Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Eur Radiol ; 34(2): 810-822, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37606663

ABSTRACT

OBJECTIVES: Non-contrast computed tomography of the brain (NCCTB) is commonly used to detect intracranial pathology but is subject to interpretation errors. Machine learning can augment clinical decision-making and improve NCCTB scan interpretation. This retrospective detection accuracy study assessed the performance of radiologists assisted by a deep learning model and compared the standalone performance of the model with that of unassisted radiologists. METHODS: A deep learning model was trained on 212,484 NCCTB scans drawn from a private radiology group in Australia. Scans from inpatient, outpatient, and emergency settings were included. Scan inclusion criteria were age ≥ 18 years and series slice thickness ≤ 1.5 mm. Thirty-two radiologists reviewed 2848 scans with and without the assistance of the deep learning system and rated their confidence in the presence of each finding using a 7-point scale. Differences in AUC and Matthews correlation coefficient (MCC) were calculated using a ground-truth gold standard. RESULTS: The model demonstrated an average area under the receiver operating characteristic curve (AUC) of 0.93 across 144 NCCTB findings and significantly improved radiologist interpretation performance. Assisted and unassisted radiologists demonstrated an average AUC of 0.79 and 0.73 across 22 grouped parent findings and 0.72 and 0.68 across 189 child findings, respectively. When assisted by the model, radiologist AUC was significantly improved for 91 findings (158 findings were non-inferior), and reading time was significantly reduced. CONCLUSIONS: The assistance of a comprehensive deep learning model significantly improved radiologist detection accuracy across a wide range of clinical findings and demonstrated the potential to improve NCCTB interpretation. CLINICAL RELEVANCE STATEMENT: This study evaluated a comprehensive CT brain deep learning model, which performed strongly, improved the performance of radiologists, and reduced interpretation time. The model may reduce errors, improve efficiency, facilitate triage, and better enable the delivery of timely patient care. KEY POINTS: • This study demonstrated that the use of a comprehensive deep learning system assisted radiologists in the detection of a wide range of abnormalities on non-contrast brain computed tomography scans. • The deep learning model demonstrated an average area under the receiver operating characteristic curve of 0.93 across 144 findings and significantly improved radiologist interpretation performance. • The assistance of the comprehensive deep learning model significantly reduced the time required for radiologists to interpret computed tomography scans of the brain.


Subject(s)
Deep Learning , Adolescent , Humans , Radiography , Radiologists , Retrospective Studies , Tomography, X-Ray Computed/methods , Adult
2.
JAMA Oncol ; 10(2): 193-201, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38095878

ABSTRACT

Importance: Agents targeting programmed death ligand 1 (PD-L1) have demonstrated efficacy in triple-negative breast cancer (TNBC) when combined with chemotherapy and are now the standard of care in patients with PD-L1-positive metastatic disease. In contrast to microtubule-targeting agents, the effect of combining platinum compounds with programmed cell death 1 (PD-1)/PD-L1 immunotherapy has not been extensively determined. Objective: To evaluate the efficacy of atezolizumab with carboplatin in patients with metastatic TNBC. Design, Setting, and Participants: This phase 2 randomized clinical trial was conducted in 6 centers from August 2017 to June 2021. Interventions: Patients with metastatic TNBC were randomized to receive carboplatin area under the curve (AUC) 6 alone or with atezolizumab, 1200 mg, every 3 weeks until disease progression or unacceptable toxic effects with a 3-year duration of follow-up. Main Outcome and Measures: The primary end point was investigator-assessed progression-free survival (PFS). Secondary end points included overall response rate (ORR), clinical benefit rate (CBR), and overall survival (OS). Other objectives included correlation of response with tumor PD-L1 levels, tumor-infiltrating lymphocytes (TILs), tumor DNA- and RNA-sequenced biomarkers, TNBC subtyping, and multiplex analyses of immune markers. Results: All 106 patients with metastatic TNBC who were enrolled were female with a mean (range) age of 55 (27-79) years, of which 12 (19%) identified as African American/Black, 1 (1%) as Asian, 73 (69%) as White, and 11 (10%) as unknown. Patients were randomized and received either carboplatin (n = 50) or carboplatin and atezolizumab (n = 56). The combination improved PFS (hazard ratio [HR], 0.66; 95% CI, 0.44-1.01; P = .05) from a median of 2.2 to 4.1 months, increased ORR from 8.0% (95% CI, 3.2%-18.8%) to 30.4% (95% CI, 19.9%-43.3%), increased CBR at 6 months from 18.0% (95% CI, 9.8%-30.1%) to 37.5% (95% CI, 26.0%-50.6%), and improved OS (HR, 0.60; 95% CI, 0.37-0.96; P = .03) from a median of 8.6 to 12.6 months. Subgroup analysis showed PD-L1-positive tumors did not benefit more from adding atezolizumab (HR, 0.62; 95% CI, 0.23-1.65; P = .35). Patients with high TILs (HR, 0.12; 95% CI, 0.30-0.50), high mutation burden (HR, 0.50; 95% CI, 0.23-1.06), and prior chemotherapy (HR, 0.59; 95% CI, 0.36-0.95) received greater benefit on the combination. Patients with obesity and patients with more than 125 mg/dL on-treatment blood glucose levels were associated with better PFS (HR, 0.35; 95% CI, 0.10-1.80) on the combination. TNBC subtypes benefited from adding atezolizumab, except the luminal androgen receptor subtype. Conclusions and Relevance: In this randomized clinical trial, the addition of atezolizumab to carboplatin significantly improved survival of patients with metastatic TNBC regardless of PD-L1 status. Further, lower risk of disease progression was associated with increased TILs, higher mutation burden, obesity, and uncontrolled blood glucose levels. Trial Registration: ClinicalTrials.gov Identifier: NCT03206203.


