Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 183(1): 126-142.e17, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32961131

ABSTRACT

CD19-directed immunotherapies are clinically effective for treating B cell malignancies but also cause a high incidence of neurotoxicity. A subset of patients treated with chimeric antigen receptor (CAR) T cells or bispecific T cell engager (BiTE) antibodies display severe neurotoxicity, including fatal cerebral edema associated with T cell infiltration into the brain. Here, we report that mural cells, which surround the endothelium and are critical for blood-brain-barrier integrity, express CD19. We identify CD19 expression in brain mural cells using single-cell RNA sequencing data and confirm perivascular staining at the protein level. CD19 expression in the brain begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions. Mouse mural cells demonstrate lower levels of Cd19 expression, suggesting limitations in preclinical animal models of neurotoxicity. These data suggest an on-target mechanism for neurotoxicity in CD19-directed therapies and highlight the utility of human single-cell atlases for designing immunotherapies.


Subject(s)
Blood-Brain Barrier/metabolism , Epithelial Cells/metabolism , Immunotherapy, Adoptive/adverse effects , Animals , Antibodies, Bispecific/immunology , Antigens, CD19/immunology , B-Lymphocytes/immunology , Blood-Brain Barrier/immunology , Brain/immunology , Brain/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/metabolism , Neoplasms , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Single-Cell Analysis/methods , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
2.
J Transl Med ; 21(1): 287, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37118754

ABSTRACT

BACKGROUND: Accurate differentiation of pseudoprogression (PsP) from tumor progression (TP) in glioblastomas (GBMs) is essential for appropriate clinical management and prognostication of these patients. In the present study, we sought to validate the findings of our previously developed multiparametric MRI model in a new cohort of GBM patients treated with standard therapy in identifying PsP cases. METHODS: Fifty-six GBM patients demonstrating enhancing lesions within 6 months after completion of concurrent chemo-radiotherapy (CCRT) underwent anatomical imaging, diffusion and perfusion MRI on a 3 T magnet. Subsequently, patients were classified as TP + mixed tumor (n = 37) and PsP (n = 19). When tumor specimens were available from repeat surgery, histopathologic findings were used to identify TP + mixed tumor (> 25% malignant features; n = 34) or PsP (< 25% malignant features; n = 16). In case of non-availability of tumor specimens, ≥ 2 consecutive conventional MRIs using mRANO criteria were used to determine TP + mixed tumor (n = 3) or PsP (n = 3). The multiparametric MRI-based prediction model consisted of predictive probabilities (PP) of tumor progression computed from diffusion and perfusion MRI derived parameters from contrast enhancing regions. In the next step, PP values were used to characterize each lesion as PsP or TP+ mixed tumor. The lesions were considered as PsP if the PP value was < 50% and TP+ mixed tumor if the PP value was ≥ 50%. Pearson test was used to determine the concordance correlation coefficient between PP values and histopathology/mRANO criteria. The area under ROC curve (AUC) was used as a quantitative measure for assessing the discriminatory accuracy of the prediction model in identifying PsP and TP+ mixed tumor. RESULTS: Multiparametric MRI model correctly predicted PsP in 95% (18/19) and TP+ mixed tumor in 57% of cases (21/37) with an overall concordance rate of 70% (39/56) with final diagnosis as determined by histopathology/mRANO criteria. There was a significant concordant correlation coefficient between PP values and histopathology/mRANO criteria (r = 0.56; p < 0.001). The ROC analyses revealed an accuracy of 75.7% in distinguishing PsP from TP+ mixed tumor. Leave-one-out cross-validation test revealed that 73.2% of cases were correctly classified as PsP and TP + mixed tumor. CONCLUSIONS: Our multiparametric MRI based prediction model may be helpful in identifying PsP in GBM patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Multiparametric Magnetic Resonance Imaging , Humans , Glioblastoma/pathology , Brain Neoplasms/pathology , Disease Progression , Magnetic Resonance Imaging , Retrospective Studies
3.
J Neurooncol ; 163(1): 173-183, 2023 May.
Article in English | MEDLINE | ID: mdl-37129737

