1
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
2
|
Arriojas A, Baek LM, Berner MJ, Zhurkevich A, Hinton AO, Meyer MD, Dobrolecki LE, Lewis MT, Zarringhalam K, Echeverria GV. Artificial intelligence-enabled automated analysis of transmission electron micrographs to evaluate chemotherapy impact on mitochondrial morphology in triple negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.635300. [PMID: 40027627 PMCID: PMC11870520 DOI: 10.1101/2025.02.19.635300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Advancements in transmission electron microscopy (TEM) have enabled in-depth studies of biological specimens, offering new avenues to large-scale imaging experiments with subcellular resolution. Mitochondrial structure is of growing interest in cancer biology due to its crucial role in regulating the multi-faceted functions of mitochondria. We and others have established the crucial role of mitochondria in triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer with limited therapeutic options. Building upon our previous work demonstrating the regulatory role of mitochondrial structure dynamics in metabolic adaptation and survival of chemotherapy-refractory TNBC cells, we sought to extend those findings to a large-scale analysis of transmission electron micrographs. Here we present a UNet artificial intelligence (AI) model for automatic annotation and assessment of mitochondrial morphology and feature quantification. Our model is trained on 11,039 manually annotated mitochondria across 125 micrographs derived from a variety of orthotopic patient-derived xenograft (PDX) mouse model tumors and adherent cell cultures. The model achieves an F1 score of 0.85 on test micrographs at the pixel level. To validate the ability of our model to detect expected mitochondrial structural features, we utilized micrographs from mouse primary skeletal muscle cells genetically modified to lack Dynamin-related protein 1 (Drp1). The algorithm successfully detected a significant increase in mitochondrial elongation, in alignment with the well-established role of Drp1 as a driver of mitochondrial fission. Further, we subjected in vitro and in vivo TNBC models to conventional chemotherapy treatments commonly used for clinical management of TNBC, including doxorubicin, carboplatin, paclitaxel, and docetaxel (DTX). We found substantial within-sample heterogeneity of mitochondrial structure in both in vitro and in vivo TNBC models and observed a consistent reduction in mitochondrial elongation in DTX-treated specimens. We went on to compare mammary tumors and matched lung metastases in a highly metastatic PDX model of TNBC, uncovering significant reduction in mitochondrial length in metastatic lesions. Our large, curated dataset provides high statistical power to detect frequent chemotherapy-induced shifts in mitochondrial shapes and sizes in residual cells left behind after treatment. The successful application of our AI model to capture mitochondrial structure marks a step forward in high-throughput analysis of mitochondrial structures, enhancing our understanding of how morphological changes may relate to chemotherapy efficacy and mechanism of action. Finally, our large, manually curated electron micrograph dataset - now publicly available - serves as a unique gold-standard resource for developing, benchmarking, and applying computational models, while further advancing investigations into mitochondrial morphology and its impact on cancer biology.
Collapse
|
3
|
Wang Q, Zhao F, Zhang H, Chu T, Wang Q, Pan X, Chen Y, Zhou H, Zheng T, Li Z, Lin F, Xie H, Ma H, Liu L, Zhang L, Li Q, Wang W, Dai Y, Tang R, Wang J, Yang P, Mao N. Deep learning-based multi-task prediction of response to neoadjuvant chemotherapy using multiscale whole slide images in breast cancer: A multicenter study. Chin J Cancer Res 2025; 37:28-47. [PMID: 40078559 PMCID: PMC11893347 DOI: 10.21147/j.issn.1000-9604.2025.01.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Early predicting response before neoadjuvant chemotherapy (NAC) is crucial for personalized treatment plans for locally advanced breast cancer patients. We aim to develop a multi-task model using multiscale whole slide images (WSIs) features to predict the response to breast cancer NAC more finely. Methods This work collected 1,670 whole slide images for training and validation sets, internal testing sets, external testing sets, and prospective testing sets of the weakly-supervised deep learning-based multi-task model (DLMM) in predicting treatment response and pCR to NAC. Our approach models two-by-two feature interactions across scales by employing concatenate fusion of single-scale feature representations, and controls the expressiveness of each