1
|
Zhang X, Wang Z, Zhao Y, Ye H, Li T, Wang H, Sun G, Liang F, Dai L, Wang P, Liu X. Multi-omics analysis unveils a four-gene prognostic signature in esophageal squamous carcinoma and the therapeutic potential of PKP1. BMC Cancer 2025; 25:777. [PMID: 40281492 PMCID: PMC12032815 DOI: 10.1186/s12885-025-14150-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies, characterized by high heterogeneity and poor outcomes. Effective classification for patient stratification and identifying reliable markers for prognosis prediction and treatment choice are crucial. METHODS Integration of single-cell RNA-sequencing (RNA-seq) and bulk RNA-seq analyses were used to characterize ESCC. Non-negative matrix factorization (NMF) clustering was performed to stratify the ESCC patients into different subtypes and the clinical and pathological features of the ESCC subtypes were compared. Cox regression analysis and LASSO regression analysis were used to select key genes and construct a risk model for ESCC. The associations of the key genes with anti-cancer drug sensitivities in ESCC cell lines were investigated. RT-qRCR experiments, proteomics analysis, and multiplex immunohistochemistry (mIHC) experiments were used to validate the results. Furthermore, one identified gene was selected to investigate its correlation with EGFR expression and the gene effect scores of various potential gene targets across pan-cancer. RESULTS The study identified the dysregulated distributions of epithelial cells and fibroblasts as characteristic of ESCC. ESCC patients could be classified into four distinct subtypes with unique cell type features and prognoses. With the gene makers of the cell type features, a four-gene prognostic signature for ESCC was constructed. The CCND1-PKP1-JUP-ANKRD12 model could effectively discriminate the survival status of ESCC patients, independent of various pathological and clinical features. The risk score for the samples was correlated with the expression levels of immunoregulatory genes. The prognostic effects of CCND1, PKP1, and JUP were confirmed at the protein level. The phosphorylation levels of PKP1, JUP, and ANKRD12 were found to be dysregulated in ESCC tumors. Their expression dysregulation and heterogeneity were demonstrated in ESCC cell lines. All four genes were significantly correlated with at least one of the anti-cancer drug sensitivities in ESCC cell lines. PKP1 expression was significantly correlated with EGFR expression and gene effect scores in multiple cancers. CONCLUSIONS We conclude that the CCND1-PKP1-JUP-ANKRD12 signature may serve as a novel indicator for ESCC prognosis and diagnosis. PKP1 expression might provide new clues for gene therapy efficacy in multiple cancers.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Zhi Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yutong Zhao
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Han Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Guiying Sun
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Feifei Liang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China.
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Xiaoli Liu
- Laboratory Department, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
2
|
Mao Y, Shangguan D, Huang Q, Xiao L, Cao D, Zhou H, Wang YK. Emerging artificial intelligence-driven precision therapies in tumor drug resistance: recent advances, opportunities, and challenges. Mol Cancer 2025; 24:123. [PMID: 40269930 PMCID: PMC12016295 DOI: 10.1186/s12943-025-02321-x] [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: 02/07/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Drug resistance is one of the main reasons for cancer treatment failure, leading to a rapid recurrence/disease progression of the cancer. Recently, artificial intelligence (AI) has empowered physicians to use its powerful data processing and pattern recognition capabilities to extract and mine valuable drug resistance information from large amounts of clinical or omics data, to study drug resistance mechanisms, to evaluate and predict drug resistance, and to develop innovative therapeutic strategies to reduce drug resistance. In this review, we proposed a feasible workflow for incorporating AI into tumor drug resistance research, highlighted current AI-driven tumor drug resistance applications, and discussed the opportunities and challenges encountered in the process. Based on a comprehensive literature analysis, we systematically summarized the role of AI in tumor drug resistance research, including drug development, resistance mechanism elucidation, drug sensitivity prediction, combination therapy optimization, resistance phenotype identification, and clinical biomarker discovery. With the continuous advancement of AI technology and rigorous validation of clinical data, AI models are expected to fuel the development of precision oncology by improving efficacy, guiding therapeutic decisions, and optimizing patient prognosis. In summary, by leveraging clinical and omics data, AI models are expected to pioneer new therapy strategies to mitigate tumor drug resistance, improve efficacy and patient survival, and provide novel perspectives and tools for oncology treatment.
