1
|
Xie Y, Chen H, Zhang X, Zhang J, Zhang K, Wang X, Min S, Wang X, Lian C. Integration of the bulk transcriptome and single-cell transcriptome reveals efferocytosis features in lung adenocarcinoma prognosis and immunotherapy by combining deep learning. Cancer Cell Int 2024; 24:388. [PMID: 39580462 PMCID: PMC11585238 DOI: 10.1186/s12935-024-03571-3] [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: 04/17/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Efferocytosis (ER) refers to the process of phagocytic clearance of programmed dead cells, and studies have shown that it is closely related to tumor immune escape. METHODS This study was based on a comprehensive analysis of TCGA, GEO and CTRP databases. ER-related genes were collected from previous literature, univariate Cox regression was performed and consistent clustering was performed to categorize lung adenocarcinoma (LUAD) patients into two subgroups. Lasso regression and multivariate Cox regression analyses were used to construct ER-related prognostic features, and multiple immune infiltration algorithms were used to assess the correlation between the extracellular burial-related risk score (ERGRS) and tumor microenvironment (TME). And the key gene HAVCR1 was identified by deep learning, etc. Finally, pan-cancer analysis of the key genes was performed and in vitro experiments were conducted to verify the promotional effect of HAVCR1 on LUAD progression. RESULTS A total of 33 ER-related genes associated with the prognosis of LUAD were identified, and the prognostic signature of ERGRS was successfully constructed to predict the overall survival (OS) and treatment response of LUAD patients. The high-risk group was highly enriched in some oncogenic pathways, while the low-ERGRS group was highly enriched in some immune-related pathways. In addition, the high ERGRS group had higher TMB, TNB and TIDE scores and lower immune scores. The low-risk group had better immunotherapeutic response and less likelihood of immune escape. Drug sensitivity analysis revealed that BRD-K92856060, monensin and hexaminolevulinate may be potential therapeutic agents for the high-risk group. And ERGRS was validated in several cohorts. In addition, HAVCR1 is one of the key genes, and knockdown of HAVCR1 in vitro significantly reduced the proliferation, migration and invasion ability of lung adenocarcinoma cells. CONCLUSION Our study developed a novel prognostic signature of efferocytosis-related genes. This prognostic signature accurately predicted survival prognosis as well as treatment outcome in LUAD patients and explored the role of HAVCR1 in lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Yiluo Xie
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Xueying Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Kai Zhang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Xinyu Wang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Shengping Min
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
2
|
Yan B, Chen Y, Wang Z, Li J, Wang R, Pan X, Li B, Li R. Analysis and identification of mRNAsi‑related expression signatures via RNA sequencing in lung cancer. Oncol Lett 2024; 28:549. [PMID: 39319211 PMCID: PMC11420643 DOI: 10.3892/ol.2024.14682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
High stemness index scores are associated with poor survival in patients with lung cancer. Studies on the mRNA expression-based stemness index (mRNAsi) are typically conducted using tumor tissues; however, mRNAsi-related expression signatures based on cell-free RNA (cfRNA) are yet to be comprehensively investigated. The present study aimed to elucidate the gene expression profiles of tumor stemness in lung cancer tissues and corresponding cfRNAs in blood, and to assess their links with immune infiltration. Tumor tissue, paracancerous tissue, peripheral blood and lymph node samples were collected from patients with stage I-III non-small cell lung cancer and RNA sequencing was performed. The TCGAbiolinks package was used to calculate the mRNAsi for each of these four types of sample. Weighted gene co-expression network analysis and differentially expressed gene analyses were performed to investigate mRNAsi-related genes, and pathway enrichment analysis was performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) orthology-based annotation system. In addition, the STAR-Fusion tool was used to detect fusion variants, and CIBERSORT was used to analyze the correlations of stemness signatures in tissues and blood with immune cell infiltration. The mRNAsi values in peripheral blood and lymph nodes were found to be higher than those in cancer tissues. 'Hematopoietic cell lineage' was the only KEGG pathway enriched in mRNAsi-related genes in both lung cancer tissues and peripheral blood. In addition, the protein tyrosine phosphatase receptor type C associated protein gene was the only gene commonly associated with the mRNAsi in these two types of sample. The expression of mRNAsi-related genes was increased in the dendritic and Treg cells in tumor tissues, but was elevated in Treg and CD8 cells in the blood. In conclusion, cfRNAs in the blood exhibit unique stemness signatures that have potential for use in the diagnosis of lung cancer.
