1
|
Wei W, Dang Y, Chen G, Han C, Zhang S, Zhu Z, Bie X, Xue J. Comprehensive analysis of senescence-related genes identifies prognostic clusters with distinct characteristics in glioma. Sci Rep 2025; 15:9540. [PMID: 40108265 PMCID: PMC11923138 DOI: 10.1038/s41598-025-93482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Cellular senescence, defined as a state of permanent arrest in cell growth, is regarded as a crucial tumor suppression mechanism. However, accumulating scientific evidence suggests that senescent cells play a detrimental role in the progression of cancer. Unfortunately, the current lack of reliable markers that specifically reflect the level of senescence in cancer greatly hinders our in-depth understanding of this important biological foundation. Therefore, the search for more specific and reliable markers to reveal the specific role of senescent cells in cancer progression is particularly urgent and important. To uncover the role of senescence in gliomas, we collected senescence-related genes for integrated analysis. Consensus clustering was used to subtype gliomas based on the senescence gene set, and we identified two robust prognostic clusters of gliomas with distinct survival outcomes, multi-omics landscapes, immune characteristics, and differential drug responses. Multiple external datasets were used to validate the stability of our subtypes. Various computational and experimental methods, including WGCNA (Weighted Gene Co-expression Network Analysis), ssGSEA (single-sample Gene Set Enrichment Analysis), and machine learning algorithms (lasso regression, support vector machines, random forests), were employed for analysis. We found that CEBPB and LMNA are associated with poor prognosis in gliomas and may mediate immunosuppression and tumor proliferation. Drug prediction indicated that dasatinib is a potential therapeutic agent. Our findings provide insights into the role of the senescence gene set in patient stratification and precision medicine.
Collapse
Affiliation(s)
- Wenyuan Wei
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ying Dang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Gang Chen
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Chao Han
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Siwei Zhang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ziqiang Zhu
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Xiaohua Bie
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Jungang Xue
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
2
|
Yang B, Li W, Xu Z, Li W, Hu G. NetSDR: Drug repurposing for cancers based on subtype-specific network modularization and perturbation analysis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167688. [PMID: 39862994 DOI: 10.1016/j.bbadis.2025.167688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/04/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Cancer, a heterogeneous disease, presents significant challenges for drug development due to its complex etiology. Drug repurposing, particularly through network medicine approaches, offers a promising avenue for cancer treatment by analyzing how drugs influence cellular networks on a systemic scale. The advent of large-scale proteomics data provides new opportunities to elucidate regulatory mechanisms specific to cancer subtypes. Herein, we present NetSDR, a Network-based Subtype-specific Drug Repurposing framework for prioritizing repurposed drugs specific to certain cancer subtypes, guided by subtype-specific proteomic signatures and network perturbations. First, by integrating cancer subtype information into a network-based method, we developed a pipeline to recognize subtype-specific functional modules. Next, we conducted drug response analysis for each module to identify the "therapeutic module" and then used deep learning to construct weighted drug response network for the particular subtype. Finally, we employed a perturbation response scanning-based drug repurposing method, which incorporates dynamic information, to facilitate the prioritization of candidate drugs. Applying the framework to gastric cancer, we attested the significance of the extracellular matrix module in treatment strategies and discovered a promising potential drug target, LAMB2, as well as a series of possible repurposed drugs. This study demonstrates a systems biology framework for precise drug repurposing in cancer and other complex diseases.
Collapse
Affiliation(s)
- Bin Yang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wanshi Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zhen Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Guang Hu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; Key Laboratory of Alkene-carbon Fibres-based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China.
| |
Collapse
|
3
|
Liu S, Meng Y, Zhang Y, Qiu L, Wan X, Yang X, Zhang Y, Liu X, Wen L, Lei X, Zhang B, Han J. Integrative analysis of senescence-related genes identifies robust prognostic clusters with distinct features in hepatocellular carcinoma. J Adv Res 2025; 69:107-123. [PMID: 38614215 PMCID: PMC11954806 DOI: 10.1016/j.jare.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024] Open
Abstract
INTRODUCTION Senescence refers to a state of permanent cell growth arrest and is regarded as a tumor suppressive mechanism, whereas accumulative evidence demonstrate that senescent cells play an adverse role during cancer progression. The scarcity of specific and reliable markers reflecting senescence level in cancer impede our understanding of this biological basis. OBJECTIVES Senescence-related genes (SRGs) were collected for integrative analysis to reveal the role of senescence in hepatocellular carcinoma (HCC). METHODS Consensus clustering was used to subtype HCC based on SRGs. Several computational methods, including single sample gene set enrichment analysis (ssGSEA), fuzzy c-means algorithm, were performed. Data of drug sensitivities were utilized to screen potential therapeutic agents for different senescence patients. Additionally, we developed a method called signature-related gene analysis (SRGA) for identification of markers relevant to phenotype of interest. Experimental strategies consisting quantitative real-time PCR (qRT-PCR), β-galactosidase assay, western blot, and tumor-T cell co-culture system were used to validate the findings in vitro. RESULTS We identified three robust prognostic clusters of HCC patients with distinct survival outcome, mutational landscape, and immune features. We further extracted signature genes of senescence clusters to construct the senescence scoring system and profile senescence level in HCC at bulk and single-cell resolution. Senescence-induced stemness reprogramming was confirmed both in silico and in vitro. HCC patients with high senescence were immune suppressed and sensitive to Tozasertib and other drugs. We suggested that MAFG, PLIN3, and 4 other genes were pertinent to HCC senescence, and MAFG potentially mediated immune suppression, senescence, and stemness. CONCLUSION Our findings provide insights into the role of SRGs in patients stratification and precision medicine.
Collapse
Affiliation(s)
- Sicheng Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Meng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaguang Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaowen Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linda Wen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xue Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- Research Laboratory of Cancer Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Manoukian P, Kuhnen LC, van Laarhoven HWM, Bijlsma MF. Association of epigenetic landscapes with heterogeneity and plasticity in pancreatic cancer. Crit Rev Oncol Hematol 2025; 206:104573. [PMID: 39581245 DOI: 10.1016/j.critrevonc.2024.104573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Due to a lack of clear symptoms, patients often present with advanced disease, with limited clinical intervention options. The high mortality rate of PDAC is, however, also a result of several other factors that include a high degree of heterogeneity and treatment resistant cellular phenotypes. Molecular subtypes of PDAC have been identified that are thought to represent cellular phenotypes at the tissue level. The epigenetic landscape is an important factor that dictates these subtypes. Permissive epigenetic landscapes serve as drivers of molecular heterogeneity and cellular plasticity in developing crypts as well as metaplastic lesions. Drawing parallels with other cancers, we hypothesize that epigenetic permissiveness is a potential driver of cellular plasticity in PDAC. In this review will explore the epigenetic alterations that underlie PDAC cell states and relate them to cellular plasticity from other contexts. In doing so, we aim to highlight epigenomic drivers of PDAC heterogeneity and plasticity and, with that, offer some insight to guide pre-clinical research.
Collapse
Affiliation(s)
- Paul Manoukian
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - Leo C Kuhnen
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Gibbs DL, Cioffi G, Aguilar B, Waite KA, Pan E, Mandel J, Umemura Y, Luo J, Rubin JB, Pot D, Barnholtz-Sloan J. Robust Cluster Prediction Across Data Types Validates Association of Sex and Therapy Response in GBM. Cancers (Basel) 2025; 17:445. [PMID: 39941811 PMCID: PMC11815886 DOI: 10.3390/cancers17030445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Previous studies have described sex-specific patient subtyping in glioblastoma. The cluster labels associated with these "legacy data" were used to train a predictive model capable of recapitulating this clustering in contemporary contexts. METHODS We used robust ensemble machine learning to train a model using gene microarray data to perform multi-platform predictions including RNA-seq and potentially scRNA-seq. RESULTS The engineered feature set was composed of many previously reported genes that are associated with patient prognosis. Interestingly, these well-known genes formed a predictive signature only for female patients, and the application of the predictive signature to male patients produced unexpected results. CONCLUSIONS This work demonstrates how annotated "legacy data" can be used to build robust predictive models capable of multi-target predictions across multiple platforms.
Collapse
Affiliation(s)
- David L. Gibbs
- Thorsson-Shmulevich Lab, Institute of Systems Biology, Seattle, WA 98109, USA
| | - Gino Cioffi
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
| | - Boris Aguilar
- Thorsson-Shmulevich Lab, Institute of Systems Biology, Seattle, WA 98109, USA
| | - Kristin A. Waite
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
| | - Edward Pan
- Global Oncology Research & Development, Daiichi-Sankyo, Inc., Basking Ridge, NJ 07920, USA
| | - Jacob Mandel
- Department of Neurology and Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yoshie Umemura
- IVY Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jingqin Luo
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center Biostatistics and Qualitative Research Shared Resource, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua B. Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Pot
- General Dynamics Information Technology, Falls Church, VA 22042, USA
| | - Jill Barnholtz-Sloan
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA (J.B.-S.)
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Wang J, Wang L, Liu Y, Li X, Ma J, Li M, Zhu Y. Comprehensive Evaluation of Multi-Omics Clustering Algorithms for Cancer Molecular Subtyping. Int J Mol Sci 2025; 26:963. [PMID: 39940732 PMCID: PMC11816650 DOI: 10.3390/ijms26030963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
As a highly heterogeneous and complex disease, the identification of cancer's molecular subtypes is crucial for accurate diagnosis and personalized treatment. The integration of multi-omics data enables a comprehensive interpretation of the molecular characteristics of cancer at various biological levels. In recent years, an increasing number of multi-omics clustering algorithms for cancer molecular subtyping have been proposed. However, the absence of a definitive gold standard makes it challenging to evaluate and compare these methods effectively. In this study, we developed a general framework for the comprehensive evaluation of multi-omics clustering algorithms and introduced an innovative metric, the accuracy-weighted average index, which simultaneously considers both clustering performance and clinical relevance. Using this framework, we performed a thorough evaluation and comparison of 11 state-of-the-art multi-omics clustering algorithms, including deep learning-based methods. By integrating the accuracy-weighted average index with computational efficiency, our analysis reveals that PIntMF demonstrates the best overall performance, making it a promising tool for molecular subtyping across a wide range of cancers.