Collapse
Affiliation(s)
- Yuan Mao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dangang Shangguan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Xiao
- Department of Histology and Embryology of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dongsheng Cao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Zhou
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, Changsha, Hunan, People's Republic of China.
| | - Yi-Kun Wang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Huang K, Liu H. Identification of drug-resistant individual cells within tumors by semi-supervised transfer learning from bulk to single-cell transcriptome. Commun Biol 2025; 8:530. [PMID: 40164749 PMCID: PMC11958800 DOI: 10.1038/s42003-025-07959-3] [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: 09/25/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
The presence of pre-existing or acquired drug-resistant cells within the tumor often leads to tumor relapse and metastasis. Single-cell RNA sequencing (scRNA-seq) enables elucidation of the subtle differences in drug responsiveness among distinct cell subpopulations within tumors. A few methods have employed scRNA-seq data to predict the drug response of individual cells to date, but their performance is far from satisfactory. In this study, we propose SSDA4Drug, a semi-supervised few-shot transfer learning method for inferring drug-resistant cancer cells. SSDA4Drug extracts pharmacogenomic features from both bulk and single-cell transcriptomic data using semi-supervised adversarial domain adaptation. This allows us to transfer knowledge of drug sensitivity from bulk-level cell lines to single cells. We conduct extensive performance evaluation experiments across multiple independent scRNA-seq datasets, demonstrating SSDA4Drug's superior performance over current state-of-the-art methods. Remarkably, with only one or two labeled target-domain samples, SSDA4Drug significantly boosts the predictive performance of single-cell drug responses. Moreover, SSDA4Drug accurately recapitulates the temporally dynamic changes of drug responses during continuous drug exposure of tumor cells, and successfully identifies reversible drug-responsive states in lung cancer cells, which initially acquire resistance through drug exposure but later restore sensitivity during drug holidays. Also, our predicted drug responses consistently align with the developmental patterns of drug sensitivity observed along the evolutionary trajectory of oral squamous cell carcinoma cells. In addition, our derived SHAP values and integrated gradients effectively pinpoint the key genes involved in drug resistance in prostate cancer cells. These findings highlight the exceptional performance of our method in determining single-cell drug responses. This powerful tool holds the potential for identifying drug-resistant tumor cell subpopulations, paving the way for advancements in precision medicine and novel drug development.
Collapse
Affiliation(s)
- Kaishun Huang
- College of Computer and Information Engineering, Nanjing Tech University, Nanjing, 211800, Jiangsu, China
| | - Hui Liu
- College of Computer and Information Engineering, Nanjing Tech University, Nanjing, 211800, Jiangsu, China.
| |
Collapse
|
4
|
Monfort-Lanzas P, Rungger K, Madersbacher L, Hackl H. Machine learning to dissect perturbations in complex cellular systems. Comput Struct Biotechnol J 2025; 27:832-842. [PMID: 40103613 PMCID: PMC11915099 DOI: 10.1016/j.csbj.2025.02.028] [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: 11/15/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Understanding the responses of biological systems to various perturbations, such as genetic, chemical, or environmental challenges, is essential for reconstructing causal network models. Emerging single-cell technologies have become instrumental in elucidating cell states and phenotypes and they have been used in combination with genetic screening. Recent advances in machine learning and artificial intelligence architectures have stimulated the development of computational tools for modeling perturbations and the response to compounds. This study outlined core principles underpinning perturbation analysis and discussed the methodologies and analytical frameworks used to decode drug and genetic perturbation responses, complex multicellular interactions, and network dynamics. The current tools used for various applications were overviewed. These developments hold great promise for improving drug development and personalized medicine. Foundation models and perturbation cell and tissue atlases offer immense potential for advancing our understanding of cellular behavior and disease mechanisms.
