1
|
Dai H, Zhang X, Zhao Y, Nie J, Hang Z, Huang X, Ma H, Wang L, Li Z, Wu M, Fan J, Jiang K, Luo W, Qin C. ADME gene-driven prognostic model for bladder cancer: a breakthrough in predicting survival and personalized treatment. Hereditas 2025; 162:42. [PMID: 40108724 PMCID: PMC11921678 DOI: 10.1186/s41065-025-00409-4] [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: 01/15/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Genes that participate in the absorption, distribution, metabolism, excretion (ADME) processes occupy a central role in pharmacokinetics. Meanwhile, variability in clinical outcomes and responses to treatment is notable in bladder cancer (BLCA). METHODS Our study utilized expansive datasets from TCGA and the GEO to explore prognostic factors in bladder cancer. Utilizing both univariate Cox regression and the lasso regression techniques, we identified ADME genes critical for patient outcomes. Utilizing genes identified in our study, a model for assessing risk was constructed. The evaluation of this model's predictive precision was conducted using Kaplan-Meier survival curves and assessments based on ROC curves. Furthermore, we devised a predictive nomogram, offering a straightforward visualization of crucial prognostic indicators. To explore the potential factors mediating the differences in outcomes between high and low risk groups, we performed comprehensive analyses including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG)-based enrichment analyses, immune infiltration variations, somatic mutation landscapes, and pharmacological sensitivity response assessment etc. Immediately following this, we selected core genes based on the PPI network and explored the prognostic potential of the core genes as well as immune modulation, and pathway activation. And the differential expression was verified by immunohistochemistry and qRT-PCR. Finally we explored the potential of the core genes as pan-cancer biomarkers. RESULTS Our efforts culminated in the establishment of a validated 17-gene ADME-centered risk prediction model, displaying remarkable predictive accuracy for BLCA prognosis. Through separate cox regression analyses, the importance of the model's risk score in forecasting BLCA outcomes was substantiated. Furthermore, a novel nomogram incorporating clinical variables alongside the risk score was introduced. Comprehensive studies established a strong correlation between the risk score and several key indicators: patterns of immune cell infiltration, reactions to immunotherapy, landscape of somatic mutation and profiles of drug sensitivity. We screened the core prognostic gene CYP2C8, explored its role in tumor bioregulation and validated its upregulated expression in bladder cancer. Furthermore, we found that it can serve as a reliable biomarker for pan-cancer. CONCLUSION The risk assessment model formulated in our research stands as a formidable instrument for forecasting BLCA prognosis, while also providing insights into the disease's progression mechanisms and guiding clinical decision-making strategies.
Collapse
Affiliation(s)
- Haojie Dai
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xi Zhang
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
- Department of Urology, The First Affliated Hospital of Nanjing Medical University, Nanjing, China
| | - You Zhao
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jun Nie
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zhenyu Hang
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xin Huang
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hongxiang Ma
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Li Wang
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zihao Li
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ming Wu
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jun Fan
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ke Jiang
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Weiping Luo
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Chao Qin
- The Affliated Liyang People's Hospital of Kangda College of Nanjing Medical University, Changzhou, Jiangsu, China
- Department of Urology, The First Affliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Wang H, Li F, Wang Q, Guo X, Chen X, Zou X, Yuan J. Identifying ADME-related gene signature for immune landscape and prognosis in KIRC by single-cell and spatial transcriptome analysis. Sci Rep 2025; 15:1294. [PMID: 39779746 PMCID: PMC11711672 DOI: 10.1038/s41598-024-84018-7] [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: 06/07/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is the most prevalent subtype of kidney cancer. Although multiple therapeutic agents have been proven effective in KIRC, their clinical application has been hindered by a lack of reliable biomarkers. This study focused on the prognostic value and function of drug absorption, distribution, metabolism, and excretion- (ADME-) related genes (ARGs) in KIRC to enhance personalized therapy. The critical role of ARGs in KIRC microenvironment was confirmed by single cell RNA-seq analysis and spatial transcriptome sequencing analysis for the first time. Then, an ADME-related prognostic signature (ARPS) was developed by the bulk RNA-seq analysis. The ARPS, created through Cox regression, LASSO, and stepAIC analyses, identified eight ARGs that stratified patients into high-risk and low-risk groups. High-risk patients had significantly poorer overall survival. Multivariate analysis confirmed the independent predictive ability of ARPS, and an ARPS-based nomogram was constructed for clinical application. Gene ontology and KEGG pathway analyses revealed immune-related functions and pathways enriched in these groups, with low-risk patients showing better responses to immunotherapy. Finally, the expression of ARGs was validated by qRT-PCR and Western blotting experiments. These findings underscore the prognostic significance of ARPS in KIRC and its potential application in guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Hongyun Wang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Feizhou Li
- Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Qiong Wang
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xinyuan Guo
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xinbing Chen
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xinrong Zou
- Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.
