- Browse by Author
Browsing by Author "Su, Jing"
Now showing 1 - 10 of 32
Results Per Page
Sort Options
Item Aberrant cholesterol metabolism in colorectal cancer represents a targetable vulnerability(Elsevier, 2023-07) Xie, Jingwu; Nguyen, Chi Mai; Turk, Anita; Nan, Hongmei; Imperiale, Thomas F.; House, Michael; Huang, Kun; Su, Jing; Biostatistics, School of Public HealthItem ARDaC Common Data Model Facilitates Data Dissemination and Enables Data Commons for Modern Clinical Studies(IOS Press, 2024) Jin, Nanxin; Li, Zuotian; Kettler, Carla; Yang, Baijian; Tu, Wanzhu; Su, Jing; Biostatistics and Health Data Science, Richard M. Fairbanks School of Public HealthModern clinical studies collect longitudinal and multimodal data about participants, treatments and responses, biospecimens, and molecular and multiomics data. Such rich and complex data requires new common data models (CDM) to support data dissemination and research collaboration. We have developed the ARDaC CDM for the Alcoholic Hepatitis Network (AlcHepNet) Research Data Commons (ARDaC) to support clinical studies and translational research in the national AlcHepNet consortium. The ARDaC CDM bridges the gap between the data models used by the AlcHepNet electronic data capture platform (REDCap) and the Genomic Data Commons (GDC) data model used by the Gen3 data commons framework. It extends the GDC data model for clinical studies; facilitates the harmonization of research data across consortia and programs; and supports the development of the ARDaC. ARDaC CDM is designed as a general and extensible CDM for addressing the needs of modern clinical studies. The ARDaC CDM is available at https://dev.ardac.org/DD.Item Bulk and Single-Cell Profiling of Breast Tumors Identifies TREM-1 as a Dominant Immune Suppressive Marker Associated With Poor Outcomes(Frontiers Media, 2021-12-08) Pullikuth, Ashok K.; Routh, Eric D.; Zimmerman, Kip D.; Chifman, Julia; Chou, Jeff W.; Soike, Michael H.; Jin, Guangxu; Su, Jing; Song, Qianqian; Black, Michael A.; Print, Cristin; Bedognetti, Davide; Howard-McNatt, Marissa; O’Neill, Stacey S.; Thomas, Alexandra; Langefeld, Carl D.; Sigalov, Alexander B.; Lu, Yong; Miller, Lance D.; Biostatistics and Health Data Science, School of MedicineBackground: Triggering receptor expressed on myeloid cells (TREM)-1 is a key mediator of innate immunity previously associated with the severity of inflammatory disorders, and more recently, the inferior survival of lung and liver cancer patients. Here, we investigated the prognostic impact and immunological correlates of TREM1 expression in breast tumors. Methods: Breast tumor microarray and RNAseq expression profiles (n=4,364 tumors) were analyzed for associations between gene expression, tumor immune subtypes, distant metastasis-free survival (DMFS) and clinical response to neoadjuvant chemotherapy (NAC). Single-cell (sc)RNAseq was performed using the 10X Genomics platform. Statistical associations were assessed by logistic regression, Cox regression, Kaplan-Meier analysis, Spearman correlation, Student's t-test and Chi-square test. Results: In pre-treatment biopsies, TREM1 and known TREM-1 inducible cytokines (IL1B, IL8) were discovered by a statistical ranking procedure as top genes for which high expression was associated with reduced response to NAC, but only in the context of immunologically "hot" tumors otherwise associated with a high NAC response rate. In surgical specimens, TREM1 expression varied among tumor molecular subtypes, with highest expression in the more aggressive subtypes (Basal-like, HER2-E). High TREM1 significantly and reproducibly associated with inferior distant metastasis-free survival (DMFS), independent of conventional prognostic markers. Notably, the association between high TREM1 and inferior DMFS was most prominent in the subset of immunogenic tumors that exhibited the immunologically hot phenotype and otherwise associated with superior DMFS. Further observations from bulk and single-cell RNAseq analyses indicated that TREM1 expression was significantly enriched in polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and M2-like macrophages, and correlated with downstream transcriptional targets of TREM-1 (IL8, IL-1B, IL6, MCP-1, SPP1, IL1RN, INHBA) which have been previously associated with pro-tumorigenic and immunosuppressive functions. Conclusions: Together, these findings indicate that increased TREM1 expression is prognostic of