Subject(s)
Antibodies, Monoclonal, Humanized , Triple Negative Breast Neoplasms , Humans , Female , Middle Aged , Aged , Male , Carboplatin/therapeutic use , Triple Negative Breast Neoplasms/pathology , B7-H1 Antigen/immunology , Blood Glucose , Ligands , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Disease Progression , Obesity , Apoptosis
3.
Med Image Anal ; 90: 102939, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37725868

ABSTRACT

Transformer-based models, capable of learning better global dependencies, have recently demonstrated exceptional representation learning capabilities in computer vision and medical image analysis. Transformer reformats the image into separate patches and realizes global communication via the self-attention mechanism. However, positional information between patches is hard to preserve in such 1D sequences, and loss of it can lead to sub-optimal performance when dealing with large amounts of heterogeneous tissues of various sizes in 3D medical image segmentation. Additionally, current methods are not robust and efficient for heavy-duty medical segmentation tasks such as predicting a large number of tissue classes or modeling globally inter-connected tissue structures. To address such challenges and inspired by the nested hierarchical structures in vision transformer, we proposed a novel 3D medical image segmentation method (UNesT), employing a simplified and faster-converging transformer encoder design that achieves local communication among spatially adjacent patch sequences by aggregating them hierarchically. We extensively validate our method on multiple challenging datasets, consisting of multiple modalities, anatomies, and a wide range of tissue classes, including 133 structures in the brain, 14 organs in the abdomen, 4 hierarchical components in the kidneys, inter-connected kidney tumors and brain tumors. We show that UNesT consistently achieves state-of-the-art performance and evaluate its generalizability and data efficiency. Particularly, the model achieves whole brain segmentation task complete ROI with 133 tissue classes in a single network, outperforming prior state-of-the-art method SLANT27 ensembled with 27 networks. Our model performance increases the mean DSC score of the publicly available Colin and CANDI dataset from 0.7264 to 0.7444 and from 0.6968 to 0.7025, respectively. Code, pre-trained models, and use case pipeline are available at: https://github.com/MASILab/UNesT.

5.
Proc IEEE Int Symp Biomed Imaging ; 2021: 1491-1494, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34667487

ABSTRACT

Performing coarse-to-fine abdominal multi-organ segmentation facilitates extraction of high-resolution segmentation minimizing the loss of spatial contextual information. However, current coarse-to-refine approaches require a significant number of models to perform single organ segmentation. We propose a coarse-to-fine pipeline RAP-Net, which starts from the extraction of the global prior context of multiple organs from 3D volumes using a low-resolution coarse network, followed by a fine phase that uses a single refined model to segment all abdominal organs instead of multiple organ corresponding models. We combine the anatomical prior with corresponding extracted patches to preserve the anatomical locations and boundary information for performing high-resolution segmentation across all organs in a single model. To train and evaluate our method, a clinical research cohort consisting of 100 patient volumes with 13 organs well-annotated is used. We tested our algorithms with 4-fold cross-validation and computed the Dice score for evaluating the segmentation performance of the 13 organs. Our proposed method using single auto-context outperforms the state-of-the-art on 13 models with an average Dice score 84.58% versus 81.69% (p<0.0001).