Collapse
Affiliation(s)
- Bo Yan
- Clinical Research Unit, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Yong Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Zhouyu Wang
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Jing Li
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Ruiru Wang
- Berry Oncology Corporation, Beijing 100102, P.R. China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| | - Boyi Li
- Kanghui Biotechnology Corporation, Shenyang, Liaoning 110042, P.R. China
| | - Rong Li
- Clinical Research Unit, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200050, P.R. China
| |
Collapse
|
3
|
Nielsen AJ, Albert GK, Sanchez A, Chen J, Liu J, Davalos AS, Geng D, Bradeen X, Hintzsche JD, Robinson W, McCarter M, Amato C, Tobin R, Couts K, Wilky BA, Davila E. DNA-PK inhibition enhances neoantigen diversity and increases T cell responses to immunoresistant tumors. J Clin Invest 2024; 134:e180278. [PMID: 39436696 PMCID: PMC11645140 DOI: 10.1172/jci180278] [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: 02/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Effective antitumor T cell activity relies on the expression and MHC presentation of tumor neoantigens. Tumor cells can evade T cell detection by silencing the transcription of antigens or by altering MHC machinery, resulting in inadequate neoantigen-specific T cell activation. We identified the DNA-protein kinase inhibitor (DNA-PKi) NU7441 as a promising immunomodulator that reduced immunosuppressive proteins, while increasing MHC-I expression in a panel of human melanoma cell lines. In tumor-bearing mice, combination therapy using NU7441 and the immune adjuvants stimulator of IFN genes (STING) ligand and the CD40 agonist NU-SL40 substantially increased and diversified the neoantigen landscape, antigen-presenting machinery, and, consequently, substantially increased both the number and repertoire of neoantigen-reactive, tumor-infiltrating lymphocytes (TILs). DNA-PK inhibition or KO promoted transcription and protein expression of various neoantigens in human and mouse melanomas and induced sensitivity to immune checkpoint blockade (ICB) in resistant tumors. In patients, protein kinase, DNA-activated catalytic subunit (PRKDC) transcript levels were inversely correlated with MHC-I expression and CD8+ TILs but positively correlated with increased neoantigen loads and improved responses to ICB. These studies suggest that inhibition of DNA-PK activity can restore tumor immunogenicity by increasing neoantigen expression and presentation and broadening the neoantigen-reactive T cell population.
Collapse
Affiliation(s)
- Allison J. Nielsen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Gabriella K. Albert
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Amelia Sanchez
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jiangli Chen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Jing Liu
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Andres S. Davalos
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Degui Geng
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Xander Bradeen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - William Robinson
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Martin McCarter
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Carol Amato
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Richard Tobin
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kasey Couts
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Breelyn A. Wilky
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Eduardo Davila
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| |
Collapse
|
4
|
Shen Z, Feng C, Chen X, Jiang Y, Chen J. Prognostic model of lung adenocarcinoma based on immunoprognosis-related genes and related drug prediction. J Thorac Dis 2024; 16:5860-5877. [PMID: 39444861 PMCID: PMC11494575 DOI: 10.21037/jtd-24-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/26/2024] [Indexed: 10/25/2024]
Abstract
Background Lung cancer (LC) is the most common malignant tumor in the world, and lung adenocarcinoma (LUAD) is the most common type of LC. Immune microenvironment plays a critical role in cancer from onset to relapse. We aimed to identify an effective immune-related prediction model for assessing prognosis and predicting the relevant tumor therapeutic drugs. Methods According to the RNA sequencing data of LUAD transcriptome in The Cancer Genome Atlas (TCGA) database and the immune-related genes obtained from IMMPORT (The Immunology Database and Analysis Portal) database, immune prognosis-related genes were screened. Weighted gene co-expression network analysis (WGCNA) identified hub genes in differentially expressed immune-related genes (DEIRGs). Least absolute shrinkage and selection operator (LASSO) Cox and ten rounds of cross-validation were used to screen core genes to establish a prognostic model, and in situ hybridization was used to verify the expression of core genes in LUAD. Then the patients from the TCGA database were divided into high-risk group and low-risk group. The survival, tumor microenvironment (TME) and immune cell infiltration of different groups were further analyzed, and the differential genes between the two groups were analyzed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) enrichment analyses. Finally, the small molecular drugs that can inhibit the prognosis of LUAD were screened by Connectivity Map (CMAP), and the therapeutic mechanism of small molecular drug oxibendazole was verified by Cell Counting Kit-8 (CCK-8) experiment. Results A four-immunoprognosis-related gene model was established to forecast the overall survival (OS) of LUAD through LASSO Cox regression and ten rounds of cross-validation analysis. This prognostic model stratified LUAD patients into low-risk and high-risk groups. According to the findings of the survival analysis, the low-risk group had a greater OS than the high-risk group and the content of immune cells in LUAD was corrected with the survival prognosis of patients. Univariate and multivariate Cox regression also revealed that the prognostic model was an independent prognosis factor in LUAD. Five kinds of small molecular drugs which can inhibit the prognosis of LUAD were screened by CMAP. As shown by CCK-8 test, the small molecular drug "oxibendazole" can effectively inhibit the proliferation of LUAD cells. Conclusions Based on immune-related prognostic genes, a new prognostic model for LUAD was constructed. Oxibendazole can inhibit the proliferation of LUAD cells, which provides a new idea for the treatment of LUAD.
