1
|
Chen R, Yang L, Han L, Hu Z, Gu Z, Deng X. Prevalence of synonymous mutations in m 6A modification sites in human cancers. Genes Dis 2025; 12:101373. [PMID: 39524540 PMCID: PMC11549976 DOI: 10.1016/j.gendis.2024.101373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Richard Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- South Pasadena High School, 1401 Fremont Ave, South Pasadena, CA 91030, USA
- Center for RNA Biology and Therapeutics, City of Hope, Duarte, CA 91010, USA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Center for RNA Biology and Therapeutics, City of Hope, Duarte, CA 91010, USA
| | - Li Han
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110001, China
| | - Zunsong Hu
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Center for RNA Biology and Therapeutics, City of Hope, Duarte, CA 91010, USA
| | - Zhaohui Gu
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Center for RNA Biology and Therapeutics, City of Hope, Duarte, CA 91010, USA
| | - Xiaolan Deng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
- Center for RNA Biology and Therapeutics, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Freire MV, Thissen R, Martin M, Fasquelle C, Helou L, Durkin K, Artesi M, Lumaka A, Leroi N, Segers K, Deberg M, Gatot JS, Habran L, Palmeira L, Josse C, Bours V. Genetic evaluation of patients with multiple primary cancers. Oncol Lett 2025; 29:4. [PMID: 39492936 PMCID: PMC11526284 DOI: 10.3892/ol.2024.14750] [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: 04/10/2024] [Accepted: 08/22/2024] [Indexed: 11/05/2024] Open
Abstract
Regarding inherited cancer predisposition, single gene carriers of pathogenic variants (PVs) have been extensively reported on in the literature, whereas the oligogenic coinheritance of heterozygous PVs in cancer-related genes is a poorly studied event. Currently, due to the increased number of cancer survivors, the probability of patients presenting with multiple primary cancers (MPCs) is higher. The present study included patients with MPCs aged ≤45 years without known PVs in common cancer predisposition genes. This study used whole exome sequencing (WES) of germline and tumoral DNA, chromosomal microarray analysis (CMA) of germline DNA (patients 1-7, 9 and 10), and a karyotype test of patient 8 to detect variants associated with the disease. The 10 patients included in the study presented a mean of 3 cancers per patient. CMA showed two microduplications and one microdeletion, while WES of the germline DNA identified 1-3 single nucleotide variants of potential interest to the disease in each patient and two additional copy number variants. Most of the identified variants were classified as variants of uncertain significance. The mapping of the germline variants into their pathways showed a possible additive effect of these as the cause of the cancer. A total of 12 somatic samples from 5 patients were available for sequencing. All of the germline variants were also present in the somatic samples, while no second hits were identified in the same genes. The sequencing of patients with early cancers, family history and multiple tumors is already a standard of care. However, growing evidence has suggested that the assessment of patients should not stop at the identification of one PV in a cancer predisposition gene.
Collapse
Affiliation(s)
- Maria Valeria Freire
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Romain Thissen
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Marie Martin
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | | | - Laura Helou
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Keith Durkin
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Maria Artesi
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Aimé Lumaka
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Natacha Leroi
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Karin Segers
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Michelle Deberg
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | | | - Lionel Habran
- Department of Pathology, CHU Liège, 4000 Liège, Belgium
| | - Leonor Palmeira
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| | - Claire Josse
- Department of Medical Oncology, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
| | - Vincent Bours
- Department of Human Genetics, GIGA Research Center-University of Liège and CHU Liège, 4000 Liège, Belgium
- Department of Human Genetics, CHU Liège, 4000 Liège, Belgium
| |
Collapse
|
3
|
Karihtala P, Kilpivaara O, Porvari K. Mutational signatures and their association with cancer survival and gene expression in multiple cancer types. Int J Cancer 2025; 156:114-129. [PMID: 39194330 DOI: 10.1002/ijc.35148] [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: 03/21/2024] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024]
Abstract
Different endogenous and exogenous mutational processes cause specific patterns of somatic mutations and mutational signatures. Although their biological research has been intensive, there are only rare studies assessing the possible prognostic role of mutational signatures. We used data from The Cancer Genome Atlas to study the associations between the activity of the mutational signatures and four survival endpoints in 18 types of malignancies. We further explored the prognostic differences according to, for example, the HPV status in head and neck squamous cell carcinomas and smoking status in lung cancers. The predictive power of the signatures over time was evaluated with a dynamic area under the curve model, and the links between mutational signature activities and differences in gene expression patterns were analyzed. In 12 of 18 studied cancer types, we identified at least one mutational signature whose activity predicted survival outcomes after adjusting for the established prognostic factors. For example, overall survival was associated with the activity of mutational signatures in nine cancer types and disease-specific survival in seven cancer types. The clock-like signatures SBS5 and SBS40 were most commonly associated with survival endpoints. The genes of the myosin binding protein and melanoma antigen families were among the most substantially dysregulated genes between the signatures of low and high activity. The differences in gene expression also revealed various enriched pathways. Based on these data, specific mutational signatures associate with the gene expression and have the potential to serve as strong prognostic factors in several cancer types.
Collapse
Affiliation(s)
- Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center and University of Helsinki, Helsinki, Finland
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUSLAB Laboratory of Genetics, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- K. Albin Johansson Cancer Research Fellow, Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Katja Porvari
- Department of Pathology, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
4
|
Yao J, Lin X, Zhang X, Xie M, Ma X, Bao X, Song J, Liang Y, Wang Q, Xue X. Predictive biomarkers for immune checkpoint inhibitors therapy in lung cancer. Hum Vaccin Immunother 2024; 20:2406063. [PMID: 39415535 PMCID: PMC11487980 DOI: 10.1080/21645515.2024.2406063] [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: 04/23/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment mode of lung cancer, extending the survival time of patients unprecedentedly. Once patients respond to ICIs, the median duration of response is usually longer than that achieved with cytotoxic or targeted drugs. Unfortunately, there is still a large proportion of lung cancer patients do not respond to ICI. Effective biomarkers are crucial for identifying lung cancer patients who can benefit from them. The first predictive biomarker is programmed death-ligand 1 (PD-L1), but its predictive value is limited to specific populations. With the development of single-cell sequencing and spatial imaging technologies, as well as the use of deep learning and artificial intelligence, the identification of predictive biomarkers has been greatly expanded. In this review, we will dissect the biomarkers used to predict ICIs efficacy in lung cancer from the tumor-immune microenvironment and host perspectives, and describe cutting-edge technologies to further identify biomarkers.
Collapse
Affiliation(s)
- Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
5
|
Lin PH, Chan JY, Guan P, Hong JH, Lim AH, Ng CCY, Yeong JPS, Lee JY, Liu W, Lim JCT, Pang ST, Teh BT. Aristolochic acid-related renal cell carcinoma exhibits a distinct tumor-immune microenvironment favoring response to immune checkpoint blockade. J Pathol 2024; 264:371-382. [PMID: 39360336 DOI: 10.1002/path.6349] [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: 02/18/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024]
Abstract
Immune checkpoint blockade (ICB) is currently the standard of care for metastatic renal cell carcinoma (RCC), but treatment responses remain unpredictable. Aristolochic acid (AA), a prevalent supplement additive in Taiwan, has been associated with RCC and induces signature mutations, although its effect on the tumor-immune microenvironment (TIME) is unclear. We aimed to investigate the immune profile of AA-positive RCCs and explore its potential role as a susceptible candidate for ICB. Tissue samples from 22 patients with clear cell RCC (ccRCC) were collected for whole-exome sequencing to determine the genetic features and AA mutational signature (the discovery cohort). The corresponding RNA was sent for NanoString PanCancer IO 360 gene expression analysis to explore the immunological features. The formalin-fixed, parafilm-embedded slides of ccRCCs were sent for multiplex immunohistochemistry/immunofluorescence stain using Vectra system to evaluate the TIME. Tissues from two patients with metastatic RCC demonstrating complete response to ICB were sent for studies to validate the findings (the index patients). The results showed that AA mutational signatures with high tumor mutational burden (TMB) were present in 31.81% of the tumors in the discovery cohort. Three distinct clusters were observed through NanoString analysis. Clusters 1 and 3 were composed mainly of AA-positive RCCs. Cluster 3 RCCs exhibited higher tumor inflammation signature scores and higher immune cell type scores. Vectra analysis revealed a higher percentage of CD15+ and BATF3+ cells in cluster 1, whereas the percentage of CD8+ cells was potentially higher in cluster 3. Strong AA mutational signatures were found in the tumors of two index patients, and both were grouped to cluster 3. In conclusion, AA may induce higher TMB and alter the immune microenvironment in RCCs, which makes the tumors more susceptible to ICB. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Po-Hung Lin
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Peiyong Guan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jing Han Hong
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Joe Poh Sheng Yeong
- Integrative Biology for Theranostics Lab, Cancer Signaling & Therapies Programme, Institute of Molecular and Cell Biology, Singapore, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Pathology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Wei Liu
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Jeffrey Chun Tatt Lim
- Integrative Biology for Theranostics Lab, Cancer Signaling & Therapies Programme, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - See-Tong Pang
- Division of Urology, Department of Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Bin Tean Teh
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
6
|
Weng Z, Mai Z, Yuan J, Liu Q, Deng F, Yang H, Ling Y, Xie X, Lin X, Lin T, Chen J, Wei X, Luo K, Fu J, Wen J. Evolution of genome and immunogenome in esophageal squamous cell carcinomas driven by neoadjuvant chemoradiotherapy. Int J Cancer 2024; 155:2021-2035. [PMID: 39081132 DOI: 10.1002/ijc.35118] [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: 03/25/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 10/04/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery is a standard treatment for locally advanced esophageal squamous cell carcinomas (ESCCs). However, the evolution of genome and immunogenome in ESCCs driven by NCRT remains incompletely elucidated. We performed whole-exome sequencing of 51 ESCC tumors collected before and after NCRT, 36 of which were subjected to transcriptome sequencing. Clonal analysis identified clonal extinction in 13 ESCC patients wherein all pre-NCRT clones disappeared after NCRT, and clonal persistence in 9 patients wherein clones endured following NCRT. The clone-persistent patients showed higher pre-NCRT genomic intratumoral heterogeneity and worse prognosis than the clone-extinct ones. In contrast to the clone-extinct patients, the clone-persistent patients demonstrated a high proportion of subclonal neoantigens within pre-treatment specimens. Transcriptome analysis revealed increased immune infiltrations and up-regulated immune-related pathways after NCRT, especially in the clone-extinct patients. The number of T cell receptor-neoantigen interactions was higher in the clone-extinct patients than in the clone-persistent ones. The decrease in T cell repertoire evenness positively correlated to the decreased number of clonal neoantigens after NCRT, especially in the clone-extinct patients. In conclusion, we identified two prognosis-related clonal dynamic modes driven by NCRT in ESCCs. This study extended our knowledge of the ESCC genome and immunogenome evolutions driven by NCRT.
Collapse
Affiliation(s)
- Zelin Weng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zihang Mai
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianye Yuan
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Qianwen Liu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangqi Deng
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yihong Ling
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuying Xie
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaodan Lin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Lin
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiyang Chen
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoli Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kongjia Luo
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Wen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
7
|
Pfeifer GP, Jin SG. Methods and applications of genome-wide profiling of DNA damage and rare mutations. Nat Rev Genet 2024; 25:846-863. [PMID: 38918545 PMCID: PMC11563917 DOI: 10.1038/s41576-024-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
DNA damage is a threat to genome integrity and can be a cause of many human diseases, owing to either changes in the chemical structure of DNA or conversion of the damage into a mutation, that is, a permanent change in DNA sequence. Determining the exact positions of DNA damage and ensuing mutations in the genome are important for identifying mechanisms of disease aetiology when characteristic mutations are prevalent and probably causative in a particular disease. However, this approach is challenging particularly when levels of DNA damage are low, for example, as a result of chronic exposure to environmental agents or certain endogenous processes, such as the generation of reactive oxygen species. Over the past few years, a comprehensive toolbox of genome-wide methods has been developed for the detection of DNA damage and rare mutations at single-nucleotide resolution in mammalian cells. Here, we review and compare these methods, describe their current applications and discuss future research questions that can now be addressed.