inferior breast cancer outcomes and may contribute to myeloid-mediated breast cancer progression and immune suppression.Item c-Met Mediated Cytokine Network Promotes Brain Metastasis of Breast Cancer by Remodeling Neutrophil Activities(MDPI, 2023-05-05) Liu, Yin; Smith, Margaret R.; Wang, Yuezhu; D’Agostino, Ralph, Jr.; Ruiz, Jimmy; Lycan, Thomas; Kucera, Gregory L.; Miller, Lance D.; Li, Wencheng; Chan, Michael D.; Farris, Michael; Su, Jing; Song, Qianqian; Zhao, Dawen; Chandrasekaran, Arvind; Xing, Fei; Biostatistics and Health Data Science, School of MedicineThe brain is one of the most common metastatic sites among breast cancer patients, especially in those who have Her2-positive or triple-negative tumors. The brain microenvironment has been considered immune privileged, and the exact mechanisms of how immune cells in the brain microenvironment contribute to brain metastasis remain elusive. In this study, we found that neutrophils are recruited and influenced by c-Met high brain metastatic cells in the metastatic sites, and depletion of neutrophils significantly suppressed brain metastasis in animal models. Overexpression of c-Met in tumor cells enhances the secretion of a group of cytokines, including CXCL1/2, G-CSF, and GM-CSF, which play critical roles in neutrophil attraction, granulopoiesis, and homeostasis. Meanwhile, our transcriptomic analysis demonstrated that conditioned media from c-Met high cells significantly induced the secretion of lipocalin 2 (LCN2) from neutrophils, which in turn promotes the self-renewal of cancer stem cells. Our study unveiled the molecular and pathogenic mechanisms of how crosstalk between innate immune cells and tumor cells facilitates tumor progression in the brain, which provides novel therapeutic targets for treating brain metastasis.Item Comprehensive and Computable Molecular Diagnostic Panel (C2Dx) From Small Volume Specimens for Precision Oncology: Molecular Subtyping of Non-Small Cell Lung Cancer From Fine Needle Aspirates(Frontiers Media, 2021-04-16) Su, Jing; Huang, Lynn S.; Barnard, Ryan; Parks, Graham; Cappellari, James; Bellinger, Christina; Dotson, Travis; Craddock, Lou; Prakash, Bharat; Hovda, Jonathan; Clark, Hollins; Petty, William Jeffrey; Pasche, Boris; Chan, Michael D.; Miller, Lance D.; Ruiz, Jimmy; Biostatistics, School of Public HealthThe Comprehensive, Computable NanoString Diagnostic gene panel (C2Dx) is a promising solution to address the need for a molecular pathological research and diagnostic tool for precision oncology utilizing small volume tumor specimens. We translate subtyping-related gene expression patterns of Non-Small Cell Lung Cancer (NSCLC) derived from public transcriptomic data which establish a highly robust and accurate subtyping system. The C2Dx demonstrates supreme performance on the NanoString platform using microgram-level FNA samples and has excellent portability to frozen tissues and RNA-Seq transcriptomic data. This workflow shows great potential for research and the clinical practice of cancer molecular diagnosis.Item DeePaN: deep patient graph convolutional network integrating clinico-genomic evidence to stratify lung cancers for immunotherapy(Nature, 2021) Fang, Chao; Xu, Dong; Su, Jing; Dry, Jonathan R.; Linghu, Bolan; Biostatistics, School of Public HealthImmuno-oncology (IO) therapies have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC). However, patient responses to IO are variable and influenced by a heterogeneous combination of health, immune, and tumor factors. There is a pressing need to discover the distinct NSCLC subgroups that influence response. We have developed a deep patient graph convolutional network, we call “DeePaN”, to discover NSCLC complexity across data modalities impacting IO benefit. DeePaN employs high-dimensional data derived from both real-world evidence (RWE)-based electronic health records (EHRs) and genomics across 1937 IO-treated NSCLC patients. DeePaN demonstrated effectiveness to stratify patients into subgroups with significantly different (P-value of 2.2 × 10−11) overall median survival of 20.35 months and 9.42 months post-IO therapy. Significant differences in IO outcome were not seen from multiple non-graph-based unsupervised methods. Furthermore, we demonstrate that patient stratification from DeePaN has the potential to augment the emerging IO biomarker of tumor mutation burden (TMB). Characterization of the subgroups discovered by DeePaN indicates