Collapse
Affiliation(s)
- Zihao Shen
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chen Feng
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Xingyou Chen
- Medical School of Nantong University, Nantong, China
| | - Yun Jiang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianle Chen
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
5
|
Pascal M, Bax HJ, Bergmann C, Bianchini R, Castells M, Chauhan J, De Las Vecillas L, Hartmann K, Álvarez EI, Jappe U, Jimenez-Rodriguez TW, Knol E, Levi-Schaffer F, Mayorga C, Poli A, Redegeld F, Santos AF, Jensen-Jarolim E, Karagiannis SN. Granulocytes and mast cells in AllergoOncology-Bridging allergy to cancer: An EAACI position paper. Allergy 2024; 79:2319-2345. [PMID: 39036854 DOI: 10.1111/all.16246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/23/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Derived from the myeloid lineage, granulocytes, including basophils, eosinophils, and neutrophils, along with mast cells, play important, often disparate, roles across the allergic disease spectrum. While these cells and their mediators are commonly associated with allergic inflammation, they also exhibit several functions either promoting or restricting tumor growth. In this Position Paper we discuss common granulocyte and mast cell features relating to immunomodulatory functions in allergy and in cancer. We highlight key mechanisms which may inform cancer treatment and propose pertinent areas for future research. We suggest areas where understanding the communication between granulocytes, mast cells, and the tumor microenvironment, will be crucial for identifying immune mechanisms that may be harnessed to counteract tumor development. For example, a comprehensive understanding of allergic and immune factors driving distinct neutrophil states and those mechanisms that link mast cells with immunotherapy resistance, might enable targeted manipulation of specific subpopulations, leading to precision immunotherapy in cancer. We recommend specific areas of investigation in AllergoOncology and knowledge exchange across disease contexts to uncover pertinent reciprocal functions in allergy and cancer and allow therapeutic manipulation of these powerful cell populations. These will help address the unmet needs in stratifying and managing patients with allergic diseases and cancer.
Collapse
Affiliation(s)
- Mariona Pascal
- Immunology Department, CDB, Hospital Clínic de Barcelona; Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, Barcelona, Spain
- RETICS Asma, reacciones adversas y alérgicas (ARADYAL) and RICORS Red De Enfermedades Inflamatorias (REI), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Heather J Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, London, UK
| | - Christoph Bergmann
- Department of Otorhinolaryngology, RKM740 Interdisciplinary Clinics, Düsseldorf, Germany
| | - Rodolfo Bianchini
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
- The interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, Vienna, Austria
| | - Mariana Castells
- Division of Allergy and Clinical Immunology, Drug Hypersensitivity and Desensitization Center, Mastocytosis Center, Brigham and Women's Hospital; Harvard Medical School, Boston, USA
| | - Jitesh Chauhan
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, London, UK
| | | | - Karin Hartmann
- Division of Allergy, Department of Dermatology, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Elena Izquierdo Álvarez
- Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Institute of Applied Molecular Medicine Instituto de Medicina Molecular Aplicada Nemesio Díez (IMMA), Madrid, Spain
| | - Uta Jappe
- Division of Clinical and Molecular Allergology, Priority Research Area Chronic Lung Diseases, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL), Airway Research Center North (ARCN), Borstel, Germany
- Interdisciplinary Allergy Outpatient Clinic, Department of Pneumology, University of Luebeck, Luebeck, Germany
| | | | - Edward Knol
- Departments Center of Translational Immunology and Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine. The Hebrew University of Jerusalem, Ein Kerem Campus, Jerusalem, Israel
| | - Cristobalina Mayorga
- RETICS Asma, reacciones adversas y alérgicas (ARADYAL) and RICORS Red De Enfermedades Inflamatorias (REI), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Allergy Unit and Research Laboratory, Hospital Regional Universitario de Málaga-HRUM, Instituto de investigación Biomédica de Málaga -IBIMA-Plataforma BIONAND, Málaga, Spain
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Frank Redegeld
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, UK
| | - Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
- The interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, Vienna, Austria
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King's College London, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| |
Collapse
|
6
|
Moutafi MK, Bates KM, Aung TN, Milian RG, Xirou V, Vathiotis IA, Gavrielatou N, Angelakis A, Schalper KA, Salichos L, Rimm DL. High-throughput transcriptome profiling indicates ribosomal RNAs to be associated with resistance to immunotherapy in non-small cell lung cancer (NSCLC). J Immunother Cancer 2024; 12:e009039. [PMID: 38857914 PMCID: PMC11168162 DOI: 10.1136/jitc-2024-009039] [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: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Despite the impressive outcomes with immune checkpoint inhibitor (ICI) in non-small cell lung cancer (NSCLC), only a minority of the patients show long-term benefits from ICI. In this study, we used retrospective cohorts of ICI treated patients with NSCLC to discover and validate spatially resolved protein markers associated with resistance to programmed cell death protein-1 (PD-1) axis inhibition. METHODS Pretreatment samples from 56 patients with NSCLC treated with ICI were collected and analyzed in a tissue microarray (TMA) format in including four different tumor regions per patient using the GeoMx platform for spatially informed transcriptomics. 34 patients had assessable tissue with tumor compartment in all 4 TMA spots, 22 with leukocyte compartment and 12 with CD68 compartment. The patients' tissue that was not assessable in fourfold redundancy in each compartment was designated as the validation cohort; cytokeratin (CK) (N=22), leukocytes CD45 (N=31), macrophages, CD68 (N=43). The human whole transcriptome, represented by~18,000 individual genes assessed by oligonucleotide-tagged in situ hybridization, was sequenced on the NovaSeq platform to quantify the RNAs present in each region of interest. RESULTS 54,000 gene variables were generated per case, from them 25,740 were analyzed after removing targets with expression lower than a prespecified frequency. Cox proportional-hazards model analysis was performed for overall and progression-free survival (OS, PFS, respectively). After identifying genes significantly associated with limited survival benefit (HR>1)/progression per spot per patient, we used the intersection of them across the four TMA spots per patient. This resulted in a list of 12 genes in the tumor-cell compartment (RPL13A, GNL3, FAM83A, CYBA, ACSL4, SLC25A6, EPAS1, RPL5, APOL1, HSPD1, RPS4Y1, ADI1). RPL13A, GNL3 in tumor-cell compartment were also significantly associated with OS and PFS, respectively, in the validation cohort (CK: HR, 2.48; p=0.02 and HR, 5.33; p=0.04). In CD45 compartment, secreted frizzled-related protein 2, was associated with OS in the discovery cohort but not in the validation cohort. Similarly, in the CD68 compartment ARHGAP and PNN interacting serine and arginine rich protein were significantly associated with PFS and OS, respectively, in the majority but not all four spots per patient. CONCLUSION This work highlights RPL13A and GNL3 as potential indicative biomarkers of resistance to PD-1 axis blockade that might help to improve precision immunotherapy strategies for lung cancer.