representation via a gating-based attention mechanism. Results In the retrospective analysis, DLMM exhibited excellent predictive performance for the prediction of treatment response, with area under the receiver operating characteristic curves (AUCs) of 0.869 [95% confidence interval (95% CI): 0.806-0.933] in the internal testing set and 0.841 (95% CI: 0.814-0.867) in the external testing sets. For the pCR prediction task, DLMM reached AUCs of 0.865 (95% CI: 0.763-0.964) in the internal testing and 0.821 (95% CI: 0.763-0.878) in the pooled external testing set. In the prospective testing study, DLMM also demonstrated favorable predictive performance, with AUCs of 0.829 (95% CI: 0.754-0.903) and 0.821 (95% CI: 0.692-0.949) in treatment response and pCR prediction, respectively. DLMM significantly outperformed the baseline models in all testing sets (P<0.05). Heatmaps were employed to interpret the decision-making basis of the model. Furthermore, it was discovered that high DLMM scores were associated with immune-related pathways and cells in the microenvironment during biological basis exploration. Conclusions The DLMM represents a valuable tool that aids clinicians in selecting personalized treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Qin Wang
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Feng Zhao
- School of Computer Science and Technology, Shandong Technology and Business University, Yantai 264005, China
| | - Haicheng Zhang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tongpeng Chu
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Qi Wang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin 541004, China
| | - Yuqian Chen
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Zhou
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tiantian Zheng
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Ziyin Li
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Fan Lin
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Haizhu Xie
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Ma
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, the Second Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Lina Zhang
- Department of Radiology, the First Affiliated Hospital of China Medical University, Shenyang 400042, China
| | - Qin Li
- Department of Radiology, Weifang Hospital of Traditional Chinese Medicine, Weifang 262600, China
| | - Weiwei Wang
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Yi Dai
- Department of Radiology, the Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Ruijun Tang
- Department of Pathology, Guilin Traditional Chinese Medicine Hospital, Guilin 541002, China
| | - Jigang Wang
- Department of Pathology, the Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Ping Yang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Ning Mao
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| |
Collapse
|
4
|
Omenn GS, Orchard S, Lane L, Lindskog C, Pineau C, Overall CM, Budnik B, Mudge JM, Packer NH, Weintraub ST, Roehrl MHA, Nice E, Guo T, Van Eyk JE, Völker U, Zhang G, Bandeira N, Aebersold R, Moritz RL, Deutsch EW. The 2024 Report on the Human Proteome from the HUPO Human Proteome Project. J Proteome Res 2024; 23:5296-5311. [PMID: 39514846 PMCID: PMC11781352 DOI: 10.1021/acs.jproteome.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The Human Proteome Project (HPP), the flagship initiative of the Human Proteome Organization (HUPO), has pursued two goals: (1) to credibly identify at least one isoform of every protein-coding gene and (2) to make proteomics an integral part of multiomics studies of human health and disease. The past year has seen major transitions for the HPP. neXtProt was retired as the official HPP knowledge base, UniProtKB became the reference proteome knowledge base, and Ensembl-GENCODE provides the reference protein target list. A function evidence FE1-5 scoring system has been developed for functional annotation of proteins, parallel to the PE1-5 UniProtKB/neXtProt scheme for evidence of protein expression. This report includes updates from neXtProt (version 2023-09) and UniProtKB release 2024_04, with protein expression detected (PE1) for 18138 of the 19411 GENCODE protein-coding genes (93%). The number of non-PE1 proteins ("missing proteins") is now 1273. The transition to GENCODE is a net reduction of 367 proteins (19,411 PE1-5 instead of 19,778 PE1-4 last year in neXtProt). We include reports from the Biology and Disease-driven HPP, the Human Protein Atlas, and the HPP Grand Challenge Project. We expect the new Functional Evidence FE1-5 scheme to energize the Grand Challenge Project for functional annotation of human proteins throughout the global proteomics community, including π-HuB in China.