Collapse
Affiliation(s)
- Juan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China;
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Lingxiao Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Yi Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Xiao Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Jie Ma
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Mansheng Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| | - Yunping Zhu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China;
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; (L.W.); (Y.L.); (X.L.); (J.M.)
| |
Collapse
|
7
|
Zheng M, Xu J, Yu S, Zhao Z, Zhang Y, Wei M. Utility of Multiparametric Breast MRI Radiomics to Predict Cyclin D1 and TGF-β1 Expression. J Comput Assist Tomogr 2025:00004728-990000000-00408. [PMID: 39794899 DOI: 10.1097/rct.0000000000001717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVE To develop a machine learning model that integrates clinical features and multisequence MRI radiomics for noninvasively predicting the expression status of prognostic-related factors cyclin D1 and TGF-β1 in breast cancer, providing additional information for the clinical development of personalized treatment plans. METHODS A total of 123 breast cancer patients confirmed by surgical pathology were retrospectively enrolled in our Hospital from January 2016 to July 2022. The patients were randomly divided into a training group (87 cases) and a validation group (36 cases). Preoperative routine and dynamic contrast-enhanced magnetic resonance imaging scans of the breast were performed for treatment subjects. The region of interest was manually outlined, and texture features were extracted using AK software. Subsequently, the LASSO algorithm was employed for dimensionality reduction and feature selection to establish the MRI radiomics labels. The diagnostic efficacy and clinical value were assessed through receiver operating characteristic (ROC) curve analysis and decision curve analysis (DCA). RESULTS In the cyclin D1 cohort, the area under the receiver operating characteristic (ROC) curve in the clinical prediction model training and validation groups was 0.738 and 0.656, respectively. The multisequence MRI radiomics prediction model achieved an AUC of 0.874 and 0.753 in these respective groups, while the combined prediction model yielded an AUC of 0.892 and 0.785. In the TGF-β1 cohort, the ROC AUC for the clinical prediction model was found to be 0.693 and 0.645 in the training and validation groups, respectively. For the multiseries MRI radiomics prediction model, it achieved an AUC of 0.875 and 0.760 in these respective groups; whereas for the combined prediction model, it reached an AUC of 0.904 and 0.833. Decision curve analysis (DCA) demonstrated that both cohorts indicated a higher clinical application value for the combined prediction model compared with both individual models-clinical prediction model alone or radiomics model. CONCLUSION The integration of clinical features and multisequence MRI radiomics in a combined modeling approach holds significant predictive value for the expression status of cyclin D1 and TGF-β1. The model provides a noninvasive, dynamic evaluation method that provides effective guidance for clinical treatment.
Collapse
Affiliation(s)
| | - Jiaqi Xu
- School of Medicine, Shaoxing University
| | - Shujie Yu
- School of Medicine, Shaoxing University
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Yu Zhang
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| | - Mingzhu Wei
- Department of Radiology, Shaoxing People's Hospital (Zhejiang University Shaoxing Hospital), Shaoxing, Zhejiang, China
| |
Collapse
|
8
|
Guo D, Chen J, Wang Y, Liu X. Survival prediction and molecular subtyping of squamous cell lung cancer based on network embedding. Sci Rep 2024; 14:29474. [PMID: 39604473 PMCID: PMC11603150 DOI: 10.1038/s41598-024-81199-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: 08/05/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Squamous cell lung cancer (SQCLC), which is fatal to humans, is heterogeneous with different genetic and histological features. We used SBMOI, a multi-omics data integration method from previous study, to integrate clinical, gene expression, and somatic mutation data of SQCLC to construct new patient features. Next, random survival forest (RSF) model and SimpleMKL model were constructed to predict the survival of SQCLC patients, and K-means model was constructed to perform molecular subtyping. The results of the RSF model showed that when the dimension of the patient features were 11 × 364 and the hard threshold was 0.2, we obtained the best results, and the AUC value of the 1-year time-dependent ROC curve was 0.706. The SimpleMKL model, constructed using the same patient features, performed exceptionally well, with 1-year, 5-year, and 10-year survival prediction AUC values of 0.944, 0.947 and 0.950, respectively. We used K-means analysis to identify three SQCLC molecular subtypes with significant survival differences. The patient features constructed by SBMOI were used to effectively predict the survival and molecular subtyping of SQCLC patients. In addition, our study further confirmed the effectiveness in multi-omics data integration task and broad applicability of SBMOI.
Collapse
Affiliation(s)
- Dingjie Guo
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Jing Chen
- Academy for Advanced Interdisciplinary Studies, Northeast Normal University, Changchun, 130024, China
| | - Yixian Wang
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xin Liu
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
9
|
Akand M, Jatsenko T, Muilwijk T, Gevaert T, Joniau S, Van der Aa F. Deciphering the molecular heterogeneity of intermediate- and (very-)high-risk non-muscle-invasive bladder cancer using multi-layered -omics studies. Front Oncol 2024; 14:1424293. [PMID: 39497708 PMCID: PMC11532112 DOI: 10.3389/fonc.2024.1424293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/13/2024] [Indexed: 11/07/2024] Open
Abstract
Bladder cancer (BC) is the most common malignancy of the urinary tract. About 75% of all BC patients present with non-muscle-invasive BC (NMIBC), of which up to 70% will recur, and 15% will progress in stage and grade. As the recurrence and progression rates of NMIBC are strongly associated with some clinical and pathological factors, several risk stratification models have been developed to individually predict the short- and long-term risks of disease recurrence and progression. The NMIBC patients are stratified into four risk groups as low-, intermediate-, high-risk, and very high-risk by the European Association of Urology (EAU). Significant heterogeneity in terms of oncological outcomes and prognosis has been observed among NMIBC patients within the same EAU risk group, which has been partly attributed to the intrinsic heterogeneity of BC at the molecular level. Currently, we have a poor understanding of how to distinguish intermediate- and (very-)high-risk NMIBC with poor outcomes from those with a more benign disease course and lack predictive/prognostic tools that can specifically stratify them according to their pathologic and molecular properties. There is an unmet need for developing a more accurate scoring system that considers the treatment they receive after TURBT to enable their better stratification for further follow-up regimens and treatment selection, based also on a better response prediction to the treatment. Based on these facts, by employing a multi-layered -omics (namely, genomics, epigenetics, transcriptomics, proteomics, lipidomics, metabolomics) and immunohistopathology approach, we hypothesize to decipher molecular heterogeneity of intermediate- and (very-)high-risk NMIBC and to better stratify the patients with this disease. A combination of different -omics will provide a more detailed and multi-dimensional characterization of the tumor and represent the broad spectrum of NMIBC phenotypes, which will help to decipher the molecular heterogeneity of intermediate- and (very-)high-risk NMIBC. We think that this combinatorial multi-omics approach has the potential to improve the prediction of recurrence and progression with higher precision and to develop a molecular feature-based algorithm for stratifying the patients properly and guiding their therapeutic interventions in a personalized manner.
Collapse
Affiliation(s)
- Murat Akand
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Tatjana Jatsenko
- Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Tim Muilwijk
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Frank Van der Aa
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Urology, Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Gong D, Arbesfeld-Qiu JM, Perrault E, Bae JW, Hwang WL. Spatial oncology: Translating contextual biology to the clinic. Cancer Cell 2024; 42:1653-1675. [PMID: 39366372 PMCID: PMC12051486 DOI: 10.1016/j.ccell.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Accepted: 09/06/2024] [Indexed: 10/06/2024]
Abstract
Microscopic examination of cells in their tissue context has been the driving force behind diagnostic histopathology over the past two centuries. Recently, the rise of advanced molecular biomarkers identified through single cell profiling has increased our understanding of cellular heterogeneity in cancer but have yet to significantly impact clinical care. Spatial technologies integrating molecular profiling with microenvironmental features are poised to bridge this translational gap by providing critical in situ context for understanding cellular interactions and organization. Here, we review how spatial tools have been used to study tumor ecosystems and their clinical applications. We detail findings in cell-cell interactions, microenvironment composition, and tissue remodeling for immune evasion and therapeutic resistance. Additionally, we highlight the emerging role of multi-omic spatial profiling for characterizing clinically relevant features including perineural invasion, tertiary lymphoid structures, and the tumor-stroma interface. Finally, we explore strategies for clinical integration and their augmentation of therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Dennis Gong
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeanna M Arbesfeld-Qiu
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ella Perrault
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jung Woo Bae
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William L Hwang
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Xiong B, Liu W, Liu Y, Chen T, Lin A, Song J, Qu L, Luo P, Jiang A, Wang L. A Multi-Omics Prognostic Model Capturing Tumor Stemness and the Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Biomedicines 2024; 12:2171. [PMID: 39457484 PMCID: PMC11504857 DOI: 10.3390/biomedicines12102171] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer stem-like cells (CSCs), a distinct subset recognized for their stem cell-like abilities, are intimately linked to the resistance to radiotherapy, metastatic behaviors, and self-renewal capacities in tumors. Despite their relevance, the definitive traits and importance of CSCs in the realm of oncology are still not fully comprehended, particularly in the context of clear cell renal cell carcinoma (ccRCC). A comprehensive understanding of these CSCs' properties in relation to stemness, and their impact on the efficacy of treatment and resistance to medication, is of paramount importance. Methods: In a meticulous research effort, we have identified new molecular categories designated as CRCS1 and CRCS2 through the application of an unsupervised clustering algorithm. The analysis of these subtypes included a comprehensive examination of the tumor immune environment, patterns of metabolic activity, progression of the disease, and its response to immunotherapy. In addition, we have delved into understanding these subtypes' distinctive clinical presentations, the landscape of their genomic alterations, and the likelihood of their response to various pharmacological interventions. Proceeding from these insights, prognostic models were developed that could potentially forecast the outcomes for patients with ccRCC, as well as inform strategies for the surveillance of recurrence after treatment and the handling of drug-resistant scenarios. Results: Compared with CRCS1, CRCS2 patients had a lower clinical stage/grading and a better prognosis. The CRCS2 subtype was in a hypoxic state and was characterized by suppression and exclusion of immune function, which was sensitive to gefitinib, erlotinib, and saracatinib. The constructed prognostic risk model performed well in both training and validation cohorts, helping to identify patients who may benefit from specific treatments or who are at risk of recurrence and drug resistance. A novel therapeutic target, SAA2, regulating neutrophil and fibroblast infiltration, and, thus promoting ccRCC progression, was identified. Conclusions: Our findings highlight the key role of CSCs in shaping the ccRCC tumor microenvironment, crucial for therapy research and clinical guidance. Recognizing tumor stemness helps to predict treatment efficacy, recurrence, and drug resistance, informing treatment strategies and enhancing ccRCC patient outcomes.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu 610200, China;
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Ying Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Tong Chen
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Jiaao Song
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China;
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| |
Collapse
|
12
|
Patrício A, Costa RS, Henriques R. Pattern-centric transformation of omics data grounded on discriminative gene associations aids predictive tasks in TCGA while ensuring interpretability. Biotechnol Bioeng 2024; 121:2881-2892. [PMID: 38859573 DOI: 10.1002/bit.28758] [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: 10/04/2023] [Revised: 02/07/2024] [Accepted: 05/18/2024] [Indexed: 06/12/2024]
Abstract
The increasing prevalence of omics data sources is pushing the study of regulatory mechanisms underlying complex diseases such as cancer. However, the vast quantities of molecular features produced and the inherent interplay between them lead to a level of complexity that hampers both descriptive and predictive tasks, requiring custom-built algorithms that can extract relevant information from these sources of data. We propose a transformation that moves data centered on molecules (e.g., transcripts and proteins) to a new data space focused on putative regulatory modules given by statistically relevant co-expression patterns. To this end, the proposed transformation extracts patterns from the data through biclustering and uses them to create new variables with guarantees of interpretability and discriminative power. The transformation is shown to achieve dimensionality reductions of up to 99% and increase predictive performance of various classifiers across multiple omics layers. Results suggest that omics data transformations from gene-centric to pattern-centric data supports both prediction tasks and human interpretation, notably contributing to precision medicine applications.