Collapse
Affiliation(s)
- Pablo Monfort-Lanzas
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Austria
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Austria
| | - Katja Rungger
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Austria
| | - Leonie Madersbacher
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Austria
| |
Collapse
|
5
|
Li X, Loscalzo J, Mahmud AKMF, Aly DM, Rzhetsky A, Zitnik M, Benson M. Digital twins as global learning health and disease models for preventive and personalized medicine. Genome Med 2025; 17:11. [PMID: 39920778 PMCID: PMC11806862 DOI: 10.1186/s13073-025-01435-7] [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: 05/13/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Ineffective medication is a major healthcare problem causing significant patient suffering and economic costs. This issue stems from the complex nature of diseases, which involve altered interactions among thousands of genes across multiple cell types and organs. Disease progression can vary between patients and over time, influenced by genetic and environmental factors. To address this challenge, digital twins have emerged as a promising approach, which have led to international initiatives aiming at clinical implementations. Digital twins are virtual representations of health and disease processes that can integrate real-time data and simulations to predict, prevent, and personalize treatments. Early clinical applications of DTs have shown potential in areas like artificial organs, cancer, cardiology, and hospital workflow optimization. However, widespread implementation faces several challenges: (1) characterizing dynamic molecular changes across multiple biological scales; (2) developing computational methods to integrate data into DTs; (3) prioritizing disease mechanisms and therapeutic targets; (4) creating interoperable DT systems that can learn from each other; (5) designing user-friendly interfaces for patients and clinicians; (6) scaling DT technology globally for equitable healthcare access; (7) addressing ethical, regulatory, and financial considerations. Overcoming these hurdles could pave the way for more predictive, preventive, and personalized medicine, potentially transforming healthcare delivery and improving patient outcomes.
Collapse
Affiliation(s)
- Xinxiu Li
- Medical Digital Twin Research Group, Department of Clinical Sciences Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Joseph Loscalzo
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - A K M Firoj Mahmud
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75105, Uppsala, Sweden
| | - Dina Mansour Aly
- Medical Digital Twin Research Group, Department of Clinical Sciences Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Andrey Rzhetsky
- Departments of Medicine and Human Genetics, Institute for Genomics and Systems Biology, University of Chicago, Chicago, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA
| | - Mikael Benson
- Medical Digital Twin Research Group, Department of Clinical Sciences Intervention and Technology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
6
|
Bhayana S, Schytz PA, Bisgaard Olesen ET, Soh K, Das V. Single-Cell Advances in Investigating and Understanding Chronic Kidney Disease and Diabetic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:55-68. [PMID: 39097167 DOI: 10.1016/j.ajpath.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
Chronic kidney disease (CKD) and its subset diabetic kidney disease are progressive conditions that affect >850 million people worldwide. Diabetes, hypertension, and glomerulonephritis are the most common causes of CKD, which is associated with significant patient morbidity and an increased risk of cardiovascular events, such as heart failure, ultimately leading to premature death. Despite newly approved drugs, increasing evidence shows that patients respond to treatment differently given the complexity of disease heterogeneity and complicated pathophysiology. This review article presents an integrative approach to understanding and addressing CKD through the lens of precision medicine and therapeutics. Advancements in single-cell omics technologies and artificial intelligence can be leveraged to explore the intricate cellular mechanisms underlying CKD and diabetic kidney disease pathogenesis. Dissecting the cellular heterogeneity and identifying rare cell populations using single-cell approaches will facilitate uncovering novel therapeutic targets and biomarkers for personalized treatment strategies. Finally, we discuss the potential of artificial intelligence-driven analyses in predicting disease progression and treatment response, thereby paving the way for tailored interventions.