- Hubei University of Chinese Medicine, Wuhan, 430065, China.
- Institute of Chinese Medicine Nephrology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
| | - Jun Yuan
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
Tremmel R, Hübschmann D, Schaeffeler E, Pirmann S, Fröhling S, Schwab M. Innovation in cancer pharmacotherapy through integrative consideration of germline and tumor genomes. Pharmacol Rev 2025; 77:100014. [PMID: 39952686 DOI: 10.1124/pharmrev.124.001049] [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: 04/03/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 01/22/2025] Open
Abstract
Precision cancer medicine is widely established, and numerous molecularly targeted drugs for various tumor entities are approved or are in development. Personalized pharmacotherapy in oncology has so far been based primarily on tumor characteristics, for example, somatic mutations. However, the response to drug treatment also depends on pharmacological processes summarized under the term ADME (absorption, distribution, metabolism, and excretion). Variations in ADME genes have been the subject of intensive research for >5 decades, considering individual patients' genetic makeup, referred to as pharmacogenomics (PGx). The combined impact of a patient's tumor and germline genome is only partially understood and often not adequately considered in cancer therapy. This may be attributed, in part, to the lack of methods for combined analysis of both data layers. Optimized personalized cancer therapies should, therefore, aim to integrate molecular information, which derives from both the tumor and the germline genome, and taking into account existing PGx guidelines for drug therapy. Moreover, such strategies should provide the opportunity to consider genetic variants of previously unknown functional significance. Bioinformatic analysis methods and corresponding algorithms for data interpretation need to be developed to integrate PGx data in cancer therapy with a special meaning for interdisciplinary molecular tumor boards, in which cancer patients are discussed to provide evidence-based recommendations for clinical management based on individual tumor profiles. SIGNIFICANCE STATEMENT: The era of personalized oncology has seen the emergence of drugs tailored to genetic variants associated with cancer biology. However, the full potential of targeted therapy remains untapped owing to the predominant focus on acquired tumor-specific alterations. Optimized cancer care must integrate tumor and patient genomes, guided by pharmacogenomic principles. An essential prerequisite for realizing truly personalized drug treatment of cancer patients is the development of bioinformatic tools for comprehensive analysis of all data layers generated in modern precision oncology programs.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Daniel Hübschmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between the German Cancer Research Center (DKFZ) and Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Innovation and Service Unit for Bioinformatics and Precision Medicine, DKFZ, Heidelberg, Germany; Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
| | - Sebastian Pirmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between the German Cancer Research Center (DKFZ) and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany; Departments of Clinical Pharmacology, and Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany; DKTK, DKFZ, Partner Site Tuebingen, Tuebingen, Germany; NCT SouthWest, a partnership between DKFZ and University Hospital Tuebingen, Tuebingen, Germany.
| |
Collapse
|
4
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. A transcriptomic comparison of in vitro models of the human placenta. Placenta 2025; 159:52-61. [PMID: 39637677 PMCID: PMC11857522 DOI: 10.1016/j.placenta.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. The aim of this research was to compare the transcriptomes of common in vitro models of the human placenta compared to bulk human placental tissue. METHODS We performed differential gene expression analysis on publicly available transcriptomic data from 7 in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, Villous Explants, and Trophoblast Stem Cells) and compared to bulk placental tissue from 2 cohort studies (CANDLE and GAPPS) or individual trophoblast cell types derived from bulk placental tissue. RESULTS All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs = 10,624), and the individual trophoblast cell types (Average DEGs = 5413), indicating that there are vast differences in gene expression. Hierarchical clustering identified 54 gene clusters with distinct expression profiles across placental models, with 23 clusters enriched for specific KEGG pathways. Placental cell lines were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as female origin, while JEG-3, JAR, and BeWo cells are of male origin. DISCUSSION None of the models were a close approximation of the human bulk placental transcriptome, highlighting the challenges with model selection. To enable appropriate model selection, we adapted our data into a web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)".