Collapse
Affiliation(s)
- Gilbert S. Omenn
- University of Michigan, Ann Arbor, Michigan 48109, United States
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK, CB10 1SD
| | - Lydie Lane
- CALIPHO Group, SIB Swiss Institute of Bioinformatics and University of Geneva, 1015 Lausanne, Switzerland
| | - Cecilia Lindskog
- Department of Immunology Genetics and Pathology, Cancer Precision Medicine, Uppsala University, 752 36 Uppsala, Sweden
| | - Charles Pineau
- Univ Rennes, Inserm, EHESP, Irset, UMR_S 1085,35000 Rennes, France
| | - Christopher M. Overall
- University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Yonsei Frontier Lab, Yonsei University, 50 Yonsei-ro, Sudaemoon-ku, Seoul, 03722, Republic of Korea
| | - Bogdan Budnik
- Hansjörg Wyss Institute for Biologically Inspired Engineering at Harvard University
| | - Jonathan M. Mudge
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK, CB10 1SD
| | | | - Susan T. Weintraub
- University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, United States
| | - Michael H. A. Roehrl
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States
| | | | - Tiannan Guo
- Center for Intelligent Proteomics, Westlake Laboratory, Westlake University, Hangzhou 310024, Zhejiang Province, China
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Pavilion, 9th Floor, Los Angeles, CA, 90048, United States
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Gong Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Nuno Bandeira
- University of California, San Diego, La Jolla, CA, 92093, United States
| | | | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Eric W. Deutsch
- Institute for Systems Biology, Seattle, Washington 98109, United States
| |
Collapse
|
5
|
Xiaojian Y, Zhanbo Q, Jian C, Zefeng W, Jian L, Jin L, Yuefen P, Shuwen H. Deep learning application in prediction of cancer molecular alterations based on pathological images: a bibliographic analysis via CiteSpace. J Cancer Res Clin Oncol 2024; 150:467. [PMID: 39422817 PMCID: PMC11489169 DOI: 10.1007/s00432-024-05992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The advancements in artificial intelligence (AI) technology for image recognition were propelling molecular pathology research into a new era. OBJECTIVE To summarize the hot spots and research trends in the field of molecular pathology image recognition. METHODS Relevant articles from January 1st, 2010, to August 25th, 2023, were retrieved from the Web of Science Core Collection. Subsequently, CiteSpace was employed for bibliometric and visual analysis, generating diverse network diagrams illustrating keywords, highly cited references, hot topics, and research trends. RESULTS A total of 110 relevant articles were extracted from a pool of 10,205 articles. The overall publication count exhibited a rising trend each year. The leading contributors in terms of institutions, countries, and authors were Maastricht University (11 articles), the United States (38 articles), and Kather Jacob Nicholas (9 articles), respectively. Half of the top ten research institutions, based on publication volume, were affiliated with Germany. The most frequently cited article was authored by Nicolas Coudray et al. accumulating 703 citations. The keyword "Deep learning" had the highest frequency in 2019. Notably, the highlighted keywords from 2022 to 2023 included "microsatellite instability", and there were 21 articles focusing on utilizing algorithms to recognize microsatellite instability (MSI) in colorectal cancer (CRC) pathological images. CONCLUSION The use of DL is expected to provide a new strategy to effectively solve the current problem of time-consuming and expensive molecular pathology detection. Therefore, further research is needed to address issues, such as data quality and standardization, model interpretability, and resource and infrastructure requirements.
Collapse
Affiliation(s)
- Yu Xiaojian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Wang Zefeng
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jin
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Pan Yuefen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
6
|
Li T, Sun G, Ye H, Song C, Shen Y, Cheng Y, Zou Y, Fang Z, Shi J, Wang K, Dai L, Wang P. ESCCPred: a machine learning model for diagnostic prediction of early esophageal squamous cell carcinoma using autoantibody profiles. Br J Cancer 2024; 131:883-894. [PMID: 38956246 PMCID: PMC11369250 DOI: 10.1038/s41416-024-02781-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a deadly cancer with no clinically ideal biomarkers for early diagnosis. The objective of this study was to develop and validate a user-friendly diagnostic tool for early ESCC detection. METHODS The study encompassed three phases: discovery, verification, and validation, comprising a total of 1309 individuals. Serum autoantibodies were profiled using the HuProtTM human proteome microarray, and autoantibody levels were measured using the enzyme-linked immunosorbent assay (ELISA). Twelve machine learning algorithms were employed to construct diagnostic models, and evaluated using the area under the receiver operating characteristic curve (AUC). The model application was facilitated through R Shiny, providing a graphical interface. RESULTS Thirteen autoantibodies targeting TAAs (CAST, FAM131A, GABPA, HDAC1, HDGFL1, HSF1, ISM2, PTMS, RNF219, SMARCE1, SNAP25, SRPK2, and ZPR1) were identified in the discovery phase. Subsequent verification and validation phases identified five TAAbs (anti-CAST, anti-HDAC1, anti-HSF1, anti-PTMS, and anti-ZPR1) that exhibited significant differences between ESCC and control subjects (P < 0.05). The support vector machine (SVM) model demonstrated robust performance, with AUCs of 0.86 (95% CI: 0.82-0.89) in the training set and 0.83 (95% CI: 0.78-0.88) in the test set. For early-stage ESCC, the SVM model achieved AUCs of 0.83 (95% CI: 0.79-0.88) in the training set and 0.83 (95% CI: 0.77-0.90) in the test set. Notably, promising results were observed for high-grade intraepithelial neoplasia, with an AUC of 0.87 (95% CI: 0.77-0.98). The web-based implementation of the early ESCC diagnostic tool is publicly accessible at https://litdong.shinyapps.io/ESCCPred/ . CONCLUSION This study provides a promising and easy-to-use diagnostic prediction model for early ESCC detection. It holds promise for improving early detection strategies and has potential implications for public health.
