosome 1p/19q co-deletion status, tumor progressions, and other metabolites.

METHOD AND MATERIALS

2HG concentrations were measured from tissue specimens of 47 gliomas with mIDH by 2D Correlation Spectroscopy using solid state NMR. All tumors were categorized into three WHO grades. 1p/19q co-deletion was determined using fluorescence in situ hybridization. Progression free survival (PFS) of patients with mIDH and wild type (WT) IDH was determined. 2HG levels were compared with WHO grades, PFS, 1p19q status and selected metabolites, such as lactate (Lac), glutamate (Glu) and glutamine (Gln).Nonparametric Mann–Whitney test for comparison/Spearman for correlation were used for statistical analysis.

RESULTS

2HG level is elevated in tumors of higher grades. Patients with mIDH have increased PFS compared to those with WT IDH. However, PFS is negatively correlated with the 2HG concentration. 1p/19q co-deletion was found in 27 out of 34 mIDH tumors. mIDH gliomas with 1p/19q co-deleted produced more 2HG. The metabolite measurements showed that the increased 2HG level is related to increases of Glu and Gln, indicating that 2HG may trigger a metabolic shift since Glu/Gln can be converted to a-ketoglutarate, the substrate of mIDH. Furthermore, the levels of Lac and 2HG are found to be positively correlated, possibly due to increase of glucose consumption related to high mutant enzyme activity.

CONCLUSION

2HG level correlates with several prognostic indices, such as PFS and 1p/19q co-deletion status. MRS analysis allows for examining the metabolic flux involving 2HG. Imaging methods capable of monitoring 2HG level may enable non-invasive and longitudinal assessment of prognosis and treatment responses.

CLINICAL RELEVANCE/APPLICATION

MRS detecting 2HG enables genetic classifying gliomas with IDH mutation. The role of 2HG in prognosis and treatment and the need of measuring 2HG level are important to apply this biomarker in precision medicine. 

RC305-03
Yuelei Lyu, Beijing, China (Presenter) Nothing to Disclose
Shuai Liu, Beijing, China (Abstract Co-Author) Nothing to Disclose
Hui You, Beijing, China (Abstract Co-Author) Nothing to Disclose
Bo Hou, BA, Beijing, China (Abstract Co-Author) Nothing to Disclose
Yu Wang, Beijing, China (Abstract Co-Author) Nothing to Disclose
Wenbin Ma, Beijing, China (Abstract Co-Author) Nothing to Disclose
Feng Feng SR, MD, Beijing, China (Abstract Co-Author) Nothing to Disclose
PURPOSE

The aim of this study is to investigate the role of cerebral blood flow (CBF) from a 3D fast spin echo (FSE) pcASL sequence in predicting the response of recurrent high-grade gliomas (rHGGs) treated with bevacizumab (BEV).

METHOD AND MATERIALS

Sixteen patients with rHGGs underwent 3D FSE pcASL imaging 1-2 days before (baseline) and within one month after BEV initiation (post-BEV) were included in this study. Average (aCBF) and maximum (mCBF) cerebral blood flow of the enhanced tumor, their normalized values to contralateral normal appearing white matter (rCBF_wm and mCBF_wm) and cerebellum (rCBF_cb and mCBF_cb), and their changes between baseline and post-BEV were evaluated. ROC curve analysis was utilized to define the optimal cutoff perfusion values for PFS and OS prediction. Kaplan-Meier analysis with log-rank test was applied to assess and compare progression-free survival (PFS) and overall survival (OS) rates.

RESULTS

Cutoffs of aCBF pre-BEV for OS, rCBF_cb pre-BEV for PFS and OS, and ΔaCBF for PFS were of statistical significance in survival prediction (P=0.026, 0.044, 0.046 and 0.048 respectively). Specifically, if aCBF was >43.72(ml/100g/min) at baseline, the median OS was 140 days compared with 404 days when aCBF ≤43.72(ml/100g/min)(P=0.026). The percent changes of aCBF predicted a longer PFS than otherwise, if aCBF decreased more than 37% post-BEV (267 days vs 116 days, P=0.048). With a threshold value of 1.09, rCBF_cb predicted a longer PFS and OS in 9/16 patients when it was less than 1.09 (251 days vs 112 days for PFS; 404 days vs 194 days for OS) (P=0.044, P=0.046).