ABSTRACT

PURPOSE: Autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) is a promising treatment modality for glioblastomas. The purpose of this study was to investigate the potential utility of multiparametric MRI-based prediction model in evaluating treatment response in glioblastoma patients treated with DCVax-L. METHODS: Seventeen glioblastoma patients treated with standard-of-care therapy + DCVax-L were included. When tumor progression (TP) was suspected and repeat surgery was being contemplated, we sought to ascertain the number of cases correctly classified as TP + mixed response or pseudoprogression (PsP) from multiparametric MRI-based prediction model using histopathology/mRANO criteria as ground truth. Multiparametric MRI model consisted of predictive probabilities (PP) of tumor progression computed from diffusion and perfusion MRI-derived parameters. A comparison of overall survival (OS) was performed between patients treated with standard-of-care therapy + DCVax-L and standard-of-care therapy alone (external controls). Additionally, Kaplan-Meier analyses were performed to compare OS between two groups of patients using PsP, Ki-67, and MGMT promoter methylation status as stratification variables. RESULTS: Multiparametric MRI model correctly predicted TP + mixed response in 72.7% of cases (8/11) and PsP in 83.3% (5/6) with an overall concordance rate of 76.5% with final diagnosis as determined by histopathology/mRANO criteria. There was a significant concordant correlation coefficient between PP values and histopathology/mRANO criteria (r = 0.54; p = 0.026). DCVax-L-treated patients had significantly prolonged OS than those treated with standard-of-care therapy (22.38 ± 12.8 vs. 13.8 ± 9.5 months, p = 0.040). Additionally, glioblastomas with PsP, MGMT promoter methylation status, and Ki-67 values below median had longer OS than their counterparts. CONCLUSION: Multiparametric MRI-based prediction model can assess treatment response to DCVax-L in patients with glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Multiparametric Magnetic Resonance Imaging , Vaccines , Humans , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Ki-67 Antigen , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Dendritic Cells
4.
Neuroradiology ; 65(9): 1343-1352, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37468750

ABSTRACT

PURPOSE: While the T2-FLAIR mismatch sign is highly specific for isocitrate dehydrogenase (IDH)-mutant, 1p/19q-noncodeleted astrocytomas among lower-grade gliomas, its utility in WHO grade 4 gliomas is not well-studied. We derived the partial T2-FLAIR mismatch sign as an imaging biomarker for IDH mutation in WHO grade 4 gliomas. METHODS: Preoperative MRI scans of adult WHO grade 4 glioma patients (n = 2165) from the multi-institutional ReSPOND (Radiomics Signatures for PrecisiON Diagnostics) consortium were analyzed. Diagnostic performance of the partial T2-FLAIR mismatch sign was evaluated. Subset analyses were performed to assess associations of imaging markers with overall survival (OS). RESULTS: One hundred twenty-one (5.6%) of 2165 grade 4 gliomas were IDH-mutant. Partial T2-FLAIR mismatch was present in 40 (1.8%) cases, 32 of which were IDH-mutant, yielding 26.4% sensitivity, 99.6% specificity, 80.0% positive predictive value, and 95.8% negative predictive value. Multivariate logistic regression demonstrated IDH mutation was significantly associated with partial T2-FLAIR mismatch (odds ratio [OR] 5.715, 95% CI [1.896, 17.221], p = 0.002), younger age (OR 0.911 [0.895, 0.927], p < 0.001), tumor centered in frontal lobe (OR 3.842, [2.361, 6.251], p < 0.001), absence of multicentricity (OR 0.173, [0.049, 0.612], p = 0.007), and presence of cystic (OR 6.596, [3.023, 14.391], p < 0.001) or non-enhancing solid components (OR 6.069, [3.371, 10.928], p < 0.001). Multivariate Cox analysis demonstrated cystic components (p = 0.024) and non-enhancing solid components (p = 0.003) were associated with longer OS, while older age (p < 0.001), frontal lobe center (p = 0.008), multifocality (p < 0.001), and multicentricity (p < 0.001) were associated with shorter OS. CONCLUSION: Partial T2-FLAIR mismatch sign is highly specific for IDH mutation in WHO grade 4 gliomas.