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Evan J Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, 98101, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| |
Collapse
|
5
|
Zhang H, Huang W, Chen M, Liu Y, Yan B, Mou S, Jiang W, Mei H. Research on molecular characteristics of ADME-related genes in kidney renal clear cell carcinoma. Sci Rep 2024; 14:16834. [PMID: 39039118 PMCID: PMC11263354 DOI: 10.1038/s41598-024-67516-6] [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: 02/17/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024] Open
Abstract
Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) are named ADME genes. However, the comprehensive role of ADME genes in kidney renal clear cell carcinoma (KIRC) remains unclear. Using the clinical and gene expression data of KIRC patients downloaded from The Cancer Genome Atlas (TCGA), ArrayExpress, and the Gene Expression Omnibus (GEO) databases, we cluster patients into two patterns, and the population with a relatively poor prognosis demonstrated higher level of immunosuppressive cell infiltration and higher proportion of glycolytic subtypes. Then, 17 ADME genes combination identified through the least absolute shrinkage and selection operator algorithm (LASSO, 1000 times) was utilized to calculate the ADME score. The ADME score was found to be an independent predictor of prognosis in KIRC and to be tightly associated with the infiltration level of immune cells, metabolic properties, tumor-related signaling pathways, genetic variation, and responses to chemotherapeutics. Our work revealed the characteristics of ADME in KIRC. Assessing the ADME profiles of individual patients can deepen our comprehension of tumor microenvironment (TME) features in KIRC and can aid in developing more personalized and effective therapeutic strategies.
Collapse
Affiliation(s)
- Haiyu Zhang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weisheng Huang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mutong Chen
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Department of Urology, Shantou University Medical College, Shantou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuhan Liu
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bing Yan
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shuanzhu Mou
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wendong Jiang
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
- Department of Urology, Shantou University Medical College, Shantou, China.
- Shenzhen Second People's Hospital, Clinical Medicine College of Anhui Medical University, Shenzhen, China.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
6
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. Transcriptomic comparison of in vitro models of the human placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598695. [PMID: 38915703 PMCID: PMC11195179 DOI: 10.1101/2024.06.14.598695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Studying the human placenta through in vitro cell culture methods is necessary due to limited access and amenability of human placental tissue to certain experimental methods as well as distinct anatomical and physiological differences between animal and human placentas. Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. Therefore, the aim of this research was to create a comprehensive transcriptomic comparison of common in vitro models of the human placenta compared to bulk placental tissue from the CANDLE and GAPPS cohorts (N=1083). We performed differential gene expression analysis on publicly available RNA sequencing data from 6 common in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, and Villous Explants) and compared to CANDLE and GAPPS bulk placental tissue or cytotrophoblast, syncytiotrophoblast, and extravillous trophoblast cell types derived from bulk placental tissue. All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs=10,873), and the individual trophoblast cell types (Average DEGs=5,346), indicating that there are vast differences in gene expression compared to bulk and cell-type specific human placental tissue. Hierarchical clustering identified 53 gene clusters with distinct expression profiles across placental models, with 22 clusters enriched for specific KEGG pathways, 7 clusters enriched for high-expression placental genes, and 7 clusters enriched for absorption, distribution, metabolism, and excretion genes. In vitro placental models were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as being of female origin, while JEG-3, JAR, and BeWo cells are of male origin. Overall, none of the models were a close approximation of the transcriptome of bulk human placental tissue, highlighting the challenges with model selection. To enable researchers to select appropriate models, we have compiled data on differential gene expression, clustering, and fetal sex into an accessible web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)" which can be utilized by researchers to make informed decisions about their selection of in vitro placental models.
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Evan J. Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143 United States
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 55905 United States
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202 United States
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
- Center for Child Health, Behavior and Development, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98101 United States
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
| |
Collapse
|
7
|
Wang J, Wang G, Hu T, Wang H, Zhou Y. Identification of an ADME-related gene for forecasting the prognosis and responding to immunotherapy in sarcomas. Eur J Med Res 2024; 29:45. [PMID: 38212774 PMCID: PMC10782529 DOI: 10.1186/s40001-023-01624-3] [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: 09/15/2023] [Accepted: 12/25/2023] [Indexed: 01/13/2024] Open
Abstract
There are more than 170 subtypes of sarcomas (SARC), which pose a challenge for diagnosis and patient management. Relatively simple or complex karyotypes play an indispensable role in the early diagnosis and effective treatment of SARC. The genes related to absorption, distribution, metabolism, and excretion (ADME) of a drug can serve as prognostic biomarkers of cancer and potential drug targets. In this study, a risk score signature was created. The SARC cohort was downloaded from The Cancer Genome Atlas (TCGA) database, and divided into high-risk group and low-risk group according to the median value of risk score. Compared with high-risk group, low-risk group has a longer survival time, which is also verified in osteosarcoma cohort from Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. In addition, the relationship between the signature and immunophenotypes, including status of immune cell infiltration and immune checkpoint expression, was explored. Then, we found that high-risk group is in immunosuppressive status. Finally, we verified that PPARD played a role as a carcinogen in osteosarcoma, which provided a direction for targeted treatment of osteosarcoma in the future. Generally speaking, the signature can not only help clinicians predict the prognosis of patients with SARC, but also provide a theoretical basis for developing more effective targeted drugs in the future.