6.
J Med Imaging (Bellingham) ; 8(1): 014004, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33634205

ABSTRACT

Purpose: Deep learning is a promising technique for spleen segmentation. Our study aims to validate the reproducibility of deep learning-based spleen volume estimation by performing spleen segmentation on clinically acquired computed tomography (CT) scans from patients with myeloproliferative neoplasms. Approach: As approved by the institutional review board, we obtained 138 de-identified abdominal CT scans. A sum of voxel volume on an expert annotator's segmentations establishes the ground truth (estimation 1). We used our deep convolutional neural network (estimation 2) alongside traditional linear estimations (estimation 3 and 4) to estimate spleen volumes independently. Dice coefficient, Hausdorff distance, R 2 coefficient, Pearson R coefficient, the absolute difference in volume, and the relative difference in volume were calculated for 2 to 4 against the ground truth to compare and assess methods' performances. We re-labeled on scan-rescan on a subset of 40 studies to evaluate method reproducibility. Results: Calculated against the ground truth, the R 2 coefficients for our method (estimation 2) and linear method (estimation 3 and 4) are 0.998, 0.954, and 0.973, respectively. The Pearson R coefficients for the estimations against the ground truth are 0.999, 0.963, and 0.978, respectively (paired t -tests produced p < 0.05 between 2 and 3, and 2 and 4). Conclusion: The deep convolutional neural network algorithm shows excellent potential in rendering more precise spleen volume estimations. Our computer-aided segmentation exhibits reasonable improvements in splenic volume estimation accuracy.

7.
Med Image Anal ; 69: 101894, 2021 04.
Article in English | MEDLINE | ID: mdl-33421919

ABSTRACT

Deep learning for three dimensional (3D) abdominal organ segmentation on high-resolution computed tomography (CT) is a challenging topic, in part due to the limited memory provide by graphics processing units (GPU) and large number of parameters and in 3D fully convolutional networks (FCN). Two prevalent strategies, lower resolution with wider field of view and higher resolution with limited field of view, have been explored but have been presented with varying degrees of success. In this paper, we propose a novel patch-based network with random spatial initialization and statistical fusion on overlapping regions of interest (ROIs). We evaluate the proposed approach using three datasets consisting of 260 subjects with varying numbers of manual labels. Compared with the canonical "coarse-to-fine" baseline methods, the proposed method increases the performance on multi-organ segmentation from 0.799 to 0.856 in terms of mean DSC score (p-value < 0.01 with paired t-test). The effect of different numbers of patches is evaluated by increasing the depth of coverage (expected number of patches evaluated per voxel). In addition, our method outperforms other state-of-the-art methods in abdominal organ segmentation. In conclusion, the approach provides a memory-conservative framework to enable 3D segmentation on high-resolution CT. The approach is compatible with many base network structures, without substantially increasing the complexity during inference. Given a CT scan with at high resolution, a low-res section (left panel) is trained with multi-channel segmentation. The low-res part contains down-sampling and normalization in order to preserve the complete spatial information. Interpolation and random patch sampling (mid panel) is employed to collect patches. The high-dimensional probability maps are acquired (right panel) from integration of all patches on field of views.


Subject(s)
Imaging, Three-Dimensional , Neural Networks, Computer , Abdomen/diagnostic imaging , Humans , Image Processing, Computer-Assisted , Tomography, X-Ray Computed
8.
J Med Imaging (Bellingham) ; 7(4): 044002, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32775501