Collapse
Affiliation(s)
- Myrto K Moutafi
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Katherine M Bates
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thazin Nwe Aung
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Rolando Garcia Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, Connecticut, USA
| | - Vasiliki Xirou
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Ioannis A Vathiotis
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Niki Gavrielatou
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Athanasios Angelakis
- Epidemiology and Data Science, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Department of Methodology, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Leonidas Salichos
- Biomedical Data Science Center Director, Center for Cancer Research, Department of Computational Biology at New York Institute of Technology, New York Institute of Technology, Old Westbury, New York, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Hu D, Zhang Z, Liu X, Wu Y, An Y, Wang W, Yang M, Pan Y, Qiao K, Du C, Zhao Y, Li Y, Bao J, Qin T, Pan Y, Xia Z, Zhao X, Sun K. Generalizable transcriptome-based tumor malignant level evaluation and molecular subtyping towards precision oncology. J Transl Med 2024; 22:512. [PMID: 38807223 PMCID: PMC11134716 DOI: 10.1186/s12967-024-05326-0] [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: 02/21/2024] [Accepted: 05/19/2024] [Indexed: 05/30/2024] Open
Abstract
In cancer treatment, therapeutic strategies that integrate tumor-specific characteristics (i.e., precision oncology) are widely implemented to provide clinical benefits for cancer patients. Here, through in-depth integration of tumor transcriptome and patients' prognoses across cancers, we investigated dysregulated and prognosis-associated genes and catalogued such important genes in a cancer type-dependent manner. Utilizing the expression matrices of these genes, we built models to quantitatively evaluate the malignant levels of tumors across cancers, which could add value to the clinical staging system for improved prediction of patients' survival. Furthermore, we performed a transcriptome-based molecular subtyping on hepatocellular carcinoma, which revealed three subtypes with significantly diversified clinical outcomes, mutation landscapes, immune microenvironment, and dysregulated pathways. As tumor transcriptome was commonly profiled in clinical practice with low experimental complexity and cost, this work proposed easy-to-perform approaches for practical clinical promotion towards better healthcare and precision oncology of cancer patients.
Collapse
Affiliation(s)
- Dingxue Hu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ziteng Zhang
- Hepato-Biliary Surgery Division, The Second Affiliated Hospital, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Xiaoyi Liu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Youchun Wu
- Hepato-Biliary Surgery Division, The Second Affiliated Hospital, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Yunyun An
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Wanqiu Wang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Mengqi Yang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yuqi Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Kun Qiao
- Thoracic Surgical Department, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518100, China
| | - Changzheng Du
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Beijing Tsinghua Changgung Hospital, Tsinghua University School of Medicine, Beijing, 102218, China
| | - Yu Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yan Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
- Integrative Microecology Clinical Center, Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Technology Research Center of Gut Microbiota Transplantation, Shenzhen Hospital, Southern Medical University, Shenzhen, 510086, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Tao Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat- Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat- Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhaohua Xia
- Thoracic Surgical Department, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518100, China.
| | - Xin Zhao
- Hepato-Biliary Surgery Division, The Second Affiliated Hospital, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China.
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
8
|
Zhang L, Zhang X, Guan M, Zeng J, Yu F, Lai F. Machine-learning developed an iron, copper, and sulfur-metabolism associated signature predicts lung adenocarcinoma prognosis and therapy response. Respir Res 2024; 25:206. [PMID: 38745285 PMCID: PMC11092068 DOI: 10.1186/s12931-024-02839-6] [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: 11/08/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Previous studies have largely neglected the role of sulfur metabolism in LUAD, and no study has combine iron, copper, and sulfur-metabolism associated genes together to create prognostic signatures. METHODS This study encompasses 1564 LUAD patients, 1249 NSCLC patients, and over 10,000 patients with various cancer types from diverse cohorts. We employed the R package ConsensusClusterPlus to separate patients into different ICSM (Iron, Copper, and Sulfur-Metabolism) subtypes. Various machine-learning methods were utilized to develop the ICSMI. Enrichment analyses were conducted using ClusterProfiler and GSVA, while IOBR quantified immune cell infiltration. GISTIC2.0 and maftools were utilized for CNV and SNV data analysis. The Oncopredict package predicted drug information based on GDSC1. TIDE algorithm and cohorts GSE91061 and IMvigor210 evaluated patient response to immunotherapy. Single-cell data was processed using the Seurat package, AUCell package calculated cells geneset activity scores, and the Scissor algorithm identified ICSMI-associated cells. In vitro experiments was conducted to explore the role of ICSMRGs in LUAD. RESULTS Unsupervised clustering identified two distinct ICSM subtypes of LUAD, each with unique clinical characteristics. The ICSMI, comprising 10 genes, was constructed using integrated machine-learning methods. Its prognostic power was validated in 10 independent datasets, revealing that LUAD patients with higher ICSMI levels had poorer prognoses. Furthermore, ICSMI demonstrated superior predictive abilities compared to 102 previously published signatures. A nomogram incorporating ICSMI and clinical features exhibited high predictive performance. ICSMI positively correlated with patients gene mutations, and integrated analysis of bulk and single-cell transcriptome data revealed its association with TME modulators. Cells representing the high-ICSMI phenotype exhibited more malignant features. LUAD patients with high ICSMI levels exhibited sensitivity to chemotherapy and targeted therapy but displayed resistance to immunotherapy. In a comprehensive analysis across various cancers, ICSMI retained significant prognostic value and emerged as a risk factor for the majority of cancer patients. CONCLUSIONS ICSMI provides critical prognostic insights for LUAD patients, offering valuable insights into the tumor microenvironment and predicting treatment responsiveness.