Collapse
Affiliation(s)
- Sagar Bhayana
- Kidney Biology, Global Drug Development, Novo Nordisk A/S, Søborg, Denmark
| | - Philip A Schytz
- Cardiovascular, Kidney and Alzheimer Disease, Medical and Science, Novo Nordisk A/S, Søborg, Denmark
| | - Emma T Bisgaard Olesen
- Cardiovascular, Kidney and Alzheimer Disease, Medical and Science, Novo Nordisk A/S, Søborg, Denmark
| | - Keng Soh
- Integrated Omics, AI and Analytics, Development, Novo Nordisk A/S, Søborg, Denmark
| | - Vivek Das
- Integrated Omics, AI and Analytics, Development, Novo Nordisk A/S, Søborg, Denmark.
| |
Collapse
|
7
|
Che J, Jin Y, Gragnoli C, Yau ST, Wu R. IdopNetwork as a genomic predictor of drug response. Drug Discov Today 2025; 30:104252. [PMID: 39603519 DOI: 10.1016/j.drudis.2024.104252] [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: 07/24/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Despite being challenging, elucidating the systematic control mechanisms of multifactorial drug responses is crucial for pharmacogenomic research. We describe a new form of statistical mechanics to reconstruct informative, dynamic, omnidirectional, and personalized networks (idopNetworks), which cover all pharmacogenomic factors and their interconnections, interdependence, and mechanistic roles. IdopNetworks can characterize how cell-cell crosstalk is mediated by genes and proteins to shape body-drug interactions and identify key roadmaps of information flow and propagation for determining drug efficacy and toxicity. We argue that idopNetworks could potentially provide insight into the genomic machinery of drug responses and provide scientific guidance for the design of drugs whose potency is maximized at lower doses.
Collapse
Affiliation(s)
- Jincan Che
- School of Grassland Science, Beijing Forestry University, Beijing 100083, China; Beijing Institute of Mathematical Sciences and Applications, Beijing 101408, China
| | - Yuebo Jin
- Department of Mathematics, Brandeis University, Waltham, MA 02453, USA
| | - Claudia Gragnoli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA; Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA; Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome 00197, Italy
| | - Shing-Tung Yau
- Beijing Institute of Mathematical Sciences and Applications, Beijing 101408, China; Yau Mathematical Sciences Center, Tsinghua University, Beijing 100084, China; Shanghai Institute for Mathematics and Interdisciplinary Sciences, Shanghai 200433, China
| | - Rongling Wu
- Beijing Institute of Mathematical Sciences and Applications, Beijing 101408, China; Yau Mathematical Sciences Center, Tsinghua University, Beijing 100084, China; Shanghai Institute for Mathematics and Interdisciplinary Sciences, Shanghai 200433, China.
| |
Collapse
|
8
|
Jiménez-Santos M, García-Martín S, Rubio-Fernández M, Gómez-López G, Al-Shahrour F. Spatial transcriptomics in breast cancer reveals tumour microenvironment-driven drug responses and clonal therapeutic heterogeneity. NAR Cancer 2024; 6:zcae046. [PMID: 39703753 PMCID: PMC11655296 DOI: 10.1093/narcan/zcae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
Breast cancer patients are categorized into three subtypes with distinct treatment approaches. Precision oncology has increased patient outcomes by targeting the specific molecular alterations of tumours, yet challenges remain. Treatment failure persists due to the coexistence of several malignant subpopulations with different drug sensitivities within the same tumour, a phenomenon known as intratumour heterogeneity (ITH). This heterogeneity has been extensively studied from a tumour-centric view, but recent insights underscore the role of the tumour microenvironment in treatment response. Our research utilizes spatial transcriptomics data from breast cancer patients to predict drug sensitivity. We observe diverse response patterns across tumour, interphase and microenvironment regions, unveiling a sensitivity and functional gradient from the tumour core to the periphery. Moreover, we find tumour therapeutic clusters with different drug responses associated with distinct biological functions driven by unique ligand-receptor interactions. Importantly, we identify genetically identical subclones with different responses depending on their location within the tumour ducts. This research underscores the significance of considering the distance from the tumour core and microenvironment composition when identifying suitable treatments to target ITH. Our findings provide critical insights into optimizing therapeutic strategies, highlighting the necessity of a comprehensive understanding of tumour biology for effective cancer treatment.