Collapse
Affiliation(s)
- Jianlong Wang
- Department of Orthopedics, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Guowei Wang
- Department of Orthopedics, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Tianrui Hu
- Department of Orthopedics, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Hongyi Wang
- Medical College, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Yong Zhou
- Department of Orthopedics, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
8
|
Yan Y, Liu Y, Liang Q, Xu Z. Drug metabolism-related gene ABCA1 augments temozolomide chemoresistance and immune infiltration abundance of M2 macrophages in glioma. Eur J Med Res 2023; 28:373. [PMID: 37749600 PMCID: PMC10518970 DOI: 10.1186/s40001-023-01370-6] [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: 07/14/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
Gliomas are the most prevalent primary tumor in the central nervous system, with an abysmal 5-year survival rate and alarming mortality. The current standard management of glioma is maximum resection of tumors followed by postoperative chemotherapy with temozolomide (TMZ) or radiotherapy. Low chemosensitivity of TMZ in glioma treatment eventuates limited therapeutic efficacy or treatment failure. Hence, overcoming the resistance of glioma to TMZ is a pressing question. Our research centered on identifying the drug metabolism-related genes potentially involved in TMZ-treated resistance of glioma through several bioinformatics datasets and cell experiments. One efflux transporter, ATP-binding cassette transporter subfamily A1 (ABCA1), was discovered with an upregulated expression level and signaled poor clinical outcomes for glioma patients. The transcript level of ABCA1 significantly elevated across the TMZ-resistant glioma cells in contrast with non-resistant cells. Over-expressed ABCA1 restrained the drug activity of TMZ, and ABCA1 knockdown improved the treatment efficacy. Meanwhile, the results of molecular docking between ABCA1 protein and TMZ showed a high binding affinity. Additionally, co-expression and immunological analysis revealed that ABCA1 facilitates the immune infiltration of M2 macrophages in glioma, thereby stimulating tumor growth and aggravating the poor survival of patients. Altogether, we discovered that the ABCA1 transporter was involved in TMZ chemoresistance and the immune infiltration of M2 macrophages in glioma. Treatment with TMZ after ABCA1 knockdown enhances the chemosensitivity, suggesting that inhibition of ABCA1 may be a potential strategy for improving the therapeutic efficacy of gliomas.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
9
|
The Somatic Mutation Landscape of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers. Cancers (Basel) 2022; 14:cancers14225708. [PMID: 36428799 PMCID: PMC9688768 DOI: 10.3390/cancers14225708] [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: 10/07/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has a critical role in the metabolism of anticancer drugs and numerous pro/anti-cancer molecules (e.g., steroids, lipids, fatty acids, bile acids and carcinogens). Recent studies have shown wide and abundant expression of UGT genes in human cancers. However, the extent to which UGT genes acquire somatic mutations within tumors remains to be systematically investigated. In the present study, our comprehensive analysis of the somatic mutation profiles of 10,069 tumors from 33 different TCGA cancer types identified 3427 somatic mutations in UGT genes. Overall, nearly 18% (1802/10,069) of the assessed tumors had mutations in UGT genes with huge variations in mutation frequency across different cancer types, ranging from over 25% in five cancers (COAD, LUAD, LUSC, SKCM and UCSC) to less than 5% in eight cancers (LAML, MESO, PCPG, PAAD, PRAD, TGCT, THYM and UVM). All 22 UGT genes showed somatic mutations in tumors, with UGT2B4, UGT3A1 and UGT3A2 showing the largest number of mutations (289, 307 and 255 mutations, respectively). Nearly 65% (2260/3427) of the mutations were missense, frame-shift and nonsense mutations that have been predicted to code for variant UGT proteins. Furthermore, about 10% (362/3427) of the mutations occurred in non-coding regions (5' UTR, 3' UTR and splice sites) that may be able to alter the efficiency of translation initiation, miRNA regulation or the splicing of UGT transcripts. In conclusion, our data show widespread somatic mutations of UGT genes in human cancers that may affect the capacity of cancer cells to metabolize anticancer drugs and endobiotics that control pro/anti-cancer signaling pathways. This highlights their potential utility as biomarkers for predicting therapeutic efficacy and clinical outcomes.
