Papillary thyroid carcinoma (PTC) has the highest incidence rate of all thyroid cancers for the last several decades. Although this particular disease tends to be fairly indolent overall, classical and tall cell variants exhibit more aggressive behavior due to a higher progression and mortality rate. However among classical PTCs, there is a need for improvement of clinical management of patients who are at higher risk of progression/death and who will potentially benefit from more aggressive management, and similarly for lower risk patients who are unnecessarily overtreated. In this study, we aimed to evaluate the prognostic role of nuclear features for disease-specific and disease-free survival within classical and follicular histological subtypes of PTC. A set of features describing the nuclear shape, architecture, and texture of both tumor and lymphocyte cells were used to train a Lasso-Cox regression model with 199 patients. Coefficients of the model were then used to assign a quantitative risk score (QuRiS) per patient. Patients were further subdivided into high and low risk of recurrence to compare survival probabilities within a 15 year study period using Kaplan-Meier estimates (C-index of 0.786 (p = 0.009 on log-rank test). Survival probabilities of both risk groups were computed with a hazard ratio 1.54 and compared using the log-rank test (p = 0.004). The trained model was then validated on the remaining 149 patients, consisting of 3 independent sets of classical (N = 107), follicular (N = 74), and tall cell (N = 16) variants of PTC, with HR=8.83, 4.21, and 0.583, respectively. We then identified specific genes that were differentially expressed between the risk groups identified by the pathomic model. Using gene set enrichment and mutation analysis, we discovered that the signaling pathways TCA cycle and amino acid metabolism are associated with poorer outcome, while mutation in the BRAF oncogene is significantly associated with better survival in cPTC patients. We also discovered that the DTX4 gene was prognostic for disease-specific survival (DSS) and further stratified the risk of disease-related death of patients with high and low expression of DTX4 using the image-based nuclear morphology features.
Stage III Colorectal cancer (CRC) is treated with surgery followed by chemotherapy. Yet >20% of clinically low-risk patients develop recurrence. It is critical to identify high-risk stage III patients who can benefit from closer monitoring and escalation of therapy. Previous studies showed promising results in predicting risk from H&E slides in CRC using deep learning based algorithms. One biomarker which was heavily studied and showed good results in predicting risk is Tumor Infiltrating Lymphocytes (TILs). In CRC, TIL density has been shown to be significantly and independently associated with overall patient survival1. Furthermore, additional studies have demonstrated that analyzing spatial organization of TILs could be more informative than density alone2. Hence, our study aimed to stratify stage III CRC patients into distinct risk groups based on features derived from TILs and to determine if this classification could have independent prognostic significance. The training set (D1) included 50 patients and validation set (D2) consisted of 70 patients from an independent site. A survival model was trained to predict the risk of recurrence in stage III CRC patients. First, a deep learning (DL) model was used to segment TILs on WSIs. Next, 1036 features related to spatial architecture (SpaTIL) and density of TILs (DenTIL) were extracted. A Cox proportional hazards regression model in conjunction with the least absolute shrinkage and selection operator (Lasso) was used to find top 5 features and the feature coefficients associated with progression free survival (PFS) and provide risk scores to each patient. The risk scores for the training dataset were computed using the selected features with their respective coefficients. A cut-off value was determined according to these risk scores, above which patients were labelled high-risk and below was low risk. In the validation set, the median PFS for the high-risk group was 15.1mos and in the low-risk group was 27mos. The model was able to accurately predict higher incidence of progression in patients in the high-risk group (HR = 3.76, 95% CI 1.3-10.9, p-value=0.0053, c-index=0.687) in the validation set. Future work will entail additional multi-site, multi-institutional validation of our biomarker to further understand its strengths and applications.
Purpose: We used computerized image analysis and machine learning approaches to characterize spatial arrangement features of the immune response from digitized autopsied H&E tissue images of the lung in coronavirus disease 2019 (COVID-19) patients. Additionally, we applied our approach to tease out potential morphometric differences from autopsies of patients who succumbed to COVID-19 versus H1N1.
Approach: H&E lung whole slide images from autopsy specimens of nine COVID-19 and two H1N1 patients were computationally interrogated. 606 image patches (∼55 per patient) of 1024 × 882 pixels were extracted from the 11 autopsied patient studies. A watershed-based segmentation approach in conjunction with a machine learning classifier was employed to identify two types of
nuclei families: lymphocytes and non-lymphocytes (i.e., other nucleated cells such as pneumocytes, macrophages, and neutrophils). Based off the proximity of the individual nuclei, clusters for each nuclei family were constructed. For each of the resulting clusters, a series of quantitative measurements relating to architecture and density of nuclei clusters were calculated. A receiver operating characteristics-based feature selection method, violin plots, and the t-distributed stochastic neighbor embedding algorithm were employed to study differences in immune patterns.
Results: In COVID-19, the immune response consistently showed multiple small-size lymphocyte clusters, suggesting that lymphocyte response is rather modest, possibly due to lymphocytopenia. In H1N1, we found larger lymphocyte clusters that were proximal to large clusters of non-lymphocytes, a possible reflection of increased prevalence of macrophages and other immune cells.
Conclusion: Our study shows the potential of computational pathology to uncover immune response features that may not be obvious by routine histopathology visual inspection.