CONCLUSION

Three-dimensional FSE pcASL technique has the potential to predict the OS and PFS in patients with rHGGs treated with bevacizumab. Average CBF at baseline is a promising imaging biomarker to evaluate the prognosis of recurrent high-grade gliomas.

CLINICAL RELEVANCE/APPLICATION

Three-dimensional pcASL can predict the OS and PFS in patients with recurrent high-grade gliomas treated with bevacizumab and is recommended to evaluate the prognosis of recurrent high-grade gliomas.

RC305-04
Yan Ren, MD, Shanghai, China (Presenter) Nothing to Disclose
Haopeng Pang, Shanghai, China (Abstract Co-Author) Nothing to Disclose
Hong Chen, Shanghai, China (Abstract Co-Author) Nothing to Disclose
Zhenwei Yao, Shanghai, China (Abstract Co-Author) Nothing to Disclose
Jinsong Wu, PhD, Shanghai, China (Abstract Co-Author) Nothing to Disclose
Xiaoyuan Feng, MD, Shanghai, China (Abstract Co-Author) Nothing to Disclose
PURPOSE

To explore the feasibility of non-invasively detecting the status of IDH1 mutation (isocitrate dehydrogenase 1) using non-Gaussian-derived diffusion MR imaging in brain gliomas.

METHOD AND MATERIALS

Fifty-three cases with pathologically confirmed low-grade glioma (WHO gradeⅡ) were enrolled in this prospective study, who performed MR scans with the imaging protocol of T1WI, T2WI, T2-Flair , DWI (0,1000 s/mm2), eDWI (22 b-value DWI of 0-5000 s/mm2) and enhanced-T1WI. In tumor regions of T2-Flair hypertense, excluding remarkable cystic parts, four diffusion parameters of monoexponential -derived apparent diffusion parameter (ADC), biexponential-derived slow diffusion coefficient (Dslow) , stretched exponential-derived distributed diffusion coefficient (DDC) and α were measured to correlate with the status of IDH1 mutation, which was determined by immunohistologic chemistry staining and molecular sequencing. Based on the status of IDH1 mutation, 53 gliomas were divided into two groups of IDH1mut (n=37) and IDH1wild (n=16). Four parameters of ADC, Dslow, DDC and α were employed to make comparisons between two groups of IDH1 mut and IDH1 wild. And ROC curve analyses were used to compare the capability of differentiating gliomas of IDH1 mut from IDH1 wild.

RESULTS

There were significantly increased values of Dslow (mm2/s) and α for groups of IDH1mut than IDH1wild (Dslow: 0.69±0.114 vs. 0.61±0.124; α: 0.87±0.053 vs. 0.82±0.058) with P values of 0.019 and 0.003, respectively. Meanwhile, the increasing signal was shown in diffuse astrocytoma with IDH1 mut than that with IDH1wild for α and Dslow. Otherwise, no significant difference for ADC and DDC were demonstrated between groups of IDH1mut and IDH1wild. ROC curve analyses showed slightly higher AUC of 0.743 (P=0.011) for α than that of 0.722 (P=0.005) for Dslow, respectively.

CONCLUSION

Non-Gaussian-derived parameters of α and Dslow may identify the status of IDH1 mutation in low-grade gliomas. In low-grade gliomas, the water molecules behave more freely and heterogeneously in groups of IDH1mut than IDH1wild.

CLINICAL RELEVANCE/APPLICATION

Non-gaussian diffusion MR imaging may non-invasively identify the status of IDH mutation (isocitrate dehydrogenase) in low-grade gliomas and this exam is recommended when the patients with gliomas hope to take conservative management and pathological samples are not available.

RC305-05
Ju G. Nam, MD, Seoul, Korea, Republic Of (Presenter) Nothing to Disclose
Koung Mi Kang, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
Seung Hong Choi, MD, PhD, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
Tae Jin Yoon, MD, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
Roh-Eul Yoo, MD, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
Ji-Hoon Kim, MD, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
Chul-Ho Sohn, MD, Seoul, Korea, Republic Of (Abstract Co-Author) Nothing to Disclose
PURPOSE

To evaluate the diagnostic performance of the DCE pharmacokinetic parameters and the impact of tissue T1 measurement versus fixed T1 value in differentiating true progression from pseudoprogression of glioblastoma after concurrent radiation therapy and chemotherapy (CCRT) with Temozolomide