Collapse
|
10
|
Li F, Yin J, Lu M, Mou M, Li Z, Zeng Z, Tan Y, Wang S, Chu X, Dai H, Hou T, Zeng S, Chen Y, Zhu F. DrugMAP: molecular atlas and pharma-information of all drugs. Nucleic Acids Res 2022; 51:D1288-D1299. [PMID: 36243961 PMCID: PMC9825453 DOI: 10.1093/nar/gkac813] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/30/2022] [Accepted: 10/12/2022] [Indexed: 02/06/2023] Open
Abstract
The efficacy and safety of drugs are widely known to be determined by their interactions with multiple molecules of pharmacological importance, and it is therefore essential to systematically depict the molecular atlas and pharma-information of studied drugs. However, our understanding of such information is neither comprehensive nor precise, which necessitates the construction of a new database providing a network containing a large number of drugs and their interacting molecules. Here, a new database describing the molecular atlas and pharma-information of drugs (DrugMAP) was therefore constructed. It provides a comprehensive list of interacting molecules for >30 000 drugs/drug candidates, gives the differential expression patterns for >5000 interacting molecules among different disease sites, ADME (absorption, distribution, metabolism and excretion)-relevant organs and physiological tissues, and weaves a comprehensive and precise network containing >200 000 interactions among drugs and molecules. With the great efforts made to clarify the complex mechanism underlying drug pharmacokinetics and pharmacodynamics and rapidly emerging interests in artificial intelligence (AI)-based network analyses, DrugMAP is expected to become an indispensable supplement to existing databases to facilitate drug discovery. It is now fully and freely accessible at: https://idrblab.org/drugmap/.
Collapse
Affiliation(s)
| | | | - Mingkun Lu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhaorong Li
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba–Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Zhenyu Zeng
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba–Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Ying Tan
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Shanshan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Xinyi Chu
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Haibin Dai
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- Correspondence may also be addressed to Su Zeng.
| | - Yuzong Chen
- Correspondence may also be addressed to Yuzong Chen.
| | - Feng Zhu
- To whom correspondence should be addressed.
| |
Collapse
|
11
|
Tang X, Li R, Wu D, Wang Y, Zhao F, Lv R, Wen X. Development and Validation of an ADME-Related Gene Signature for Survival, Treatment Outcome and Immune Cell Infiltration in Head and Neck Squamous Cell Carcinoma. Front Immunol 2022; 13:905635. [PMID: 35874705 PMCID: PMC9304892 DOI: 10.3389/fimmu.2022.905635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
ADME genes are a set of genes which are involved in drug absorption, distribution, metabolism, and excretion (ADME). However, prognostic value and function of ADME genes in head and neck squamous cell carcinoma (HNSCC) remain largely unclear. In this study, we established an ADME-related prognostic model through the least absolute shrinkage and selection operator (LASSO) analysis in the Cancer Genome Atla (TCGA) training cohort and its robustness was validated by TCGA internal validation cohort and a Gene Expression Omnibus (GEO) external cohort. The 14-gene signature stratified patients into high- or low-risk groups. Patients with high-risk scores exhibited significantly poorer overall survival (OS) and disease-free survival (DFS) than those with low-risk scores. Receiver operating characteristic (ROC) curve analysis was used to confirm the signature’s predictive efficacy for OS and DFS. Furthermore, gene ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analyses showed that immune-related functions and pathways were enriched, such as lymphocyte activation, leukocyte cell-cell adhesion and T-helper cell differentiation. The Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and other analyses revealed that immune cell (especially B cell and T cell) infiltration levels were significantly higher in the low-risk group. Moreover, patients with low-risk scores were significantly associated with immunotherapy and chemotherapy treatment benefit. In conclusion, we constructed a novel ADME-related prognostic and therapeutic biomarker associated with immune cell infiltration of HNSCC patients.