Collapse
Affiliation(s)
- María José Jiménez-Santos
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Santiago García-Martín
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Marcos Rubio-Fernández
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
- Lung-H120 Group, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
9
|
Chen L, Hu Y, Li Y, Zhang B, Wang J, Deng M, Zhang J, Zhu W, Gu H, Zhang L. Integrated multiomics analysis identified comprehensive crosstalk between diverse programmed cell death patterns and novel molecular subtypes in Hepatocellular Carcinoma. Sci Rep 2024; 14:27529. [PMID: 39528670 PMCID: PMC11555373 DOI: 10.1038/s41598-024-78911-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with increasing global prevalence and is one of the leading causes of cancer-related mortality in the human population. Developing robust clinical prediction models and prognostic stratification strategies is crucial for developing individualized treatment plans. A range of novel forms of programmed cell death (PCD) plays a role in the pathological progression and advancement of HCC, and in-depth study of PCD is expected to further improve the prognosis of HCC patients. Sixteen patterns (apoptosis, autophagy, anoikis, lysosome-dependent cell death, immunogenic cell death, necroptosis, ferroptosis, netosis, pyroptosis, disulfidptosis, entotic cell death, cuproptosis, parthanatos, netotic cell death, alkaliptosis, and oxeiptosis) related to PCD were collected from the literatures and used for subsequent analysis. Supervised (Elastic net, Random Forest, XgBoost, and Boruta) and unsupervised (Nonnegative Matrix Factorization, NMF) clustering algorithms were applied to develop and validate a novel classifier for the individualized management of HCC patients at the transcriptomic, proteomic and single-cell levels. Multiple machine learning algorithms developed a programmed cell death index (PCDI) comprising five robust signatures (FTL, G6PD, SLC2A1, HTRA2, and DLAT) in four independent HCC cohorts, and a higher PCDI was predictive of higher pathological grades and worse prognoses. Furthermore, a higher PCDI was found to be correlated with the presence of a repressive tumor immune microenvironment (TME), as determined through an integrated examination of bulk and single-cell transcriptome data. In addition, patients with TP53 mutation had higher PCDI in comparison with TP53 WT patients. Three HCC subtypes were identified through unsupervised clustering (NMF), exhibiting distinct prognoses and significant biological processes, among the three subtypes, PCDcluster 3 was of particular interest as it contained a large proportion of patients with high risk and low metabolic activity. Construction and evaluation of the Nomogram model was drawn based on the multivariate logistic regression analysis, and highlighted the robustness of the Nomogram model in other independent HCC cohorts. Finally, to explore the prognostic value, we also validated the frequent upregulation of DLAT in a real-world cohort of human HCC specimens by qPCR, western blot, and immunohistochemical staining (IHC). Together, our work herein comprehensively emphasized PCD-related patterns and key regulators, such as DLAT, contributed to the evolution and prognosis of tumor foci in HCC patients, and strengthened our understanding of PCD characteristics and promoted more effective risk stratification strategies.