Collapse
Affiliation(s)
- Gerd P Pfeifer
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA.
| | - Seung-Gi Jin
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
8
|
Hashemi M, Rezaei M, Rezaeiaghdam H, Jamali B, Koohpar ZK, Tanha M, Bizhanpour A, Asadi S, Jafari AM, Khosroshahi EM, Eslami M, Salimimoghadam S, Nabavi N, Rashidi M, Fattah E, Taheriazam A, Entezari M. Highlighting function of Wnt signalling in urological cancers: Molecular interactions, therapeutic strategies, and (nano)strategies. Transl Oncol 2024; 50:102145. [PMID: 39357465 PMCID: PMC11474201 DOI: 10.1016/j.tranon.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer is a complex, multistep process characterized by abnormal cell growth and metastasis as well as the capacity of the tumor cells in therapy resistance development. The urological system is particularly susceptible to a group of malignancies known as urological cancers, where an accumulation of genetic alterations drives carcinogenesis. In various human cancers, Wnt singalling is dysregulated; following nuclear transfer of β-catenin, it promotes tumor progression and affects genes expression. Elevated levels of Wnt have been documented in urological cancers, where its overexpression enhances growth and metastasis. Additionally, increased Wnt singalling contributes to chemoresistance in urological cancers, leading to reduced sensitivity to chemotherapy agents like cisplatin, doxorubicin, and paclitaxel. Wnt upregulation can change radiotherapy response of urological cancers. The regulation of Wnt involves various molecular pathways, including Akt, miRNAs, lncRNAs, and circRNAs, all of which play roles in carcinogenesis. Targeting and silencing Wnt or its associated pathways can mitigate tumorigenesis in urological cancers. Anti-cancer compounds such as curcumin and thymoquinone have shown efficacy in suppressing tumorigenesis through the downregulation of Wnt singalling. Notably, nanoparticles have proven effective in treating urological cancers, with several studies in prostate cancer (PCa) using nanoparticles to downregulate Wnt and suppress tumor growth. Future research should focus on developing small molecules that inhibit Wnt singalling to further suppress tumorigenesis and advance the treatment of urological cancers. Moreover, Wnt can be used as reliable biomarker for the diagnosis and prognosis of urological cancers.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Rezaei
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Hadi Rezaeiaghdam
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Behdokht Jamali
- Department of Microbiology and Genetics, Kherad Institute of Higher Education, Bushehr, Iran
| | - Zeinab Khazaei Koohpar
- Department Of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mahsa Tanha
- Department Of Biological Sciences, University Of Alabama, Tuscaloosa, Al, United States
| | - Anahita Bizhanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Maedeh Eslami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8V 1P7, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Eisa Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
9
|
Grützmann K, Kraft T, Meinhardt M, Meier F, Westphal D, Seifert M. Network-based analysis of heterogeneous patient-matched brain and extracranial melanoma metastasis pairs reveals three homogeneous subgroups. Comput Struct Biotechnol J 2024; 23:1036-1050. [PMID: 38464935 PMCID: PMC10920107 DOI: 10.1016/j.csbj.2024.02.013] [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: 11/06/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 03/12/2024] Open
Abstract
Melanoma, the deadliest form of skin cancer, can metastasize to different organs. Molecular differences between brain and extracranial melanoma metastases are poorly understood. Here, promoter methylation and gene expression of 11 heterogeneous patient-matched pairs of brain and extracranial metastases were analyzed using melanoma-specific gene regulatory networks learned from public transcriptome and methylome data followed by network-based impact propagation of patient-specific alterations. This innovative data analysis strategy allowed to predict potential impacts of patient-specific driver candidate genes on other genes and pathways. The patient-matched metastasis pairs clustered into three robust subgroups with specific downstream targets with known roles in cancer, including melanoma (SG1: RBM38, BCL11B, SG2: GATA3, FES, SG3: SLAMF6, PYCARD). Patient subgroups and ranking of target gene candidates were confirmed in a validation cohort. Summarizing, computational network-based impact analyses of heterogeneous metastasis pairs predicted individual regulatory differences in melanoma brain metastases, cumulating into three consistent subgroups with specific downstream target genes.
Collapse
Affiliation(s)
- Konrad Grützmann
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Theresa Kraft
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Matthias Meinhardt
- Department of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| | - Dana Westphal
- Department of Dermatology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| |
Collapse
|
10
|
Boiarsky D, Tewari AK, Gulhan DC, Bakouny Z, Ananda G, Savignano H, Lakshminarayanan G, McClure HM, Silver R, Choueiri TK, Taplin ME, Park PJ, Berchuck JE. A panel-based mutational signature of homologous recombination deficiency associates with response to PARP inhibition in metastatic castration-resistant prostate cancer. Prostate 2024; 84:1479-1489. [PMID: 39252459 DOI: 10.1002/pros.24788] [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: 06/26/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The PARP inhibitor (PARPi) olaparib is approved for homologous recombination repair (HRR) gene-altered metastatic castration-resistant prostate cancer (mCRPC). However, there is significant heterogeneity in response to PARPi in patients with mCRPC. Better clinical biomarkers are needed to identify patients likely to benefit from PARPi. METHODS Patients with prostate adenocarcinoma and panel sequencing at Dana-Farber Cancer Institute were identified. Mutational signature analysis was performed using SigMA to characterize tumors as HRR deficient (HRD). The validity of SigMA to identify patients likely to benefit from olaparib was compared to the current FDA label (presence of a deleterious alteration in one of 14 HRR genes). RESULTS 546 patients were identified, of which 34% were HRD. Among patients with HRR gene alterations, only patients with BRCA2 two-copy loss (2CL) were more likely to be HRD compared to patients without HRR gene alterations (74% vs 31%; P = 9.1 × 10-7). 28 patients with mCRPC received olaparib, of which 13 were HRD and 9 had BRCA2 2CL. SigMA improved upon the current FDA label for predicting PSA50 (sensitivity: 100% vs 90%; specificity: 83% vs 44%; PPV: 77% vs 47%; NPV: 100% vs 89%) and rPFS > 6 months (sensitivity: both 92%; specificity: 93% vs 53%; PPV: 92% vs 63%; NPV: 93% vs 89%). On multivariate analysis, incorporating prognostic clinical factors and HR gene alterations, SigMA-predicted HRD independently associated with improved PSA-PFS (HR = 0.086, p = 0.00082) and rPFS (HR = 0.078, p = 0.0070). CONCLUSIONS SigMA-predicted HRD may better identify patients likely to benefit from olaparib as compared to the current FDA label. Larger studies are needed for further validation.
Collapse
Affiliation(s)
- Daniel Boiarsky
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Alok K Tewari
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Doga C Gulhan
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Guruprasad Ananda
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hunter Savignano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Heather M McClure
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rebecca Silver
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Rosalind Franklin University of Medicine and Science, Chicago, Illinois, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob E Berchuck
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Ma W, Tang W, Kwok JS, Tong AH, Lo CW, Chu AT, Chung BH. A review on trends in development and translation of omics signatures in cancer. Comput Struct Biotechnol J 2024; 23:954-971. [PMID: 38385061 PMCID: PMC10879706 DOI: 10.1016/j.csbj.2024.01.024] [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: 10/27/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
The field of cancer genomics and transcriptomics has evolved from targeted profiling to swift sequencing of individual tumor genome and transcriptome. The steady growth in genome, epigenome, and transcriptome datasets on a genome-wide scale has significantly increased our capability in capturing signatures that represent both the intrinsic and extrinsic biological features of tumors. These biological differences can help in precise molecular subtyping of cancer, predicting tumor progression, metastatic potential, and resistance to therapeutic agents. In this review, we summarized the current development of genomic, methylomic, transcriptomic, proteomic and metabolic signatures in the field of cancer research and highlighted their potentials in clinical applications to improve diagnosis, prognosis, and treatment decision in cancer patients.
Collapse
Affiliation(s)
- Wei Ma
- Hong Kong Genome Institute, Hong Kong, China
| | - Wenshu Tang
- Hong Kong Genome Institute, Hong Kong, China
| | | | | | | | | | - Brian H.Y. Chung
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Kong Genome Project
- Hong Kong Genome Institute, Hong Kong, China
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Backman S, Botling J, Nord H, Ghosal S, Stålberg P, Juhlin CC, Almlöf J, Sundin A, Zhang L, Moens L, Eriksson B, Welin S, Hellman P, Skogseid B, Pacak K, Mollazadegan K, Åkerström T, Crona J. The evolutionary history of metastatic pancreatic neuroendocrine tumours reveals a therapy driven route to high-grade transformation. J Pathol 2024; 264:357-370. [PMID: 39360347 DOI: 10.1002/path.6348] [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] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024]
Abstract
Tumour evolution with acquisition of more aggressive disease characteristics is a hallmark of disseminated cancer. Metastatic pancreatic neuroendocrine tumours (PanNETs) in particular may progress from a low/intermediate to a high-grade disease. The aim of this work was to understand the molecular mechanisms underlying metastatic progression as well as PanNET transformation from a low/intermediate to a high-grade disease. We performed multi-omics analysis (genome/exome sequencing, total RNA-sequencing and methylation array) of 32 longitudinal samples from six patients with metastatic low/intermediate grade PanNET. The clonal composition of tumour lesions and underlying phylogeny of each patient were determined with bioinformatics analyses. Findings were validated in post-alkylating chemotherapy samples from 24 patients with PanNET using targeted next generation sequencing. We validate the current PanNET evolutionary model with MEN1 inactivation that occurs very early in tumourigenesis. This was followed by pronounced genetic diversity on both spatial and temporal levels, with parallel and convergent tumour evolution involving the ATRX/DAXX and mechanistic target of the rapamycin (mTOR) pathways. Following alkylating chemotherapy treatment, some PanNETs developed mismatch repair deficiency and acquired a hypermutational phenotype. This was validated among 16 patients with PanNET who had high-grade progression after alkylating chemotherapy, of whom eight had a tumour mutational burden >50 (50%). In comparison, among the eight patients who did not show high-grade progression, 0 had a tumour mutational burden >50 (0%; odds ratio 'infinite', 95% confidence interval 1.8 to 'infinite', p = 0.02). Our findings contribute to broaden the understanding of metastatic/high-grade PanNETs and suggests that therapy driven disease evolution is an important hallmark of this disease. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Samuel Backman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Helena Nord
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Suman Ghosal
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - C Christofer Juhlin
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Almlöf
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anders Sundin
- Section of Radiology, Molecular Imaging, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Liang Zhang
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Lotte Moens
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Barbro Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Staffan Welin
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Britt Skogseid
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - Tobias Åkerström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Joakim Crona
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Spear S, Le Saux O, Mirza HB, Iyer N, Tyson K, Grundland Freile F, Walton JB, Woodman C, Jarvis S, Ennis DP, Aguirre Hernandez C, Xu Y, Spiliopoulou P, Brenton JD, Costa-Pereira AP, Cook DP, Vanderhyden BC, Keun HC, Triantafyllou E, Arnold JN, McNeish IA. PTEN Loss Shapes Macrophage Dynamics in High-Grade Serous Ovarian Carcinoma. Cancer Res 2024; 84:3772-3787. [PMID: 39186679 PMCID: PMC7616669 DOI: 10.1158/0008-5472.can-23-3890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 07/11/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
High-grade serous ovarian carcinoma (HGSC) remains a disease with poor prognosis that is unresponsive to current immune checkpoint inhibitors. Although PI3K pathway alterations, such as PTEN loss, are common in HGSC, attempts to target this pathway have been unsuccessful. We hypothesized that aberrant PI3K pathway activation may alter the HGSC immune microenvironment and present a targeting opportunity. Single-cell RNA sequencing identified populations of resident macrophages specific to Pten-null omental tumors in murine models, which were confirmed by flow cytometry. These macrophages were derived from peritoneal fluid macrophages and exhibited a unique gene expression program, marked by high expression of the enzyme heme oxygenase-1 (HMOX1). Targeting resident peritoneal macrophages prevented the appearance of HMOX1hi macrophages and reduced tumor growth. In addition, direct inhibition of HMOX1 extended survival in vivo. RNA sequencing identified IL33 in Pten-null tumor cells as a likely candidate driver, leading to the appearance of HMOX1hi macrophages. Human HGSC tumors also contained HMOX1hi macrophages with a corresponding gene expression program. Moreover, the presence of these macrophages was correlated with activated tumoral PI3K/mTOR signaling and poor overall survival in patients with HGSC. In contrast, tumors with low numbers of HMOX1hi macrophages were marked by increased adaptive immune response gene expression. These data suggest targeting HMOX1hi macrophages as a potential therapeutic strategy for treating poor prognosis HGSC. Significance: Macrophages with elevated HMOX1 expression are enriched in PTEN-deficient high-grade serous ovarian carcinoma, promote tumor growth, and represent a potential therapeutic target.