Collapse
Affiliation(s)
- Xinran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dehua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yikai Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Zhao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruxue Lv
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xin Wen,
| |
Collapse
|
12
|
Risk Alleles for Multiple Myeloma Susceptibility in ADME Genes. Cells 2022; 11:cells11020189. [PMID: 35053305 PMCID: PMC8773885 DOI: 10.3390/cells11020189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
The cause of multiple myeloma (MM) remains largely unknown. Several pieces of evidence support the involvement of genetic and multiple environmental factors (i.e., chemical agents) in MM onset. The inter-individual variability in the bioactivation, detoxification, and clearance of chemical carcinogens such as asbestos, benzene, and pesticides might increase the MM risk. This inter-individual variability can be explained by the presence of polymorphic variants in absorption, distribution, metabolism, and excretion (ADME) genes. Despite the high relevance of this issue, few studies have focused on the inter-individual variability in ADME genes in MM risk. To identify new MM susceptibility loci, we performed an extended candidate gene approach by comparing high-throughput genotyping data of 1936 markers in 231 ADME genes on 64 MM patients and 59 controls from the CEU population. Differences in genotype and allele frequencies were validated using an internal control group of 35 non-cancer samples from the same geographic area as the patient group. We detected an association between MM risk and ADH1B rs1229984 (OR = 3.78; 95% CI, 1.18–12.13; p = 0.0282), PPARD rs6937483 (OR = 3.27; 95% CI, 1.01–10.56; p = 0.0479), SLC28A1 rs8187737 (OR = 11.33; 95% CI, 1.43–89.59; p = 0.005), SLC28A2 rs1060896 (OR = 6.58; 95% CI, 1.42–30.43; p = 0.0072), SLC29A1 rs8187630 (OR = 3.27; 95% CI, 1.01–10.56; p = 0.0479), and ALDH3A2 rs72547554 (OR = 2.46; 95% CI, 0.64–9.40; p = 0.0293). The prognostic value of these genes in MM was investigated in two public datasets showing that shorter overall survival was associated with low expression of ADH1B and SLC28A1. In conclusion, our proof-of-concept findings provide novel insights into the genetic bases of MM susceptibility.
Collapse
|
13
|
Yin HM, He Q, Chen J, Li Z, Yang W, Hu X. Drug metabolism-related eight-gene signature can predict the prognosis of gastric adenocarcinoma. J Clin Lab Anal 2021; 35:e24085. [PMID: 34773716 PMCID: PMC8649372 DOI: 10.1002/jcla.24085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/28/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Background Metabolic abnormalities in patients with gastric adenocarcinoma lead to drug resistance and poor prognosis. Therefore, this study aimed to explore biomarkers that can predict the prognostic risk of gastric adenocarcinoma by analyzing drug metabolism‐related genes. Methods The RNA‐seq and clinical information on gastric adenocarcinoma were downloaded from the UCSC and gene expression omnibus databases. Univariate and least absolute shrinkage and selection operator regression analyses were used to identify the prognostic gene signature of gastric adenocarcinoma. The relationships between gastric adenocarcinoma prognostic risk and tumor microenvironment were assessed using CIBERSORT, EPIC, QUANTISEQ, MCPCounter, xCell, and TIMER algorithms. The potential drugs that could target the gene signatures were predicted in WebGestalt, and molecular docking analysis verified their binding stabilities. Results Combined with clinical information, an eight‐gene signature, including GPX3, ABCA1, NNMT, NOS3, SLCO4A1, ADH4, DHRS7, and TAP1, was identified from the drug metabolism‐related gene set. Based on their expressions, risk scores were calculated, and patients were divided into high‐ and low‐risk groups, which had significant differences in survival status and immune infiltrations. Risk group was also identified as an independent prognostic factor of gastric adenocarcinoma, and the established prognostic and nomogram models exhibited excellent capacities for predicting prognosis. Finally, miconazole and niacin were predicted as potential therapeutic drugs for gastric adenocarcinoma that bond stably with NOS3 and NNMT through hydrogen interactions. Conclusions This study proposed a drug metabolism‐related eight‐gene signature as a potential biomarker to predict the gastric adenocarcinoma prognosis risks.