Subject(s)
Brain Neoplasms , Glioma , Adult , Humans , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Retrospective Studies , Glioma/diagnostic imaging , Glioma/genetics , Magnetic Resonance Imaging/methods , Mutation , World Health Organization
5.
J Digit Imaging ; 36(1): 11-16, 2023 02.
Article in English | MEDLINE | ID: mdl-36279026

ABSTRACT

Technological tools can redesign traditional approaches to radiology education, for example, with simulation cases and via computer-generated feedback. In this study, we investigated the use of an AI-powered, Bayesian inference-based clinical decision support (CDS) software to provide automated "real-time" feedback to trainees during interpretation of clinical and simulation brain MRI examinations. Radiology trainees participated in sessions in which they interpreted 3 brain MRIs: two cases from a routine clinical worklist (one without and one with CDS) and a teaching file-based simulation case with CDS. The CDS software required trainees to input imaging features and differential diagnoses, after which inferred diagnoses were displayed, and the case was reviewed with an attending neuroradiologist. An observer timed each case, including time spent on education, and trainees completed a survey rating their confidence in their findings and the educational value of the case. Ten trainees reviewed 75 brain MRI examinations during 25 reading sessions. Trainees had slightly lower confidence in their findings and diagnosis and rated the educational value slightly higher for simulation cases with CDS compared to clinical cases without CDS (p < 0.05). There were no significant differences in ratings of clinical cases with or without CDS. No differences in overall timing were found among the reading scenarios. Simulation cases with "CDS-provided feedback" may improve the educational value of interpreting imaging studies at a workstation without adding additional time. Further investigation will help drive innovation in trainee education, which may be particularly relevant in this era of increasing remote work and asynchronous attending review.


Subject(s)
Decision Support Systems, Clinical , Internship and Residency , Radiology , Humans , Artificial Intelligence , Bayes Theorem , Radiology/education , Radiography , Clinical Competence
6.
J Neuroradiol ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37652263

ABSTRACT

PURPOSE: To determine if machine learning (ML) or deep learning (DL) pipelines perform better in AI-based three-class classification of glioblastoma (GBM), intracranial metastatic disease (IMD) and primary CNS lymphoma (PCNSL). METHODOLOGY: Retrospective analysis included 502 cases for training (208 GBM, 67 PCNSL and 227 IMD), with external validation on 86 cases (27:27:32). Multiparametric MRI images (T1W, T2W, FLAIR, DWI and T1-CE) were co-registered, resampled, denoised and intensity normalized, followed by semiautomatic 3D segmentation of the enhancing tumor (ET) and peritumoral region (PTR). Model performance was assessed using several ML pipelines and 3D-convolutional neural networks (3D-CNN) using sequence specific masks, as well as combination of masks. All pipelines were trained and evaluated with 5-fold nested cross-validation on internal data followed by external validation using multi-class AUC. RESULTS: Two ML models achieved similar performance on test set, one using T2-ET and T2-PTR masks (AUC: 0.885, 95% CI: [0.816, 0.935] and another using T1-CE-ET and FLAIR-PTR mask (AUC: 0.878, CI: [0.804, 0.930]). The best performing DL models achieved an AUC of 0.854, (CI [0.774, 0.914]) on external data using T1-CE-ET and T2-PTR masks, followed by model derived from T1-CE-ET, ADC-ET and FLAIR-PTR masks (AUC: 0.851, CI [0.772, 0.909]). CONCLUSION: Both ML and DL derived pipelines achieved similar performance. T1-CE mask was used in three of the top four overall models. Additionally, all four models had some mask derived from PTR, either T2WI or FLAIR.

7.
NMR Biomed ; 35(7): e4719, 2022 07.
Article in English | MEDLINE | ID: mdl-35233862

ABSTRACT

Pseudoprogression (PsP) refers to treatment-related clinico-radiologic changes mimicking true progression (TP) that occurs in patients with glioblastoma (GBM), predominantly within the first 6 months after the completion of surgery and concurrent chemoradiation therapy (CCRT) with temozolomide. Accurate differentiation of TP from PsP is essential for making informed decisions on appropriate therapeutic intervention as well as for prognostication of these patients. Conventional neuroimaging findings are often equivocal in distinguishing between TP and PsP and present a considerable diagnostic dilemma to oncologists and radiologists. These challenges have emphasized the need for developing alternative imaging techniques that may aid in the accurate diagnosis of TP and PsP. In this review, we encapsulate the current state of knowledge in the clinical applications of commonly used metabolic and physiologic magnetic resonance (MR) imaging techniques such as diffusion and perfusion imaging and proton spectroscopy in distinguishing TP from PsP. We also showcase the potential of promising imaging techniques, such as amide proton transfer and amino acid-based positron emission tomography, in providing useful information about the treatment response. Additionally, we highlight the role of "radiomics", which is an emerging field of radiology that has the potential to change the way in which advanced MR techniques are utilized in assessing treatment response in GBM patients. Finally, we present our institutional experiences and discuss future perspectives on the role of multiparametric MR imaging in identifying PsP in GBM patients treated with "standard-of-care" CCRT as well as novel/targeted therapies.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Disease Progression , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Magnetic Resonance Imaging/methods , Protons
8.
AJR Am J Roentgenol ; 218(5): 831-832, 2022 05.
Article in English | MEDLINE | ID: mdl-34910536