METHOD AND MATERIALS

This retrospective study included thirty four histopathologically proven glioblastoma patients who had developed any new enhancing lesion after CCRT, which was defined to be either true (n=14) or pseudoprogression (n=20) following RANO criteria. DCE pharmacokinetic parameters, including the volume transfer constant (Ktrans), the rate transfer constant (Kep), the extravascular extracellular space per unit volume of tissue(Ve), and the blood plasma volume per unit volume of tissue(Vp), for each lesion were calculated twice, using both a fixed T1 of 1000ms and tissue T1 measurement using variable flip angle method. Intraobserver reproducibility was assessed using intraclass correlation coefficient (ICC). The mean of each parameter was compared between two groups using unpaired t test and multivariate analysis. The diagnostic performance was evaluated by receiver operating characteristic (ROC) analysis with Bonferroni correction.

RESULTS

ICC of all eight parameters was excellent (0.760 to 0.999). The mean Ktrans from both fixed T1 and T1 measurement and mean Kep from fixed T1 were significantly higher in the true progression group (true vs. pseudoprogression: 0.14 /min ± 0.15 vs. 0.07 /min ± 0.04, 0.12 /min ± 0.14 vs. 0.06 /min ± 0.04, and 0.30 /min ± 0.26 vs. 0.18 /min ± 0.09; P = .037, .039, and .026, respectively). Multivariable analysis revealed that Kep from fixed T1 was the only independent differentiating variable (P = .019). In ROC analysis, Kep and Ktrans from fixed T1 showed significant diagnostic power distinguishing true from pseudoprogression (AUC 0.718 and 0.714; P = .015 and .016, respectively). None from T1 measurement showed proper diagnostic performance (all Ps>.05).

CONCLUSION

Semiquantitative DCE-derived parameters, Kep and Ktrans, calculated from a fixed T1 act as preferable tools to differentiate true progression from pseudoprogression compared with the parameters from tissue T1 measurement.

CLINICAL RELEVANCE/APPLICATION

Early differentiation of true progression against Temozolomide might help nonreactive patients stop suffering from side effects and give them opportunity for another treatment.

RC305-06
Rajan Jain, MD, Hartsdale, NY (Presenter) Consultant, Cancer Panels; Royalties, Thieme Medical Publishers, Inc
LEARNING OBJECTIVES

1) Describe The Cancer Imaging Archive, Glioma phenotype research group organization and VASARI feature set. 2) Role of imaging in the new age of tumor genomics: How it can improve understanding of tumor biology. 3) Describe imaging characteristics of gliomas based on genomic differences: Phenotype-genotype correlation (Imaging Genomics/Radio-genomics).

ABSTRACT

Recent advances in glioma genomics have significantly changed our understanding of tumor biology and hence, affected how these patients are treated. Similarly, integrating imaging data with genomic markers has also helped create better prognostic and predictive biomarkers which offer promising future for personalized medicine. This session will highlight a multi-disciplinary approach with the focus on advanced imaging and genomics markers before and after therapy in gliomas.     

RC305-07
Bradley J. Erickson, MD, PhD, Rochester, MN, (bje@mayo.edu) (Presenter) Stockholder, OneMedNet Corporation; Stockholder, VoiceIt Technologies, LLC; Stockholder, FlowSigma
LEARNING OBJECTIVES

1) Become familiar with common image processing techniques applied to MRI images obtained in brain tumor patients, and the ways in which they can lead to incorrect conclusions. This will primarily focus on diffusion and perfusion imaging.

ABSTRACT

Diffusion and perfusion imaging is commonly used for brain tumor patients. These methods provide unique insights into what is happening in tumors, particularly during and after therapeutic intervention. Understanding how these methods work can substatially impact interpretation of MRI examiantions in these patients. Hwoever, the processing of diffusion and perfusion images is complex, and can often lead to incorrect conclusions if artifacts and other differences are not understood.While the computation of ADC images from DWI acquistions is straightforward, one must be careful when making conclusions based on minimum values seen in an ROI due to filtering that may or may not be applied to the images. Registration and subtraction can also be useful, but can also have errors in them.Perfusion images, particuarly DSC images, involve very complex processing based on assumptions about what is happening in tissues. Those assuptions substantially impact calculations that are used to produce rCBV and other types of perfusion images. Even seemingly simple things like selection of the normal-appearing white matter can substantially change measured values. Understanding basic principles used in processing can help to avoid or minimize errors in interpretation.