Collapse
Affiliation(s)
- Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Guiying Sun
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
- Henan Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Caijuan Song
- The Institution for Chronic and Noncommunicable Disease Control and Prevention, Zhengzhou Center for Disease Control and Prevention, Zhengzhou, 450052, Henan Provinc, China
| | - Yajing Shen
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Yifan Cheng
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Yuanlin Zou
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Zhaoyang Fang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Jianxiang Shi
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Keyan Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liping Dai
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan Provinc, China.
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan Province, China.
| |
Collapse
|
7
|
Majumder B, Nataraj NB, Maitreyi L, Datta S. Mismatch repair-proficient tumor footprints in the sands of immune desert: mechanistic constraints and precision platforms. Front Immunol 2024; 15:1414376. [PMID: 39100682 PMCID: PMC11294168 DOI: 10.3389/fimmu.2024.1414376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/17/2024] [Indexed: 08/06/2024] Open
Abstract
Mismatch repair proficient (MMRp) tumors of colorectal origin are one of the prevalent yet unpredictable clinical challenges. Despite earnest efforts, optimal treatment modalities have yet to emerge for this class. The poor prognosis and limited actionability of MMRp are ascribed to a low neoantigen burden and a desert-like microenvironment. This review focuses on the critical roadblocks orchestrated by an immune evasive mechanistic milieu in the context of MMRp. The low density of effector immune cells, their weak spatiotemporal underpinnings, and the high-handedness of the IL-17-TGF-β signaling are intertwined and present formidable challenges for the existing therapies. Microbiome niche decorated by Fusobacterium nucleatum alters the metabolic program to maintain an immunosuppressive state. We also highlight the evolving strategies to repolarize and reinvigorate this microenvironment. Reconstruction of anti-tumor chemokine signaling, rational drug combinations eliciting T cell activation, and reprograming the maladapted microbiome are exciting developments in this direction. Alternative vulnerability of other DNA damage repair pathways is gaining momentum. Integration of liquid biopsy and ex vivo functional platforms provide precision oncology insights. We illustrated the perspectives and changing landscape of MMRp-CRC. The emerging opportunities discussed in this review can turn the tide in favor of fighting the treatment dilemma for this elusive cancer.
Collapse
|
8
|
Kimura A, Tsujikawa T, Morimoto H, Saburi S, Mitsuda J, Mukudai S, Nagao H, Shibata S, Ogi H, Miyagawa-Hayashino A, Konishi E, Itoh K, Hirano S. Rapid multiplex immunohistochemistry for characterizing tumor-immune microenvironment. Heliyon 2024; 10:e33830. [PMID: 39050465 PMCID: PMC11268184 DOI: 10.1016/j.heliyon.2024.e33830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 04/28/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Intratumoral immune profiles are related to prognosis and therapeutic efficacy, and could result in personalized treatments based on biomarkers. To develop a multiplex, quantitative, and rapid tissue evaluation method based on the clinically established standard immunohistochemistry (IHC), a 6-marker rapid multiplex IHC was developed based on our previously reported 14-marker multiplex IHC by reducing the number of labels and accelerating the staining procedure. First, fewer labels were required to identify the same immunological features linked to prognosis in 14-marker multiplex IHC analyses. The six selected markers showed a significant correlation with the 14 markers in the immune classification. Next, a rapid staining protocol was developed by optimizing the reaction temperature, chromogen, and washing time, allowing the completion of 6-marker analysis in 5 h and 49 min, as opposed to the several days required for conventional multiplex IHC. Validation of benign tonsil and head and neck cancer tissues revealed a significant correlation between rapid and conventional 6-makrer multiplex IHC in terms of staining intensities, densities of T cells, macrophages, lymphoid/myeloid immune cell ratios, and spatial profiles of intratumoral immune infiltrates. This method may enable quantitative assessment of the tumor-immune microenvironment on a clinically feasible time scale, which promotes the development of tissue biomarker-guided therapeutic strategies.