ABSTRACT

Although professional societies now support MRI in patients with nonconditional (legacy) cardiac implanted electronic devices (CIEDs), concern remains regarding potential cumulative effects of serial examinations. We evaluated 481 patients with CIEDs who underwent 599 1.5-T MRI examinations (44.6% cardiac examinations), including 68 patients who underwent multiple examinations (maximum, seven examinations). No major events occurred. The minor adverse event rate was 5.7%. Multiple statistical evaluations showed no increase in adverse event rate with increasing number of previous examinations.


Subject(s)
Defibrillators, Implantable , Pacemaker, Artificial , Electronics , Humans , Magnetic Resonance Imaging/adverse effects , Physical Examination
9.
J Clin Ultrasound ; 50(9): 1353-1359, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36205388

ABSTRACT

In view of the inherent limitations associated with performing dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI) in clinical settings, current study was designed to provide a proof of principle that Doppler sonography and DCE-MRI derived perfusion parameters yield similar hemodynamic information from metastatic lymph nodes in squamous cell carcinomas of head and neck (HNSCCs). Strong positive correlations between volume fraction of plasma space in tissues (Vp ) and blood volume (r = 0.72, p = 0.02) and between Vp and %area perfused (r = 0.65, p = 0.04) were observed. Additionally, a moderate positive correlation trending towards significance was obtained between volume transfer constant (Ktrans ) and %area perfused (r = 0.49, p = 0.09).


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/drug therapy , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/drug therapy , Contrast Media , Induction Chemotherapy , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Magnetic Resonance Imaging/methods
10.
AJR Am J Roentgenol ; 216(4): 1046-1047, 2021 04.
Article in English | MEDLINE | ID: mdl-32903058

ABSTRACT

Among 2820 inpatients with coronavirus disease (COVID-19), 59 (2.1%) underwent brain MRI. Of them, six (10.2%) had MRI findings suspicious for COVID-19-related disseminated leukoencephalopathy (CRDL), which is characterized by extensive confluent or multifocal white matter lesions (with characteristics and locations atypical for other causes), microhemorrhages, diffusion restriction, and enhancement. CRDL is an uncommon but important differential consideration in patients with neurologic manifestations of COVID-19.


Subject(s)
Brain/diagnostic imaging , COVID-19/complications , Leukoencephalopathies/etiology , Magnetic Resonance Imaging/methods , Pandemics , SARS-CoV-2 , Adult , Aged , Aged, 80 and over , COVID-19/epidemiology , Female , Humans , Leukoencephalopathies/diagnosis , Male , Middle Aged , Retrospective Studies
11.
AJR Am J Roentgenol ; 217(4): 959-974, 2021 10.
Article in English | MEDLINE | ID: mdl-33236647

ABSTRACT

Neurologic involvement is well-recognized in COVID-19. This article reviews the neuroimaging manifestations of COVID-19 on CT and MRI, presenting cases from the New York City metropolitan region encountered by the authors during the first surge of the pandemic. The most common neuroimaging manifestations are acute infarcts with large clot burden and intracranial hemorrhage, including microhemorrhages. However, a wide range of additional imaging patterns occur, including leukoencephalopathy, global hypoxic injury, acute disseminated encephalomyelitis, cytotoxic lesions of the corpus callosum, olfactory bulb involvement, cranial nerve enhancement, and Guillain-Barré syndrome. The described CNS abnormalities largely represent secondary involvement from immune activation that leads to a prothrombotic state and cytokine storm; evidence for direct neuroinvasion is scant. Comorbidities such as hypertension, complications of prolonged illness and hospitalization, and associated supportive treatments also contribute to the CNS involvement in COVID-19. Routine long-term neurologic follow-up may be warranted, given emerging evidence of long-term microstructural and functional changes on brain imaging after COVID-19 recovery.