The presence of tumor-infiltrating lymphocytes (TILs) is correlated with outcome and prognosis in epithelial ovarian cancer (EOC). In this study, automated image analysis was used to analyze the association between overall survival (OS) and TIL spatial arrangement and density in a multi-site cohort of 102 EOC patients who received adjuvant chemotherapy following debulking surgery. Features of the spatial arrangement of TILs (SpaTIL) were used to quantify the spatial co-localization of TILs and tumor cells on digitized pathology slides of the malignant neoplasm of excised specimens. A multivariable Cox regression model of SpaTIL features was fit on the n1 = 51 patient training set and was evaluated in the n2 = 51 patient validation set. The SpaTIL signature was significantly associated with OS, both in the training set (hazard ratio (HR) = 2.81, 95% confidence interval (CI) = 1.33 − 5.92, and p = 0.003) and the validation set (HR = 2.06, 95% CI = 1.04 − 4.07, and p = 0.008). In addition, fusing our spaTIL risk score and the clinical staging further improved the results of the predictive model (HR = 4.045, 95% CI = 4.11−5.41, and p = 0.0002 in the validation set) and outperformed clinical staging alone. This finding illustrates that a spaTIL risk score is not only able to predict OS independent of clinical data, but also offers prognostic value complementary to current clinical standard-of-care. Patients with longer survival times had significantly higher heterogeneity of non-TIL cluster area, while shorter time survivors had mostly same-sized, evenly-distributed non-TIL clusters and smaller average TIL cluster area. These findings suggest that dispersion of TILs throughout the tumor is associated with better treatment response to post-treatment adjuvant chemotherapy, and therefore longer survival time.
Recently immune-checkpoint inhibitors have demonstrated promising clinical efficacy in patients with advanced non-small cell lung cancer (NSCLC). However, the response rates to immune checkpoint blockade drugs remain modest (45% in the front line setting and 20% in the second line setting). Consequently, there is an unmet need to develop accurate, validated biomarkers to predict which NSCLC patients will benefit from immunotherapy. While there has been recent interest in evaluating the role of texture and shape patterns of the nodule on CT scans to predict response to checkpoint inhibitors for NSCLC, our group has shown that nodule vessel morphology might also play a role in determining tumor aggressiveness and behavior. In this work we present a new approach using quantitative vessel tortuosity (QVT) radiomics, to predict response to checkpoint inhibitors and overall survival for patients with NSCLC treated with Nivolumab (a PD1 inhibitor) on a retrospective data set of 111 patients (D1) including 56 responders and 45 non-responders. Patients who did not receive Nivolumab after 2 cycles due to a lack of response or progression as per Response Evaluation Criteria in Solid Tumors (RECIST) were classified as non-responders, patients who had radiological response or stable disease as per RECIST were classified as responders. On D1, in conjunction with a linear discriminant analysis (LDA) classifier the QVT features were able to predict response to immunotherapy with an AUC of 0.73_0.04. Kaplan Meier analysis showed significant difference of overall survival between patients with low risk and high risk defined by the radiomics classifier (p-value = 0.004, HR= 2.29, 95% CI= 1.35 - 3.87).
Immune checkpoint inhibitors targeting the programmed cell death (PD)1/ L1 axis have been approved for treatment of chemotherapy refractory advanced non-small cell lung cancer (NSCLC) for a few years. While higher PD-L1 expression is associated with better outcomes after monotherapy with immune checkpoint inhibitors, it is not a perfect predictive biomarker for clinical benefit from immunotherapy, because some patients with low PD-L1 expression have sustained responses. In clinical practice, using radiological tools like Response Evaluation Criteria in Solid Tumors (RECIST), tends to underestimate the benefit of therapy. For instance, some patients treated with immunotherapy suffer from pseudoprogression while actually having a favorable response, RECIST in this setting is inadequate to capture the response. In this study we sought to explore whether radiomic texture features extracted from both inside and outside of the tumor from baseline CT scans were associated with overall patient survival (OS) in 139 NSCLC patients being treated with IO from two separate sites. Patients were divided into a discovery (D1 = 50; nivolumab from Cleveland Clinic) and two validation sets (D2 = 62 from Cleveland Clinic, D3 = 27 from University of Pennsylvania Health System. Patients in the validation sets had been treated with different types of checkpoint inhibitor drugs including nivolumab, pembrolizumab, and atezolizumab. 454 radiomic texture features from within (intra-tumoral) and outside the tumor (peri-tumoral) were extracted from baseline contrast CT images. Following feature selection on the discovery set, a radiomic risk-score signature was generated by using least absolute shrinkage and selection operator. Using a Cox regression model, the association of the radiomic signature with overall survival (OS) was evaluated in the discovery and two validation sets. In addition, 95% confidence intervals (CI) and relative hazard ratios (HR) were calculated. Our results revealed that the radiomics signature was significantly associated with OS, both in the discovery set (HR = 5.06, 95%CI = 3, 8.55; p-value < 0.0001) and the two validation data sets (D2: HR = 5.88, 95% CI = 2.19, 21.63, p-value = 0.0009; D3: HR = 5.37, 95% CI = 1.74, 16.57, p-value = 0.0034). Our initial results appear to suggest that our radiomic signature could serve as a non-invasive way of predicting and monitoring response to checkpoint inhibitors for patients with non-small cell lung cancer.
Access to the requested content is limited to institutions that have purchased or subscribe to SPIE eBooks.
You are receiving this notice because your organization may not have SPIE eBooks access.*
*Shibboleth/Open Athens users─please
sign in
to access your institution's subscriptions.
To obtain this item, you may purchase the complete book in print or electronic format on
SPIE.org.
INSTITUTIONAL Select your institution to access the SPIE Digital Library.
PERSONAL Sign in with your SPIE account to access your personal subscriptions or to use specific features such as save to my library, sign up for alerts, save searches, etc.