ABSTRACT

Purpose: Deep learning methods have become essential tools for quantitative interpretation of medical imaging data, but training these approaches is highly sensitive to biases and class imbalance in the available data. There is an opportunity to increase the available training data by combining across different data sources (e.g., distinct public projects); however, data collected under different scopes tend to have differences in class balance, label availability, and subject demographics. Recent work has shown that importance sampling can be used to guide training selection. To date, these approaches have not considered imbalanced data sources with distinct labeling protocols. Approach: We propose a sampling policy, known as adaptive stochastic policy (ASP), inspired by reinforcement learning to adapt training based on subject, data source, and dynamic use criteria. We apply ASP in the context of multiorgan abdominal computed tomography segmentation. Training was performed with cross validation on 840 subjects from 10 data sources. External validation was performed with 20 subjects from 1 data source. Results: Four alternative strategies were evaluated with the state-of-the-art baseline as upper confident bound (UCB). ASP achieves average Dice of 0.8261 compared to 0.8135 UCB ( p < 0.01 , paired t -test) across fivefold cross validation. On withheld testing datasets, the proposed ASP achieved 0.8265 mean Dice versus 0.8077 UCB ( p < 0.01 , paired t -test). Conclusions: ASP provides a flexible reweighting technique for training deep learning models. We conclude that the proposed method adapts the sample importance, which leverages the performance on a challenging multisite, multiorgan, and multisize segmentation task.

9.
Tomography ; 6(2): 170-176, 2020 06.
Article in English | MEDLINE | ID: mdl-32548293

ABSTRACT

Positron emission tomography (PET) is typically performed in the supine position. However, breast magnetic resonance imaging (MRI) is performed in prone, as this improves visibility of deep breast tissues. With the emergence of hybrid scanners that integrate molecular information from PET and functional information from MRI, it is of great interest to determine if the prognostic utility of prone PET is equivalent to supine. We compared PERCIST (PET Response Criteria in Solid Tumors) measurements between prone and supine FDG-PET in patients with breast cancer and the effect of orientation on predicting pathologic complete response (pCR). In total, 47 patients were enrolled and received up to 6 cycles of neoadjuvant therapy. Prone and supine FDG-PET were performed at baseline (t0 ; n = 46), after cycle 1 (t1 ; n = 1) or 2 (t2 ; n = 10), or after all neoadjuvant therapy (t3 ; n = 19). FDG uptake was quantified by maximum and peak standardized uptake value (SUV) with and without normalization to lean body mass; that is, SUVmax , SUVpeak , SULmax , and SULpeak . PERCIST measurements were performed for each paired baseline and post-treatment scan. Receiver operating characteristic analysis for the prediction of pCR was performed using logistic regression that included age and tumor size as covariates. SUV and SUL metrics were significantly different between orientation (P < .001), but were highly correlated (P > .98). Importantly, no differences were observed with the PERCIST measurements (P > .6). Overlapping 95% confidence intervals for the receiver operating characteristic analysis suggested no difference at predicting pCR. Therefore, prone and supine PERCIST in this data set were not statistically different.


Subject(s)
Breast Neoplasms , Fluorodeoxyglucose F18 , Positron-Emission Tomography , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Female , Humans , Radiopharmaceuticals , Tomography, X-Ray Computed
10.
Article in English | MEDLINE | ID: mdl-34040277

ABSTRACT

Segmentation of abdominal computed tomography (CT) provides spatial context, morphological properties, and a framework for tissue-specific radiomics to guide quantitative Radiological assessment. A 2015 MICCAI challenge spurred substantial innovation in multi-organ abdominal CT segmentation with both traditional and deep learning methods. Recent innovations in deep methods have driven performance toward levels for which clinical translation is appealing. However, continued cross-validation on open datasets presents the risk of indirect knowledge contamination and could result in circular reasoning. Moreover, "real world" segmentations can be challenging due to the wide variability of abdomen physiology within patients. Herein, we perform two data retrievals to capture clinically acquired deidentified abdominal CT cohorts with respect to a recently published variation on 3D U-Net (baseline algorithm). First, we retrieved 2004 deidentified studies on 476 patients with diagnosis codes involving spleen abnormalities (cohort A). Second, we retrieved 4313 deidentified studies on 1754 patients without diagnosis codes involving spleen abnormalities (cohort B). We perform prospective evaluation of the existing algorithm on both cohorts, yielding 13% and 8% failure rate, respectively. Then, we identified 51 subjects in cohort A with segmentation failures and manually corrected the liver and gallbladder labels. We re-trained the model adding the manual labels, resulting in performance improvement of 9% and 6% failure rate for the A and B cohorts, respectively. In summary, the performance of the baseline on the prospective cohorts was similar to that on previously published datasets. Moreover, adding data from the first cohort substantively improved performance when evaluated on the second withheld validation cohort.