Collapse
Affiliation(s)
- André Patrício
- INESC-ID and Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Rafael S Costa
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Rui Henriques
- INESC-ID and Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
13
|
Deng EZ, Marino GB, Clarke DJB, Diamant I, Resnick AC, Ma W, Wang P, Ma'ayan A. Multiomics2Targets identifies targets from cancer cohorts profiled with transcriptomics, proteomics, and phosphoproteomics. CELL REPORTS METHODS 2024; 4:100839. [PMID: 39127042 PMCID: PMC11384097 DOI: 10.1016/j.crmeth.2024.100839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/06/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
The availability of data from profiling of cancer patients with multiomics is rapidly increasing. However, integrative analysis of such data for personalized target identification is not trivial. Multiomics2Targets is a platform that enables users to upload transcriptomics, proteomics, and phosphoproteomics data matrices collected from the same cohort of cancer patients. After uploading the data, Multiomics2Targets produces a report that resembles a research publication. The uploaded matrices are processed, analyzed, and visualized using the tools Enrichr, KEA3, ChEA3, Expression2Kinases, and TargetRanger to identify and prioritize proteins, genes, and transcripts as potential targets. Figures and tables, as well as descriptions of the methods and results, are automatically generated. Reports include an abstract, introduction, methods, results, discussion, conclusions, and references and are exportable as citable PDFs and Jupyter Notebooks. Multiomics2Targets is applied to analyze version 3 of the Clinical Proteomic Tumor Analysis Consortium (CPTAC3) pan-cancer cohort, identifying potential targets for each CPTAC3 cancer subtype. Multiomics2Targets is available from https://multiomics2targets.maayanlab.cloud/.
Collapse
Affiliation(s)
- Eden Z Deng
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Giacomo B Marino
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, 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, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Ido Diamant
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Adam C Resnick
- Center for Data Driven Discovery in Biomedicine, Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| |
Collapse
|
14
|
Zhu W, Huang H, Hu Z, Gu Y, Zhang R, Shu H, Liu H, Sun X. Comprehensive Transcriptome Analysis Expands lncRNA Functional Profiles in Breast Cancer. Int J Mol Sci 2024; 25:8456. [PMID: 39126025 PMCID: PMC11313538 DOI: 10.3390/ijms25158456] [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: 07/09/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer is a heterogeneous disease that arises as a multi-stage process involving multiple cell types. Patients diagnosed with the same clinical stage and pathological classification may have different prognoses and therapeutic responses due to alterations in molecular genetics. As an essential marker for the molecular subtyping of breast cancer, long non-coding RNAs (lncRNAs) play a crucial role in gene expression regulation, cell differentiation, and the maintenance of genomic stability. Here, we developed a modular framework for lncRNA identification and applied it to a breast cancer cohort to identify novel lncRNAs not previously annotated. To investigate the potential biological function, regulatory mechanisms, and clinical relevance of the novel lncRNAs, we elucidated the genomic and chromatin features of these lncRNAs, along with the associated protein-coding genes and putative enhancers involved in the breast cancer regulatory networks. Furthermore, we uncovered that the expression patterns of novel and annotated lncRNAs identified in breast cancer were related to the hormone response in the PAM50 subtyping criterion, as well as the immune response and progression states of breast cancer across different immune cells and immune checkpoint genes. Collectively, the comprehensive identification and functional analysis of lncRNAs revealed that these lncRNAs play an essential role in breast cancer by altering gene expression and participating in the regulatory networks, contributing to a better insight into breast cancer heterogeneity and potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
15
|
Li S, Garb BF, Qin T, Soppe S, Lopez E, Patil S, D’Silva NJ, Rozek LS, Sartor MA. Tumor Subtype Classification Tool for HPV-associated Head and Neck Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.601906. [PMID: 39026719 PMCID: PMC11257489 DOI: 10.1101/2024.07.05.601906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Importance Molecular subtypes of HPV-associated Head and Neck Squamous Cell Carcinoma (HNSCC), named IMU (immune strong) and KRT (highly keratinized), are well-recognized and have been shown to have distinct mechanisms of carcinogenesis, clinical outcomes, and potentially differing optimal treatment strategies. Currently, no standardized method exists to subtype a new HPV+ HNSCC tumor. Our paper introduces a machine learning-based classifier and webtool to reliably subtype HPV+ HNSCC tumors using the IMU/KRT paradigm and highlights the importance of subtype in HPV+ HNSCC. Objective To develop a robust, accurate machine learning-based classification tool that standardizes the process of subtyping HPV+ HNSCC, and to investigate the clinical, demographic, and molecular features associated with subtype in a meta-analysis of four patient cohorts. Data Sources We conducted RNA-seq on 67 HNSCC FFPE blocks from University of Michigan hospital. Combining this with three publicly available datasets, we utilized a total of 229 HPV+ HNSCC RNA-seq samples. All participants were HPV+ according to RNA expression. An ensemble machine learning approach with five algorithms and three different input training gene sets were developed, with final subtype determined by majority vote. Several additional steps were taken to ensure rigor and reproducibility throughout. Study Selection The classifier was trained and tested using 84 subtype-labeled HPV+ RNA-seq samples from two cohorts: University of Michigan (UM; n=18) and TCGA-HNC (n=66). The classifier robustness was validated with two independent cohorts: 83 samples from the HPV Virome Consortium and 62 additional samples from UM. We revealed 24 of 39 tested clinicodemographic and molecular variables significantly associated with subtype. Results The classifier achieved 100% accuracy in the test set. Validation on two additional cohorts demonstrated successful separation by known features of the subtypes. Investigating the relationship between subtype and 39 molecular and clinicodemographic variables revealed IMU is associated with epithelial-mesenchymal transition (p=2.25×10-4), various immune cell types, and lower radiation resistance (p=0.0050), while KRT is more highly keratinized (p=2.53×10-8), and more likely female than IMU (p=0.0082). Conclusions and Relevance This study provides a reliable classifier for subtyping HPV+ HNSCC tumors as either IMU or KRT based on bulk RNA-seq data, and additionally, improves our understanding of the HPV+ HNSCC subtypes.
Collapse
Affiliation(s)
- Shiting Li
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Bailey F. Garb
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Elizabeth Lopez
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Snehal Patil
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Nisha J. D’Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Laura S. Rozek
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Hablase R, Kyrou I, Randeva H, Karteris E, Chatterjee J. The "Road" to Malignant Transformation from Endometriosis to Endometriosis-Associated Ovarian Cancers (EAOCs): An mTOR-Centred Review. Cancers (Basel) 2024; 16:2160. [PMID: 38893278 PMCID: PMC11172073 DOI: 10.3390/cancers16112160] [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: 04/19/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Ovarian cancer is an umbrella term covering a number of distinct subtypes. Endometrioid and clear-cell ovarian carcinoma are endometriosis-associated ovarian cancers (EAOCs) frequently arising from ectopic endometrium in the ovary. The mechanistic target of rapamycin (mTOR) is a crucial regulator of cellular homeostasis and is dysregulated in both endometriosis and endometriosis-associated ovarian cancer, potentially favouring carcinogenesis across a spectrum from benign disease with cancer-like characteristics, through an atypical phase, to frank malignancy. In this review, we focus on mTOR dysregulation in endometriosis and EAOCs, investigating cancer driver gene mutations and their potential interaction with the mTOR pathway. Additionally, we explore the complex pathogenesis of transformation, considering environmental, hormonal, and epigenetic factors. We then discuss postmenopausal endometriosis pathogenesis and propensity for malignant transformation. Finally, we summarize the current advancements in mTOR-targeted therapeutics for endometriosis and EAOCs.
Collapse
Affiliation(s)
- Radwa Hablase
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust Hospital, Guildford GU2 7XX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK (H.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 1GB, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Harpal Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK (H.R.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
| | - Jayanta Chatterjee
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB83PH, UK; (R.H.); (E.K.)
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust Hospital, Guildford GU2 7XX, UK
| |
Collapse
|
17
|
Xie M, Kuang Y, Song M, Bao E. Subtype-MGTP: a cancer subtype identification framework based on multi-omics translation. Bioinformatics 2024; 40:btae360. [PMID: 38857453 PMCID: PMC11194476 DOI: 10.1093/bioinformatics/btae360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/30/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
MOTIVATION The identification of cancer subtypes plays a crucial role in cancer research and treatment. With the rapid development of high-throughput sequencing technologies, there has been an exponential accumulation of cancer multi-omics data. Integrating multi-omics data has emerged as a cost-effective and efficient strategy for cancer subtyping. While current methods primarily rely on genomics data, protein expression data offers a closer representation of phenotype. Therefore, integrating protein expression data holds promise for enhancing subtyping accuracy. However, the scarcity of protein expression data compared to genomics data presents a challenge in its direct incorporation into existing methods. Moreover, striking a balance between omics-specific learning and cross-omics learning remains a prevalent challenge in current multi-omics integration methods. RESULTS We introduce Subtype-MGTP, a novel cancer subtyping framework based on the translation of Multiple Genomics To Proteomics. Subtype-MGTP comprises two modules: a translation module, which leverages available protein data to translate multi-type genomics data into predicted protein expression data, and an improved deep subspace clustering module, which integrates contrastive learning to cluster the predicted protein data, yielding refined subtyping results. Extensive experiments conducted on benchmark datasets demonstrate that Subtype-MGTP outperforms nine state-of-the-art cancer subtyping methods. The interpretability of clustering results is further supported by the clinical and survival analysis. Subtype-MGTP also exhibits strong robustness against varying rates of missing protein data and demonstrates distinct advantages in integrating multi-omics data with imbalanced multi-omics data. AVAILABILITY AND IMPLEMENTATION The code and results are available at https://github.com/kybinn/Subtype-MGTP.
Collapse
Affiliation(s)
- Minzhu Xie
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
- Key Laboratory of Computing and Stochastic Mathematics (Ministry of Education), Changsha 410081, China
- College of Mathematics and Statistics, Hunan Normal University, Changsha 410081, China
| | - Yabin Kuang
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
| | - Mengyun Song
- College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China
| | - Ergude Bao
- School of Software Engineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
18
|
Zhou Z, Lin T, Chen S, Zhang G, Xu Y, Zou H, Zhou A, Zhang Y, Weng S, Han X, Liu Z. Omics-based molecular classifications empowering in precision oncology. Cell Oncol (Dordr) 2024; 47:759-777. [PMID: 38294647 DOI: 10.1007/s13402-023-00912-8] [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] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine. CONCLUSION This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
19
|
Xi Y, Zheng K, Deng F, Liu Y, Sun H, Zheng Y, Tong HHY, Ji Y, Zhang Y, Chen W, Zhang Y, Zou X, Hao J. Themis: advancing precision oncology through comprehensive molecular subtyping and optimization. Brief Bioinform 2024; 25:bbae261. [PMID: 38833322 PMCID: PMC11149663 DOI: 10.1093/bib/bbae261] [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: 01/30/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024] Open
Abstract
Recent advances in tumor molecular subtyping have revolutionized precision oncology, offering novel avenues for patient-specific treatment strategies. However, a comprehensive and independent comparison of these subtyping methodologies remains unexplored. This study introduces 'Themis' (Tumor HEterogeneity analysis on Molecular subtypIng System), an evaluation platform that encapsulates a few representative tumor molecular subtyping methods, including Stemness, Anoikis, Metabolism, and pathway-based classifications, utilizing 38 test datasets curated from The Cancer Genome Atlas (TCGA) and significant studies. Our self-designed quantitative analysis uncovers the relative strengths, limitations, and applicability of each method in different clinical contexts. Crucially, Themis serves as a vital tool in identifying the most appropriate subtyping methods for specific clinical scenarios. It also guides fine-tuning existing subtyping methods to achieve more accurate phenotype-associated results. To demonstrate the practical utility, we apply Themis to a breast cancer dataset, showcasing its efficacy in selecting the most suitable subtyping methods for personalized medicine in various clinical scenarios. This study bridges a crucial gap in cancer research and lays a foundation for future advancements in individualized cancer therapy and patient management.