Collapse
Affiliation(s)
- Liangyu Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xun Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Maohao Guan
- Department of Thoracic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jianshen Zeng
- Department of Thoracic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Fancai Lai
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
9
|
Zhang L, Zhang X, Guan M, Zeng J, Yu F, Lai F. Identification of a novel ADCC-related gene signature for predicting the prognosis and therapy response in lung adenocarcinoma. Inflamm Res 2024; 73:841-866. [PMID: 38507067 DOI: 10.1007/s00011-024-01871-y] [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: 12/15/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Previous studies have largely neglected the role of ADCC in LUAD, and no study has systematically compiled ADCC-associated genes to create prognostic signatures. METHODS In this study, 1564 LUAD patients, 2057 NSCLC patients, and more than 5000 patients with various cancer types from diverse cohorts were included. R package ConsensusClusterPlus was utilized to classify patients into different subtypes. A number of machine-learning algorithms were used to construct the ADCCRS. GSVA and ClusterProfiler were used for enrichment analyses, and IOBR was used to quantify immune cell infiltration level. GISTIC2.0 and maftools were used to analyze the CNV and SNV data. The Oncopredict package was used to predict drug information based on the GDSC1. Three immunotherapy cohorts were used to evaluate patient response to immunotherapy. The Seurat package was used to process single-cell data, the AUCell package was used to calculate cells' geneset activity scores, and the Scissor algorithm was used to identify ADCCRS-associated cells. RESULTS Through unsupervised clustering, two distinct subtypes of LUAD were identified, each exhibiting distinct clinical characteristics. The ADCCRS, consisted of 16 genes, was constructed by integrated machine-learning methods. The prognostic power of ADCCRS was validated in 28 independent datasets. Further, ADCCRS shows better predictive abilities than 102 previously published signatures in predicting LUAD patients' survival. A nomogram incorporating ADCCRS and clinical features was constructed, demonstrating high predictive performance. ADCCRS positively correlates with patients' gene mutation, and integrated analysis of bulk and single-cell transcriptome data revealed the association of ADCCRS with TME modulators. Cells representing high-ADCCRS phenotype exhibited more malignant features. LUAD patients with high ADCCRS levels exhibited sensitivity to chemotherapy and targeted therapy, while displaying resistance to immunotherapy. In pan-cancer analysis, ADCCRS still exhibited significant prognostic value and was found to be a risk factor for most cancer patients. CONCLUSIONS ADCCRS offers a critical prognostic insight for patients with LUAD, shedding light on the tumor microenvironment and forecasting treatment responsiveness.
Collapse
Affiliation(s)
- Liangyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Xun Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Maohao Guan
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Jianshen Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China.