11.
Article in English | MEDLINE | ID: mdl-34040279

ABSTRACT

Human in-the-loop quality assurance (QA) is typically performed after medical image segmentation to ensure that the systems are performing as intended, as well as identifying and excluding outliers. By performing QA on large-scale, previously unlabeled testing data, categorical QA scores (e.g. "successful" versus "unsuccessful") can be generated. Unfortunately, the precious use of resources for human in-the-loop QA scores are not typically reused in medical image machine learning, especially to train a deep neural network for image segmentation. Herein, we perform a pilot study to investigate if the QA labels can be used as supplementary supervision to augment the training process in a semi-supervised fashion. In this paper, we propose a semi-supervised multi-organ segmentation deep neural network consisting of a traditional segmentation model generator and a QA involved discriminator. An existing 3-D abdominal segmentation network is employed, while the pre-trained ResNet-18 network is used as discriminator. A large-scale dataset of 2027 volumes are used to train the generator, whose 2-D montage images and segmentation mask with QA scores are used to train the discriminator. To generate the QA scores, the 2-D montage images were reviewed manually and coded 0 (success), 1 (errors consistent with published performance), and 2 (gross failure). Then, the ResNet-18 network was trained with 1623 montage images in equal distribution of all three code labels and achieved an accuracy 94% for classification predictions with 404 montage images withheld for the test cohort. To assess the performance of using the QA supervision, the discriminator was used as a loss function in a multi-organ segmentation pipeline. The inclusion of QA-loss function boosted performance on the unlabeled test dataset from 714 patients to 951 patients over the baseline model. Additionally, the number of failures decreased from 606 (29.90%) to 402 (19.83%). The contributions of the proposed method are three-fold: We show that (1) the QA scores can be used as a loss function to perform semi-supervised learning for unlabeled data, (2) the well trained discriminator is learnt by QA score rather than traditional "true/false", and (3) the performance of multi-organ segmentation on unlabeled datasets can be fine-tuned with more robust and higher accuracy than the original baseline method. The use of QA-inspired loss functions represents a promising area of future research and may permit tighter integration of supervised and semi-supervised learning.

12.
Article in English | MEDLINE | ID: mdl-34526733

ABSTRACT

Dynamic contrast enhanced computed tomography (CT) is an imaging technique that provides critical information on the relationship of vascular structure and dynamics in the context of underlying anatomy. A key challenge for image processing with contrast enhanced CT is that phase discrepancies are latent in different tissues due to contrast protocols, vascular dynamics, and metabolism variance. Previous studies with deep learning frameworks have been proposed for classifying contrast enhancement with networks inspired by computer vision. Here, we revisit the challenge in the context of whole abdomen contrast enhanced CTs. To capture and compensate for the complex contrast changes, we propose a novel discriminator in the form of a multi-domain disentangled representation learning network. The goal of this network is to learn an intermediate representation that separates contrast enhancement from anatomy and enables classification of images with varying contrast time. Briefly, our unpaired contrast disentangling GAN(CD-GAN) Discriminator follows the ResNet architecture to classify a CT scan from different enhancement phases. To evaluate the approach, we trained the enhancement phase classifier on 21060 slices from two clinical cohorts of 230 subjects. The scans were manually labeled with three independent enhancement phases (non-contrast, portal venous and delayed). Testing was performed on 9100 slices from 30 independent subjects who had been imaged with CT scans from all contrast phases. Performance was quantified in terms of the multi-class normalized confusion matrix. The proposed network significantly improved correspondence over baseline UNet, ResNet50 and StarGAN's performance of accuracy scores 0.54. 0.55, 0.62 and 0.91, respectively (p-value<0.0001 paired t-test for ResNet versus CD-GAN). The proposed discriminator from the disentangled network presents a promising technique that may allow deeper modeling of dynamic imaging against patient specific anatomies.