Collapse
Affiliation(s)
- Sarah Spear
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Olivia Le Saux
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
- Centre Léon Bérard, Department of Medical Oncology, Lyon, France
| | - Hasan B. Mirza
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Nayana Iyer
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Katie Tyson
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Fabio Grundland Freile
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Josephine B. Walton
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Chloé Woodman
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Sheba Jarvis
- Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Darren P. Ennis
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Carmen Aguirre Hernandez
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Yuewei Xu
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Pavlina Spiliopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - James D. Brenton
- CRUK Cambridge Institute, University of Cambridge, United Kingdom
| | - Ana P. Costa-Pereira
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - David P. Cook
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Hector C. Keun
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| | - Evangelos Triantafyllou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - James N. Arnold
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Iain A. McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London; London, United Kingdom
| |
Collapse
|
14
|
Jiang Y, Xie J, Cheng Q, Cai Z, Xu K, Lu W, Wang F, Wu X, Song Y, Lv T, Zhan P. Comprehensive genomic and spatial immune infiltration analysis of survival outliers in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy. Int Immunopharmacol 2024; 141:112901. [PMID: 39151386 DOI: 10.1016/j.intimp.2024.112901] [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: 04/26/2024] [Revised: 07/20/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND A minority of patients with extensive-stage small cell lung cancer (ES-SCLC) exhibit prolonged survival following first-line chemoimmunotherapy, which warrants the use of reliable biomarkers. Here, we investigated the disparities in genomics and immune cell spatial distribution between long- and short-term survival of patients with ES-SCLC. METHODS We retrospectively recruited 11 long-term (>2 years) and 13 short-term (<9 months) ES-SCLC survivors receiving first-line chemoimmunotherapy. The samples were processed using targeted next-generation sequencing (tNGS), programmed death ligand-1 staining, multiplex immunohistochemical staining for immune cells (mIHC), tumor mutation burden (TMB), and chromosomal instability score measurements. The expression of putative genes in SCLC at the bulk and single-cell RNA-sequencing levels, as well as the role of putative genes in pan-cancer immunotherapy cohorts, were analyzed. RESULTS At the genomic level, a greater proportion of the smoking signature and higher TMB (>3.1) were associated with favorable survival. At the single-gene and pathway levels, tNGS revealed that MCL1 and STMN1 amplification and alterations in the apoptosis pathway were more common in short-term survivors, whereas alterations in the DLL3, KMT2B, HGF, EPHA3, ADGRB3, lysine deprivation, and HGF-cMET pathways were observed more frequently in long-term survivors. mIHC analysis of immune cells with different spatial distributions revealed that long-term survivors presented increased numbers of M1-like macrophages in all locations and decreased numbers of CD8+ T cells in the tumor stroma. Bulk transcriptomic analysis demonstrated that high levels of STMN1 and DLL3 represented an immune-suppressive tumor immune microenvironment (TIME), whereas HGF indicated an immune-responsive TIME. The expression levels of our putative genes were comparative in both TP53/RB1 mutant-type and TP53/RB1 wild-type. At the single-cell level, STMN1, MCL1, and DLL3 were highly expressed among all molecular subtypes (SCLC-A, SCLC-N, and SCLC-P), with STMN1 being enriched in cell division and G2M checkpoint pathways. CONCLUSIONS For ES-SCLC patients receiving first-line chemoimmunotherapy, alterations in DLL3, KMT2B, HGF, EPHA3, and ADGRB3 and a greater proportion of M1-like macrophages infiltration in all locations were predictors of favorable survival, while MCL1 and STMN1 amplification, as well as a greater proportion of CD8+ T cells infiltrating the tumor stroma, predicted worse survival.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing 210000, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zijing Cai
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China
| | - Ke Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoying Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yong Song
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| |
Collapse
|
15
|
Silveira AB, Houy A, Ganier O, Özemek B, Vanhuele S, Vincent-Salomon A, Cassoux N, Mariani P, Pierron G, Leyvraz S, Rieke D, Picca A, Bielle F, Yaspo ML, Rodrigues M, Stern MH. Base-excision repair pathway shapes 5-methylcytosine deamination signatures in pan-cancer genomes. Nat Commun 2024; 15:9864. [PMID: 39543136 DOI: 10.1038/s41467-024-54223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Transition of cytosine to thymine in CpG dinucleotides is the most frequent type of mutation in cancer. This increased mutability is commonly attributed to the spontaneous deamination of 5-methylcytosine (5mC), which is normally repaired by the base-excision repair (BER) pathway. However, the contribution of 5mC deamination in the increasing diversity of cancer mutational signatures remains poorly explored. We integrate mutational signatures analysis in a large series of tumor whole genomes with lineage-specific epigenomic data to draw a detailed view of 5mC deamination in cancer. We uncover tumor type-specific patterns of 5mC deamination signatures in CpG and non-CpG contexts. We demonstrate that the BER glycosylase MBD4 preferentially binds to active chromatin and early replicating DNA, which correlates with lower mutational burden in these domains. We validate our findings by modeling BER deficiencies in isogenic cell models. Here, we establish MBD4 as the main actor responsible for 5mC deamination repair in humans.
Collapse
Affiliation(s)
- André Bortolini Silveira
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Alexandre Houy
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Olivier Ganier
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Begüm Özemek
- Otto Warburg Laboratory "Gene Regulation and Systems Biology of Cancer", Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Sandra Vanhuele
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theranostic Medicine, Institut Curie, PSL Research University, Paris, France
| | | | - Pascale Mariani
- Department of Surgical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Gaelle Pierron
- Department of Genetics, Institut Curie, PSL Research University, Paris, France
| | - Serge Leyvraz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Damian Rieke
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alberto Picca
- Service de Neuro-oncologie, Institut de Neurologie, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Franck Bielle
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
- Service de Neuropathologie, Laboratoire Escourolle, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marie-Laure Yaspo
- Otto Warburg Laboratory "Gene Regulation and Systems Biology of Cancer", Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Manuel Rodrigues
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris, France
| | - Marc-Henri Stern
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France.
- Department of Genetics, Institut Curie, PSL Research University, Paris, France.
| |
Collapse
|
16
|
Zheng G, Chen S, Ma W, Wang Q, Sun L, Zhang C, Chen G, Zhang S, Chen S. Spatial and Single-Cell Transcriptomics Unraveled Spatial Evolution of Papillary Thyroid Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2404491. [PMID: 39540244 DOI: 10.1002/advs.202404491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Recurrence and metastasis are the major issues for papillary thyroid cancer (PTC). Current morphological and molecular classification systems are not satisfied for PTC diagnosis due to lacking variant-specific morphological criteria and high signal-to-noise in mutation-based diagnosis, respectively. Importantly, intratumor heterogeneity is largely lost in current molecular classification system, which can be resolved by single cell RNA sequencing (scRNA-seq). However, scRNA-seq loses spatial information and morphological features. Herein, scRNA-seq is integrated and spatially-resolved transcriptomics (SRT) to elaborate the mechanisms underlying the spatial heterogeneity, malignancy and metastasis of PTCs by associating transcriptome and local morphology. This results demonstrated that PTC cells evolved with multiple routes, driven by the enhanced aerobic metabolism and the suppressed mRNA translation and protein synthesis and the involvement of cell-cell interaction. Two curated malignant and metastatic footprints can discriminate PTC cells from normal thyrocytes. Ferroptosis resistance contributed to PTC evolution. This results will advance the knowledge of intratumor spatial heterogeneity and evolution of PTCs at spatial and single-cell levels, and propose better diagnostic strategy.
Collapse
Affiliation(s)
- Guangzhe Zheng
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shaobo Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100032, China
| | - Wanqi Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Quanshu Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Li Sun
- The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Changwen Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Ge Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100032, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shuguang Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100032, China
| |
Collapse
|
17
|
Hollizeck S, Wang N, Wong SQ, Litchfield C, Guinto J, Ftouni S, Rebello R, Kanwal S, Dong R, Grimmond S, Sandhu S, Mileshkin L, Tothill RW, Chandrananda D, Dawson SJ. Unravelling mutational signatures with plasma circulating tumour DNA. Nat Commun 2024; 15:9876. [PMID: 39543119 DOI: 10.1038/s41467-024-54193-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
The use of circulating tumour DNA (ctDNA) to profile mutational signatures represents a non-invasive opportunity for understanding cancer mutational processes. Here we present MisMatchFinder, a liquid biopsy approach for mutational signature detection using low-coverage whole-genome sequencing of ctDNA. Through analysis of 375 plasma samples across 9 cancers, we demonstrate that MisMatchFinder accurately infers single-base and doublet-base substitutions, as well as insertions and deletions to enhance the detection of ctDNA and clinically relevant mutational signatures.
Collapse
Affiliation(s)
- Sebastian Hollizeck
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Ning Wang
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Jerick Guinto
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sarah Ftouni
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard Rebello
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Sehrish Kanwal
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Ruining Dong
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Sean Grimmond
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Linda Mileshkin
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Richard W Tothill
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Dineika Chandrananda
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
18
|
Heard JR, Ahdoot M, Theodorescu D, Mitra AP. Biomarkers of treatment response in bladder cancer. Expert Rev Mol Diagn 2024:1-13. [PMID: 39535158 DOI: 10.1080/14737159.2024.2428747] [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: 09/22/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION There have been many recent advancements in the treatment of bladder cancer including the approval of novel intravesical agents for non-muscle-invasive disease and systemic-targeted therapeutics for muscle-invasive and advanced disease. However, treatment strategies for bladder cancer are still largely based on clinicopathologic characteristics. AREAS COVERED Based on primary literature sourced from PubMed, Embase, and Cochrane Library, we review the current status of molecular markers and biomarker panels with respective to their value in predicting response to standard chemotherapeutics and novel agents in non-muscle-invasive, muscle-invasive, and advanced bladder cancer. EXPERT OPINION Several biomarkers based on molecular characterization of tumors and quantification of circulating tumor DNA have been associated with response or resistance to standard chemotherapeutics. More recent investigations have reported on predictive biomarkers for novel therapeutics in bladder cancer, although large-scale validation is still needed. Given the increasing therapeutic options for this disease, employment of such predictive biomarkers may help guide treatment selection and sequencing.