| | - Fancai Lai
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the Fitst Affiliated Hospiral, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
10
|
Yu L, Lin N, Ye Y, Zhuang H, Zou S, Song Y, Chen X, Wang Q. The prognosis, chemotherapy and immunotherapy efficacy of the SUMOylation pathway signature and the role of UBA2 in lung adenocarcinoma. Aging (Albany NY) 2024; 16:4378-4395. [PMID: 38407971 PMCID: PMC10968705 DOI: 10.18632/aging.205594] [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: 10/18/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Lung adenocarcinoma (LUAD) is one of the most common malignant tumors worldwide. Small Ubiquitin-like Modifier (SUMO)-ylation plays a crucial role in tumorigenesis. However, the SUMOylation pathway landscape and its clinical implications in LUAD remain unclear. Here, we analyzed genes involved in the SUMOylation pathway in LUAD and constructed a SUMOylation pathway signature (SUMOPS) using the LASSO-Cox regression model, validated in independent cohorts. Our analysis revealed significant dysregulation of SUMOylation-related genes in LUAD, comprising of favorable or unfavorable prognostic factors. The SUMOPS model was associated with established molecular and histological subtypes of LUAD, highlighting its clinical relevance. The SUMOPS stratified LUAD patients into SUMOPS-high and SUMOPS-low subtypes with distinct survival outcomes and adjuvant chemotherapy responses. The SUMOPS-low subtype showed favorable responses to adjuvant chemotherapy. The correlations between SUMOPS scores and immune cell infiltration suggested that patients with the SUMOPS-high subtype exhibited favorable immune profiles for immune checkpoint inhibitor (ICI) treatment. Additionally, we identified UBA2 as a key SUMOylation-related gene with an increased expression and a poor prognosis in LUAD. Cell function experiment confirmed the role of UBA2 in promoting LUAD cell proliferation, invasion, and migration. These findings provide valuable insights into the SUMOylation pathway and its prognostic implications in LUAD, paving the way for personalized treatment strategies and the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Na Lin
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Yan Ye
- Jiangxi Health Commission Key Laboratory of Leukemia, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, China
| | - Haohan Zhuang
- Laboratory Animal Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Shumei Zou
- 900 Hospital of The Joint Logistics Team, Fuzhou, Fujian 350001, China
| | - Yingfang Song
- 900 Hospital of The Joint Logistics Team, Fuzhou, Fujian 350001, China
- Department of Pulmonary and Critical Care Medicine, Fuzong Clinical College of Fujian Medical University, Fuzhou, Fujian 350001, China
- Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China
| | - Xiaoli Chen
- Jiangxi Health Commission Key Laboratory of Leukemia, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, China
| | - Qingshui Wang
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, China
| |
Collapse
|
11
|
Serafini MS, Cavalieri S, Licitra L, Pistore F, Lenoci D, Canevari S, Airoldi M, Cossu Rocca M, Strojan P, Kuhar CG, Merlano M, Perrone F, Vingiani A, Denaro N, Perri F, Argiris A, Gurizzan C, Ghi MG, Cassano A, Allegrini G, Bossi P, De Cecco L. Association of a gene-expression subtype to outcome and treatment response in patients with recurrent/metastatic head and neck squamous cell carcinoma treated with nivolumab. J Immunother Cancer 2024; 12:e007823. [PMID: 38290766 PMCID: PMC10828850 DOI: 10.1136/jitc-2023-007823] [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: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors have been approved and currently used in the clinical management of recurrent and metastatic head and neck squamous cell carcinoma (R/M HNSCC) patients. The reported benefit in clinical trials is variable and heterogeneous. Our study aims at exploring and comparing the predictive role of gene-expression signatures with classical biomarkers for immunotherapy-treated R/M HNSCC patients in a multicentric phase IIIb trial. METHODS Clinical data were prospectively collected in Nivactor tiral (single-arm, open-label, multicenter, phase IIIb clinical trial in platinum-refractory HNSCC treated with nivolumab). Findings were validated in an external independent cohort of immune-treated HNSCC patients, divided in long-term and short-term survivors (overall survival >18 and <6 months since the start of immunotherapy, respectively). Pretreatment tumor tissue specimen from immunotherapy-treated R/M HNSCC patients was used for PD-L1 (Tumor Proportion Score; Combined Positive Score (CPS)) and Tumor Mutational Burden (Oncopanel TSO500) evaluation and gene expression profiling; classical biomarkers and immune signatures (retrieved from literature) were challenged in the NIVACTOR dataset. RESULTS Cluster-6 (Cl6) stratification of NIVACTOR cases in high score (n=16, 20%) and low score (n=64, 80%) demonstrated a statistically significant and clinically meaningful improvement in overall survival in the high-score cases (p=0.00028; HR=4.34, 95% CI 1.84 to 10.22) and discriminative ability reached area under the curve (AUC)=0.785 (95% CI 0.603 to 0.967). The association of high-score Cl6 with better outcome was also confirmed in: (1) NIVACTOR progression-free survival (p=4.93E-05; HR=3.71, 95% CI 1.92 to 7.18) and objective-response-rate (AUC=0.785; 95% CI 0.603 to 0.967); (2) long survivors versus short survivors (p=0.00544). In multivariate Cox regression analysis, Cl6 was independent from Eastern Cooperative Oncology Group performance status, PDL1-CPS, and primary tumor site. CONCLUSIONS These data highlight the presence of underlying biological differences able to predict survival and response following treatment with immunotherapy in platinum-refractory R/M HNSCC that could have translational implications improving treatment selection. TRIAL REGISTRATION NUMBER EudraCT Number: 2017-000562-30.