Collapse
Affiliation(s)
- Hong-Mei Yin
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiong He
- Pathology Department, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jia Chen
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen Li
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanli Yang
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaobo Hu
- Department of Clinical Laboratory, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Establishment of non-small-cell lung cancer risk prediction model based on prognosis-associated ADME genes. Biosci Rep 2021; 41:229783. [PMID: 34522968 PMCID: PMC8527211 DOI: 10.1042/bsr20211433] [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: 06/22/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022] Open
Abstract
PURPOSE ADME genes are those involved in the absorption, distribution, metabolism, and excretion (ADME) of drugs. In the present study, a non-small-cell lung cancer (NSCLC) risk prediction model was established using prognosis-associated ADME genes, and the predictive performance of this model was evaluated and verified. In addition, multifaceted difference analysis was performed on groups with high and low risk scores. METHODS An NSCLC sample transcriptome and clinical data were obtained from public databases. The prognosis-associated ADME genes were obtained by univariate Cox and lasso regression analyses to build a risk model. Tumor samples were divided into high-risk and low-risk score groups according to the risk score. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of the differentially expressed genes and the differences in the immune infiltration, mutation, and medication reactions in the two groups were studied in detail. RESULTS A risk prediction model was established with seven prognosis-associated ADME genes. Its good predictive ability was confirmed by studies of the model's effectiveness. Univariate and multivariate Cox regression analyses showed that the model's risk score was an independent prognostic factor for patients with NSCLC. The study also showed that the risk score closely correlated with immune infiltration, mutations, and medication reactions. CONCLUSION The risk prediction model established with seven ADME genes in the present study can predict the prognosis of patients with NSCLC. In addition, significant differences in immune infiltration, mutations, and therapeutic efficacy exist between the high- and low-risk score groups.
Collapse
|
15
|
Hu DG, Marri S, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. The Expression Profiles and Deregulation of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers and Their Association with Clinical Outcomes. Cancers (Basel) 2021; 13:4491. [PMID: 34503303 PMCID: PMC8430925 DOI: 10.3390/cancers13174491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has 22 functional enzymes that play a critical role in the metabolism of small lipophilic compounds, including carcinogens, drugs, steroids, lipids, fatty acids, and bile acids. The expression profiles of UGT genes in human cancers and their impact on cancer patient survival remains to be systematically investigated. In the present study, a comprehensive analysis of the RNAseq and clinical datasets of 9514 patients from 33 different TCGA (the Genome Cancer Atlas) cancers demonstrated cancer-specific UGT expression profiles with high interindividual variability among and within individual cancers. Notably, cancers derived from drug metabolizing tissues (liver, kidney, gut, pancreas) expressed the largest number of UGT genes (COAD, KIRC, KIRP, LIHC, PAAD); six UGT genes (1A6, 1A9, 1A10, 2A3, 2B7, UGT8) showed high expression in five or more different cancers. Kaplan-Meier plots and logrank tests revealed that six UGT genes were significantly associated with increased overall survival (OS) rates [UGT1A1 (LUSC), UGT1A6 (ACC), UGT1A7 (ACC), UGT2A3 (KIRC), UGT2B15 (BLCA, SKCM)] or decreased OS rates [UGT2B15 (LGG), UGT8 (UVM)] in specific cancers. Finally, differential expression analysis of 611 patients from 12 TCGA cancers identified 16 UGT genes (1A1, 1A3, 1A6, 1A7, 1A8, 1A9, 1A10, 2A1, 2A3, 2B4, 2B7, 2B11, 2B15, 3A1, 3A2, UGT8) that were up/downregulated in at least one cancer relative to normal tissues. In conclusion, our data show widespread expression of UGT genes in cancers, highlighting the capacity for intratumoural drug metabolism through the UGT conjugation pathway. The data also suggests the potentials for specific UGT genes to serve as prognostic biomarkers or therapeutic targets in cancers.
Collapse
Affiliation(s)
- Dong Gui Hu
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Shashikanth Marri
- Dicipline of Molecular Medicine and Pathology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Peter I. Mackenzie
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Julie-Ann Hulin
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Ross A. McKinnon
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| | - Robyn Meech
- Dicipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (P.I.M.); (J.-A.H.); (R.A.M.); (R.M.)