Subject(s)
Brain Diseases/complications , Brain Diseases/diagnostic imaging , COVID-19/complications , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Tomography, X-Ray Computed/methods , Adult , Brain/diagnostic imaging , Humans , Pandemics , SARS-CoV-2
12.
J Digit Imaging ; 34(4): 1049-1058, 2021 08.
Article in English | MEDLINE | ID: mdl-34131794

ABSTRACT

Automated quantitative and probabilistic medical image analysis has the potential to improve the accuracy and efficiency of the radiology workflow. We sought to determine whether AI systems for brain MRI diagnosis could be used as a clinical decision support tool to augment radiologist performance. We utilized previously developed AI systems that combine convolutional neural networks and expert-derived Bayesian networks to distinguish among 50 diagnostic entities on multimodal brain MRIs. We tested whether these systems could augment radiologist performance through an interactive clinical decision support tool known as Adaptive Radiology Interpretation and Education System (ARIES) in 194 test cases. Four radiology residents and three academic neuroradiologists viewed half of the cases unassisted and half with the results of the AI system displayed on ARIES. Diagnostic accuracy of radiologists for top diagnosis (TDx) and top three differential diagnosis (T3DDx) was compared with and without ARIES. Radiology resident performance was significantly better with ARIES for both TDx (55% vs 30%; P < .001) and T3DDx (79% vs 52%; P = 0.002), with the largest improvement for rare diseases (39% increase for T3DDx; P < 0.001). There was no significant difference between attending performance with and without ARIES for TDx (72% vs 69%; P = 0.48) or T3DDx (86% vs 89%; P = 0.39). These findings suggest that a hybrid deep learning and Bayesian inference clinical decision support system has the potential to augment diagnostic accuracy of non-specialists to approach the level of subspecialists for a large array of diseases on brain MRI.


Subject(s)
Deep Learning , Radiology , Bayes Theorem , Brain/diagnostic imaging , Humans , Magnetic Resonance Imaging
13.
Cancer ; 126(11): 2625-2636, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32129893

ABSTRACT

BACKGROUND: Imaging of glioblastoma patients after maximal safe resection and chemoradiation commonly demonstrates new enhancements that raise concerns about tumor progression. However, in 30% to 50% of patients, these enhancements primarily represent the effects of treatment, or pseudo-progression (PsP). We hypothesize that quantitative machine learning analysis of clinically acquired multiparametric magnetic resonance imaging (mpMRI) can identify subvisual imaging characteristics to provide robust, noninvasive imaging signatures that can distinguish true progression (TP) from PsP. METHODS: We evaluated independent discovery (n = 40) and replication (n = 23) cohorts of glioblastoma patients who underwent second resection due to progressive radiographic changes suspicious for recurrence. Deep learning and conventional feature extraction methods were used to extract quantitative characteristics from the mpMRI scans. Multivariate analysis of these features revealed radiophenotypic signatures distinguishing among TP, PsP, and mixed response that compared with similar categories blindly defined by board-certified neuropathologists. Additionally, interinstitutional validation was performed on 20 new patients. RESULTS: Patients who demonstrate TP on neuropathology are significantly different (P < .0001) from those with PsP, showing imaging features reflecting higher angiogenesis, higher cellularity, and lower water concentration. The accuracy of the proposed signature in leave-one-out cross-validation was 87% for predicting PsP (area under the curve [AUC], 0.92) and 84% for predicting TP (AUC, 0.83), whereas in the discovery/replication cohort, the accuracy was 87% for predicting PsP (AUC, 0.84) and 78% for TP (AUC, 0.80). The accuracy in the interinstitutional cohort was 75% (AUC, 0.80). CONCLUSION: Quantitative mpMRI analysis via machine learning reveals distinctive noninvasive signatures of TP versus PsP after treatment of glioblastoma. Integration of the proposed method into clinical studies can be performed using the freely available Cancer Imaging Phenomics Toolkit.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Machine Learning , Magnetic Resonance Imaging/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Brain Neoplasms/diagnostic imaging , Disease Progression , Female , Glioblastoma/diagnostic imaging , Humans , Male , Middle Aged
14.
Radiology ; 295(3): 626-637, 2020 06.
Article in English | MEDLINE | ID: mdl-32255417