13.
Article in English | MEDLINE | ID: mdl-33907347

ABSTRACT

Abdominal multi-organ segmentation of computed tomography (CT) images has been the subject of extensive research interest. It presents a substantial challenge in medical image processing, as the shape and distribution of abdominal organs can vary greatly among the population and within an individual over time. While continuous integration of novel datasets into the training set provides potential for better segmentation performance, collection of data at scale is not only costly, but also impractical in some contexts. Moreover, it remains unclear what marginal value additional data have to offer. Herein, we propose a single-pass active learning method through human quality assurance (QA). We built on a pre-trained 3D U-Net model for abdominal multi-organ segmentation and augmented the dataset either with outlier data (e.g., exemplars for which the baseline algorithm failed) or inliers (e.g., exemplars for which the baseline algorithm worked). The new models were trained using the augmented datasets with 5-fold cross-validation (for outlier data) and withheld outlier samples (for inlier data). Manual labeling of outliers increased Dice scores with outliers by 0.130, compared to an increase of 0.067 with inliers (p<0.001, two-tailed paired t-test). By adding 5 to 37 inliers or outliers to training, we find that the marginal value of adding outliers is higher than that of adding inliers. In summary, improvement on single-organ performance was obtained without diminishing multi-organ performance or significantly increasing training time. Hence, identification and correction of baseline failure cases present an effective and efficient method of selecting training data to improve algorithm performance.

14.
Clin Cancer Res ; 26(9): 2111-2123, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31822498

ABSTRACT

PURPOSE: Preclinical data demonstrating androgen receptor (AR)-positive (AR+) triple-negative breast cancer (TNBC) cells are sensitive to AR antagonists, and PI3K inhibition catalyzed an investigator-initiated, multi-institutional phase Ib/II study TBCRC032. The trial investigated the safety and efficacy of the AR-antagonist enzalutamide alone or in combination with the PI3K inhibitor taselisib in patients with metastatic AR+ (≥10%) breast cancer. PATIENTS AND METHODS: Phase Ib patients [estrogen receptor positive (ER+) or TNBC] with AR+ breast cancer received 160 mg enzalutamide in combination with taselisib to determine dose-limiting toxicities and the maximum tolerated dose (MTD). Phase II TNBC patients were randomized to receive either enzalutamide alone or in combination with 4 mg taselisib until disease progression. Primary endpoint was clinical benefit rate (CBR) at 16 weeks. RESULTS: The combination was tolerated, and the MTD was not reached. The adverse events were hyperglycemia and skin rash. Overall, CBR for evaluable patients receiving the combination was 35.7%, and median progression-free survival (PFS) was 3.4 months. Luminal AR (LAR) TNBC subtype patients trended toward better response compared with non-LAR (75.0% vs. 12.5%, P = 0.06), and increased PFS (4.6 vs. 2.0 months, P = 0.082). Genomic analyses revealed subtype-specific treatment response, and novel FGFR2 fusions and AR splice variants. CONCLUSIONS: The combination of enzalutamide and taselisib increased CBR in TNBC patients with AR+ tumors. Correlative analyses suggest AR protein expression alone is insufficient for identifying patients with AR-dependent tumors and knowledge of tumor LAR subtype and AR splice variants may identify patients more or less likely to benefit from AR antagonists.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/drug therapy , Androgen Receptor Antagonists/administration & dosage , Androgen Receptor Antagonists/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Middle Aged , Neoplasm Metastasis , Nitriles , Oxazepines/administration & dosage , Oxazepines/adverse effects , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/adverse effects , Phenylthiohydantoin/analogs & derivatives , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Survival Rate , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
15.
J Med Imaging (Bellingham) ; 6(4): 044005, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31763353

ABSTRACT

Tissue window filtering has been widely used in deep learning for computed tomography (CT) image analyses to improve training performance (e.g., soft tissue windows for abdominal CT). However, the effectiveness of tissue window normalization is questionable since the generalizability of the trained model might be further harmed, especially when such models are applied to new cohorts with different CT reconstruction kernels, contrast mechanisms, dynamic variations in the acquisition, and physiological changes. We evaluate the effectiveness of both with and without using soft tissue window normalization on multisite CT cohorts. Moreover, we propose a stochastic tissue window normalization (SWN) method to improve the generalizability of tissue window normalization. Different from the random sampling, the SWN method centers the randomization around the soft tissue window to maintain the specificity for abdominal organs. To evaluate the performance of different strategies, 80 training and 453 validation and testing scans from six datasets are employed to perform multiorgan segmentation using standard 2D U-Net. The six datasets cover the scenarios, where the training and testing scans are from (1) same scanner and same population, (2) same CT contrast but different pathology, and (3) different CT contrast and pathology. The traditional soft tissue window and nonwindowed approaches achieved better performance on (1). The proposed SWN achieved general superior performance on (2) and (3) with statistical analyses, which offers better generalizability for a trained model.