Collapse
Affiliation(s)
- John R Heard
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael Ahdoot
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anirban P Mitra
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
19
|
Tan JK, Awuah WA, Ahluwalia A, Sanker V, Ben-Jaafar A, Tenkorang PO, Aderinto N, Mehta A, Darko K, Shah MH, Roy S, Abdul-Rahman T, Atallah O. Genes to therapy: a comprehensive literature review of whole-exome sequencing in neurology and neurosurgery. Eur J Med Res 2024; 29:538. [PMID: 39523358 PMCID: PMC11552425 DOI: 10.1186/s40001-024-02063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Whole-exome sequencing (WES), a ground-breaking technology, has emerged as a linchpin in neurology and neurosurgery, offering a comprehensive elucidation of the genetic landscape of various neurological disorders. This transformative methodology concentrates on the exonic portions of DNA, which constitute approximately 1% of the human genome, thus facilitating an expedited and efficient sequencing process. WES has been instrumental in advancing our understanding of neurodegenerative diseases, neuro-oncology, cerebrovascular disorders, and epilepsy by revealing rare variants and novel mutations and providing intricate insights into their genetic complexities. This has been achieved while maintaining a substantial diagnostic yield, thereby offering novel perspectives on the pathophysiology and personalized management of these conditions. The utilization of WES boasts several advantages over alternative genetic sequencing methodologies, including cost-effectiveness, reduced incidental findings, simplified analysis and interpretation process, and reduced computational demands. However, despite its benefits, there are challenges, such as the interpretation of variants of unknown significance, cost considerations, and limited accessibility in resource-constrained settings. Additionally, ethical, legal, and social concerns are raised, particularly in the context of incidental findings and patient consent. As we look to the future, the integration of WES with other omics-based approaches could help revolutionize the field of personalized medicine through its implications in predictive models and the development of targeted therapeutic strategies, marking a significant stride toward more effective and clinically oriented solutions.
Collapse
Affiliation(s)
- Joecelyn Kirani Tan
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| | | | | | - Vivek Sanker
- Department of Neurosurgery, Trivandrum Medical College, Thiruvananthapuram, India
| | - Adam Ben-Jaafar
- University College Dublin, School of Medicine, Belfield, Dublin 4, Ireland
| | | | - Nicholas Aderinto
- Internal Medicine Department, LAUTECH Teaching Hospital, Ogbomoso, Nigeria
| | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, Hungary
| | - Kwadwo Darko
- Department of Neurosurgery, Korle Bu Teaching Hospital, Accra, Ghana
| | | | - Sakshi Roy
- School of Medicine, Queen's University Belfast, Belfast, UK
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| |
Collapse
|
20
|
Johannet P, Abdelfattah S, Wilde C, Patel S, Walch H, Rousseau B, Argiles G, Artz O, Patel M, Arfe A, Cercek A, Yaeger R, Ganesh K, Schultz N, Diaz LA, Foote MB. Molecular and Clinicopathologic Impact of GNAS Variants Across Solid Tumors. J Clin Oncol 2024; 42:3847-3857. [PMID: 39121438 PMCID: PMC11540749 DOI: 10.1200/jco.24.00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 08/11/2024] Open
Abstract
PURPOSE The molecular drivers underlying mucinous tumor pathogenicity are poorly understood. GNAS mutations predict metastatic burden and treatment resistance in mucinous appendiceal adenocarcinoma. We investigated the pan-cancer clinicopathologic relevance of GNAS variants. METHODS We assessed 58,043 patients with Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (IMPACT)-sequenced solid tumors to identify oncogenic variants, including GNAS, associated with mucinous tumor phenotype. We then performed comprehensive molecular analyses to compare GNAS-mutant (mut) and wild-type tumors across cancers. Gene expression patterns associated with GNAS-mut tumors were assessed in a The Cancer Genome Atlas cohort. Associations between GNAS variant status and peritoneal metastasis, first-line systemic therapy response, progression-free survival (PFS), and overall survival (OS) were determined using a propensity-matched subcohort of patients with metastatic disease. RESULTS Mucinous tumors were enriched for oncogenic GNAS variants. GNAS was mutated in >1% of small bowel, cervical, colorectal, pancreatic, esophagogastric, hepatobiliary, and GI neuroendocrine cancers. Across these cancers, GNAS-mut tumors exhibited a generally conserved C-to-T mutation-high, aneuploidy-low molecular profile with co-occurring prevalent KRAS variants (65% of GNAS-mut tumors) and fewer TP53 alterations. GNAS-mut tumors exhibited recurrently comutated alternative tumor suppressors (RBM10, INPPL1) and upregulation of MAPK and cell surface modulators. GNAS-mut tumors demonstrate an increased prevalence of peritoneal metastases (odds ratio [OR], 1.7 [95% CI, 1.1 to 2.5]; P = .006), worse response to first-line systemic therapy (OR, 2.2 [95% CI, 1.3 to 3.8]; P = .003), and shorter PFS (median, 5.6 v 7.0 months; P = .047). In a multivariable analysis, GNAS mutated status was independently prognostic of worse OS (hazard ratio, 1.25 [95% CI, 1.01 to 1.56]; adjusted P = .04). CONCLUSION Across the assessed cancers, GNAS-mut tumors exhibit a conserved molecular and clinical phenotype defined by mucinous tumor status, increased peritoneal metastasis, poor response to first-line systemic therapy, and worse survival.
Collapse
Affiliation(s)
- Paul Johannet
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Somer Abdelfattah
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Callahan Wilde
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Shrey Patel
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Henry Walch
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Guillem Argiles
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Oliver Artz
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Miteshkumar Patel
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Andrea Arfe
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrea Cercek
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Rona Yaeger
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Karuna Ganesh
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luis A. Diaz
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| | - Michael B. Foote
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York, NY
| |
Collapse
|
21
|
Kim KT, Lee MH, Shin SJ, Cho I, Kuk JC, Yun J, Choi YY. Decorin as a key marker of desmoplastic cancer-associated fibroblasts mediating first-line immune checkpoint blockade resistance in metastatic gastric cancer. Gastric Cancer 2024:10.1007/s10120-024-01567-6. [PMID: 39520589 DOI: 10.1007/s10120-024-01567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Gastric cancer (GC) remains a significant cause of cancer-related mortality worldwide. Despite the transformative impact of immune checkpoint blockade (ICB) therapies across various cancers, only a minority of patients with metastatic GC (mGC) benefit, emphasizing the urgent need for precise biomarkers to predict therapeutic responses and optimize patient selection. METHODS In this multi-omics study, we conducted whole exome and transcriptome sequencing on 12 tumors from mGC patients treated with nivolumab as first-line therapy. To validate our findings, we performed whole transcriptome sequencing on 17 additional tumors and analyzed 45 tumors from public dataset (PRJEB25780) of patients who received ICB therapy as second or third-line treatment. Comprehensive multi-omics analyses were conducted using single-cell RNA sequencing (n = 5, GSE167297) and spatial transcriptome sequencing (n = 2, independent internal dataset). RESULTS ICB-sensitive tumors exhibited robust activation of the interferon response pathway, while ICB-resistant tumors displayed epithelial-mesenchymal transition signatures. Intriguingly, at the single-cell level, genes associated with ICB sensitivity were predominantly expressed in immune cells, whereas genes associated with resistance were primarily found in cancer-associated fibroblasts (CAFs), particularly the desmoplastic CAF (dCAF) subtype. We identified DCN as a hallmark dCAF marker, and high DCN expression was inversely correlated with PD-L1 levels, ICB resistance, and poor prognosis in mGC (log-rank p = 0.027). CONCLUSION This study elucidates the critical influence of the tumor microenvironment, specifically dCAFs, in mediating ICB resistance in mGC. Our findings highlight DCN as a representative marker for dCAF and a promising negative predictive biomarker for ICB response. These findings highlight the complex stromal-immune interactions and open avenues for personalized treatment for mGC.
Collapse
Affiliation(s)
- Ki Tae Kim
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry, Seoul National University, Seoul, Korea
- Dental Research Institute and Dental Multi-omics Center, Seoul National University, Seoul, South Korea
| | - Min Hee Lee
- Department of Radiology, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Su-Jin Shin
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - In Cho
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Jung Cheol Kuk
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Jina Yun
- Department of Medicine, Division of Hematology/Oncology, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Yoon Young Choi
- Department of Surgery, Soonchunhyang Bucheon Hospital, Soonchunhyang University College of Medicine, 170 Jomaru-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
22
|
Forsthuber A, Aschenbrenner B, Korosec A, Jacob T, Annusver K, Krajic N, Kholodniuk D, Frech S, Zhu S, Purkhauser K, Lipp K, Werner F, Nguyen V, Griss J, Bauer W, Soler Cardona A, Weber B, Weninger W, Gesslbauer B, Staud C, Nedomansky J, Radtke C, Wagner SN, Petzelbauer P, Kasper M, Lichtenberger BM. Cancer-associated fibroblast subtypes modulate the tumor-immune microenvironment and are associated with skin cancer malignancy. Nat Commun 2024; 15:9678. [PMID: 39516494 PMCID: PMC11549091 DOI: 10.1038/s41467-024-53908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in cancer progression and treatment outcome. This study dissects the intra-tumoral diversity of CAFs in basal cell carcinoma, squamous cell carcinoma, and melanoma using molecular and spatial single-cell analysis. We identify three distinct CAF subtypes: myofibroblast-like RGS5+ CAFs, matrix CAFs (mCAFs), and immunomodulatory CAFs (iCAFs). Large-cohort tissue analysis reveals significant shifts in CAF subtype patterns with increasing malignancy. Two CAF subtypes exhibit immunomodulatory properties via different mechanisms. mCAFs sythesize extracellular matrix and may restrict T cell invasion in low-grade tumors via ensheathing tumor nests, while iCAFs are enriched in late-stage tumors, and express high levels of cytokines and chemokines to aid immune cell recruitment and activation. This is supported by the induction of an iCAF-like phenotype with immunomodulatory functions in primary healthy fibroblasts exposed to skin cancer cell secretomes. Thus, targeting CAF variants holds promise to enhance immunotherapy efficacy in skin cancers.
Collapse
Affiliation(s)
- Agnes Forsthuber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bertram Aschenbrenner
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Korosec
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tina Jacob
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Krajic
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Daria Kholodniuk
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sophie Frech
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Shaohua Zhu
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Kim Purkhauser
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Lipp
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Franziska Werner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Vy Nguyen
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Soler Cardona
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Benedikt Weber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Gesslbauer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Jakob Nedomansky
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan N Wagner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Beate M Lichtenberger
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
23
|
Xie F, Luo S, Liu D, Lu X, Wang M, Liu X, Jia F, Pang Y, Shen Y, Zeng C, Ma X, Tang D, Tu L, Yang L, Cheng Y, Luo Y, Xie F, Hou H, Huang T, Ni B, Zhuang C, Zhao W, Li K, Zheng X, Bi W, Jia X, He Y, Wang S, Cao H, Wu K, Wang Y. Genomic and transcriptomic landscape of human gastrointestinal stromal tumors. Nat Commun 2024; 15:9495. [PMID: 39489749 PMCID: PMC11532483 DOI: 10.1038/s41467-024-53821-1] [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: 12/23/2022] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Gastrointestinal stromal tumor (GISTs) are clinically heterogenous exhibiting varying degrees of disease aggressiveness in individual patients. We comprehensively describe the genomic and transcriptomic landscape of a cohort of 117 GISTs including 31 low-risk, 18 intermediate-risk, 29 high-risk, 34 metastatic and 5 neoadjuvant GISTs from 105 patients. GISTs have notably low tumor mutation burden but widespread copy number variations. Aggressive GISTs harbor remarkably more genomic aberrations than low-/intermediate-risk GISTs. Complex genomic alterations, chromothripsis and kataegis, occur selectively in aggressive GISTs. Despite the paucity of mutations, recurrent inactivating YLPM1 mutations are identified (10.3%, 7 of 68 patients), enriched in high-risk/metastatic GIST and functional study further demonstrates YLPM1 inactivation promotes GIST proliferation, growth and oxidative phosphorylation. Spatially and temporally separated GISTs from individual patients demonstrate complex tumor heterogeneity in metastatic GISTs. Finally, four prominent subtypes are proposed with different genomic features, expression profiles, immune characteristics, clinical characteristics and subtype-specific treatment strategies. This large-scale analysis depicts the landscape and provides further insights into GIST pathogenesis and precise treatment.