Collapse
Affiliation(s)
- Yue Xi
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kun Zheng
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Fulan Deng
- School of Materials Science and Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yujun Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hourong Sun
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yingxia Zheng
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Henry H Y Tong
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China
| | - Yuan Ji
- Molecular Pathology center, Dept. Pathology, Zhongshan Hospital, Fudan University
| | - Yingchun Zhang
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
| | - Wantao Chen
- Ninth People's Hospital, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yiming Zhang
- Department of Reproductive Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
| | - Xin Zou
- National Engineering Center for Biochip at Shanghai, China
| | - Jie Hao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Liu X, Wu L, Wang L, Li Y. Identification and classification of glioma subtypes based on RNA-binding proteins. Comput Biol Med 2024; 174:108404. [PMID: 38582000 DOI: 10.1016/j.compbiomed.2024.108404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Glioma is a common and aggressive primary malignant cancer known for its high morbidity, mortality, and recurrence rates. Despite this, treatment options for glioma are currently restricted. The dysregulation of RBPs has been linked to the advancement of several types of cancer, but their precise role in glioma evolution is still not fully understood. This study sought to investigate how RBPs may impact the development and prognosis of glioma, with potential implications for prognosis and therapy. METHODS RNA-seq profiles of glioma and corresponding clinical data from the CGGA database were initially collected for analysis. Unsupervised clustering was utilized to identify crucial tumor subtypes in glioma development. Subsequent time-series analysis and MS model were employed to track the progression of these identified subtypes. RBPs playing a significant role in glioma progression were then pinpointed using WGCNA and Lasso Cox regression models. Functional analysis of these key RBP-related genes was conducted through GSEA. Additionally, the CIBERSORT algorithm was utilized to estimate immune infiltrating cells, while the STRING database was consulted to uncover potential mechanisms of the identified biomarkers. RESULTS Six tumor subgroups were identified and found to be highly homogeneous within each subgroup. The progression stages of these tumor subgroups were determined using time-series analysis and a MS model. Through WGCNA, Lasso Cox, and multivariate Cox regression analysis, it was confirmed that BCLAF1 is correlated with survival in glioma patients and is closely linked to glioma progression. Functional annotation suggests that BCLAF1 may impact glioma progression by influencing RNA splicing, which in turn affects the cell cycle, Wnt signaling pathway, and other cancer development pathways. CONCLUSIONS The study initially identified six subtypes of glioma progression and assessed their malignancy ranking. Furthermore, it was determined that BCLAF1 could serve as an RBP-related prognostic marker, offering significant implications for the clinical diagnosis and personalized treatment of glioma.
Collapse
Affiliation(s)
- Xudong Liu
- School of Medicine, Chongqing University, Chongqing, 400044, China; Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lei Wang
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China.
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
21
|
Zhou Q, Ye W, Yu X, Bao YJ. A pathway-based computational framework for identification of a new modal of multi-omics biomarkers and its application in esophageal cancer. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 247:108077. [PMID: 38382307 DOI: 10.1016/j.cmpb.2024.108077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/14/2024] [Accepted: 02/10/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND The pathway-based strategy has been recently proposed for identifying biomarkers with the advantages of higher biological interpretability and cross-data robustness than the conventional gene-based strategy. However, its utility in clinical applications has been limited due to the high computational complexity and ill-defined performance. OBJECTIVE The current study presents a machine learning-based computational framework using multi-omics data for identifying a new modal of biomarkers, called pathway-derived core biomarkers, which have the advantages of both gene-based and pathway-based biomarkers. METHODS Machine-learning methods and gene-pathway network were integrated to select the pathway-derived core biomarkers. Multiple machine-learning algorithms were used to construct and validate the diagnostic models of the biomarkers based on more than 1400 multi-omics clinical samples of esophageal squamous cell carcinoma (ESCC). RESULTS The results showed that the classifier models based on the new modal biomarkers achieved superior performance in the training datasets with an average AUC/accuracy of 0.98/0.95 and 0.89/0.81 for mRNAs and miRNA, respectively, higher than the currently known classifier models based on the conventional gene-based strategy and pathway-based strategy. In the testing cohorts, the AUC/accuracy increased by 6.1 %/7.3 % than the models based on the native gene-based biomarkers. The improved performance was further confirmed in independent validation cohorts. Specifically, the sensitivity/specificity increased by ∼3 % and the variance significantly decreased by ∼69 % compared with that of the native gene-based biomarkers. Importantly, the pathway-derived core biomarkers also recovered 45 % more previously reported biomarkers than the gene-based biomarkers and are more functionally relevant to the ESCC etiology (involved in 14 versus 7 pathways related with ESCC or other cancer), highlighting the cross-data robustness of this new modal of biomarkers via enhanced functional relevance. CONCLUSIONS The results demonstrated that the new modal of biomarkers not only have improved predicting performance and robustness, but also exhibit higher functional interpretability thus leading to the potential application in cancer diagnosis.
Collapse
Affiliation(s)
- Qi Zhou
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Weicai Ye
- School of Computer Science and Engineering, Guangdong Province Key Laboratory of Computational Science, and National Engineering Laboratory for Big Data Analysis and Application, Sun Yat-sen University, Guangzhou, China
| | - Xiaolan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China; Hubei Jiangxia Laboratory, Wuhan, China
| | - Yun-Juan Bao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China.
| |
Collapse
|
22
|
Ma T, Wang J. GraphPath: a graph attention model for molecular stratification with interpretability based on the pathway-pathway interaction network. Bioinformatics 2024; 40:btae165. [PMID: 38530778 PMCID: PMC11007237 DOI: 10.1093/bioinformatics/btae165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/22/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024] Open
Abstract
MOTIVATION Studying the molecular heterogeneity of cancer is essential for achieving personalized therapy. At the same time, understanding the biological processes that drive cancer development can lead to the identification of valuable therapeutic targets. Therefore, achieving accurate and interpretable clinical predictions requires paramount attention to thoroughly characterizing patients at both the molecular and biological pathway levels. RESULTS Here, we present GraphPath, a biological knowledge-driven graph neural network with multi-head self-attention mechanism that implements the pathway-pathway interaction network. We train GraphPath to classify the cancer status of patients with prostate cancer based on their multi-omics profiling. Experiment results show that our method outperforms P-NET and other baseline methods. Besides, two external cohorts are used to validate that the model can be generalized to unseen samples with adequate predictive performance. We reduce the dimensionality of latent pathway embeddings and visualize corresponding classes to further demonstrate the optimal performance of the model. Additionally, since GraphPath's predictions are interpretable, we identify target cancer-associated pathways that significantly contribute to the model's predictions. Such a robust and interpretable model has the potential to greatly enhance our understanding of cancer's biological mechanisms and accelerate the development of targeted therapies. AVAILABILITY AND IMPLEMENTATION https://github.com/amazingma/GraphPath.
Collapse
Affiliation(s)
- Teng Ma
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 41083, Hunan, China
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 41083, Hunan, China
| |
Collapse
|
23
|
Unger M, Kather JN. Deep learning in cancer genomics and histopathology. Genome Med 2024; 16:44. [PMID: 38539231 PMCID: PMC10976780 DOI: 10.1186/s13073-024-01315-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2024] [Indexed: 07/08/2024] Open
Abstract
Histopathology and genomic profiling are cornerstones of precision oncology and are routinely obtained for patients with cancer. Traditionally, histopathology slides are manually reviewed by highly trained pathologists. Genomic data, on the other hand, is evaluated by engineered computational pipelines. In both applications, the advent of modern artificial intelligence methods, specifically machine learning (ML) and deep learning (DL), have opened up a fundamentally new way of extracting actionable insights from raw data, which could augment and potentially replace some aspects of traditional evaluation workflows. In this review, we summarize current and emerging applications of DL in histopathology and genomics, including basic diagnostic as well as advanced prognostic tasks. Based on a growing body of evidence, we suggest that DL could be the groundwork for a new kind of workflow in oncology and cancer research. However, we also point out that DL models can have biases and other flaws that users in healthcare and research need to know about, and we propose ways to address them.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
24
|
Li A, Wang S, Nie J, Xiao S, Xie X, Zhang Y, Tong W, Yao G, Liu N, Dan F, Shu Z, Liu J, Liu Z, Yang F. USP3 promotes osteosarcoma progression via deubiquitinating EPHA2 and activating the PI3K/AKT signaling pathway. Cell Death Dis 2024; 15:235. [PMID: 38531846 PMCID: PMC10965993 DOI: 10.1038/s41419-024-06624-7] [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: 10/02/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Ubiquitin-specific protease 3 (USP3) plays an important role in the progression of various tumors. However, the role of USP3 in osteosarcoma (OS) remains poorly understood. The aim of this study was to explore the biological function of USP3 in OS and the underlying molecular mechanism. We found that OS had higher USP3 expression compared with that of normal bone tissue, and high expression of USP3 was associated with poor prognosis in patients with OS. Overexpression of USP3 significantly increased OS cell proliferation, migration, and invasion. Mechanistically, USP3 led to the activation of the PI3K/AKT signaling pathway in OS by binding to EPHA2 and then reducing its protein degradation. Notably, the truncation mutant USP3-F2 (159-520) interacted with EPHA2, and amino acid 203 was found to play an important role in this process. And knockdown of EPHA2 expression reversed the pro-tumour effects of USP3-upregulating. Thus, our study indicates the USP3/EPHA2 axis may be a novel potential target for OS treatment.
Collapse
Affiliation(s)
- Anan Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shijiang Wang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiangbo Nie
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shining Xiao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinsheng Xie
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu Zhang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weilai Tong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Geliang Yao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ning Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Dan
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhiguo Shu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiaming Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Feng Yang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
25
|
Altorki NK, Bhinder B, Borczuk AC, Elemento O, Mittal V, McGraw TE. A signature of enhanced proliferation associated with response and survival to anti-PD-L1 therapy in early-stage non-small cell lung cancer. Cell Rep Med 2024; 5:101438. [PMID: 38401548 PMCID: PMC10982989 DOI: 10.1016/j.xcrm.2024.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/20/2023] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
In early-stage non-small cell lung cancer, the combination of neoadjuvant anti-PD-L1 and subablative stereotactic body radiation therapy (SBRT) is associated with higher rates of major pathologic response compared to anti-PD-L1 alone. Here, we identify a 140-gene set, enriched in genes characteristic of highly proliferating cells, associated with response to the dual therapy. Analysis of on-treatment transcriptome data indicate roles for T and B cells in response. The 140-gene set is associated with disease-free survival when applied to the combined trial arms. This 140-gene set identifies a subclass of tumors in all 7 of The Cancer Genome Atlas tumor types examined. Worse survival is associated with the 140-gene signature in 5 of these tumor types. Collectively, our data support that this 140-gene set, discovered in association with response to combined anti-PD-L1 and SBRT, identifies a clinically aggressive subclass of solid tumors that may be more likely to respond to immunotherapies.
Collapse
Affiliation(s)
- Nasser K Altorki
- Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA.