16.
Cancer Immunol Res ; 7(11): 1755-1759, 2019 11.
Article in English | MEDLINE | ID: mdl-31462410

ABSTRACT

Pneumonitis may complicate anti-programmed death-1 (PD-1) therapy, although symptoms usually resolve with steroids. The long-term effects on respiratory function, however, are not well defined. We screened melanoma patients treated with anti-PD-1, with and without ipilimumab (anti-CTLA-4), and identified 31 patients with pneumonitis. Median time to radiographic findings was 4.8 months. Twenty-three patients (74%) presented with respiratory symptoms, whereas 8 (26%) were asymptomatic, and 11 (35%) were hospitalized. With 22.1 months median follow-up, 27 patients (87%) had resolution of symptoms, whereas 4 had persistent cough, dyspnea, and/or wheezing. By contrast, the rate of radiographic resolution was lower: Only 11 (35%) had complete radiographic resolution, whereas 14 (45%) had improvement of pneumonitis with persistent scarring or opacities, and 6 (19%) had persistent or worsened ground-glass opacities and/or nodular densities. Persistence (vs. resolution) of radiographic findings was associated with older age and initial need for steroids but not with need for hospitalization, timing of onset, or treatment regimen (combination vs. monotherapy). Among patients with serial pulmonary function tests, lung function improved with time. Although symptoms of anti-PD-1-induced pneumonitis resolved quickly, scarring or inflammation frequently persisted on computerized tomography. Therefore, further study of subclinical pulmonary effects of anti-PD-1 is needed.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Melanoma/therapy , Pneumonia/chemically induced , Pneumonia/diagnostic imaging , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , CTLA-4 Antigen/antagonists & inhibitors , Humans , Middle Aged , Pneumonia/pathology , Pneumonia/physiopathology , Risk Factors , Tomography, X-Ray Computed
17.
Tomography ; 5(1): 99-109, 2019 03.
Article in English | MEDLINE | ID: mdl-30854447

ABSTRACT

This multicenter study evaluated the effect of variations in arterial input function (AIF) determination on pharmacokinetic (PK) analysis of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data using the shutter-speed model (SSM). Data acquired from eleven prostate cancer patients were shared among nine centers. Each center used a site-specific method to measure the individual AIF from each data set and submitted the results to the managing center. These AIFs, their reference tissue-adjusted variants, and a literature population-averaged AIF, were used by the managing center to perform SSM PK analysis to estimate Ktrans (volume transfer rate constant), ve (extravascular, extracellular volume fraction), kep (efflux rate constant), and τi (mean intracellular water lifetime). All other variables, including the definition of the tumor region of interest and precontrast T1 values, were kept the same to evaluate parameter variations caused by variations in only the AIF. Considerable PK parameter variations were observed with within-subject coefficient of variation (wCV) values of 0.58, 0.27, 0.42, and 0.24 for Ktrans, ve, kep, and τi, respectively, using the unadjusted AIFs. Use of the reference tissue-adjusted AIFs reduced variations in Ktrans and ve (wCV = 0.50 and 0.10, respectively), but had smaller effects on kep and τi (wCV = 0.39 and 0.22, respectively). kep is less sensitive to AIF variation than Ktrans, suggesting it may be a more robust imaging biomarker of prostate microvasculature. With low sensitivity to AIF uncertainty, the SSM-unique τi parameter may have advantages over the conventional PK parameters in a longitudinal study.