potential to inform IO therapeutic insight, including the enrichment of mutated KRAS and high blood monocyte count in the IO beneficial and IO non-beneficial subgroups, respectively. Our work has proven the concept that graph-based AI is feasible and can effectively integrate high-dimensional genomic and EHR data to meaningfully stratify cancer patients on distinct clinical outcomes, with potential to inform precision oncology.Item DEPOT: graph learning delineates the roles of cancers in the progression trajectories of chronic kidney disease using electronic medical records(medRxiv, 2023-08-16) Song, Qianqian; Liu, Xiang; Li, Zuotian; Zhang, Pengyue; Eadon, Michael; Su, Jing; Biostatistics and Health Data Science, School of MedicineChronic kidney disease (CKD) is a common, complex, and heterogeneous disease impacting aging populations. Determining the landscape of disease progression trajectories from midlife to senior age in a real-world context allows us to better understand the progression of CKD, the heterogeneity of progression patterns among the risk population, and the interactions with other clinical conditions like cancers. In this study, we use electronic health records (EHRs) to outline the CKD progression trajectory roadmap for the Wake Forest Baptist Medical Center (WFBMC) patient population. We establish an EHR cohort (n = 79,434) with patients' health status identified by 18 Essential Clinical Indices across 508,732 clinical encounters. We develop the DisEase PrOgression Trajectory (DEPOT) approach to model CKD progression trajectories and individualize clinical decision support. The DEPOT is an evidence-driven, graph-based clinical informatics approach that addresses the unique challenges in longitudinal EHR data by systematically using the graph artificial intelligence (graph-AI) model for representation learning and reverse graph embedding for trajectory reconstruction. Moreover, DEPOT includes a prediction model to assign new patients along the progression trajectory. We successfully establish the EHR-based CKD progression trajectories with DEPOT in the WFUBMC cohort. We annotate the trajectories with clinical features, including kidney function, age, and other indices, including cancer. This CKD progression trajectory roadmap reveals diverse kidney failure pathways associated with different clinical conditions. Specifically, we have identified one high-risk trajectory and two low-risk trajectories. Switching pathways from low-risk trajectories to the high-risk one is associated with accelerated decline in kidney function. On this roadmap, high-risk patients are enriched in the skin and GU cancers, which differs from low-risk patients, suggesting fundamentally different disease progression mechanisms. Overall, the CKD progression trajectory roadmap reveals novel diverse renal failure pathways in type 2 diabetes mellitus and highlights disease progression patterns associated with cancer phenotypes.Item DSTG: deconvoluting spatial transcriptomics data through graph-based artificial intelligence(Oxford University Press, 2021-09-02) Song, Qianqian; Su, Jing; Biostatistics, School of Public HealthRecent development of spatial transcriptomics (ST) is capable of associating spatial information at different spots in the tissue section with RNA abundance of cells within each spot, which is particularly important to understand tissue cytoarchitectures and functions. However, for such ST data, since a spot is usually larger than an individual cell, gene expressions measured at each spot are from a mixture of cells with heterogenous cell types. Therefore, ST data at each spot needs to be disentangled so as to reveal the cell compositions at that spatial spot. In this study, we propose a novel method, named deconvoluting spatial transcriptomics data through graph-based convolutional networks (DSTG), to accurately deconvolute the observed gene expressions at each spot and recover its cell constitutions, thus achieving high-level segmentation and revealing spatial architecture of cellular heterogeneity within tissues. DSTG not only demonstrates superior performance on synthetic spatial data generated from different protocols, but also effectively identifies spatial compositions of cells in mouse cortex layer, hippocampus slice and pancreatic tumor tissues. In conclusion, DSTG accurately uncovers the cell states and subpopulations based on spatial localization. DSTG is available