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alain C Borczuk
- Department of Pathology and Laboratory Medicine, Northwell Health Cancer Institute, Northwell Health, Greenvale, NY 10042, USA
| | - Olivier Elemento
- Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Vivek Mittal
- Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA
| | - Timothy E McGraw
- Meyer Cancer Center, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine and New York Presbyterian Hospital, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
26
|
Lenoci D, Resteghini C, Serafini MS, Pistore F, Canevari S, Ma B, Cavalieri S, Alfieri S, Trama A, Licitra L, De Cecco L. Tumor molecular landscape of Epstein-Barr virus (EBV) related nasopharyngeal carcinoma in EBV-endemic and non-endemic areas: Implications for improving treatment modalities. Transl Res 2024; 265:1-16. [PMID: 37949350 DOI: 10.1016/j.trsl.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 10/14/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Epstein-Barr virus (EBV) related- nasopharyngeal carcinoma (NPC) is a squamous carcinoma of the nasopharyngeal mucosal lining. Endemic areas (EA) are east and Southeast Asia, were NPC was recorded with higher incidence and longer estimated survival than in non-endemic area (NEA) such as Europe, We analyzed the gene expression and microenvironment properties of NPC in both areas to identify molecular subtypes and assess biological and clinical correlates that might explain the differences in incidence and outcome between EA- and NEA-NPCs. Six EA-NPC transcriptomic datasets, including tumor and normal samples, were integrated in a meta-analysis to identify molecular subtypes using a ConsensusClusterPlus bioinformatic approach. Based on the biological/functional characterization of four identified clusters were identified: Cl1, Immune-active; Cl2, defense-response; Cl3, proliferation; Cl4, perineural-interaction/EBV-exhaustion. Kaplan-Meier survival analysis, applied to the single dataset with available disease-free survival indicated Cl3 as the cluster with the worst prognosis (P = 0.0476), confirmed when applying four previously published prognostic signatures. A Cl3 classifier signature was generated and its prognostic performance was confirmed (P = 0.0368) on a validation dataset. Prediction of treatment response suggested better responses to: radiotherapy and immune checkpoint inhibitors immune-active and defense-response clusters; chemotherapy proliferation cluster; cisplatin perineural-interaction/EBV-exhaustion cluster. RNA sequencing for gene expression profiling was performed on 50 NEA-NPC Italian samples. In the NEA cohort, Cl1, Cl2 and Cl3 were represented, while perineural-interaction/EBV-exhaustion was almost absent. The immune/biological characterization and treatment-response prediction analyses of NEA-NPC partially replicated the EA-NPC results. Well characterized EA- and NEA-NPC retrospective and prospective cohorts are needed to validate the obtained results and can help designing future clinical studies.
Collapse
Affiliation(s)
- Deborah Lenoci
- Molecular Mechanisms Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via GA. Amadeo, 42-20133 Milano, Italy
| | - Carlo Resteghini
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Mara S Serafini
- Molecular Mechanisms Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via GA. Amadeo, 42-20133 Milano, Italy
| | - Federico Pistore
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Silvana Canevari
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Brigette Ma
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Annalisa Trama
- Evaluative Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Loris De Cecco
- Molecular Mechanisms Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via GA. Amadeo, 42-20133 Milano, Italy.
| |
Collapse
|
27
|
Angeloni M, van Doeveren T, Lindner S, Volland P, Schmelmer J, Foersch S, Matek C, Stoehr R, Geppert CI, Heers H, Wach S, Taubert H, Sikic D, Wullich B, van Leenders GJLH, Zaburdaev V, Eckstein M, Hartmann A, Boormans JL, Ferrazzi F, Bahlinger V. A deep-learning workflow to predict upper tract urothelial carcinoma protein-based subtypes from H&E slides supporting the prioritization of patients for molecular testing. J Pathol Clin Res 2024; 10:e12369. [PMID: 38504364 PMCID: PMC10951050 DOI: 10.1002/2056-4538.12369] [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: 11/24/2023] [Revised: 02/08/2024] [Accepted: 02/26/2024] [Indexed: 03/21/2024]
Abstract
Upper tract urothelial carcinoma (UTUC) is a rare and aggressive, yet understudied, urothelial carcinoma (UC). The more frequent UC of the bladder comprises several molecular subtypes, associated with different targeted therapies and overlapping with protein-based subtypes. However, if and how these findings extend to UTUC remains unclear. Artificial intelligence-based approaches could help elucidate UTUC's biology and extend access to targeted treatments to a wider patient audience. Here, UTUC protein-based subtypes were identified, and a deep-learning (DL) workflow was developed to predict them directly from routine histopathological H&E slides. Protein-based subtypes in a retrospective cohort of 163 invasive tumors were assigned by hierarchical clustering of the immunohistochemical expression of three luminal (FOXA1, GATA3, and CK20) and three basal (CD44, CK5, and CK14) markers. Cluster analysis identified distinctive luminal (N = 80) and basal (N = 42) subtypes. The luminal subtype mostly included pushing, papillary tumors, whereas the basal subtype diffusely infiltrating, non-papillary tumors. DL model building relied on a transfer-learning approach by fine-tuning a pre-trained ResNet50. Classification performance was measured via three-fold repeated cross-validation. A mean area under the receiver operating characteristic curve of 0.83 (95% CI: 0.67-0.99), 0.8 (95% CI: 0.62-0.99), and 0.81 (95% CI: 0.65-0.96) was reached in the three repetitions. High-confidence DL-based predicted subtypes showed significant associations (p < 0.001) with morphological features, i.e. tumor type, histological subtypes, and infiltration type. Furthermore, a significant association was found with programmed cell death ligand 1 (PD-L1) combined positive score (p < 0.001) and FGFR3 mutational status (p = 0.002), with high-confidence basal predictions containing a higher proportion of PD-L1 positive samples and high-confidence luminal predictions a higher proportion of FGFR3-mutated samples. Testing of the DL model on an independent cohort highlighted the importance to accommodate histological subtypes. Taken together, our DL workflow can predict protein-based UTUC subtypes, associated with the presence of targetable alterations, directly from H&E slides.
Collapse
Affiliation(s)
- Miriam Angeloni
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Thomas van Doeveren
- Department of UrologyErasmus MC Urothelial Cancer Research GroupRotterdamThe Netherlands
| | - Sebastian Lindner
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Patrick Volland
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Jorina Schmelmer
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | | | - Christian Matek
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Robert Stoehr
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Carol I Geppert
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Hendrik Heers
- Department of UrologyPhilipps‐Universität MarburgMarburgGermany
| | - Sven Wach
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of Urology and Pediatric UrologyUniversity Hospital Erlangen, Friedrich‐Alexander Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Helge Taubert
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of Urology and Pediatric UrologyUniversity Hospital Erlangen, Friedrich‐Alexander Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Danijel Sikic
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of Urology and Pediatric UrologyUniversity Hospital Erlangen, Friedrich‐Alexander Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Bernd Wullich
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of Urology and Pediatric UrologyUniversity Hospital Erlangen, Friedrich‐Alexander Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Geert JLH van Leenders
- Department of PathologyErasmus MC Cancer Institute, University Medical CentreRotterdamthe Netherlands
| | - Vasily Zaburdaev
- Department of BiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Joost L Boormans
- Department of UrologyErasmus MC Urothelial Cancer Research GroupRotterdamThe Netherlands
| | - Fulvia Ferrazzi
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of NephropathologyInstitute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Veronika Bahlinger
- Institute of Pathology, University Hospital Erlangen‐Nürnberg, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Department of Pathology and NeuropathologyUniversity Hospital and Comprehensive Cancer Center TübingenTübingenGermany
| |
Collapse
|
28
|
Bouwman L, Sepodes B, Leufkens H, Torre C. Trends in orphan medicinal products approvals in the European Union between 2010-2022. Orphanet J Rare Dis 2024; 19:91. [PMID: 38413985 PMCID: PMC10900541 DOI: 10.1186/s13023-024-03095-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Over the last twenty years of orphan drug regulation in Europe, the regulatory framework has increased its complexity, with different regulatory paths and tools engineered to facilitate the innovation and accelerate approvals. Recently, the proposal of the new Pharmaceutical Legislation for the European Union, which will replace at least three Regulations and one Directive, was released and its new framework is raising many questions. The aim of this study was to present a characterisation of the Orphan Medicinal Products (OMPs) authorised by the European Commission (EC), between 2010 and 2022, looking into eighteen variables, contributing to the ongoing discussion on the proposal and implementation of the new Pharmaceutical Legislation proposed. METHODS Data of the OMPs identified and approved between 2010 and 2022 were extracted from the European Public Assessment Reports (EPARs) produced by the European Medicines Agency. Information regarding legal basis of the application, applicant, protocol assistance received, type of authorization, registration status, type of molecule, ATC code, therapeutic area, target age, disease prevalence, number of pivotal clinical trials supporting the application, clinical trial designs, respective efficacy endpoints and number of patients enrolled in the pivotal clinical trials were extracted. A descriptive statistical analysis was applied. RESULTS We identified 192 OMPs approved in the period between 2010 and 2022. 89% of the OMPs have legal basis of "full application". 86% of the sponsors received protocol assistance whereas 64% of the MAA benefited from the accelerated assessment. 53% of the active substances are small molecules; about 1 in 5 molecules are repurposed. 40% of the OMPs have oncological therapeutic indications and 56% of the OMPs are intended to treat only adults. 71% of the products were approved based on a single pivotal trial. CONCLUSIONS This analysis of OMPs approved between 2010 and 2022 shows that a shift has occurred in the rare disease medicine development space. Through the period studied we observe an increase of non-small molecules approved, accelerated assessment received and non-standard MA's granted.
Collapse
Affiliation(s)
- Luísa Bouwman
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal.
| | - Bruno Sepodes
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal
| | - Hubert Leufkens
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Carla Torre
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Lisbon, Portugal
| |
Collapse
|
29
|
Essouma M. Autoimmune inflammatory myopathy biomarkers. Clin Chim Acta 2024; 553:117742. [PMID: 38176522 DOI: 10.1016/j.cca.2023.117742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The autoimmune inflammatory myopathy disease spectrum, commonly known as myositis, is a group of systemic diseases that mainly affect the muscles, skin and lungs. Biomarker assessment helps in understanding disease mechanisms, allowing for the implementation of precise strategies in the classification, diagnosis, and management of these diseases. This review examines the pathogenic mechanisms and highlights current data on blood and tissue biomarkers of autoimmune inflammatory myopathies.
Collapse
Affiliation(s)
- Mickael Essouma
- Network of Immunity in Infections, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Cameroon
| |
Collapse
|
30
|
Hasan MN, Rahman MM, Husna AA, Nozaki N, Yamato O, Miura N. YRNA and tRNA fragments can differentiate benign from malignant canine mammary gland tumors. Biochem Biophys Res Commun 2024; 691:149336. [PMID: 38039834 DOI: 10.1016/j.bbrc.2023.149336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Mammary gland tumors (MGT) are the most common tumors in sexually intact female dogs. The functional regulation of miRNAs, a type of noncoding RNAs (ncRNAs), in canine MGT has been extensively investigated. However, the expression of other ncRNAs, such as YRNAs and transfer RNA-derived fragments (tRFs) in canine MGT is unknown. We investigated ncRNAs other than miRNAs from our small RNA project (PRJNA716131) in different canine MGT histologic subtypes. This study included benign tumors (benign mixed tumor, complex adenoma) and malignant tumors (carcinoma in benign tumor and carcinoma with metastasis) samples. Aberrantly expressed ncRNAs were examined by comparisons among MGT subtypes. The relative expression trends were validated in canine MGT tissues, plasma, extracellular vesicles, and MGT cell lines using quantitative reverse transcription PCR. Three aberrantly expressed ncRNAs were identified by comparisons among MGT subtypes. YRNA and tRNA-Gly-GCC distinguished benign mixed tumor from other MGT histologic subtypes, while tRNA-Val differentiated complex adenoma, carcinoma in benign tumors, and carcinoma with metastasis. The ROC curve of the three ncRNAs showed they might be potential biomarkers to discriminate malignant from benign MGT. YRNA and tRFs expression levels were decreased in metastatic compared with primary canine MGT cell lines. To the best of our knowledge, this is the first investigation of YRNA and tRFs in canine MGT. The three identified ncRNAs may be biomarkers for differentiating MGT histologic subtypes. Suggested Reviewers: Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporatio.