RC305-08
Jerrold L. Boxerman, MD, PhD, Providence, RI (Presenter) Medical Advisor, Imaging Biometrics, LLC
Zheng Zhang, PhD, Providence, RI (Abstract Co-Author) Nothing to Disclose
Yair Safriel, MBBCh, Clearwater, FL (Abstract Co-Author) Principal, PharmaScan Clinical Trials
Jeffrey M. Rogg, MD, Providence, RI (Abstract Co-Author) Nothing to Disclose
Ronald L. Wolf, MD, PhD, Philadelphia, PA (Abstract Co-Author) Nothing to Disclose
Helga Marques, MS, Providence, RI (Abstract Co-Author) Nothing to Disclose
James Gimpel, Philadelphia, PA (Abstract Co-Author) Nothing to Disclose
A. Gregory Sorensen, MD, Malvern, PA (Abstract Co-Author) CEO, Siemens USA Consultant, sanofi-aventis Group Research support, sanofi-aventis Group Consultant, Bayer AG Research support, Exelixis, Inc Research support, Schering-Plough Corporation Consultant, Mitsubishi Corporation Consultant, Biogen Idec Inc Research support, Takeda Pharmaceutical Company Limited
Mark Gilbert, Houston, TX (Abstract Co-Author) Nothing to Disclose
Daniel P. Barboriak, MD, Durham, NC (Abstract Co-Author) Advisory Board, General Electric Company
PURPOSE

ACRIN 6686/RTOG 0825 is a phase III double-blinded placebo-controlled trial of conventional chemoradiation and adjuvant temozolomide with or without bevacizumab (BEV) in newly diagnosed glioblastoma. We aim to assess the association between changes in tumor size measured by contrast enhancement (post-Gd T1) or FLAIR after 3 treatment cycles and overall survival (OS).

METHOD AND MATERIALS

284 patients (171 men; ages 19-79 years, median 57; 159 on BEV arm) had post-op (baseline) and 22-week (pre-cycle 4) MRI. Central readers measured unidimensional (1D), bidimensional (2D) and volumetric (3D) lesion post-Gd T1 and FLAIR hyperintensity at both time points, and determined change from baseline at week 22 for all six imaging markers. Hazard ratio (HR) from Cox proportional hazards model either with or without treatment and marker interaction term and Kaplan-Meier survival estimates with log-rank test were used for inference. 

RESULTS

T1: Increasing 2D-T1 was significantly associated with worse survival (n=261, HR=2.19, 95% CI 1.49-2.93, p<0.0001 adjusting for treatment). Median OS (days) was significantly shorter for patients with increasing versus decreasing 2D-T1 in both BEV (443 vs. 535, p=0.004) and non-BEV (526 vs. 887, p=0.001) arms. T2: Increasing 2D-FLAIR was significantly associated with worse survival (n=272, HR=1.72, 95% CI 1.24-2.39, p=0.001 adjusting for treatment). Median OS was significantly shorter for patients with increasing versus decreasing 2D-FLAIR in the non-BEV arm (595 vs. 872, p=0.001), but not in the BEV arm (499 vs. 535, p=0.12). Results for 1D and 3D measures were similar. T1+T2: Increasing 2D-FLAIR for patients without increasing 1D-T1 tumor enhancement was associated with worse survival overall (HR=1.63, 95% CI 1.15-2.32, p=0.006 adjusting for treatment), with similar association for non-BEV and BEV arms (HR=2.07 vs. 1.65, respectively, p=0.64). 

CONCLUSION

Post-Gd T1 has predictive value for OS in BEV and non-BEV subgroups. FLAIR has independent prognostic value for OS overall, and may further dichotomize patients by OS in both subgroups when there is no increased enhancement on post-Gd T1.

CLINICAL RELEVANCE/APPLICATION

Post-therapy increased T1 enhancement is a useful MRI marker for failed BEV and non-BEV therapy, with added prognostic value of FLAIR particularly in non-BEV therapies.Funded by NCI U01-CA080098 and U01-CA079778.