Collapse
Affiliation(s)
- Feifei Xie
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Shuzhen Luo
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Dongbing Liu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fujian Jia
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Yuzhi Pang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yanying Shen
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chunling Zeng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xinli Ma
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Daoqiang Tang
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Lin Tu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Linxi Yang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fanfan Xie
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Hao Hou
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Tao Huang
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, 200031, Shanghai, China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chun Zhuang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Wenyi Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Ke Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Wenbo Bi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yi He
- Department of Urology, No.1 Hospital of Jiaxing, 314000, Jiaxing, China
| | - Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| | - Kui Wu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China.
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
24
|
Medo M, Ng CKY, Medová M. A comprehensive comparison of tools for fitting mutational signatures. Nat Commun 2024; 15:9467. [PMID: 39487150 PMCID: PMC11530434 DOI: 10.1038/s41467-024-53711-6] [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: 10/06/2023] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
Mutational signatures connect characteristic mutational patterns in the genome with biological or chemical processes that take place in cancers. Analysis of mutational signatures can help elucidate tumor evolution, prognosis, and therapeutic strategies. Although tools for extracting mutational signatures de novo have been extensively benchmarked, a similar effort is lacking for tools that fit known mutational signatures to a given catalog of mutations. We fill this gap by comprehensively evaluating twelve signature fitting tools on synthetic mutational catalogs with empirically driven signature weights corresponding to eight cancer types. On average, SigProfilerSingleSample and SigProfilerAssignment/MuSiCal perform best for small and large numbers of mutations per sample, respectively. We further show that ad hoc constraining the list of reference signatures is likely to produce inferior results. Evaluation of real mutational catalogs suggests that the activity of signatures that are absent in the reference catalog poses considerable problems to all evaluated tools.
Collapse
Affiliation(s)
- Matúš Medo
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Charlotte K Y Ng
- Department for BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Michaela Medová
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Gupta R, Das CK, Nair SS, Pedraza-Bermeo AM, Zahalka AH, Kyprianou N, Bhardwaj N, Tewari AK. From foes to friends: rethinking the role of lymph nodes in prostate cancer. Nat Rev Urol 2024; 21:687-700. [PMID: 39095580 DOI: 10.1038/s41585-024-00912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/04/2024]
Abstract
Clinically localized prostate cancer is often treated with radical prostatectomy combined with pelvic lymph node dissection. Data suggest that lymph node dissection does improve disease staging, but its therapeutic value has often been debated, with few studies showing that lymph node removal directly improves oncological outcomes; however, lymph nodes are an important first site of antigen recognition and immune system activation and the success of many currently used immunological therapies hinges on this dogma. Evidence, particularly in the preclinical setting, has demonstrated that the success of immune checkpoint inhibitors is dampened by the removal of tumour-draining lymph nodes. Thus, whether lymph nodes are truly 'foes' or whether they are actually 'friends' in oncological care is an important idea to discuss.
Collapse
Affiliation(s)
- Raghav Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan K Das
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sujit S Nair
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ali H Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashutosh K Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Stephan C, Al Assaad M, Levine MF, Deshpande A, Sigouros M, Manohar J, Sboner A, Elemento O, Pavlick AC, Mosquera JM. Whole genome sequencing elucidates etiological differences in MCPyV-negative Merkel cell carcinoma. Pathol Res Pract 2024; 263:155668. [PMID: 39427588 DOI: 10.1016/j.prp.2024.155668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
Merkel cell carcinoma (MCC) is an aggressive neuroendocrine neoplasm of the skin. Immunosuppression, ultraviolet radiation and the integration of Merkel cell polyomavirus (MCPyV) have all been shown to be involved in the pathogenesis of this malignancy. We performed whole genome sequencing on two MCPyV-negative cases of MCC that demonstrated very different clinical presentations and outcomes, and mutational profiles. The first case exhibited a highly aggressive clinical course, absence of UV-signature mutations and a low tumor mutational burden. A rearrangement in the tumor suppressor gene SUFU was identified, a likely driver and potential target of the Hedgehog signaling pathway. Meanwhile, the second case exhibited a less aggressive behavior, harbored UV-signature mutations, and a high mutational burden including mutations in TP53 and RB1.
Collapse
Affiliation(s)
- Carla Stephan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Majd Al Assaad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Anna C Pavlick
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Yuan P, Ma N, Xu B. Poly (adenosine diphosphate-ribose) polymerase inhibitors in the treatment of triple-negative breast cancer with homologous repair deficiency. Med Res Rev 2024; 44:2774-2792. [PMID: 38922930 DOI: 10.1002/med.22058] [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: 02/22/2023] [Revised: 04/23/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
Breast cancer (BC) is a highly heterogeneous disease, and the presence of germline breast cancer gene mutation (gBRCAm) is associated with a poor prognosis. Triple-negative breast cancer (TNBC) is a BC subtype, characterized by the absence of hormone and growth factor receptor expression, making therapeutic decisions difficult. Defects in the DNA damage response pathway due to mutation in breast cancer genes (BRCA 1/2) lead to homologous recombination deficiency (HRD). However, in HRD conditions, poly (adenosine diphosphate-ribose) polymerase (PARP) proteins repair DNA damage and lead to tumor cell survival. Biological understanding of HRD leads to the development of PARP inhibitors (PARPi), which trap PARP proteins and cause genomic instability and tumor cell lysis. HRD assessment can be an important biomarker in identifying gBRCAm patients with BC who could benefit from PARPi therapy. HRD can be identified by homologous recombination repair (HRR) gene-based assays, genomic-scarring assays and mutational signatures, transcription and protein expression profiles, and functional assays. However, gold standard methodologies that are robust and reliable to assess HRD are not available currently. Hence, there is a pressing need to develop accurate biomarkers identifying HRD tumors to guide targeted therapies such as PARPi in patients with BC. HRD assessment has shown fruitful outcomes in chemotherapy studies and preliminary evidence on PARPi intervention as monotherapy and combination therapy in HRD-stratified patients. Furthermore, ongoing trials are exploring the potential of PARPi in BC and clinically complex TNBC settings, where HRD testing is used as an adjunct to stratify patients based on BRCA mutations.
Collapse
Affiliation(s)
- Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Ma
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Yoon KA, Kim Y, Jung SY, Ryu JS, Kim KH, Lee EG, Chae H, Kwon Y, Kim J, Park JB, Kong SY. Proteogenomic analysis dissects early-onset breast cancer patients with prognostic relevance. Exp Mol Med 2024:10.1038/s12276-024-01332-w. [PMID: 39482530 DOI: 10.1038/s12276-024-01332-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 07/05/2024] [Accepted: 07/29/2024] [Indexed: 11/03/2024] Open
Abstract
Early-onset breast cancer is known for its aggressive clinical characteristics and high prevalence in East Asian countries, but a comprehensive understanding of its molecular features is still lacking. In this study, we conducted a proteogenomic analysis of 126 treatment-naïve primary tumor tissues obtained from Korean patients with young breast cancer (YBC) aged ≤40 years. By integrating genomic, transcriptomic, and proteomic data, we identified five distinct functional subgroups that accurately represented the clinical characteristics and biological behaviors of patients with YBC. Our integrated approach could be used to determine the proteogenomic status of HER2, enhancing its clinical significance and prognostic value. Furthermore, we present a proteome-based homologous recombination deficiency (HRD) analysis that has the potential to overcome the limitations of conventional genomic HRD tests, facilitating the identification of new patient groups requiring targeted HR deficiency treatments. Additionally, we demonstrated that protein-RNA correlations can be used to predict the late recurrence of hormone receptor-positive breast cancer. Within each molecular subtype of breast cancer, we identified functionally significant protein groups whose differential abundance was closely correlated with the clinical progression of breast cancer. Furthermore, we derived a recurrence predictive index capable of predicting late recurrence, specifically in luminal subtypes, which plays a crucial role in guiding decisions on treatment durations for YBC patients. These findings improve the stratification and clinical implications for patients with YBC by contributing to the optimal adjuvant treatment and duration for favorable clinical outcomes.
Collapse
Affiliation(s)
- Kyong-Ah Yoon
- Department of Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Youngwook Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
| | - So-Youn Jung
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Jin-Sun Ryu
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Korea
- Laboratory Animal Research Facility, Research Institute, National Cancer Center, Goyang, Korea
| | - Kyung-Hee Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea
- Proteomics Core Facility, Research Core Center, Research Institute, National Cancer Center, Goyang, Korea
| | - Eun-Gyeong Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Heejung Chae
- Cancer Data Center, Control Institute, National Cancer Center, Goyang, Korea
- Division of Medical Oncology, Hospital, National Cancer Center, Goyang, Korea
| | - Youngmee Kwon
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | | | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
| | - Sun-Young Kong
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
- Department of Laboratory Medicine, Research Institute, National Cancer Center Korea, Goyang, Korea.
- Department of Targeted Therapy Branch, Research Institute, National Cancer Center, Goyang, Korea.
| |
Collapse
|
29
|
Basin MF, Miguel CM, Jacob JM, Goldberg H, Grivas P, Spiess PE, Necchi A, Kamat AM, Pavlick DC, Huang RSP, Lin DI, Danziger N, Sokol ES, Sivakumar S, Graf R, Cheng L, Vasan N, Ross J, Basnet A, Bratslavsky G. Single-Hit and Multi-hit PIK3CA Short Variant Genomic Alterations in Clinically Advanced Prostate Cancer: A Genomic Landscape Study. Target Oncol 2024; 19:981-990. [PMID: 39369133 PMCID: PMC11557715 DOI: 10.1007/s11523-024-01100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Tumors harboring two or more PIK3CA short variant (SV) ("multi-hit") mutations have been linked to improved outcomes with anti-PIK3CA-targeted therapies in breast cancer. The landscape and clinical implications of multi-hit PIK3CA alterations in clinically advanced prostate cancer (CAPC) remains elusive. OBJECTIVE To evaluate the genomic landscape of single-hit and multi-hit PIK3CA genomic alterations in CAPC. PATIENTS AND METHODS The Foundation Medicine FoundationCore database was used to identify 19,978 CAPC tumors that underwent hybrid capture-based comprehensive genomic profiling to evaluate all classes of genomic alterations (GA) and determine tumor mutational burden (TMB), microsatellite instability (MSI), genomic ancestry, single-base substitution mutational signatures, and homologous recombination deficiency signature (HRDsig). Tumor cell PD-L1 expression was determined by IHC (Dako 22C3). RESULTS 18,741 (93.8%) tumors were PIK3CA wild type (WT), 1155 (5.8%) featured single PIK3CA SV, and 82 (0.4%) featured multi-hit PIK3CA SVs. Single-hit (6.6 versus 3.8; p < 0.0001) and multi-hit (12.8 versus 3.8; p < 0.0001) featured more driver GA per tumor than PIK3CA WT CAPC, as well as higher prevalence of MMR mutational signature, MSI high status, and TMB levels versus PIK3CA WT (p < 0.0001). Other differences in GA included higher frequencies of GA in BRCA2 in multi-hit versus WT (18.3% versus 8.5%; p = 0.0191), ATM in multi-hit versus WT (13.4% versus 5.6%; p = 0.02) and PTEN in single-hit versus WT (40.2% versus 30.1%; p < 0.0001). Homologous recombination deficiency signatures were higher in PIK3CA WT versus single-hit (11.2% versus 7.6%; p = 0.0002). There were no significant differences in PD-L1 expression among the three groups. CONCLUSIONS Identification of multi-hit PIK3CA GA in CAPC highlights a potentially unique phenotype that may be associated with response to anti-PIK3CA targeted therapy and checkpoint inhibition, supporting relevant clinical trial designs.