Collapse
Affiliation(s)
- Md Nazmul Hasan
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan; Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan.
| | - Md Mahfuzur Rahman
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Al Asmaul Husna
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan.
| | - Nobuhiro Nozaki
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan.
| | - Osamu Yamato
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan; Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan.
| | - Naoki Miura
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan; Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24, Korimoto, Kagoshima, 890-0065, Japan.
| |
Collapse
|
31
|
Martín-García D, Téllez T, Redondo M, García-Aranda M. The use of SP/Neurokinin-1 as a Therapeutic Target in Colon and Rectal Cancer. Curr Med Chem 2024; 31:6487-6509. [PMID: 37861026 DOI: 10.2174/0109298673261625230924114406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 10/21/2023]
Abstract
Different studies have highlighted the role of Substance P / Neurokinin 1 Receptor (SP/NK-1R) axis in multiple hallmarks of cancer including cell transformation, proliferation, and migration as well as angiogenesis and metastasis of a wide range of solid tumors including colorectal cancer. Until now, the selective high-affinity antagonist of human SP/NK1-R aprepitant (Emend) has been authorized by the Food and Drug Administration as a low dosage medication to manage and treat chemotherapy-induced nausea. However, increasing evidence in recent years support the potential utility of high doses of aprepitant as an antitumor agent and thus, opening the possibility to the pharmacological repositioning of SP/NK1-R antagonists as an adjuvant therapy to conventional cancer treatments. In this review, we summarize current knowledge on the molecular basis of colorectal cancer as well as the pathophysiological importance of SP/NK1-R and the potential utility of SP/NK-1R axis as a therapeutic target in this malignancy.
Collapse
Affiliation(s)
| | - Teresa Téllez
- Surgical Specialties, Biochemistry and Immunology, University of Malaga, Spain
| | - Maximino Redondo
- Surgical Specialties, Biochemistry and Immunology, University of Malaga, Spain
| | - Marilina García-Aranda
- Surgical Specialties, Biochemistry and Immunology, University of Malaga, Spain
- Research and Innovation Unit, Hospital Costa del Sol, 29602 Marbella, Spain
| |
Collapse
|
32
|
Zhang JZ, Wang C. A comparative study of clustering methods on gene expression data for lung cancer prognosis. BMC Res Notes 2023; 16:319. [PMID: 37941025 PMCID: PMC10630994 DOI: 10.1186/s13104-023-06604-8] [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: 05/07/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
Lung cancer subtyping based on gene expression data is important for identifying patient subgroups with differing survival prognosis to facilitate customized treatment strategies for each subtype of patients. Unsupervised clustering methods are the traditional approach for clustering patients into subtypes. However, since those methods cluster patients based only on gene expression data, the resulting clusters may not always be relevant to the survival outcome of interest. In recent years, semi-supervised and supervised methods have been proposed, which leverage the survival outcome data to identify clusters more relevant to survival prognosis. This paper aims to compare the performance of different clustering methods for identifying clinically prognostic lung cancer subtypes based on two lung adenocarcinoma datasets. For each method, we clustered patients into two clusters and assessed the difference in patient survival time between clusters. Unsupervised methods were found to have large logrank p-values and no significant results in most cases. Semi-supervised and supervised methods had improved performance over unsupervised methods and very significant p-values. These results indicate that unsupervised methods are not capable of identifying clusters with significant differences in survival prognosis in most cases, while supervised and semi-supervised methods can better cluster patients into clinically useful subtypes.
Collapse
Affiliation(s)
- Jason Z Zhang
- Wake Forest University, Winston-Salem, NC, United States of America
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
33
|
Li K, Sun L, Wang Y, Cen Y, Zhao J, Liao Q, Wu W, Sun J, Zhou M. Single-cell characterization of macrophages in uveal melanoma uncovers transcriptionally heterogeneous subsets conferring poor prognosis and aggressive behavior. Exp Mol Med 2023; 55:2433-2444. [PMID: 37907747 PMCID: PMC10689813 DOI: 10.1038/s12276-023-01115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
Uveal melanoma (UM) is the most frequent primary intraocular malignancy with high metastatic potential and poor prognosis. Macrophages represent one of the most abundant infiltrating immune cells with diverse functions in cancers. However, the cellular heterogeneity and functional diversity of macrophages in UM remain largely unexplored. In this study, we analyzed 63,264 single-cell transcriptomes from 11 UM patients and identified four transcriptionally distinct macrophage subsets (termed MΦ-C1 to MΦ-C4). Among them, we found that MΦ-C4 exhibited relatively low expression of both M1 and M2 signature genes, loss of inflammatory pathways and antigen presentation, instead demonstrating enhanced signaling for proliferation, mitochondrial functions and metabolism. We quantified the infiltration abundance of MΦ-C4 from single-cell and bulk transcriptomes across five cohorts and found that increased MΦ-C4 infiltration was relevant to aggressive behaviors and may serve as an independent prognostic indicator for poor outcomes. We propose a novel subtyping scheme based on macrophages by integrating the transcriptional signatures of MΦ-C4 and machine learning to stratify patients into MΦ-C4-enriched or MΦ-C4-depleted subtypes. These two subtypes showed significantly different clinical outcomes and were validated through bulk RNA sequencing and immunofluorescence assays in both public multicenter cohorts and our in-house cohort. Following further translational investigation, our findings highlight a potential therapeutic strategy of targeting macrophage subsets to control metastatic disease and consistently improve the outcome of patients with UM.
Collapse
Affiliation(s)
- Ke Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Lanfang Sun
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Yanan Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Yixin Cen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Jingting Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Qianling Liao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
| | - Jie Sun
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
| | - Meng Zhou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, 325027, Wenzhou, China.
| |
Collapse
|
34
|
Bao J, Yu Y. Identification of a prognostic evaluator from glutamine metabolic heterogeneity studies within and between tissues in hepatocellular carcinoma. Front Pharmacol 2023; 14:1241677. [PMID: 37954858 PMCID: PMC10637396 DOI: 10.3389/fphar.2023.1241677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/21/2023] [Indexed: 11/14/2023] Open
Abstract
Background: The liver is the major metabolic organ of the human body, and abnormal metabolism is the main factor influencing hepatocellular carcinoma (HCC). This study was designed to determine the effect of glutamine metabolism on HCC heterogeneity and to develop a prognostic evaluator based on the heterogeneity study of glutamine metabolism within HCC tumors and between tissues. Methods: Single-cell transcriptome data were extracted from the GSE149614 dataset and processed using the Seurat package in R for quality control of these data. HCC subtypes in the Cancer Genome Atlas and the GSE14520 dataset were identified via consensus clustering based on glutamine family amino acid metabolism (GFAAM) process genes. The machine learning algorithms gradient boosting machine, support vector machine, random forest, eXtreme gradient boosting, decision trees, and least absolute shrinkage and selection operator were utilized to develop the prognosis model of differentially expressed genes among the molecular gene subtypes. Results: The samples in the GSE149614 dataset included 10 cell types, and there was no significant difference in the GFAAM pathway. HCC was classified into three molecular subtypes according to GFAAM process genes, showing molecular heterogeneity in prognosis, clinicopathological features, and immune cell infiltration. C1 showed the worst survival rate and the highest immune score and immune cell infiltration. A six-gene model for prognostic and immunotherapy responses was constructed among subtypes, and the calculated high-risk score was significantly correlated with poor prognosis, high immune abundance, and a low response rate of immunotherapy in HCC. Conclusion: Our discovery of GFAAM-associated marker genes may help to further decipher the role in HCC occurrence and progression. In particular, this six-gene prognostic model may serve as a predictor of treatment and prognosis in HCC patients.
Collapse
Affiliation(s)
- Jie Bao
- Digestive System Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Wei L, Niraula D, Gates EDH, Fu J, Luo Y, Nyflot MJ, Bowen SR, El Naqa IM, Cui S. Artificial intelligence (AI) and machine learning (ML) in precision oncology: a review on enhancing discoverability through multiomics integration. Br J Radiol 2023; 96:20230211. [PMID: 37660402 PMCID: PMC10546458 DOI: 10.1259/bjr.20230211] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 09/05/2023] Open
Abstract
Multiomics data including imaging radiomics and various types of molecular biomarkers have been increasingly investigated for better diagnosis and therapy in the era of precision oncology. Artificial intelligence (AI) including machine learning (ML) and deep learning (DL) techniques combined with the exponential growth of multiomics data may have great potential to revolutionize cancer subtyping, risk stratification, prognostication, prediction and clinical decision-making. In this article, we first present different categories of multiomics data and their roles in diagnosis and therapy. Second, AI-based data fusion methods and modeling methods as well as different validation schemes are illustrated. Third, the applications and examples of multiomics research in oncology are demonstrated. Finally, the challenges regarding the heterogeneity data set, availability of omics data, and validation of the research are discussed. The transition of multiomics research to real clinics still requires consistent efforts in standardizing omics data collection and analysis, building computational infrastructure for data sharing and storing, developing advanced methods to improve data fusion and interpretability, and ultimately, conducting large-scale prospective clinical trials to fill the gap between study findings and clinical benefits.
Collapse
Affiliation(s)
- Lise Wei
- Department of Radiation Oncology, University of Michigan, Michigan, United States
| | - Dipesh Niraula
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Evan D. H. Gates
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Jie Fu
- Department of Radiation Oncology, Stanford University, Stanford, California, United States
| | - Yi Luo
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Matthew J. Nyflot
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Stephen R. Bowen
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Issam M El Naqa
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Sunan Cui
- Department of Radiation Oncology, University of Washington, Washington, United States
| |
Collapse
|
36
|
Bond A, Mccay K, Lal S. Artificial intelligence & clinical nutrition: What the future might have in store. Clin Nutr ESPEN 2023; 57:542-549. [PMID: 37739704 DOI: 10.1016/j.clnesp.2023.07.082] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/02/2023] [Accepted: 07/17/2023] [Indexed: 09/24/2023]
Abstract
Artificial Intelligence (AI) is a rapidly emerging technology in healthcare that has the potential to revolutionise clinical nutrition. AI can assist in analysing complex data, interpreting medical images, and providing personalised nutrition interventions for patients. Clinical nutrition is a critical aspect of patient care, and AI can help clinicians make more informed decisions regarding patients' nutritional requirements, disease prevention, and management. AI algorithms can analyse large datasets to identify novel associations between diet and disease outcomes, enabling clinicians to make evidence-based nutritional recommendations. AI-powered devices and applications can also assist in tracking dietary intake, providing feedback, and motivating patients towards healthier food choices. However, the adoption of AI in clinical nutrition raises several ethical and regulatory concerns, such as data privacy and bias. Further research is needed to assess the clinical effectiveness and safety of AI-powered nutrition interventions. In conclusion, AI has the potential to transform clinical nutrition, but its integration into clinical practice should be carefully monitored to ensure patient safety and benefit. This article discusses the current and future applications of AI in clinical nutrition and highlights its potential benefits.