Honored Educators

Presenters or authors on this event have been recognized as RSNA Honored Educators for participating in multiple qualifying educational activities. Honored Educators are invested in furthering the profession of radiology by delivering high-quality educational content in their field of study. Learn how you can become an honored educator by visiting the website at: https://www.rsna.org/Honored-Educator-Award/

Daniel P. Barboriak, MD - 2013 Honored Educator

RC305-09
Shanshan Jiang, MD, Baltimore, MD (Presenter) Nothing to Disclose
Jinyuan Zhou, PhD, Baltimore, MD (Abstract Co-Author) License agreement, Koninklijke Philips NV
Charles Eberhart, MD,PhD, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Michael Lim, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Alfredo Quinones-Hinojosa, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Huamin Qin, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Hye-Young Heo, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Yi Zhang, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Lindsay Blair, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Zhibo Wen, Guangzhou, China (Abstract Co-Author) Nothing to Disclose
Peter B. Barker, DPhil, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Martin G. Pomper, MD, PhD, Baltimore, MD (Abstract Co-Author) Shareholder, CTS, Inc; Board Member, CTS, Inc; Research Grant, CTS, Inc; Advisor, CTS, Inc; Institutional license agreement, Progenics Pharmaceuticals, Inc; Institutional license agreement, Advanced Accelerator Applications SA; Institutional license agreement, LI-COR, Inc; Institutional license agreement, BIND Therapeutics, Inc
John Laterra, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
Peter C. Van Zijl, PhD, Baltimore, MD (Abstract Co-Author) Speakers Bureau, Koninklijke Philips NV; License agreement, Koninklijke Philips NV
Jaishri Blakeley, Baltimore, MD (Abstract Co-Author) Nothing to Disclose
PURPOSE

The surveillance of post-treatment gliomas remains a formidable radiologic dilemma. Protein-based amide proton transfer-weighted (APTw) MRI has showed promising diagnostic values for gliomas. Here, we performed a radiopathologic correlation analysis via APTw image-directed stereotactic biopsy to determine the accuracy of the APTw signal as an imaging biomarker of active tumor.

METHOD AND MATERIALS

21 patients with suspected malignant glioma recurrence underwent a volumetric APTw imaging sequence at 3T. 64 APTw imaging-guided biopsy specimens were obtained and analyzed for tumor cellularity, necrosis, and proliferation. Specimens were classified as active, quiescent, or mixed by neuropathology. Active and mixed were grouped as recurrence, while quiescent and no tumor were grouped as treatment effect.

RESULTS

20/64 specimens were active tumor, 13 quiescent tumor, and 27 mixed. No tumor was found in four specimens. 9/21 patients had multiple categories within the same lesion. The mean APTw signal intensities were 3.14 ± 0.68% in active specimens, 1.97 ± 1.04% in mixed specimens, and 1.22 ± 0.60% specimens, independent of Gd enhancement. There were moderate to strong positive correlations between APTw and tumor status (R = 0.728), cellularity (R = 0.523), and proliferation (R = 0.407), a mild negative correlation between APTw and necrosis (R = -0.328). Multiple linear regression showed that tumor status was the most powerful predictor of APTw intensities (R2 = 0.575). The APTw intensities showed significant difference between the treatment effect group (1.61 ± 0.93%) and recurrence group (3.14 ± 0.68%), having a cut-off value of 1.64% and an AUC of 0.962 to differentiate these two groups.

CONCLUSION

APTw hyperintensity within heterogeneous tumors is associated with active recurrent tumor. The APTw imaging signal as a biomarker of active glioma has the potential to non-invasively identify the status of malignant gliomas post-treatment.

CLINICAL RELEVANCE/APPLICATION

APTw imaging signal has the potential to non-invasively differentiate tumor recurrence from treatment effect that may eventually avoid surgery where tissues sampling is only for diagnosis.

RC305-10
Mohamed Zaid, MBBCh, Houston, TX (Abstract Co-Author) Nothing to Disclose
Islam S. Hassan, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Wei Wei, Houston, TX (Abstract Co-Author) Nothing to Disclose
Ashok J. Kumar, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Sujit Prabhu, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Jeffrey S. Weinberg, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Raymond Sawaya, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Pascal O. Zinn, MD, Boston, MA (Abstract Co-Author) Nothing to Disclose
Rivka R. Colen, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Nabil A. Elshafeey, MD, Houston, TX (Presenter) Nothing to Disclose
PURPOSE

To evaluate the ability of combined  fMRI and tractography to predict postoperative focal deficits in GBM patients undergoing surgery. 

METHOD AND MATERIALS

We enrolled 220 patients who underwent presurgical fMRI in this IRB approved study. Our inclusion criteria were pathologically proven diagnosis of GBM, full pre and post‐operative neurological examination, pre‐operative task based motor fMRI as well as diffusion tensor imaging (DTI).We obtained multiple fMRI and DTI parameters including Lesion to activation distance (LAD) in T1+C (T1LAD) and T2 FLAIR (FLAIR‐LAD). both from center of activity to center of lesion and from margin of activity to margin of lesion. DTI was done to measure distance between neoplasms margin, edema margin and corticospinal tract (CST) in T1+C and T2 FLAIR respectively. Kruskal‐Wallis test was used to compare distance between patient groups, all tests were 2 sided and p‐values of 0.05 or less we reconsidered statistically significant.