Collapse
Affiliation(s)
- Li Chen
- Department of Blood Transfusion, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yuanbo Hu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Center for Reproductive Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Bingyu Zhang
- School of Public Health, China Medical University, Shenyang, China
| | - Jiale Wang
- School of International Education, Henan University of Technology, Zhengzhou, China
| | - Mengmeng Deng
- Department of Laboratory Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jinlian Zhang
- Department of Pathology, the Second Affiliated Hospital of Bengbu Medical University, Benbgu, China
| | - Wenyao Zhu
- Department of Urology, the Central Hospital of Bengbu, Bengbu, China
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Lingyu Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| |
Collapse
|
10
|
Yang GN, Sun YBY, Roberts PK, Moka H, Sung MK, Gardner-Russell J, El Wazan L, Toussaint B, Kumar S, Machin H, Dusting GJ, Parfitt GJ, Davidson K, Chong EW, Brown KD, Polo JM, Daniell M. Exploring single-cell RNA sequencing as a decision-making tool in the clinical management of Fuchs' endothelial corneal dystrophy. Prog Retin Eye Res 2024; 102:101286. [PMID: 38969166 DOI: 10.1016/j.preteyeres.2024.101286] [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: 01/17/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has enabled the identification of novel gene signatures and cell heterogeneity in numerous tissues and diseases. Here we review the use of this technology for Fuchs' Endothelial Corneal Dystrophy (FECD). FECD is the most common indication for corneal endothelial transplantation worldwide. FECD is challenging to manage because it is genetically heterogenous, can be autosomal dominant or sporadic, and progress at different rates. Single-cell RNA sequencing has enabled the discovery of several FECD subtypes, each with associated gene signatures, and cell heterogeneity. Current FECD treatments are mainly surgical, with various Rho kinase (ROCK) inhibitors used to promote endothelial cell metabolism and proliferation following surgery. A range of emerging therapies for FECD including cell therapies, gene therapies, tissue engineered scaffolds, and pharmaceuticals are in preclinical and clinical trials. Unlike conventional disease management methods based on clinical presentations and family history, targeting FECD using scRNA-seq based precision-medicine has the potential to pinpoint the disease subtypes, mechanisms, stages, severities, and help clinicians in making the best decision for surgeries and the applications of therapeutics. In this review, we first discuss the feasibility and potential of using scRNA-seq in clinical diagnostics for FECD, highlight advances from the latest clinical treatments and emerging therapies for FECD, integrate scRNA-seq results and clinical notes from our FECD patients and discuss the potential of applying alternative therapies to manage these cases clinically.
Collapse
Affiliation(s)
- Gink N Yang
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Yu B Y Sun
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Philip Ke Roberts
- Department of Ophthalmology, Medical University Vienna, 18-20 Währinger Gürtel, Vienna, Austria
| | - Hothri Moka
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Min K Sung
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Jesse Gardner-Russell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Layal El Wazan
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Bridget Toussaint
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Satheesh Kumar
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Heather Machin
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia
| | - Gregory J Dusting
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Geraint J Parfitt
- Mogrify Limited, 25 Cambridge Science Park Milton Road, Milton, Cambridge, UK
| | - Kathryn Davidson
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Elaine W Chong
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Department of Ophthalmology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Karl D Brown
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| | - Jose M Polo
- Department of Anatomy and Development Biology, Monash University, Clayton, Australia
| | - Mark Daniell
- Centre for Eye Research Australia, Level 7, Peter Howson Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia; Ophthalmology, Department of Surgery, University of Melbourne and Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia; Lions Eye Donation Service, Level 7, Smorgon Family Wing, 32 Gisborne Street, East Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Cao X, Huang YA, You ZH, Shang X, Hu L, Hu PW, Huang ZA. scPriorGraph: constructing biosemantic cell-cell graphs with prior gene set selection for cell type identification from scRNA-seq data. Genome Biol 2024; 25:207. [PMID: 39103856 DOI: 10.1186/s13059-024-03357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Cell type identification is an indispensable analytical step in single-cell data analyses. To address the high noise stemming from gene expression data, existing computational methods often overlook the biologically meaningful relationships between genes, opting to reduce all genes to a unified data space. We assume that such relationships can aid in characterizing cell type features and improving cell type recognition accuracy. To this end, we introduce scPriorGraph, a dual-channel graph neural network that integrates multi-level gene biosemantics. Experimental results demonstrate that scPriorGraph effectively aggregates feature values of similar cells using high-quality graphs, achieving state-of-the-art performance in cell type identification.