| |
Collapse
|
16
|
Matheux A, Gassiot M, Fromont G, Leenhardt F, Boulahtouf A, Fabbrizio E, Marchive C, Garcin A, Agherbi H, Combès E, Evrard A, Houédé N, Balaguer P, Gongora C, Mbatchi LC, Pourquier P. PXR Modulates the Prostate Cancer Cell Response to Afatinib by Regulating the Expression of the Monocarboxylate Transporter SLC16A1. Cancers (Basel) 2021; 13:cancers13143635. [PMID: 34298852 PMCID: PMC8305337 DOI: 10.3390/cancers13143635] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Many kinase inhibitors have been tested as potential alternatives for the treatment of castration-resistant prostate cancers. However, none of these clinical trials led to drug approval despite interesting responses. Our study reveals that genes involved in drug metabolism and their master regulator PXR (Pregnane X Receptor) could be responsible, at least in part, for these disappointing results as they can modulate tumor cell response to specific kinase inhibitors. We found that stable expression of PXR sensitized prostate cancer cells to erlotinib, dabrafenib, and afatinib, while it rendered cells resistant to dasatinib and had no effect for other inhibitors tested. We also report for the first time that sensitization to afatinib is due to an alteration in drug transport that involves the SLC16A1 monocarboxylate transporter. Together, our results further indicate that PXR might be considered as a biomarker of response to kinase inhibitors in castration-resistant prostate cancers. Abstract Resistance to castration is a crucial issue in the treatment of metastatic prostate cancer. Kinase inhibitors (KIs) have been tested as potential alternatives, but none of them are approved yet. KIs are subject of extensive metabolism at both the hepatic and the tumor level. Here, we studied the role of PXR (Pregnane X Receptor), a master regulator of metabolism, in the resistance to KIs in a prostate cancer setting. We confirmed that PXR is expressed in prostate tumors and is more frequently detected in advanced forms of the disease. We showed that stable expression of PXR in 22Rv1 prostate cancer cells conferred a resistance to dasatinib and a higher sensitivity to erlotinib, dabrafenib, and afatinib. Higher sensitivity to afatinib was due to a ~ 2-fold increase in its intracellular accumulation and involved the SLC16A1 transporter as its pharmacological inhibition by BAY-8002 suppressed sensitization of 22Rv1 cells to afatinib and was accompanied with reduced intracellular concentration of the drug. We found that PXR could bind to the SLC16A1 promoter and induced its transcription in the presence of PXR agonists. Together, our results suggest that PXR could be a biomarker of response to kinase inhibitors in castration-resistant prostate cancers.
Collapse
Affiliation(s)
- Alice Matheux
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Carémeau, F-30029 Nîmes, France
| | - Matthieu Gassiot
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Gaëlle Fromont
- Département de Pathologie, CHU de Tours, Université François Rabelais, Inserm UMR 1069, F-37044 Tours, France;
| | - Fanny Leenhardt
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, F-34090 Montpellier, France
| | - Abdelhay Boulahtouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Eric Fabbrizio
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Candice Marchive
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Aurélie Garcin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Hanane Agherbi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Eve Combès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Alexandre Evrard
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Carémeau, F-30029 Nîmes, France
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, F-34090 Montpellier, France
| | - Nadine Houédé
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Département d’Oncologie Médicale, Institut de Cancérologie du Gard—CHU Carémeau, F-30029 Nîmes, France
| | - Patrick Balaguer
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Céline Gongora
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
| | - Litaty C. Mbatchi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Carémeau, F-30029 Nîmes, France
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, F-34090 Montpellier, France
| | - Philippe Pourquier
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, ICM, F-34298 Montpellier, France; (A.M.); (M.G.); (F.L.); (A.B.); (E.F.); (C.M.); (A.G.); (H.A.); (E.C.); (A.E.); (N.H.); (P.B.); (C.G.); (L.C.M.)
- Correspondence: ; Tel.: +33-4-66-68-32-31
| |
Collapse
|
17
|
Identification and validation of ADME genes as prognosis and therapy markers for hepatocellular carcinoma patients. Biosci Rep 2021; 41:228648. [PMID: 33988674 PMCID: PMC8164111 DOI: 10.1042/bsr20210583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/01/2021] [Accepted: 05/10/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose: ADME genes are genes involved in drug absorption, distribution, metabolism, and excretion (ADME). Previous studies report that expression levels of ADME-related genes correlate with prognosis of hepatocellular carcinoma (HCC) patients. However, the role of ADME gene expression on HCC prognosis has not been fully explored. The present study sought to construct a prediction model using ADME-related genes for prognosis of HCC. Methods: Transcriptome and clinical data were retrieved from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC), which were used as training and validation cohorts, respectively. A prediction model was constructed using univariate Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) analysis. Patients were divided into high- and low-risk groups based on the median risk score. The predictive ability of the risk signature was estimated through bioinformatics analyses. Results: Six ADME-related genes (CYP2C9, ABCB6, ABCC5, ADH4, DHRS13, and SLCO2A1) were used to construct the prediction model with a good predictive ability. Univariate and multivariate Cox regression analyses showed the risk signature was an independent predictor of overall survival (OS). A single-sample gene set enrichment analysis (ssGSEA) strategy showed a significant relationship between risk signature and immune status. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed differentially expressed genes (DEGs) in the high- and low-risk groups were enriched in biological process (BP) associated with metabolic and cell cycle pathways. Conclusion: A prediction model was constructed using six ADME-related genes for prediction of HCC prognosis. This signature can be used to improve HCC diagnosis, treatment, and prognosis in clinical use.
Collapse
|