ABSTRACT

Background Although artificial intelligence (AI) shows promise across many aspects of radiology, the use of AI to create differential diagnoses for rare and common diseases at brain MRI has not been demonstrated. Purpose To evaluate an AI system for generation of differential diagnoses at brain MRI compared with radiologists. Materials and Methods This retrospective study tested performance of an AI system for probabilistic diagnosis in patients with 19 common and rare diagnoses at brain MRI acquired between January 2008 and January 2018. The AI system combines data-driven and domain-expertise methodologies, including deep learning and Bayesian networks. First, lesions were detected by using deep learning. Then, 18 quantitative imaging features were extracted by using atlas-based coregistration and segmentation. Third, these image features were combined with five clinical features by using Bayesian inference to develop probability-ranked differential diagnoses. Quantitative feature extraction algorithms and conditional probabilities were fine-tuned on a training set of 86 patients (mean age, 49 years ± 16 [standard deviation]; 53 women). Accuracy was compared with radiology residents, general radiologists, neuroradiology fellows, and academic neuroradiologists by using accuracy of top one, top two, and top three differential diagnoses in 92 independent test set patients (mean age, 47 years ± 18; 52 women). Results For accuracy of top three differential diagnoses, the AI system (91% correct) performed similarly to academic neuroradiologists (86% correct; P = .20), and better than radiology residents (56%; P < .001), general radiologists (57%; P < .001), and neuroradiology fellows (77%; P = .003). The performance of the AI system was not affected by disease prevalence (93% accuracy for common vs 85% for rare diseases; P = .26). Radiologists were more accurate at diagnosing common versus rare diagnoses (78% vs 47% across all radiologists; P < .001). Conclusion An artificial intelligence system for brain MRI approached overall top one, top two, and top three differential diagnoses accuracy of neuroradiologists and exceeded that of less-specialized radiologists. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Zaharchuk in this issue.


Subject(s)
Artificial Intelligence , Brain Diseases/diagnostic imaging , Brain/diagnostic imaging , Diagnosis, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Adult , Aged , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Rare Diseases , Retrospective Studies , Sensitivity and Specificity
15.
J Magn Reson Imaging ; 52(4): 978-997, 2020 10.
Article in English | MEDLINE | ID: mdl-32190946

ABSTRACT

Glioblastoma is the most common and most malignant primary brain tumor. Despite aggressive multimodal treatment, its prognosis remains poor. Even with continuous developments in MRI, which has provided us with newer insights into the diagnosis and understanding of tumor biology, response assessment in the posttherapy setting remains challenging. We believe that the integration of additional information from advanced neuroimaging techniques can further improve the diagnostic accuracy of conventional MRI. In this article, we review the utility of advanced neuroimaging techniques such as diffusion-weighted imaging, diffusion tensor imaging, perfusion-weighted imaging, proton magnetic resonance spectroscopy, and chemical exchange saturation transfer in characterizing and evaluating treatment response in patients with glioblastoma. We will also discuss the existing challenges and limitations of using these techniques in clinical settings and possible solutions to avoiding pitfalls in study design, data acquisition, and analysis for future studies. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE: 3 J. Magn. Reson. Imaging 2020;52:978-997.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Diffusion Tensor Imaging , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Humans , Magnetic Resonance Imaging
16.
Br J Cancer ; 120(1): 54-56, 2019 01.
Article in English | MEDLINE | ID: mdl-30478409

ABSTRACT

EGFRvIII targeted chimeric antigen receptor T (CAR-T) cell therapy has recently been reported for treating glioblastomas (GBMs); however, physiology-based MRI parameters have not been evaluated in this setting. Ten patients underwent multiparametric MRI at baseline, 1, 2 and 3 months after CAR-T therapy. Logistic regression model derived progression probabilities (PP) using imaging parameters were used to assess treatment response. Four lesions from "early surgery" group demonstrated high PP at baseline suggestive of progression, which was confirmed histologically. Out of eight lesions from remaining six patients, three lesions with low PP at baseline remained stable. Two lesions with high PP at baseline were associated with large decreases in PP reflecting treatment response, whereas other two lesions with high PP at baseline continued to demonstrate progression. One patient didn't have baseline data but demonstrated progression on follow-up. Our findings indicate that multiparametric MRI may be helpful in monitoring CAR-T related early therapeutic changes in GBM patients.