Subject(s)
Prostatic Neoplasms/blood supply , Prostatic Neoplasms/diagnostic imaging , Algorithms , Arteries/diagnostic imaging , Contrast Media/pharmacokinetics , Humans , Image Interpretation, Computer-Assisted/methods , Information Dissemination , Magnetic Resonance Imaging/methods , Male , Models, Biological , Neovascularization, Pathologic/diagnostic imaging , Reproducibility of Results
18.
J Magn Reson Imaging ; 50(5): 1377-1392, 2019 11.
Article in English | MEDLINE | ID: mdl-30925001

ABSTRACT

The complexity of modern in vivo magnetic resonance imaging (MRI) methods in oncology has dramatically changed in the last 10 years. The field has long since moved passed its (unparalleled) ability to form images with exquisite soft-tissue contrast and morphology, allowing for the enhanced identification of primary tumors and metastatic disease. Currently, it is not uncommon to acquire images related to blood flow, cellularity, and macromolecular content in the clinical setting. The acquisition of images related to metabolism, hypoxia, pH, and tissue stiffness are also becoming common. All of these techniques have had some component of their invention, development, refinement, validation, and initial applications in the preclinical setting using in vivo animal models of cancer. In this review, we discuss the genesis of quantitative MRI methods that have been successfully translated from preclinical research and developed into clinical applications. These include methods that interrogate perfusion, diffusion, pH, hypoxia, macromolecular content, and tissue mechanical properties for improving detection, staging, and response monitoring of cancer. For each of these techniques, we summarize the 1) underlying biological mechanism(s); 2) preclinical applications; 3) available repeatability and reproducibility data; 4) clinical applications; and 5) limitations of the technique. We conclude with a discussion of lessons learned from translating MRI methods from the preclinical to clinical setting, and a presentation of four fundamental problems in cancer imaging that, if solved, would result in a profound improvement in the lives of oncology patients. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;50:1377-1392.


Subject(s)
Magnetic Resonance Imaging/methods , Medical Oncology/trends , Neoplasms/diagnostic imaging , Animals , Brain Neoplasms/diagnostic imaging , Diffusion Magnetic Resonance Imaging/methods , Humans , Hydrogen-Ion Concentration , Hypoxia , Image Processing, Computer-Assisted , Immunotherapy , Macromolecular Substances , Neoplasm Metastasis , Neoplasm Transplantation , Oxygen/metabolism , Reproducibility of Results , Theranostic Nanomedicine , Translational Research, Biomedical/trends
19.
Comput Biol Med ; 107: 109-117, 2019 04.
Article in English | MEDLINE | ID: mdl-30798219

ABSTRACT

Delineation of Computed Tomography (CT) abdominal anatomical structure, specifically spleen segmentation, is useful for not only measuring tissue volume and biomarkers but also for monitoring interventions. Recently, segmentation algorithms using deep learning have been widely used to reduce time humans spend to label CT data. However, the computerized segmentation has two major difficulties: managing intermediate results (e.g., resampled scans, 2D sliced image for deep learning), and setting up the system environments and packages for autonomous execution. To overcome these issues, we propose an automated pipeline for the abdominal spleen segmentation. This pipeline provides an end-to-end synthesized process that allows users to avoid installing any packages and to deal with the intermediate results locally. The pipeline has three major stages: pre-processing of input data, segmentation of spleen using deep learning, 3D reconstruction with the generated labels by matching the segmentation results with the original image dimensions, which can then be used later and for display or demonstration. Given the same volume scan, the approach described here takes about 50 s on average whereas the manual segmentation takes about 30 min on the average. Even if it includes all subsidiary processes such as preprocessing and necessary setups, the whole pipeline process requires on the average 20 min from beginning to end.


Subject(s)
Deep Learning , Imaging, Three-Dimensional/methods , Spleen/diagnostic imaging , Tomography, X-Ray Computed/methods , Algorithms , Humans , Time Factors
20.
NPJ Precis Oncol ; 3: 5, 2019.
Article in English | MEDLINE | ID: mdl-30793038

ABSTRACT

Proliferating tricholemmal tumors (PTTs) are rare benign neoplasms that arise from the outer sheath of a hair follicle. Occasionally, these PTTs undergo malignant transformation to become malignant proliferating tricholemmal tumors (MPTTs). Little is known about the molecular alterations, malignant progression, and management of MPTTs. Here, we describe the case of a 58-year-old female that had a widely metastatic MPTT that harbored an activating PIK3CA mutation and was sensitive to the PI3K inhibitor, alpelisib (BYL719). We review the available literature on metastatic MPTT, detail the patient's course, and present a whole genome analysis of this rare tumor.

SELECTION OF CITATIONS
SEARCH DETAIL
...