Collapse
Affiliation(s)
- Michael F Basin
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Carla M Miguel
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Joseph M Jacob
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Hanan Goldberg
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Petros Grivas
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Andrea Necchi
- IRCCS San Raffaele Hospital and Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | - Ryon Graf
- Foundation Medicine, Inc., Cambridge, MA, USA
| | - Liang Cheng
- Brown University Warren Alpert Medical School and the Legorreta Cancer Center at Brown University, Providence, RI, USA
| | | | - Jeffrey Ross
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
- Foundation Medicine, Inc., Cambridge, MA, USA
| | - Alina Basnet
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Gennady Bratslavsky
- Department of Urology, Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA.
| |
Collapse
|
30
|
Shia J, Sanchez-Vega F, Cho S, Chen JF, Chen CT, Bhanot U, Urganci N, Firat C, Ntiamoah P, Isidro RA, Srivastava A, Weiser MR, Mandelker D, Vakiani E, Boland CR, Garcia-Aguilar J, Stadler ZK. MSH6-proficient crypt foci in MSH6 constitutional mismatch repair deficiency: reversion of a frameshifted coding microsatellite to its wild-type sequence. Fam Cancer 2024; 23:569-577. [PMID: 39387980 DOI: 10.1007/s10689-024-00423-x] [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: 07/05/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024]
Abstract
The discovery of "mismatch repair deficient (MMRd)-crypt foci" in non-neoplastic intestinal mucosa in Lynch syndrome (LS) has significantly enhanced our understanding of how tumors and tumor immunity form and evolve in LS. In this study, we report the frequent presence of "mismatch repair proficient (MMRp)-crypt foci" in both non-neoplastic and neoplastic intestinal mucosa in a patient with constitutional MMR deficiency (CMMRD), who carried a germline MSH6 pathogenic variant (c.3261dupC) in trans with an MSH6 likely pathogenic variant (c.3724_3726del) and whose tissues were otherwise deficient in MMR globally. The MMRp-crypts occurred at a rate of 1.1/100 crypts in non-neoplastic intestinal mucosa and were readily discernible in adenomas > 1 cm. Sequencing analysis revealed normalization of the MSH6c.3261dupC variant in MMRp-adenoma crypts, indicating reverse frameshifting of the exon 5 C8 microsatellite. Interestingly but not surprisingly, the MMRp-adenoma crypts remained microsatellite-instability-high (MSI-H), and shared oncogenic APC mutations with the background MMRd-adenoma. Contrasting with MSH6-CMMRD, no PMS2-CMMRD individuals (0/5) harbored MMRp-crypts. In conclusion, our study documents distinct MMRp-crypts in MSH6-CMMRD, a phenomenon in keeping with MSH6 being a frequent target of MSI-H due to its coding microsatellite and suggesting that MSH6-CMMRD can potentially serve as a unique model system to further our understanding of MSH6's role in MSI-H tumor formation and evolution. Our findings also bear diagnostic implications; when using MMR immunohistochemistry as an ancillary tool in detecting CMMRD, awareness of these MMRp crypts can help avoid diagnostic pitfalls.
Collapse
Affiliation(s)
- Jinru Shia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Francisco Sanchez-Vega
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stanley Cho
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jie-Fu Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chin-Tung Chen
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umesh Bhanot
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nil Urganci
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Canan Firat
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter Ntiamoah
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raymond A Isidro
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amitabh Srivastava
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin R Weiser
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Efsevia Vakiani
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Julio Garcia-Aguilar
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Colorectal Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
31
|
Wei TT, Blanc E, Peidli S, Bischoff P, Trinks A, Horst D, Sers C, Blüthgen N, Beule D, Morkel M, Obermayer B. High-confidence calling of normal epithelial cells allows identification of a novel stem-like cell state in the colorectal cancer microenvironment. Int J Cancer 2024; 155:1655-1669. [PMID: 39031967 DOI: 10.1002/ijc.35079] [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: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/22/2024]
Abstract
Single-cell analyses can be confounded by assigning unrelated groups of cells to common developmental trajectories. For instance, cancer cells and admixed normal epithelial cells could adopt similar cell states thus complicating analyses of their developmental potential. Here, we develop and benchmark CCISM (for Cancer Cell Identification using Somatic Mutations) to exploit genomic single nucleotide variants for the disambiguation of cancer cells from genomically normal non-cancer cells in single-cell data. We find that our method and others based on gene expression or allelic imbalances identify overlapping sets of colorectal cancer versus normal colon epithelial cells, depending on molecular characteristics of individual cancers. Further, we define consensus cell identities of normal and cancer epithelial cells with higher transcriptome cluster homogeneity than those derived using existing tools. Using the consensus identities, we identify significant shifts of cell state distributions in genomically normal epithelial cells developing in the cancer microenvironment, with immature states increased at the expense of terminal differentiation throughout the colon, and a novel stem-like cell state arising in the left colon. Trajectory analyses show that the new cell state extends the pseudo-time range of normal colon stem-like cells in a cancer context. We identify cancer-associated fibroblasts as sources of WNT and BMP ligands potentially contributing to increased plasticity of stem cells in the cancer microenvironment. Our analyses advocate careful interpretation of cell heterogeneity and plasticity in the cancer context and the consideration of genomic information in addition to gene expression data when possible.
Collapse
Affiliation(s)
- Tzu-Ting Wei
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Peidli
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
| | - Philip Bischoff
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| | - Alexandra Trinks
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioportal Single Cells, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| | - Christine Sers
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
- German Cancer Consortium Partner Site Berlin, German Cancer Research Center, Heidelberg, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Morkel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioportal Single Cells, Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
32
|
Tang C, Castillon VJ, Waters M, Fong C, Park T, Boscenco S, Kim S, Pekala K, Carrot-Zhang J, Hakimi AA, Schultz N, Ostrovnaya I, Gusev A, Jee J, Reznik E. Obesity-dependent selection of driver mutations in cancer. Nat Genet 2024; 56:2318-2321. [PMID: 39468367 PMCID: PMC11549034 DOI: 10.1038/s41588-024-01969-3] [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: 01/09/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024]
Abstract
Obesity is a risk factor for cancer, but whether obesity is linked to specific genomic subtypes of cancer is unknown. We examined the relationship between obesity and tumor genotype in two clinicogenomic corpora. Obesity was associated with specific driver mutations in lung adenocarcinoma, endometrial carcinoma and cancers of unknown primaries, independent of clinical covariates, demographic factors and genetic ancestry. Obesity is therefore a driver of etiological heterogeneity in some cancers.
Collapse
Affiliation(s)
- Cerise Tang
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Venise Jan Castillon
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michele Waters
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chris Fong
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tricia Park
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sonia Boscenco
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susie Kim
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelly Pekala
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jian Carrot-Zhang
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikolaus Schultz
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Ostrovnaya
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham & Women's Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Justin Jee
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ed Reznik
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
33
|
Domingo-Sabugo C, Willis-Owen SA, Mandal A, Nastase A, Dwyer S, Brambilla C, Gálvez JH, Zhuang Q, Popat S, Eveleigh R, Munter M, Lim E, Nicholson AG, Lathrop GM, Cookson WO, Moffatt MF. Genomic analysis defines distinct pancreatic and neuronal subtypes of lung carcinoid. J Pathol 2024; 264:332-343. [PMID: 39329437 DOI: 10.1002/path.6352] [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: 02/02/2024] [Revised: 06/14/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024]
Abstract
Lung carcinoids (L-CDs) are rare, poorly characterised neuroendocrine tumours (NETs). L-CDs are more common in women and are not the consequence of cigarette smoking. They are classified histologically as typical carcinoids (TCs) or atypical carcinoids (ACs). ACs confer a worse survival. Histological classification is imperfect, and there is increasing interest in molecular markers. We therefore investigated global transcriptomic and epigenomic profiles of 15 L-CDs resected with curative intent at Royal Brompton Hospital. We identified underlying mutations and structural abnormalities through whole-exome sequencing (WES) and single nucleotide polymorphism (SNP) genotyping. Transcriptomic clustering algorithms identified two distinct L-CD subtypes. These showed similarities either to pancreatic or neuroendocrine tumours at other sites and so were named respectively L-CD-PanC and L-CD-NeU. L-CD-PanC tumours featured upregulation of pancreatic and metabolic pathway genes matched by promoter hypomethylation of genes for beta cells and insulin secretion (p < 1 × 10-6). These tumours were centrally located and showed mutational signatures of activation-induced deaminase/apolipoprotein B editing complex activity, together with genome-wide DNA methylation loss enriched in repetitive elements (p = 2.2 × 10-16). By contrast, the L-CD-NeU group exhibited upregulation of neuronal markers (adjusted p < 0.01) and was characterised by focal spindle cell morphology (p = 0.04), peripheral location (p = 0.01), high mutational load (p = 2.17 × 10-4), recurrent copy number alterations, and enrichment for ACs. Mutations affected chromatin remodelling and SWI/SNF complex pathways. L-CD-NeU tumours carried a mutational signature attributable to aflatoxin and aristolochic acid (p = 0.05), suggesting a possible environmental exposure in their pathogenesis. Immunologically, myeloid and T-cell markers were enriched in L-CD-PanC and B-cell markers in L-CD-NeU tumours. The substantial epigenetic and non-coding differences between L-CD-PanC and L-CD-NeU open new possibilities for biomarker selection and targeted treatment of L-CD. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | - Amit Mandal
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anca Nastase
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah Dwyer
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Cecilia Brambilla
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Histopathology, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - José Héctor Gálvez
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Qinwei Zhuang
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Sanjay Popat
- Royal Marsden Hospital NHS Foundation Trust, London and Surrey, UK
- The Institute of Cancer Research, London, UK
| | - Robert Eveleigh
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Markus Munter
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | - Eric Lim
- Department of Thoracic Surgery, Royal Brompton Hospital, London, UK
| | - Andrew G Nicholson
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Histopathology, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - G Mark Lathrop
- Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montréal, QC, Canada
| | | | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
34
|
Baron M, Labreche K, Veyri M, Désiré N, Bouzidi A, Seck-Thiam F, Charlotte F, Rousseau A, Morin V, Nakid-Cordero C, Abbar B, Picca A, Le Cann M, Balegroune N, Gauthier N, Theodorou I, Touat M, Morel V, Bielle F, Samri A, Alentorn A, Sanson M, Roos-Weil D, Haioun C, Poullot E, De Septenville AL, Davi F, Guihot A, Boelle PY, Leblond V, Coulet F, Spano JP, Choquet S, Autran B. Epstein-Barr virus and immune status imprint the immunogenomics of non-Hodgkin lymphomas occurring in immune-suppressed environments. Haematologica 2024; 109:3615-3630. [PMID: 38841782 PMCID: PMC11532699 DOI: 10.3324/haematol.2023.284332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Non-Hodgkin lymphomas (NHL) commonly occur in immunodeficient patients, both those infected by human immunodeficiency virus (HIV) and those who have been transplanted, and are often driven by Epstein-Barr virus (EBV) with cerebral localization, raising the question of tumor immunogenicity, a critical issue for treatment responses. We investigated the immunogenomics of 68 lymphoproliferative disorders from 51 immunodeficient (34 post-transplant, 17 HIV+) and 17 immunocompetent patients. Overall, 72% were large B-cell lymphoma and 25% were primary central nervous system lymphoma, while 40% were EBV+. Tumor whole-exome and RNA sequencing, along with a bioinformatics pipeline allowed analysis of tumor mutational burden, tumor landscape and tumor microenvironment and prediction of tumor neoepitopes. Both tumor mutational burden (2.2 vs. 3.4/Mb, P=0.001) and numbers of neoepitopes (40 vs. 200, P=0.00019) were lower in EBV+ than in EBV- NHL, regardless of the immune status. In contrast both EBV and the immune status influenced the tumor mutational profile, with HNRNPF and STAT3 mutations observed exclusively in EBV+ and immunodeficient NHL, respectively. Peripheral blood T-cell responses against tumor neoepitopes were detected in all EBV- cases but in only half of the EBV+ ones, including responses against IgH-derived MHC-class-II restricted neoepitopes. The tumor microenvironment analysis showed higher CD8 T-cell infiltrates in EBV+ versus EBV- NHL, together with a more tolerogenic profile composed of regulatory T cells, type-M2 macrophages and an increased expression of negative immune-regulators. Our results highlight that the immunogenomics of NHL in patients with immunodeficiency primarily relies on the tumor EBV status, while T-cell recognition of tumor- and IgH-specific neoepitopes is conserved in EBV- patients, offering potential opportunities for future T-cell-based immune therapies.