RESULTS

Our cohort was 53% females, 47% males; majority showed Rt. handedness (90%). GBMs were located in Lt. hemisphere in 63% of patients, 38% in the right, 48% in frontal lobe, 30% and 22.5% in parietal and temporal lobes respectively. Gross total resection was achieved in 70 %; 30% underwent subtotal.The most important imaging marker for determining the postoperative status of motor focal neurological deficit (MFND) was Distance between lesion edge and Edema edge with the worst prognosis in development of MFND in patients with FLAIR‐LAD of less than 1 mm (P=0.0004). The second best marker was T1‐LAD measured from edge of tumor to edge of activation; distance below 6.6 mm was associated with deterioration in motor functions (P= 0.002). Similarly, a statistically significant deterioration occurred in T1‐LAD less than 2.9 mm measured from center of tumor to center of activity (p‐value= 0.007).The distance of contrast enhancement and FLAIR signal abnormality to corticospinal tract showed no statistical significance.

CONCLUSION

LAD is an important functional imaging marker for predicting Motor FND in GBM patients. GBM has a significant edema portion that contributes to the potential outcome of patients

CLINICAL RELEVANCE/APPLICATION

Combination of both DTI and fMRI in presurgical mapping for patients with Glioblastoma in close vicinity to motor area is of supreme importance to avoid postoperative deficits while maximizing resection. 

RC305-11
Ahmed T. Shaaban, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Nabil A. Elshafeey, MD, Houston, TX (Presenter) Nothing to Disclose
Ahmed A. Hassan, MBBCh, Houston, TX (Abstract Co-Author) Nothing to Disclose
Ho-Ling Liu, PhD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Ping Hou, PhD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Vinodh A. Kumar, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Ashok J. Kumar, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Norman E. Leeds, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
Rivka R. Colen, MD, Houston, TX (Abstract Co-Author) Nothing to Disclose
PURPOSE

To evaluate the predictive value of Magnetic Resonance Spectroscopy (MRS) as well as different magnetic
resonance perfusion (MRP) parameters including dynamic susceptibility contrast (DSC), Dynamic contrast
enhanced (DCE), arterial spin labeling (ASL) in discriminating Pseudoprogression (PsP) from true
progression (PD) in Glioblastoma (GBM) patients.

METHOD AND MATERIALS

Our Institutional review board has approved this HIPAA-compliant retrospective study. We identified a total
of 69 patients (45 Males: 24 females) (average age =50, median age = 51) with pathologically proven GBM.
All patients underwent advanced MRI studies (DSC, DCE, ASL and MRS). All patients had pathological
proof of either PD or PsP after the advanced MRI studies. For each patient, three board-certified
neuroradiologists, blinded to the pathology report, evaluated all the advanced imaging features using a
designed qualitative questionnaire to determine PD and PsP. The questionnaire included the following
parameters: DCE (PEI 3, 60, Curves), DSC (rCBV, NEI, Curves), ASL (CBF), MRS (Nacetylaspartate,
choline/creatine, lipid). Statistical analysis was performed to evaluate the ability of each imaging feature in
discriminating PD from PsD.

RESULTS

According to the pathology reports, 7 patients had PsP while remaining 62 patients had PD. While MRS was
the most superior imaging features in accurately discriminating PD from PsP followed by DSC, ASL and
DCE respectively, yet, combination of the aforementioned features yielded the best discriminatory results.
The ability to predict Psp versus true progression as compared to the gold standard (pathological
confirmation) was 97%

CONCLUSION

DCE, DSC, ASL, MRS can be valid reliable imaging markers with statistically significant predictive values to
differentiate PD versus PsP. The combination of those different advanced MRI technique can eventually
yield an accurate platform for diagnosis of PD vs PsP

CLINICAL RELEVANCE/APPLICATION

Advanced Imaging analysis can be used as a distinguishable method between the true and pseudoprogression in GBM Patients

RC305-12
Fernando E Boada, New York, NY, (Fernando.Boada@nyumc.org) (Presenter) Nothing to Disclose