Collapse
Affiliation(s)
- Alisa Kimura
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Tsujikawa
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Oregon, USA
| | - Hiroki Morimoto
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sumiyo Saburi
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junichi Mitsuda
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeyuki Mukudai
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hikaru Nagao
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Hiroshi Ogi
- SCREEN Holdings Co., Ltd., Kyoto, Japan
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aya Miyagawa-Hayashino
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Hirano
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
9
|
Petralia F, Ma W, Yaron TM, Caruso FP, Tignor N, Wang JM, Charytonowicz D, Johnson JL, Huntsman EM, Marino GB, Calinawan A, Evangelista JE, Selvan ME, Chowdhury S, Rykunov D, Krek A, Song X, Turhan B, Christianson KE, Lewis DA, Deng EZ, Clarke DJB, Whiteaker JR, Kennedy JJ, Zhao L, Segura RL, Batra H, Raso MG, Parra ER, Soundararajan R, Tang X, Li Y, Yi X, Satpathy S, Wang Y, Wiznerowicz M, González-Robles TJ, Iavarone A, Gosline SJC, Reva B, Robles AI, Nesvizhskii AI, Mani DR, Gillette MA, Klein RJ, Cieslik M, Zhang B, Paulovich AG, Sebra R, Gümüş ZH, Hostetter G, Fenyö D, Omenn GS, Cantley LC, Ma'ayan A, Lazar AJ, Ceccarelli M, Wang P. Pan-cancer proteogenomics characterization of tumor immunity. Cell 2024; 187:1255-1277.e27. [PMID: 38359819 PMCID: PMC10988632 DOI: 10.1016/j.cell.2024.01.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/29/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
Despite the successes of immunotherapy in cancer treatment over recent decades, less than <10%-20% cancer cases have demonstrated durable responses from immune checkpoint blockade. To enhance the efficacy of immunotherapies, combination therapies suppressing multiple immune evasion mechanisms are increasingly contemplated. To better understand immune cell surveillance and diverse immune evasion responses in tumor tissues, we comprehensively characterized the immune landscape of more than 1,000 tumors across ten different cancers using CPTAC pan-cancer proteogenomic data. We identified seven distinct immune subtypes based on integrative learning of cell type compositions and pathway activities. We then thoroughly categorized unique genomic, epigenetic, transcriptomic, and proteomic changes associated with each subtype. Further leveraging the deep phosphoproteomic data, we studied kinase activities in different immune subtypes, which revealed potential subtype-specific therapeutic targets. Insights from this work will facilitate the development of future immunotherapy strategies and enhance precision targeting with existing agents.
Collapse
Affiliation(s)
- Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tomer M Yaron
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Francesca Pia Caruso
- BIOGEM Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy; Department of Electrical Engineering and Information Technologies, University of Naples "Federico II", Naples, Italy
| | - Nicole Tignor
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua M Wang
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel Charytonowicz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Emily M Huntsman
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Giacomo B Marino
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Calinawan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John Erol Evangelista
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shrabanti Chowdhury
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dmitry Rykunov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiaoyu Song
- Institute for Healthcare Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen E Christianson
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - David A Lewis
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eden Z Deng
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel J B Clarke
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey R Whiteaker
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jacob J Kennedy
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lei Zhao
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Rossana Lazcano Segura
- Departments of Pathology & Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Harsh Batra
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Edwin Roger Parra
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ximing Tang
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Xinpei Yi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Ying Wang
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Maciej Wiznerowicz
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-701 Poznań, Poland; International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Heliodor Swiecicki Clinical Hospital, 60-203 Poznań, Poland
| | - Tania J González-Robles
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Antonio Iavarone
- Department of Neurological Surgery, Department of Biochemistry, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sara J C Gosline
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Michael A Gillette
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marcin Cieslik
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amanda G Paulovich
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - David Fenyö
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Gilbert S Omenn
- Departments of Computational Medicine & Bioinformatics, Internal Medicine, Human Genetics, & Environmental Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Department of Public Health Sciences, University of Miami, Miami, FL, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|