Collapse
Affiliation(s)
- Marine Baron
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris, France; Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris.
| | - Karim Labreche
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Marianne Veyri
- Sorbonne Université, INSERM, Pierre et Louis Institute of Epidemiology and Public Health, F-75013 Paris France, Theravir Team, Department of Medical Oncology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Nathalie Désiré
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Amira Bouzidi
- Sorbonne Université, INSERM, Research Unit on Cardiovascular and Metabolic Disease UMR ICAN, Department of Endocrine Biochemistry and Oncology, AP-HP, Hôpital-Pitié-Salpêtrière, F-75013 Paris
| | - Fatou Seck-Thiam
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Frédéric Charlotte
- Sorbonne Université, Department of Anatomy and Pathologic Cytology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Alice Rousseau
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Véronique Morin
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Cécilia Nakid-Cordero
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Baptiste Abbar
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Alberto Picca
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Marie Le Cann
- Department of Clinical Haematology, AP-HP, Hôpital Kremlin Bicêtre, F-94270 Le Kremlin
| | - Noureddine Balegroune
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Nicolas Gauthier
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | | | - Mehdi Touat
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Véronique Morel
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Franck Bielle
- Sorbonne Université, Department of Neuropathology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013, Paris
| | - Assia Samri
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Agusti Alentorn
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Marc Sanson
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Damien Roos-Weil
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Corinne Haioun
- Lymphoid malignancies Unit, AP-HP, Mondor Hospital, F-94000 Créteil
| | - Elsa Poullot
- Department of Anatomy and Pathologic Cytology, AP-HP, Mondor Hospital, F-94000 Créteil
| | - Anne Langlois De Septenville
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Department of Biological Hematology, AP-HP, Hôpital Pitié-Salpêtrière, Paris
| | - Frédéric Davi
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Department of Biological Hematology, AP-HP, Hôpital Pitié-Salpêtrière, Paris
| | - Amélie Guihot
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Pierre-Yves Boelle
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Véronique Leblond
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Florence Coulet
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Microsatellites Instability and Cancer, CRSA, Department of Medical Genetics, AP-HP, Pitié-Salpêtrière Hospital, F-75013 Paris
| | - Jean-Philippe Spano
- Sorbonne Université, INSERM, Pierre et Louis Institute of Epidemiology and Public Health, F-75013 Paris France, Theravir Team, Department of Medical Oncology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Sylvain Choquet
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Brigitte Autran
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| |
Collapse
|
35
|
Berzero G, Pieri V, Palazzo L, Finocchiaro G, Filippi M. Liquid biopsy in brain tumors: moving on, slowly. Curr Opin Oncol 2024; 36:521-529. [PMID: 39011725 DOI: 10.1097/cco.0000000000001079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW Due to limited access to the tumor, there is an obvious clinical potential for liquid biopsy in patients with primary brain tumors. Here, we review current approaches, present limitations to be dealt with, and new promising data that may impact the field. RECENT FINDINGS The value of circulating tumor cell-free DNA (ctDNA) in the cerebrospinal fluid (CSF) for the noninvasive diagnosis of primary brain tumors has been confirmed in several reports. The detection of ctDNA in the peripheral blood is desirable for patient follow-up but requires ultrasensitive methods to identify low mutant allelic frequencies. Digital PCR approaches and targeted gene panels have been used to identify recurrent hotspot mutations and copy number variations (CNVs) from CSF or plasma. Tumor classification from circulating methylomes in plasma has been actively pursued, although the need of advanced bioinformatics currently hampers clinical application. The use of focused ultrasounds to open the blood-brain barrier may represent a way to enrich of ctDNA the peripheral blood and enhance plasma-based liquid biopsy. SUMMARY Monitoring CNVs and hotspot mutations by liquid biopsy is a promising tool to detect minimal residual disease and strengthen response assessment in patients with primary brain tumors. Novel methods to increase the relative and/or absolute amount of ctDNA can improve the clinical potential of plasma-based liquid biopsies.
Collapse
Affiliation(s)
- Giulia Berzero
- Neurology Unit, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University
| | - Valentina Pieri
- Neurology Unit, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University
| | - Leonardo Palazzo
- Neurology Unit, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University
| | | | - Massimo Filippi
- Neurology Unit, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University
- Neurorehabilitation Unit, Neurophysiology Unit, Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
36
|
Randerson-Moor J, Davies J, Harland M, Nsengimana J, Bigirumurame T, Walker C, Laye J, Appleton ES, Ball G, Cook GP, Bishop DT, Salmond RJ, Newton-Bishop J. Systemic Inflammation, the Peripheral Blood Transcriptome, and Primary Melanoma. J Invest Dermatol 2024; 144:2513-2529.e17. [PMID: 38583742 DOI: 10.1016/j.jid.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 04/09/2024]
Abstract
Peripheral blood transcriptomes from 383 patients with newly diagnosed melanoma were subjected to differential gene expression analysis. The hypotheses were that impaired systemic immunity is associated with poorer prognosis (thicker tumors and fewer tumor-infiltrating lymphocytes) and evidence of systemic inflammation (high-sensitivity CRP and fibrinogen levels). Higher fibrinogen levels were associated with thicker primary tumors. In single-gene analysis, high-sensitivity CRP levels were significantly associated with higher blood CD274 expression (coding for PD-L1), but each was independently prognostic, with high-sensitivity CRP associated with increased mortality and higher CD274 protective, independent of age. Pathway analysis identified downregulation of immune cell signaling pathways in the blood of people with thicker tumors and notable upregulation of signal transducer and activator of transcription 1 gene STAT1 in people with brisk tumor-infiltrating lymphocytes. Transcriptomic data provided evidence for increased NF-kB signaling with higher inflammatory markers but with reduction in expression of HLA class II molecules and higher CD274, suggesting that aberrant systemic inflammation is a significant mediator of reduced immune function in melanoma. In summary, transcriptomic data revealed evidence of reduced immune function in patients with thicker tumors and fewer tumor-infiltrating lymphocytes at diagnosis. Inflammatory markers were associated with thicker primaries and independently with death from melanoma, suggesting that systemic inflammation contributes to that reduced immune function.
Collapse
Affiliation(s)
- Juliette Randerson-Moor
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - John Davies
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Leeds Institute of Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Mark Harland
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Theophile Bigirumurame
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Christopher Walker
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jon Laye
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Elizabeth S Appleton
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom; Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Graham P Cook
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - D Timothy Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert J Salmond
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research (LIMR), School of Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
37
|
Yun J, Youn YC, Kim HR. Association Between Clonal Hematopoiesis of Indeterminate Potential and Brain β-Amyloid Deposition in Korean Patients With Cognitive Impairment. Ann Lab Med 2024; 44:576-580. [PMID: 38802262 PMCID: PMC11375198 DOI: 10.3343/alm.2024.0086] [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: 02/16/2024] [Revised: 03/12/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
Few studies have focused on the association between clonal hematopoiesis of indeterminate potential (CHIP) and β-amyloid (Aβ) deposition in the brain, which causes Alzheimer's disease. We aimed to investigate the potential role of CHIP in brain Aβ deposition in Korean patients. We enrolled 58 Korean patients over 50 yrs of age with cognitive impairment who underwent brain Aβ positron emission tomography. We explored CHIP in their peripheral blood using deep-targeted next-generation sequencing. Irrespective of the presence or absence of brain Aβ deposition, mutations in DNMT3A and the C:G>T:A single-nucleotide variants were identified as the primary characteristics, which reflect aged hematopoiesis in the study population. Multivariate logistic regression revealed that the presence of CHIP was not associated with brain Aβ deposition. As both CHIP and brain Aβ deposition are associated with aging, further research is required to elucidate their possible interplay.
Collapse
Affiliation(s)
- Jiwon Yun
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul, Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hye Ryoun Kim
- Department of Laboratory Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Yan T, Zhou W, Li C. Discovery of a T cell proliferation-associated regulator signature correlates with prognosis risk and immunotherapy response in bladder cancer. Int Urol Nephrol 2024; 56:3447-3462. [PMID: 38789872 DOI: 10.1007/s11255-024-04086-6] [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: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND The efficacy of immunotherapy is heavily influenced by T cell activity. This study aimed to examine how T cell proliferation regulators can predict the prognosis and response to immunotherapy in patients with bladder cancer (BCa). METHODS T cell proliferation-related subtypes were determined by employing the non-negative matrix factorization (NMF) algorithm that analyzed the expression patterns of T cell proliferation regulators. Subtypes were assessed for variations in prognosis, immune infiltration, and functional behaviors. Subsequently, a risk model related to T cell proliferation was created through Cox and Lasso regression analyses in the TCGA cohort and then confirmed in two GEO cohorts and an immunotherapy cohort. RESULTS BCa patients were categorized into two subtypes (C1 and C2) according to the expression profiles of 31 T cell proliferation-related genes (TRGs) with distinct prognoses and immune landscapes. The C2 subtype had a shorter overall survival (OS), with higher levels of M2 macrophage infiltration, and the activation of cancer-related pathways than the C1 subtype. Following this, thirteen prognosis-related genes that were involved in T cell proliferation were utilized to create the prognostic signature. The model's predictive accuracy was confirmed by analyzing both internal and external datasets. Individuals in the high-risk category experienced a poorer prognosis, increased immunosuppressive factors in the tumor microenvironment, and diminished responses to immunotherapy. Additionally, the immunotherapeutic prediction efficacy of the model was further confirmed by an immunotherapy cohort (anti-PD-L1 in the IMvigor210 cohort). CONCLUSIONS Our study characterized two subtypes linked to T cell proliferation in BCa patients with distinct prognoses and tumor microenvironment (TME) patterns, providing new insights into the heterogeneity of T cell proliferation in BCa and its connection to the immune landscape. The signature has prospective clinical implications for predicting outcomes and may help physicians to select prospective responders who prioritize current immunotherapy.
Collapse
Affiliation(s)
- Ting Yan
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China
| | - Wei Zhou
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, People's Republic of China
| | - Chun Li
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China.
| |
Collapse
|
39
|
Mizutani T, Boretto M, Lim S, Drost J, González DM, Oka R, Geurts MH, Begthel H, Korving J, van Es JH, van Boxtel R, Clevers H. Recapitulating the adenoma-carcinoma sequence by selection of four spontaneous oncogenic mutations in mismatch-repair-deficient human colon organoids. NATURE CANCER 2024:10.1038/s43018-024-00841-x. [PMID: 39487295 DOI: 10.1038/s43018-024-00841-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/23/2024] [Indexed: 11/04/2024]
Abstract
Carcinogenesis results from the sequential acquisition of oncogenic mutations that convert normal cells into invasive, metastasizing cancer cells. Colorectal cancer exemplifies this process through its well-described adenoma-carcinoma sequence, modeled previously using clustered regularly interspaced short palindromic repeats (CRISPR) to induce four consecutive mutations in wild-type human gut organoids. Here, we demonstrate that long-term culture of mismatch-repair-deficient organoids allows the selection of spontaneous oncogenic mutations through the sequential withdrawal of Wnt agonists, epidermal growth factor (EGF) agonists and the bone morphogenetic protein (BMP) antagonist Noggin, while TP53 mutations were selected through the addition of Nutlin-3. Thus, organoids sequentially acquired mutations in AXIN1 and AXIN2 (Wnt pathway), TP53, ACVR2A and BMPR2 (BMP pathway) and NRAS (EGF pathway), gaining complete independence from stem cell niche factors. Quadruple-pathway (Wnt, EGF receptor, p53 and BMP) mutant organoids formed solid tumors upon xenotransplantation. This demonstrates that carcinogenesis can be recapitulated in a DNA repair-mutant background through in vitro selection that targets four consecutive cancer pathways.