Collapse
Affiliation(s)
- Xiyue Cao
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Yu-An Huang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Lun Hu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Ürümqi, China
| | - Peng-Wei Hu
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Ürümqi, China
| | - Zhi-An Huang
- Research Office, City University of Hong Kong (Dongguan), Dongguan, 523000, China
| |
Collapse
|
12
|
Pellecchia S, Franchini M, Viscido G, Arnese R, Gambardella G. Single cell lineage tracing reveals clonal dynamics of anti-EGFR therapy resistance in triple negative breast cancer. Genome Med 2024; 16:55. [PMID: 38605363 PMCID: PMC11008053 DOI: 10.1186/s13073-024-01327-2] [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: 05/02/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Most primary Triple Negative Breast Cancers (TNBCs) show amplification of the Epidermal Growth Factor Receptor (EGFR) gene, leading to increased protein expression. However, unlike other EGFR-driven cancers, targeting this receptor in TNBC yields inconsistent therapeutic responses. METHODS To elucidate the underlying mechanisms of this variability, we employ cellular barcoding and single-cell transcriptomics to reconstruct the subclonal dynamics of EGFR-amplified TNBC cells in response to afatinib, a tyrosine kinase inhibitor (TKI) that irreversibly inhibits EGFR. RESULTS Integrated lineage tracing analysis revealed a rare pre-existing subpopulation of cells with distinct biological signature, including elevated expression levels of Insulin-Like Growth Factor Binding Protein 2 (IGFBP2). We show that IGFBP2 overexpression is sufficient to render TNBC cells tolerant to afatinib treatment by activating the compensatory insulin-like growth factor I receptor (IGF1-R) signalling pathway. Finally, based on reconstructed mechanisms of resistance, we employ deep learning techniques to predict the afatinib sensitivity of TNBC cells. CONCLUSIONS Our strategy proved effective in reconstructing the complex signalling network driving EGFR-targeted therapy resistance, offering new insights for the development of individualized treatment strategies in TNBC.
Collapse
Affiliation(s)
- Simona Pellecchia
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Scuola Superiore Meridionale, Genomics and Experimental Medicine Program, Naples, Italy
| | - Melania Franchini
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Gaetano Viscido
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Chemical, Materials and Industrial Engineering , University of Naples Federico II, Naples, Italy
| | - Riccardo Arnese
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
13
|
Toledo B, Deiana C, Scianò F, Brandi G, Marchal JA, Perán M, Giovannetti E. Treatment resistance in pancreatic and biliary tract cancer: molecular and clinical pharmacology perspectives. Expert Rev Clin Pharmacol 2024; 17:323-347. [PMID: 38413373 DOI: 10.1080/17512433.2024.2319340] [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: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Treatment resistance poses a significant obstacle in oncology, especially in biliary tract cancer (BTC) and pancreatic cancer (PC). Current therapeutic options include chemotherapy, targeted therapy, and immunotherapy. Resistance to these treatments may arise due to diverse molecular mechanisms, such as genetic and epigenetic modifications, altered drug metabolism and efflux, and changes in the tumor microenvironment. Identifying and overcoming these mechanisms is a major focus of research: strategies being explored include combination therapies, modulation of the tumor microenvironment, and personalized approaches. AREAS COVERED We provide a current overview and discussion of the most relevant mechanisms of resistance to chemotherapy, target therapy, and immunotherapy in both BTC and PC. Furthermore, we compare the different strategies that are being implemented to overcome these obstacles. EXPERT OPINION So far there is no unified theory on drug resistance and progress is limited. To overcome this issue, individualized patient approaches, possibly through liquid biopsies or single-cell transcriptome studies, are suggested, along with the potential use of artificial intelligence, to guide effective treatment strategies. Furthermore, we provide insights into what we consider the most promising areas of research, and we speculate on the future of managing treatment resistance to improve patient outcomes.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Chiara Deiana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabio Scianò
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Lumobiotics GmbH, Karlsruhe, Germany
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Pisa, Italy
- Cancer Pharmacology Lab, Associazione Italiana per la Ricerca sul Cancro (AIRC) Start-Up Unit, Fondazione Pisana per la Scienza, University of Pisa, Pisa, Italy
| |
Collapse
|
14
|
Pellecchia S, Viscido G, Franchini M, Gambardella G. Correction: Predicting drug response from single-cell expression profiles of tumours. BMC Med 2024; 22:70. [PMID: 38365695 PMCID: PMC10874051 DOI: 10.1186/s12916-024-03289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024] Open
Affiliation(s)
- Simona Pellecchia
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - Gaetano Viscido
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Chemical, Materials and Industrial Engineering, University of Naples Federico II, Naples, Italy
| | - Melania Franchini
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|