Collapse
Affiliation(s)
- Mara Serena Serafini
- Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milano, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milano, Italy
| | - Federico Pistore
- Head and Neck Medical Oncology, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milan, Italy
| | - Deborah Lenoci
- Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Mario Airoldi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | | | | | - Cvetka Grasic Kuhar
- University of Ljubljana, Ljubljana, Slovenia
- Institute of Oncology, Ljubljana, Slovenia
| | | | - Federica Perrone
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Vingiani
- Department of Oncology and Hemato-oncology, University of Milan, Milano, Italy
- Department of Diagnostic Pathology and Laboratory, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Francesco Perri
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Athanassios Argiris
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Cristina Gurizzan
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Maria Grazia Ghi
- Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy
| | - Alessandra Cassano
- Policlinico Universitario Agostino Gemelli Dipartimento di scienze mediche e chirurgiche, Roma, Italy
| | | | - Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Loris De Cecco
- Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
12
|
Yiong CS, Lin TP, Lim VY, Toh TB, Yang VS. Biomarkers for immune checkpoint inhibition in sarcomas - are we close to clinical implementation? Biomark Res 2023; 11:75. [PMID: 37612756 PMCID: PMC10463641 DOI: 10.1186/s40364-023-00513-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Sarcomas are a group of diverse and complex cancers of mesenchymal origin that remains poorly understood. Recent developments in cancer immunotherapy have demonstrated a potential for better outcomes with immune checkpoint inhibition in some sarcomas compared to conventional chemotherapy. Immune checkpoint inhibitors (ICIs) are key agents in cancer immunotherapy, demonstrating improved outcomes in many tumor types. However, most patients with sarcoma do not benefit from treatment, highlighting the need for identification and development of predictive biomarkers for response to ICIs. In this review, we first discuss United States (US) Food and Drug Administration (FDA)-approved and European Medicines Agency (EMA)-approved biomarkers, as well as the limitations of their use in sarcomas. We then review eight potential predictive biomarkers and rationalize their utility in sarcomas. These include gene expression signatures (GES), circulating neutrophil-to-lymphocyte ratio (NLR), indoleamine 2,3-dioxygenase (IDO), lymphocyte activation gene 3 (LAG-3), T cell immunoglobin and mucin domain-containing protein 3 (TIM-3), TP53 mutation status, B cells, and tertiary lymphoid structures (TLS). Finally, we discuss the potential for TLS as both a predictive and prognostic biomarker for ICI response in sarcomas to be implemented in the clinic.
Collapse
Affiliation(s)
- Chin Sern Yiong
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Tzu Ping Lin
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Vivian Yujing Lim
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Valerie Shiwen Yang
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Duke-NUS Medical School, Oncology Academic Clinical Program, Singapore, 169857, Singapore.
| |
Collapse
|
13
|
Larionova I, Tashireva L. Immune gene signatures as prognostic criteria for cancer patients. Ther Adv Med Oncol 2023; 15:17588359231189436. [PMID: 37547445 PMCID: PMC10399276 DOI: 10.1177/17588359231189436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Recently, the possibility of using immune gene signatures (IGSs) has been considered as a novel prognostic tool for numerous cancer types. State-of-the-art methods of genomic, transcriptomic, and protein analysis have allowed the identification of a number of immune signatures correlated to disease outcome. The major adaptive and innate immune components are the T lymphocytes and macrophages, respectively. Herein, we collected essential data on IGSs consisting of subsets of T cells and tumor-associated macrophages and indicating cancer patient outcomes. We discuss factors that can introduce errors in the recognition of immune cell types and explain why the significance of immune signatures can be interpreted with uncertainty. The unidirectional functions of cell types should be entirely addressed in the signatures constructed by the combination of innate and adaptive immune cells. The state of the antitumor immune response is the key basis for IGSs and should be considered in gene signature construction. We also analyzed immune signatures for the prediction of immunotherapy response. Finally, we attempted to explain the present-day limitations in the use of immune signatures as robust criteria for prognosis.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 36 Lenina Av., Tomsk 634050, Russia
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Liubov Tashireva
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
14
|
Sadee W, Wang D, Hartmann K, Toland AE. Pharmacogenomics: Driving Personalized Medicine. Pharmacol Rev 2023; 75:789-814. [PMID: 36927888 PMCID: PMC10289244 DOI: 10.1124/pharmrev.122.000810] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Personalized medicine tailors therapies, disease prevention, and health maintenance to the individual, with pharmacogenomics serving as a key tool to improve outcomes and prevent adverse effects. Advances in genomics have transformed pharmacogenetics, traditionally focused on single gene-drug pairs, into pharmacogenomics, encompassing all "-omics" fields (e.g., proteomics, transcriptomics, metabolomics, and metagenomics). This review summarizes basic genomics principles relevant to translation into therapies, assessing pharmacogenomics' central role in converging diverse elements of personalized medicine. We discuss genetic variations in pharmacogenes (drug-metabolizing enzymes, drug transporters, and receptors), their clinical relevance as biomarkers, and the legacy of decades of research in pharmacogenetics. All types of therapies, including proteins, nucleic acids, viruses, cells, genes, and irradiation, can benefit from genomics, expanding the role of pharmacogenomics across medicine. Food and Drug Administration approvals of personalized therapeutics involving biomarkers increase rapidly, demonstrating the growing impact of pharmacogenomics. A beacon for all therapeutic approaches, molecularly targeted cancer therapies highlight trends in drug discovery and clinical applications. To account for human complexity, multicomponent biomarker panels encompassing genetic, personal, and environmental factors can guide diagnosis and therapies, increasingly involving artificial intelligence to cope with extreme data complexities. However, clinical application encounters substantial hurdles, such as unknown validity across ethnic groups, underlying bias in health care, and real-world validation. This review address the underlying science and technologies germane to pharmacogenomics and personalized medicine, integrated with economic, ethical, and regulatory issues, providing insights into the current status and future direction of health care. SIGNIFICANCE STATEMENT: Personalized medicine aims to optimize health care for the individual patients with use of predictive biomarkers to improve outcomes and prevent adverse effects. Pharmacogenomics drives biomarker discovery and guides the development of targeted therapeutics. This review addresses basic principles and current trends in pharmacogenomics, with large-scale data repositories accelerating medical advances. The impact of pharmacogenomics is discussed, along with hurdles impeding broad clinical implementation, in the context of clinical care, ethics, economics, and regulatory affairs.