Subject(s)
ErbB Receptors/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive , Neoplasm Recurrence, Local/therapy , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Female , Glioblastoma/diagnostic imaging , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local/diagnostic imaging , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use
17.
Radiology ; 290(3): 607-618, 2019 03.
Article in English | MEDLINE | ID: mdl-30667332

ABSTRACT

Due to the exponential growth of computational algorithms, artificial intelligence (AI) methods are poised to improve the precision of diagnostic and therapeutic methods in medicine. The field of radiomics in neuro-oncology has been and will likely continue to be at the forefront of this revolution. A variety of AI methods applied to conventional and advanced neuro-oncology MRI data can already delineate infiltrating margins of diffuse gliomas, differentiate pseudoprogression from true progression, and predict recurrence and survival better than methods used in daily clinical practice. Radiogenomics will also advance our understanding of cancer biology, allowing noninvasive sampling of the molecular environment with high spatial resolution and providing a systems-level understanding of underlying heterogeneous cellular and molecular processes. By providing in vivo markers of spatial and molecular heterogeneity, these AI-based radiomic and radiogenomic tools have the potential to stratify patients into more precise initial diagnostic and therapeutic pathways and enable better dynamic treatment monitoring in this era of personalized medicine. Although substantial challenges remain, radiologic practice is set to change considerably as AI technology is further developed and validated for clinical use.


Subject(s)
Artificial Intelligence , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Magnetic Resonance Imaging , Precision Medicine , Radiology/methods , Algorithms , Biomarkers, Tumor/metabolism , Humans , Tumor Microenvironment
18.
NMR Biomed ; 32(2): e4042, 2019 02.
Article in English | MEDLINE | ID: mdl-30556932

ABSTRACT

Accurate differentiation of true progression (TP) from pseudoprogression (PsP) in patients with glioblastomas (GBMs) is essential for planning adequate treatment and for estimating clinical outcome measures and future prognosis. The purpose of this study was to investigate the utility of three-dimensional echo planar spectroscopic imaging (3D-EPSI) in distinguishing TP from PsP in GBM patients. For this institutional review board approved and HIPAA compliant retrospective study, 27 patients with GBM demonstrating enhancing lesions within six months of completion of concurrent chemo-radiation therapy were included. Of these, 18 were subsequently classified as TP and 9 as PsP based on histological features or follow-up MRI studies. Parametric maps of choline/creatine (Cho/Cr) and choline/N-acetylaspartate (Cho/NAA) were computed and co-registered with post-contrast T1 -weighted and FLAIR images. All lesions were segmented into contrast enhancing (CER), immediate peritumoral (IPR), and distal peritumoral (DPR) regions. For each region, Cho/Cr and Cho/NAA ratios were normalized to corresponding metabolite ratios from contralateral normal parenchyma and compared between TP and PsP groups. Logistic regression analyses were performed to obtain the best model to distinguish TP from PsP. Significantly higher Cho/NAA was observed from CER (2.69 ± 1.00 versus 1.56 ± 0.51, p = 0.003), IPR (2.31 ± 0.92 versus 1.53 ± 0.56, p = 0.030), and DPR (1.80 ± 0.68 versus 1.19 ± 0.28, p = 0.035) regions in TP patients compared with those with PsP. Additionally, significantly elevated Cho/Cr (1.74 ± 0.44 versus 1.34 ± 0.26, p = 0.023) from CER was observed in TP compared with PsP. When these parameters were incorporated in multivariate regression analyses, a discriminatory model with a sensitivity of 94% and a specificity of 87% was observed in distinguishing TP from PsP. These results indicate the utility of 3D-EPSI in differentiating TP from PsP with high sensitivity and specificity.


Subject(s)
Disease Progression , Echo-Planar Imaging , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Area Under Curve , Female , Humans , Logistic Models , Male , Metabolome , Middle Aged , Proton Magnetic Resonance Spectroscopy , ROC Curve
19.
J Magn Reson Imaging ; 49(1): 184-194, 2019 01.
Article in English | MEDLINE | ID: mdl-29676844