Collapse
Affiliation(s)
- Ashley Bond
- Intestinal Failure Unit, Salford Royal Foundation Trust, UK; University of Manchester, Manchester, UK.
| | - Kevin Mccay
- Manchester Metropolitan University, Manchester, UK; Northern Care Alliance NHS Foundation Trust, Salford Royal Hospital, Salford, UK
| | - Simon Lal
- Intestinal Failure Unit, Salford Royal Foundation Trust, UK; University of Manchester, Manchester, UK
| |
Collapse
|
37
|
Zhang J, Liu L, Wei X, Zhao C, Li S, Li J, Le TD. Pan-cancer characterization of ncRNA synergistic competition uncovers potential carcinogenic biomarkers. PLoS Comput Biol 2023; 19:e1011308. [PMID: 37812646 PMCID: PMC10586676 DOI: 10.1371/journal.pcbi.1011308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/19/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Non-coding RNAs (ncRNAs) act as important modulators of gene expression and they have been confirmed to play critical roles in the physiology and development of malignant tumors. Understanding the synergism of multiple ncRNAs in competing endogenous RNA (ceRNA) regulation can provide important insights into the mechanisms of malignant tumors caused by ncRNA regulation. In this work, we present a framework, SCOM, for identifying ncRNA synergistic competition. We systematically construct the landscape of ncRNA synergistic competition across 31 malignant tumors, and reveal that malignant tumors tend to share hub ncRNAs rather than the ncRNA interactions involved in the synergistic competition. In addition, the synergistic competition ncRNAs (i.e. ncRNAs involved in the synergistic competition) are likely to be involved in drug resistance, contribute to distinguishing molecular subtypes of malignant tumors, and participate in immune regulation. Furthermore, SCOM can help to infer ncRNA synergistic competition across malignant tumors and uncover potential diagnostic and prognostic biomarkers of malignant tumors. Altogether, the SCOM framework (https://github.com/zhangjunpeng411/SCOM/) and the resulting web-based database SCOMdb (https://comblab.cn/SCOMdb/) serve as a useful resource for exploring ncRNA regulation and to accelerate the identification of carcinogenic biomarkers.
Collapse
Affiliation(s)
- Junpeng Zhang
- School of Engineering, Dali University, Dali, Yunnan, People’s Republic of China
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Xuemei Wei
- School of Engineering, Dali University, Dali, Yunnan, People’s Republic of China
| | - Chunwen Zhao
- School of Engineering, Dali University, Dali, Yunnan, People’s Republic of China
| | - Sijing Li
- School of Engineering, Dali University, Dali, Yunnan, People’s Republic of China
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, South Australia, Australia
| |
Collapse
|
38
|
Nourbakhsh M, Saksager A, Tom N, Chen XS, Colaprico A, Olsen C, Tiberti M, Papaleo E. A workflow to study mechanistic indicators for driver gene prediction with Moonlight. Brief Bioinform 2023; 24:bbad274. [PMID: 37551622 PMCID: PMC10516357 DOI: 10.1093/bib/bbad274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 08/09/2023] Open
Abstract
Prediction of driver genes (tumor suppressors and oncogenes) is an essential step in understanding cancer development and discovering potential novel treatments. We recently proposed Moonlight as a bioinformatics framework to predict driver genes and analyze them in a system-biology-oriented manner based on -omics integration. Moonlight uses gene expression as a primary data source and combines it with patterns related to cancer hallmarks and regulatory networks to identify oncogenic mediators. Once the oncogenic mediators are identified, it is important to include extra levels of evidence, called mechanistic indicators, to identify driver genes and to link the observed gene expression changes to the underlying alteration that promotes them. Such a mechanistic indicator could be for example a mutation in the regulatory regions for the candidate gene. Here, we developed new functionalities and released Moonlight2 to provide the user with a mutation-based mechanistic indicator as a second layer of evidence. These functionalities analyze mutations in a cancer cohort to classify them into driver and passenger mutations. Those oncogenic mediators with at least one driver mutation are retained as the final set of driver genes. We applied Moonlight2 to the basal-like breast cancer subtype, lung adenocarcinoma and thyroid carcinoma using data from The Cancer Genome Atlas. For example, in basal-like breast cancer, we found four oncogenes (COPZ2, SF3B4, KRTCAP2 and POLR2J) and nine tumor suppressor genes (KIR2DL4, KIF26B, ARL15, ARHGAP25, EMCN, GMFG, TPK1, NR5A2 and TEK) containing a driver mutation in their promoter region, possibly explaining their deregulation. Moonlight2R is available at https://github.com/ELELAB/Moonlight2R.
Collapse
Affiliation(s)
- Mona Nourbakhsh
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
| | - Astrid Saksager
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
| | - Nikola Tom
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
| | - Xi Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Catharina Olsen
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Reproduction and Genetics
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), VUB-ULB, Brussels 1090, Belgium
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels 1050, Belgium
| | - Matteo Tiberti
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Elena Papaleo
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| |
Collapse
|
39
|
Chen W, Wang H, Liang C. Deep multi-view contrastive learning for cancer subtype identification. Brief Bioinform 2023; 24:bbad282. [PMID: 37539822 DOI: 10.1093/bib/bbad282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
Cancer heterogeneity has posed great challenges in exploring precise therapeutic strategies for cancer treatment. The identification of cancer subtypes aims to detect patients with distinct molecular profiles and thus could provide new clues on effective clinical therapies. While great efforts have been made, it remains challenging to develop powerful computational methods that can efficiently integrate multi-omics datasets for the task. In this paper, we propose a novel self-supervised learning model called Deep Multi-view Contrastive Learning (DMCL) for cancer subtype identification. Specifically, by incorporating the reconstruction loss, contrastive loss and clustering loss into a unified framework, our model simultaneously encodes the sample discriminative information into the extracted feature representations and well preserves the sample cluster structures in the embedded space. Moreover, DMCL is an end-to-end framework where the cancer subtypes could be directly obtained from the model outputs. We compare DMCL with eight alternatives ranging from classic cancer subtype identification methods to recently developed state-of-the-art systems on 10 widely used cancer multi-omics datasets as well as an integrated dataset, and the experimental results validate the superior performance of our method. We further conduct a case study on liver cancer and the analysis results indicate that different subtypes might have different responses to the selected chemotherapeutic drugs.
Collapse
Affiliation(s)
- Wenlan Chen
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250358, China
| | - Hong Wang
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250358, China
| | - Cheng Liang
- School of Information Science and Engineering, Shandong Normal University, Jinan, 250358, China
| |
Collapse
|
40
|
Ji Y, Dutta P, Davuluri R. Deep multi-omics integration by learning correlation-maximizing representation identifies prognostically stratified cancer subtypes. BIOINFORMATICS ADVANCES 2023; 3:vbad075. [PMID: 37424943 PMCID: PMC10328436 DOI: 10.1093/bioadv/vbad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/08/2023] [Indexed: 07/11/2023]
Abstract
Motivation Molecular subtyping by integrative modeling of multi-omics and clinical data can help the identification of robust and clinically actionable disease subgroups; an essential step in developing precision medicine approaches. Results We developed a novel outcome-guided molecular subgrouping framework, called Deep Multi-Omics Integrative Subtyping by Maximizing Correlation (DeepMOIS-MC), for integrative learning from multi-omics data by maximizing correlation between all input -omics views. DeepMOIS-MC consists of two parts: clustering and classification. In the clustering part, the preprocessed high-dimensional multi-omics views are input into two-layer fully connected neural networks. The outputs of individual networks are subjected to Generalized Canonical Correlation Analysis loss to learn the shared representation. Next, the learned representation is filtered by a regression model to select features that are related to a covariate clinical variable, for example, a survival/outcome. The filtered features are used for clustering to determine the optimal cluster assignments. In the classification stage, the original feature matrix of one of the -omics view is scaled and discretized based on equal frequency binning, and then subjected to feature selection using RandomForest. Using these selected features, classification models (for example, XGBoost model) are built to predict the molecular subgroups that were identified at clustering stage. We applied DeepMOIS-MC on lung and liver cancers, using TCGA datasets. In comparative analysis, we found that DeepMOIS-MC outperformed traditional approaches in patient stratification. Finally, we validated the robustness and generalizability of the classification models on independent datasets. We anticipate that the DeepMOIS-MC can be adopted to many multi-omics integrative analyses tasks. Availability and implementation Source codes for PyTorch implementation of DGCCA and other DeepMOIS-MC modules are available at GitHub (https://github.com/duttaprat/DeepMOIS-MC). Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Yanrong Ji
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pratik Dutta
- Department of Biomedical Informatics, Stony Brook Cancer Center, Stony Brook Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ramana Davuluri
- Department of Biomedical Informatics, Stony Brook Cancer Center, Stony Brook Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
41
|
Shetty KS, Jose A, Bani M, Vinod PK. Network diffusion-based approach for survival prediction and identification of biomarkers using multi-omics data of papillary renal cell carcinoma. Mol Genet Genomics 2023; 298:871-882. [PMID: 37093328 DOI: 10.1007/s00438-023-02022-4] [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: 09/08/2022] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
Identification of cancer subtypes based on molecular knowledge is crucial for improving the patient diagnosis, prognosis, and treatment. In this work, we integrated copy number variations (CNVs) and transcriptomic data of Kidney Papillary Renal Cell Carcinoma (KIRP) using a network diffusion strategy to stratify cancers into clinically and biologically relevant subtypes. We constructed GeneNet, a KIRP specific gene expression network from RNA-seq data. The copy number variation data was projected onto GeneNet and propagated on the network for clustering. We identified robust subtypes that are biologically informative and significantly associated with patient survival, tumor stage and clinical subtypes of KIRP. We performed a Singular Value Decomposition (SVD) analysis of KIRP subtypes, which revealed the genes/silent players related to poor survival. A differential gene expression analysis between subtypes showed that genes related to immune, extracellular matrix organization, and genomic instability are upregulated in the poor survival group. Overall, the network-based approach revealed the molecular subtypes of KIRP and captured the relationship between gene expression and CNVs. This framework can be further expanded to integrate other omics data.