Collapse
Affiliation(s)
- Wolfgang Sadee
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Danxin Wang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Katherine Hartmann
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Amanda Ewart Toland
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| |
Collapse
|
15
|
Wang H, Arulraj T, Kimko H, Popel AS. Generating immunogenomic data-guided virtual patients using a QSP model to predict response of advanced NSCLC to PD-L1 inhibition. NPJ Precis Oncol 2023; 7:55. [PMID: 37291190 DOI: 10.1038/s41698-023-00405-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Generating realistic virtual patients from a limited amount of patient data is one of the major challenges for quantitative systems pharmacology modeling in immuno-oncology. Quantitative systems pharmacology (QSP) is a mathematical modeling methodology that integrates mechanistic knowledge of biological systems to investigate dynamics in a whole system during disease progression and drug treatment. In the present analysis, we parameterized our previously published QSP model of the cancer-immunity cycle to non-small cell lung cancer (NSCLC) and generated a virtual patient cohort to predict clinical response to PD-L1 inhibition in NSCLC. The virtual patient generation was guided by immunogenomic data from iAtlas portal and population pharmacokinetic data of durvalumab, a PD-L1 inhibitor. With virtual patients generated following the immunogenomic data distribution, our model predicted a response rate of 18.6% (95% bootstrap confidence interval: 13.3-24.2%) and identified CD8/Treg ratio as a potential predictive biomarker in addition to PD-L1 expression and tumor mutational burden. We demonstrated that omics data served as a reliable resource for virtual patient generation techniques in immuno-oncology using QSP models.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Theinmozhi Arulraj
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Holly Kimko
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, Gaithersburg, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Oncology, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
16
|
Hua L, Wu J, Ge J, Li X, You B, Wang W, Hu B. Identification of lung adenocarcinoma subtypes and predictive signature for prognosis, immune features, and immunotherapy based on immune checkpoint genes. Front Cell Dev Biol 2023; 11:1060086. [PMID: 37234773 PMCID: PMC10206047 DOI: 10.3389/fcell.2023.1060086] [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: 10/02/2022] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is the most common variant of non-small cell lung cancer (NSCLC) across the world. Recently, the rapid development of immunotherapy has brought a new dawn for LUAD patients. Closely related to the tumor immune microenvironment and immune cell functions, more and more new immune checkpoints have been discovered, and various cancer treatment studies targeting these novel immune checkpoints are currently in full swing. However, studies on the phenotype and clinical significance of novel immune checkpoints in LUAD are still limited, and only a minority of patients with LUAD can benefit from immunotherapy. Methods: The LUAD datasets were downloaded from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases, and the immune checkpoints score of each sample were calculated based on the expression of the 82 immune checkpoints-related genes (ICGs). The weighted gene co-expression network analysis (WGCNA) was used to obtain the gene modules closely related to the score and two different LUAD clusters were identified based on these module genes by the Non-negative Matrix Factorization (NMF) Algorithm. The differentially expressed genes between the two clusters were further used to construct a predictive signature for prognosis, immune features, and the response to immunotherapy for LUAD patients through a series of regression analyses. Results: A new immune checkpoints-related signature was finally established according to the expression of 7 genes (FCER2, CD200R1, RHOV, TNNT2, WT1, AHSG, and KRTAP5-8). This signature can stratify patients into high-risk and low-risk groups with different survival outcomes and sensitivity to immunotherapy, and the signature has been well validated in different clinical subgroups and validation cohorts. Conclusion: We constructed a novel immune checkpoints-related LUAD risk assessment system, which has a good predictive ability and significance for guiding immunotherapy. We believe that these findings will not only aid in the clinical management of LUAD patients but also provide some insights into screening appropriate patients for immunotherapy.
Collapse
Affiliation(s)
- Linbin Hua
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiashu Ge
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xin Li
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bin You
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Wang H, Arulraj T, Kimko H, Popel AS. Generating immunogenomic data-guided virtual patients using a QSP model to predict response of advanced NSCLC to PD-L1 inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538191. [PMID: 37162938 PMCID: PMC10168221 DOI: 10.1101/2023.04.25.538191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Generating realistic virtual patients from a limited amount of patient data is one of the major challenges for quantitative systems pharmacology modeling in immuno-oncology. Quantitative systems pharmacology (QSP) is a mathematical modeling methodology that integrates mechanistic knowledge of biological systems to investigate dynamics in a whole system during disease progression and drug treatment. In the present analysis, we parameterized our previously published QSP model of the cancer-immunity cycle to non-small cell lung cancer (NSCLC) and generated a virtual patient cohort to predict clinical response to PD-L1 inhibition in NSCLC. The virtual patient generation was guided by immunogenomic data from iAtlas portal and population pharmacokinetic data of durvalumab, a PD-L1 inhibitor. With virtual patients generated following the immunogenomic data distribution, our model predicted a response rate of 18.6% (95% bootstrap confidence interval: 13.3-24.2%) and identified CD8/Treg ratio as a potential predictive biomarker in addition to PD-L1 expression and tumor mutational burden. We demonstrated that omics data served as a reliable resource for virtual patient generation techniques in immuno-oncology using QSP models.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Theinmozhi Arulraj
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Holly Kimko
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, Gaithersburg, MD, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|