ABSTRACT

BACKGROUND: Accurate differentiation of brain infections from necrotic glioblastomas (GBMs) may not always be possible on morphologic MRI or on diffusion tensor imaging (DTI) and dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) if these techniques are used independently. PURPOSE: To investigate the combined analysis of DTI and DSC-PWI in distinguishing brain injections from necrotic GBMs. STUDY TYPE: Retrospective. POPULATION: Fourteen patients with brain infections and 21 patients with necrotic GBMs. FIELD STRENGTH/SEQUENCE: 3T MRI, DTI, and DSC-PWI. ASSESSMENT: Parametric maps of mean diffusivity (MD), fractional anisotropy (FA), coefficient of linear (CL), and planar anisotropy (CP) and leakage corrected cerebral blood volume (CBV) were computed and coregistered with postcontrast T1 -weighted and FLAIR images. All lesions were segmented into the central core and enhancing region. For each region, median values of MD, FA, CL, CP, relative CBV (rCBV), and top 90th percentile of rCBV (rCBVmax ) were measured. STATISTICAL TESTS: All parameters from both regions were compared between brain infections and necrotic GBMs using Mann-Whitney tests. Logistic regression analyses were performed to obtain the best model in distinguishing these two conditions. RESULTS: From the central core, significantly lower MD (0.90 × 10-3 ± 0.44 × 10-3 mm2 /s vs. 1.66 × 10-3 ± 0.62 × 10-3 mm2 /s, P = 0.001), significantly higher FA (0.15 ± 0.06 vs. 0.09 ± 0.03, P < 0.001), and CP (0.07 ± 0.03 vs. 0.04 ± 0.02, P = 0.009) were observed in brain infections compared to those in necrotic GBMs. Additionally, from the contrast-enhancing region, significantly lower rCBV (1.91 ± 0.95 vs. 2.76 ± 1.24, P = 0.031) and rCBVmax (3.46 ± 1.41 vs. 5.89 ± 2.06, P = 0.001) were observed from infective lesions compared to necrotic GBMs. FA from the central core and rCBVmax from enhancing region provided the best classification model in distinguishing brain infections from necrotic GBMs, with a sensitivity of 91% and a specificity of 93%. DATA CONCLUSION: Combined analysis of DTI and DSC-PWI may provide better performance in differentiating brain infections from necrotic GBMs. LEVEL OF EVIDENCE: 1 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;49:184-194.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Glioblastoma/diagnostic imaging , Infections/diagnostic imaging , Magnetic Resonance Angiography , Necrosis/diagnostic imaging , Adult , Aged , Anisotropy , Brain/microbiology , Contrast Media/administration & dosage , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Reproducibility of Results , Retrospective Studies
20.
World J Urol ; 37(10): 2175-2182, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30560299

ABSTRACT

PURPOSE: To evaluate the efficacy and outcome of superselective vesical arterial embolization in the management of severe intractable hematuria secondary to hemorrhagic cystitis. MATERIALS AND METHODS: We retrospectively reviewed the medical records of nine patients with severe intractable hematuria treated with superselective vesical artery embolization at our institution between March 2003 and February 2015. There were six males and three females with a mean age of 56.1 years. Seven patients had transitional cell carcinoma (TCC) of urinary bladder and had undergone transurethral resection of bladder tumor and pelvic radiotherapy. One patient had synchronous renal pelvis and bladder TCC. One patient had aortoarteritis and was receiving cyclophosphamide therapy and another patient had carcinoma cervix post-pelvic radiotherapy. Following the failure of conservative management, superselective vesical artery catheterization and embolization was performed with 300-500-µ PVA particles in all patients. Coil embolization of inferior gluteal artery followed by particle embolization of vesical arteries was done in one patient in whom superior, inferior vesical and inferior gluteal arteries were arising as a trifurcation. RESULTS: The technical success rate was 100% with complete cessation of hematuria within 48 h in all patients. No significant complications were noted, except for post-embolization syndrome in one patient, which improved on symptomatic treatment. During a mean follow-up period of 14.45 months (ranging from 3-28 months), one patient had mild recurrent hematuria (at 2 months) which resolved spontaneously. CONCLUSIONS: Superselective vesical artery embolization is a safe and effective procedure in controlling intractable life-threatening hematuria in a select group of patients who have failed conventional treatment protocols. This procedure may be considered as the treatment of choice since it usually obviates the need for emergency surgery in these severely ill patients.


Subject(s)
Cystitis/complications , Embolization, Therapeutic/methods , Hematuria/etiology , Hematuria/therapy , Hemorrhage/complications , Arteries , Female , Humans , Male , Middle Aged , Retrospective Studies , Urinary Bladder/blood supply
SELECTION OF CITATIONS
SEARCH DETAIL