Collapse
Affiliation(s)
- Keerthi S Shetty
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - Aswin Jose
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - Mihir Bani
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India
| | - P K Vinod
- Center for Computational Natural Sciences and Bioinformatics, IIIT Hyderabad, Hyderabad, 500032, India.
| |
Collapse
|
42
|
Yang M, Matan-Lithwick S, Wang Y, De Jager PL, Bennett DA, Felsky D. Multi-omic integration via similarity network fusion to detect molecular subtypes of ageing. Brain Commun 2023; 5:fcad110. [PMID: 37082508 PMCID: PMC10110975 DOI: 10.1093/braincomms/fcad110] [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: 01/23/2023] [Revised: 02/17/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Molecular subtyping of brain tissue provides insights into the heterogeneity of common neurodegenerative conditions, such as Alzheimer's disease. However, existing subtyping studies have mostly focused on single data modalities and only those individuals with severe cognitive impairment. To address these gaps, we applied similarity network fusion, a method capable of integrating multiple high-dimensional multi-omic data modalities simultaneously, to an elderly sample spanning the full spectrum of cognitive ageing trajectories. We analyzed human frontal cortex brain samples characterized by five omic modalities: bulk RNA sequencing (18 629 genes), DNA methylation (53 932 CpG sites), histone acetylation (26 384 peaks), proteomics (7737 proteins) and metabolomics (654 metabolites). Similarity network fusion followed by spectral clustering was used for subtype detection, and subtype numbers were determined by Eigen-gap and rotation cost statistics. Normalized mutual information determined the relative contribution of each modality to the fused network. Subtypes were characterized by associations with 13 age-related neuropathologies and cognitive decline. Fusion of all five data modalities (n = 111) yielded two subtypes (n S1 = 53, n S2 = 58), which were nominally associated with diffuse amyloid plaques; however, this effect was not significant after correction for multiple testing. Histone acetylation (normalized mutual information = 0.38), DNA methylation (normalized mutual information = 0.18) and RNA abundance (normalized mutual information = 0.15) contributed most strongly to this network. Secondary analysis integrating only these three modalities in a larger subsample (n = 513) indicated support for both three- and five-subtype solutions, which had significant overlap, but showed varying degrees of internal stability and external validity. One subtype showed marked cognitive decline, which remained significant even after correcting for tests across both three- and five-subtype solutions (p Bonf = 5.9 × 10-3). Comparison to single-modality subtypes demonstrated that the three-modal subtypes were able to uniquely capture cognitive variability. Comprehensive sensitivity analyses explored influences of sample size and cluster number parameters. We identified highly integrative molecular subtypes of ageing derived from multiple high dimensional, multi-omic data modalities simultaneously. Fusing RNA abundance, DNA methylation, and histone acetylation measures generated subtypes that were associated with cognitive decline. This work highlights the potential value and challenges of multi-omic integration in unsupervised subtyping of post-mortem brain.
Collapse
Affiliation(s)
- Mu Yang
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Stuart Matan-Lithwick
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Philip L De Jager
- The Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10033, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Daniel Felsky
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- The Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
43
|
Rather AA, Chachoo MA. Robust correlation estimation and UMAP assisted topological analysis of omics data for disease subtyping. Comput Biol Med 2023; 155:106640. [PMID: 36774889 DOI: 10.1016/j.compbiomed.2023.106640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/08/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
Deciphering information hidden in the gene expression assays for identifying disease subtypes has significant importance in precision medicine. However, computational limitations thwart this process due to the intricacy of the biological networks and the curse of dimensionality of gene expression data. Therefore, clustering in such scenarios often becomes the first choice of exploratory data analysis to identify natural structures and intrinsic patterns in the data. However, sparse and high dimensional nature of omics data prevents conventional clustering algorithms to discover subtypes that are clinically relevant and statistically significant. Hence, non-linear dimensionality reduction techniques coupled with clustering in such scenarios often becomes imperative to improve the clustering results. In this study, we present a robust pipeline to discover disease subtypes with clinical relevance. Specifically, we focus on discovering patient sub-groups that have a residual life patterns remarkably different from other sub-groups. This is significant because by refining prognosis, subtyping can reduce uncertainty in approximating patients expected outcome. The methodology present is based on robust correlation estimation, UMAP- a non-linear dimensionality reduction method and mapper- a tool from topology. Notably, we suggest a method for improving the robustness of the correlation matrix of gene expression data for improving the clustering results. The performance of the model is evaluated by applying to five cancer datasets obtained through TCGA and comparisons are performed with some state of the art methods of NEMO, RSC-OTRI and SNF with regard to log-rank test and Restricted Life Expectancy Difference. For example in GBM dataset, the minimum separation for any two discovered subtypes is 221 days which is significantly higher than the other methodologies. We also compared the results without using the robust correlation based estimate and observed that robust correlation improves separability between survival curves significantly. From the results we infer that our methodology performs better compared to other methodologies with regard to separating survival curves of patient sub-groups despite using single omics profiles of patients compared to multiple omics profiles of SNF and NEMO. Pathway over-representation analysis is performed on the final clustering results to investigate the biological underpinnings characterizing each subtype.
Collapse
Affiliation(s)
- Arif Ahmad Rather
- Department of Computer Sciences, University of Kashmir, Srinagar, JK, India.
| | | |
Collapse
|
44
|
Zhao L, Cai Y, Cho WC. Editorial: Genomics of immunoregulation and inflammatory responses in the tumor microenvironment. Front Genet 2023; 14:1136342. [PMID: 36845401 PMCID: PMC9950731 DOI: 10.3389/fgene.2023.1136342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Affiliation(s)
- Lan Zhao
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Yunpeng Cai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| |
Collapse
|
45
|
Xie C, Hu J, Hu Q, Jiang L, Chen W. Classification of the mitochondrial ribosomal protein-associated molecular subtypes and identified a serological diagnostic biomarker in hepatocellular carcinoma. Front Surg 2023; 9:1062659. [PMID: 36684217 PMCID: PMC9853988 DOI: 10.3389/fsurg.2022.1062659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Purpose The objective of this study was to sort out innovative molecular subtypes associated with mitochondrial ribosomal proteins (MRPs) to predict clinical therapy response and determine the presence of circulating markers in hepatocellular carcinoma (HCC) patients. Methods Using an unsupervised clustering method, we categorized the relative molecular subtypes of MRPs in HCC patients. The prognosis, biological properties, immune checkpoint inhibitor and chemotherapy response of the patients were clarified. A signature and nomogram were developed to evaluate the prognosis. Enzyme-linked immunosorbent assay (ELISA) measured serum mitochondrial ribosomal protein L9 (MRPL9) levels in liver disease patients and normal individuals. Receiver operating characteristic (ROC) curves were conducted to calculate the diagnostic effect. The Cell Counting Kit 8 was carried out to examine cell proliferation, and flow cytometry was used to investigate the cell cycle. Transwell assay was applied to investigate the potential of cell migration and invasion. Western blot detected corresponding changes of biological markers. Results Participants were classified into two subtypes according to MRPs expression levels, which were characterized by different prognoses, biological features, and marked differences in response to chemotherapy and immune checkpoint inhibitors. Serum MRPL9 was significantly higher in HCC patients than in normal individuals and the benign liver disease group. ROC curve analysis showed that MRPL9 was superior to AFP and Ferritin in differentiating HCC from healthy and benign patients, or alone. Overexpressed MRPL9 could enhance aggressiveness and facilitate the G1/S progression in HCC cells. Conclusion We constructed novel molecular subtypes based on MRPs expression in HCC patients, which provided valuable strategies for the prediction of prognosis and clinical personalized treatment. MRPL9 might act as a reliable circulating diagnostic biomarker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
| | | | | | | | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
Shi X, Liang C, Wang H. Multiview Robust Graph-Based Clustering for Cancer Subtype Identification. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:544-556. [PMID: 35044919 DOI: 10.1109/tcbb.2022.3143897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cancer subtype identification is to classify cancer into groups according to their molecular characteristics and clinical manifestations and is the basis for more personalized diagnosis and therapy. Public datasets such as The Cancer Genome Atlas (TCGA) have collected a massive number of multi-omics data. The accumulation of these datasets provides unprecedented opportunities to study the mechanism of cancers and further identify cancer subtypes at a comprehensive level. In this paper, we propose a multi-view robust graph-based clustering (MRGC) method to effectively identify cancer subtypes. Our method first learns robust latent representations from the raw omics data to alleviate the influences of the noise, where a set of similarity matrices are then adaptively learned based on these new representations. Finally, a global similarity graph is obtained by exploiting the consensus structure from the graphs. As a result, the three parts in our method can reinforce each other in a mutual iterative manner. We conduct extensive experiments on both generic machine learning datasets and cancer datasets. The experimental results confirm that our model can achieve satisfactory clustering performance compared to several state-of-the-art approaches. Moreover, we convey the practicability of MRGC by carrying out a case study on hepatocellular carcinoma.
Collapse
|
47
|
Fan Y, S Chan A, Zhu J, Yi Leung S, Fan X. A Bayesian model for identifying cancer subtypes from paired methylation profiles. Brief Bioinform 2022; 24:6961790. [PMID: 36575828 PMCID: PMC9851340 DOI: 10.1093/bib/bbac568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/19/2022] [Accepted: 11/22/2022] [Indexed: 12/29/2022] Open
Abstract
Aberrant DNA methylation is the most common molecular lesion that is crucial for the occurrence and development of cancer, but has thus far been underappreciated as a clinical tool for cancer classification, diagnosis or as a guide for therapeutic decisions. Partly, this has been due to a lack of proven algorithms that can use methylation data to stratify patients into clinically relevant risk groups and subtypes that are of prognostic importance. Here, we proposed a novel Bayesian model to capture the methylation signatures of different subtypes from paired normal and tumor methylation array data. Application of our model to synthetic and empirical data showed high clustering accuracy, and was able to identify the possible epigenetic cause of a cancer subtype.
Collapse
Affiliation(s)
- Yetian Fan
- School of Mathematics and Statistics, Liaoning University, Shenyang, 110036, China,Department of Statistics, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, China
| | - April S Chan
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jun Zhu
- Sema4, Stamford, CT, 06902, USA,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suet Yi Leung
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaodan Fan
- Corresponding author: Xiaodan Fan, Department of Statistics, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, China. E-mail:
| |
Collapse
|
48
|
Shojaee Z, Shahzadeh Fazeli SA, Abbasi E, Adibnia F, Masuli F, Rovetta S. A Mutual Information Based on Ant Colony Optimization Method to Feature Selection for Categorical Data Clustering. IRANIAN JOURNAL OF SCIENCE AND TECHNOLOGY, TRANSACTIONS A: SCIENCE 2022. [DOI: 10.1007/s40995-022-01395-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Sarode P, Pullamsetti SS, Savai R. New Insights into the Epigenomic Landscape of Small-Cell Lung Cancer: A Game Changer? Am J Respir Crit Care Med 2022; 206:1441-1443. [PMID: 35947642 PMCID: PMC9757098 DOI: 10.1164/rccm.202208-1471ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung ResearchBad Nauheim, Germany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung ResearchBad Nauheim, Germany,Department of Internal MedicineGerman Center for Lung Research (DZL)Cardio-Pulmonary Institute (CPI),Institute for Lung HealthJustus Liebig UniversityGiessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung ResearchBad Nauheim, Germany,Department of Internal MedicineGerman Center for Lung Research (DZL)Cardio-Pulmonary Institute (CPI),Institute for Lung HealthJustus Liebig UniversityGiessen, Germany,Frankfurt Cancer InstituteGoethe University FrankfurtFrankfurt, Germany
| |
Collapse
|
50
|
Mendeville M, Roemer MGM, Los-de Vries GT, Chamuleau MED, de Jong D, Ylstra B. The path towards consensus genome classification of diffuse large B-cell lymphoma for use in clinical practice. Front Oncol 2022; 12:970063. [DOI: 10.3389/fonc.2022.970063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a widely heterogeneous disease in presentation, treatment response and outcome that results from a broad biological heterogeneity. Various stratification approaches have been proposed over time but failed to sufficiently capture the heterogeneous biology and behavior of the disease in a clinically relevant manner. The most recent DNA-based genomic subtyping studies are a major step forward by offering a level of refinement that could serve as a basis for exploration of personalized and targeted treatment for the years to come. To enable consistent trial designs and allow meaningful comparisons between studies, harmonization of the currently available knowledge into a single genomic classification widely applicable in daily practice is pivotal. In this review, we investigate potential avenues for harmonization of the presently available genomic subtypes of DLBCL inspired by consensus molecular classifications achieved for other malignancies. Finally, suggestions for laboratory techniques and infrastructure required for successful clinical implementation are described.
Collapse
|