as a ready-to-use open source software (https://github.com/Su-informatics-lab/DSTG) for precise interrogation of spatial organizations and functions in tissues.Item Feature Selection for Unsupervised Machine Learning(IEEE, 2023) Huang, Huyunting; Tang, Ziyang; Zhang, Tonglin; Yang, Baijian; Song, Qianqian; Su, Jing; Biostatistics and Health Data Science, Richard M. Fairbanks School of Public HealthCompared to supervised machine learning (ML), the development of feature selection for unsupervised ML is far behind. To address this issue, the current research proposes a stepwise feature selection approach for clustering methods with a specification to the Gaussian mixture model (GMM) and the k-means. Rather than the existing GMM and k-means which are carried out based on all the features, the proposed method selects a subset of features to implement the two methods, respectively. The research finds that a better result can be obtained if the existing GMM and k-means methods are modified by nice initializations. Experiments based on Monte Carlo simulations show that the proposed method is more computationally efficient and the result is more accurate than the existing GMM and k-means methods based on all the features. The experiment based on a real-world dataset confirms this finding.Item Genomic signature for oligometastatic disease in non-small cell lung cancer patients with brain metastases(Frontiers Media, 2024-09-17) Choi, Ariel R.; D’Agostino, Ralph B., Jr.; Farris, Michael K.; Abdulhaleem, Mohammed; Hunting, John C.; Wang, Yuezhu; Smith, Margaret R.; Ruiz, Jimmy; Lycan, Thomas W.; Petty, W. Jeffrey; Cramer, Christina K.; Tatter, Stephen B.; Laxton, Adrian W.; White, Jaclyn J.; Li, Wencheng; Su, Jing; Whitlow, Christopher; Xing, Fei; Chan, Michael D.; Biostatistics and Health Data Science, Richard M. Fairbanks School of Public HealthPurpose/objectives: Biomarkers for extracranial oligometastatic disease remain elusive and few studies have attempted to correlate genomic data to the presence of true oligometastatic disease. Methods: Patients with non-small cell lung cancer (NSCLC) and brain metastases were identified in our departmental database. Electronic medical records were used to identify patients for whom liquid biopsy-based comprehensive genomic profiling (Guardant Health) was available. Extracranial oligometastatic disease was defined as patients having ≤5 non-brain metastases without diffuse involvement of a single organ. Widespread disease was any spread beyond oligometastatic. Fisher's exact tests were used to screen for mutations statistically associated (p<0.1) with either oligometastatic or widespread extracranial disease. A risk score for the likelihood of oligometastatic disease was generated and correlated to the likelihood of having oligometastatic disease vs widespread disease. For oligometastatic patients, a competing risk analysis was done to assess for cumulative incidence of oligometastatic progression. Cox regression was used to determine association between oligometastatic risk score and oligoprogression. Results: 130 patients met study criteria and were included in the analysis. 51 patients (39%) had extracranial oligometastatic disease. Genetic mutations included in the Guardant panel that were associated (p<0.1) with the presence of oligometastatic disease included ATM, JAK2, MAP2K2, and NTRK1, while ARID1A and CCNE1 were associated with widespread disease. Patients with a positive, neutral and negative risk score for oligometastatic disease had a 78%, 41% and 11.5% likelihood of having oligometastatic disease, respectively (p<0.0001). Overall survival for patients with positive, neutral and negative risk scores for oligometastatic disease was 86% vs 82% vs 64% at 6 months (p=0.2). Oligometastatic risk score was significantly associated with the likelihood of oligoprogression based on the Wald chi-square test. Patients with positive, neutral and negative risk scores for oligometastatic disease had a cumulative incidence of oligometastatic progression of 77% vs 35% vs 33% at 6 months (p=0.03). Conclusions: Elucidation of a genomic signature for extracranial oligometastatic disease derived from non-invasive liquid biopsy appears feasible for NSCLC patients. Patients with this signature exhibited higher rates of early oligoprogression. External validation could lead to a biomarker that has the potential to direct local therapies in oligometastatic patients.