Collapse
Affiliation(s)
- Tomohiro Mizutani
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Matteo Boretto
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Sangho Lim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jarno Drost
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Diego Montiel González
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rurika Oka
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Maarten H Geurts
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Ruben van Boxtel
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland.
| |
Collapse
|
40
|
Amodio V, Vitiello PP, Bardelli A, Germano G. DNA repair-dependent immunogenic liabilities in colorectal cancer: opportunities from errors. Br J Cancer 2024; 131:1576-1590. [PMID: 39271762 PMCID: PMC11554791 DOI: 10.1038/s41416-024-02848-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the major causes of cancer death worldwide. Chemotherapy continues to serve as the primary treatment modality, while immunotherapy is largely ineffective for the majority of CRC patients. Seminal discoveries have emphasized that modifying DNA damage response (DDR) mechanisms confers both cell-autonomous and immune-related vulnerabilities across various cancers. In CRC, approximately 15% of tumours exhibit alterations in the mismatch repair (MMR) machinery, resulting in a high number of neoantigens and the activation of the type I interferon response. These factors, in conjunction with immune checkpoint blockades, collectively stimulate anticancer immunity. Furthermore, although less frequently, somatic alterations in the homologous recombination (HR) pathway are observed in CRC; these defects lead to genome instability and telomere alterations, supporting the use of poly (ADP-ribose) polymerase (PARP) inhibitors in HR-deficient CRC patients. Additionally, other DDR inhibitors, such as Ataxia Telangiectasia and Rad3-related protein (ATR) inhibitors, have shown some efficacy both in preclinical models and in the clinical setting, irrespective of MMR proficiency. The aim of this review is to elucidate how preexisting or induced vulnerabilities in DNA repair pathways represent an opportunity to increase tumour sensitivity to immune-based therapies in CRC.
Collapse
Affiliation(s)
- V Amodio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - P P Vitiello
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy
| | - A Bardelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Oncology, Molecular Biotechnology Center, University of Torino, 10126, Turin, Italy.
| | - G Germano
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Department of Medical Biotechnologies and Translational Medicine, University of Milano, 20133, Milan, Italy.
| |
Collapse
|
41
|
Klümper N, Grünwald V, Hartmann A, Hölzel M, Eckstein M. The Role of Microsatellite Instability/DNA Mismatch Repair Deficiency and Tumor Mutational Burden as Biomarkers in Predicting Response to Immunotherapy in Castration-resistant Prostate Cancer. Eur Urol 2024; 86:388-390. [PMID: 38744632 DOI: 10.1016/j.eururo.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
Large trials of immune checkpoint inhibitors (ICIs) in castration-resistant prostate cancer (CRPC) have mostly failed. Biomarker-selected CRPC patients, especially those with high microsatellite instability (MSI-H), mismatch repair deficiency (dMMR), or elevated tumor mutational burden (TMB), may benefit from single-agent ICIs. Despite their rarity in CRPC (∼2-5%), identification of MSI-H, dMMR, or TMB-H could improve patient selection for immunotherapy.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology, University Hospital Bonn, Bonn, Germany; Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; EMN Comprehensive Cancer Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research, Munich, Germany
| | - Michael Hölzel
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; EMN Comprehensive Cancer Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research, Munich, Germany
| |
Collapse
|
42
|
Taiyab A, Ashraf A, Sulaimani MN, Rathi A, Shamsi A, Hassan MI. Role of MTH1 in oxidative stress and therapeutic targeting of cancer. Redox Biol 2024; 77:103394. [PMID: 39418911 PMCID: PMC11532495 DOI: 10.1016/j.redox.2024.103394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Cancer cells maintain high levels of reactive oxygen species (ROS) to drive their growth, but ROS can trigger cell death through oxidative stress and DNA damage. To survive enhanced ROS levels, cancer cells activate their antioxidant defenses. One such defense is MTH1, an enzyme that prevents the incorporation of oxidized nucleotides into DNA, thus preventing DNA damage and allowing cancer to proliferate. MTH1 levels are often elevated in many cancers, and thus, inhibiting MTH1 is an attractive strategy for suppressing tumor growth and metastasis. Targeted MTH1 inhibition can induce DNA damage in cancer cells, exploiting their vulnerability to oxidative stress and selectively targeting them for destruction. Targeting MTH1 is promising for cancer treatment because normal cells have lower ROS levels and are less dependent on these pathways, making the approach both effective and specific to cancer. This review aims to investigate the potential of MTH1 as a therapeutic target, especially in cancer treatment, offering detailed insights into its structure, function, and role in disease progression. We also discussed various MTH1 inhibitors that have been developed to selectively induce oxidative damage in cancer cells, though their effectiveness varies. In addition, this review provide deeper mechanistic insights into the role of MTH1 in cancer prevention and oxidative stress management in various diseases.
Collapse
Affiliation(s)
- Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Anam Ashraf
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Md Nayab Sulaimani
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Aanchal Rathi
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, P.O. Box 346, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
43
|
Tan N, Li Y, Ying J, Chen W. Histological transformation in lung adenocarcinoma: Insights of mechanisms and therapeutic windows. J Transl Int Med 2024; 12:452-465. [PMID: 39513032 PMCID: PMC11538883 DOI: 10.1515/jtim-2024-0019] [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] [Indexed: 11/15/2024] Open
Abstract
Histological transformation from lung adenocarcinoma (ADC) to small cell lung carcinoma (SCLC), large cell neuroendocrine carcinoma (LCNEC), squamous cell carcinoma (SCC), and sarcomatoid carcinoma (PSC) after targeted therapies is recognized as a mechanism of resistance in ADC treatments. Patients with transformed lung cancer typically experience a poor prognosis and short survival time. However, effective treatment options for these patients are currently lacking. Therefore, understanding the mechanisms underlying histological transformation is crucial for the development of effective therapies. Hypotheses including intratumoral heterogeneity, cancer stem cells, and alteration of suppressor genes have been proposed to explain the mechanism of histological transformation. In this review, we provide a comprehensive overview of the known molecular features and signaling pathways of transformed tumors, and summarized potential therapies based on previous findings.
Collapse
Affiliation(s)
- Nuopei Tan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Lobo A, Collins K, Kaushal S, Acosta AM, Akgul M, Adhya AK, Al-Ahmadie HA, Al-Obaidy KI, Amin A, Amin MB, Aron M, Balzer BL, Biswal R, Mohanty S, Browning L, Chakrabarti I, Cima L, Cimadamore A, Desai S, Dhillon J, Deshwal A, Diego GG, Diwaker P, Galea LA, Magi-Galluzzi C, Giannico GA, Gupta NS, Haider A, Hirsch MS, Iczkowski KA, Arora S, Jain E, Jain D, Jha S, Kandukuri S, Kao CS, Kryvenko ON, Kumar RM, Kumari N, Kunju LP, Kuthi L, Lobo J, Lopez JI, Luthringer DJ, Maclean F, Manini C, Mannan R, Martos MG, Mehra R, Menon S, Mishra P, Moch H, Montironi R, Baisakh MR, Netto GJ, Nigam LK, Osunkoya AO, Pagliuca F, Paner GP, Panizo A, Parwani AV, Picken MM, Prendeville S, Przybycin CG, Purkait S, Queipo FJ, Rao BV, Rao P, Reuter VE, Sancheti S, Sangoi AR, Sardana R, Satturwar S, Shah RB, Sharma S, Dixit M, Verma M, Sirohi D, Smith SC, Soni S, Sundaram S, Swain M, Tretiakova M, Trpkov K, MuñizUnamunzaga G, Zhou M, Williamson SR, Lopez-Beltran A, Cheng L, Mohanty SK. Advances, recognition, and interpretation of molecular heterogeneity among conventional and subtype histology of urothelial carcinoma (UC): a survey among urologic pathologists and comprehensive review of the literature. Histopathology 2024; 85:748-759. [PMID: 39075659 DOI: 10.1111/his.15287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024]
Abstract
AIMS Urothelial carcinoma (UC) demonstrates significant molecular and histologic heterogeneity. The WHO 2022 classification has hinted at adding molecular signatures to the morphologic diagnosis. As morphology and associated molecular repertoire may potentially translate to choices of and response to therapy and relapse rate, broader acceptability of recognizing these key features among uropathologists is needed. This prompted an international survey to ascertain the practice patterns in classical/subtype UC among uropathologists across the globe. METHODS AND RESULTS A survey instrument was shared among 98 uropathologists using SurveyMonkey software. Anonymized respondent data were analysed. The response rate was 85%. A majority were in concordance with the profiles of luminal (93%) and basal (82%) types. Opinion on the FGFR3 testing platform was variable. While 95% concurred that TERT promoter mutation is the key driver in UC, 72% had the opinion that APOBEC mutagenesis is the main signature in muscle invasive bladder cancer (MIBC). Uropathologists have divergent opinions on MIBC and ERCC2 mutations. Among the participants, 94% would quantify aggressive micropapillary and sarcomatoid histology, while 88% would reevaluate another transurethral resection of the bladder tumour specimen in nonmuscle invasive tumour with micropapillary, small cell, or sarcomatoid histology. A leading number agreed to specific molecular signatures of micropapillary (93%), plasmacytoid (97%), and small cell (86%) subtypes. Ninety-six percent of participants agreed that a small-cell component portends a more aggressive course and should be treated with neoadjuvant chemotherapy and 63% would perform HER2/neu testing only on oncologist's request in advanced tumours. Ninety percent agreed that microsatellite instability testing, although not a standard protocol, should be considered in young patients with upper tract UC. Eighty-six percent agreed that UC with high tumour mutational burden would be a better candidate for immunotherapy. CONCLUSION In the era of precision medicine, enhanced understanding of molecular heterogeneity of UC will contribute to better therapeutic options, novel biomarker discovery, innovative management protocols, and outcomes. Our survey provides a broad perspective of pathologists' perceptions and experience regarding incorporation of histomolecular approaches to "personalize" therapy. Due to variable clinical adoption, there is a need for additional data using uniform study criteria. This will drive generation of best practice guidelines in this area for widespread and consistent clinical utility.
Collapse
Affiliation(s)
- Anandi Lobo
- Department of Pathology, Kapoor Centre of Urology and Pathology, Raipur, India
| | - Katrina Collins
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Andres M Acosta
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Mahmut Akgul
- Department of Pathology, Albany Medical Center, Albany, USA
| | - Amit K Adhya
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | - Ali Amin
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Mahul B Amin
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Manju Aron
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Bonnie L Balzer
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Rupanita Biswal
- Department of Pathology, Bagchi Sri Shankara Cancer Hospital, Bhubaneswar, India
| | - Subashish Mohanty
- Department of Pathology, SUM Ultimate Medicare Hospital, Bhubaneswar, India
| | - Lisa Browning
- Department of Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Indranil Chakrabarti
- Department of Pathology, All India Institute of Medical Sciences, Kalyani, India
| | - Luca Cima
- Department of Pathology, Santa Chiara Hospital of Trento, Trento, Italy
| | - Alessia Cimadamore
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | | | | | - Guillermo G Diego
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Preeti Diwaker
- Department of Pathology, University College of Medical Sciences, New Delhi, India
| | - Laurence A Galea
- Department of Pathology, Melbourne Pathology, Melbourne, Australia
| | | | | | - Nilesh S Gupta
- Department of Pathology, Henry Ford Health System, Detroit, USA
| | - Aiman Haider
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | | | | | - Samriti Arora
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Ekta Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Shilpy Jha
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| | - Shivani Kandukuri
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Chia-Sui Kao
- Department of Pathology, Cleveland Clinic, Cleveland, USA
| | - Oleksandr N Kryvenko
- Department of Pathology, University of Miami Miller School of Medicine, Miami, USA
| | - Ramani M Kumar
- Department of Pathology, Dane Diagnostics, Palakkad, India
| | - Niraj Kumari
- Department of Pathology, All India Institute of Medical Sciences, Raebareli, India
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Levente Kuthi
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - João Lobo
- Department of Pathology, Portuguese Oncology Institute - Porto, Porto, Portugal
| | - Jose I Lopez
- Department of Pathology, Cruces University Hospital, Barakaldo, Spain
| | | | - Fiona Maclean
- Department of Pathology, Douglass Hanly Moir Pathology, Sydney, Australia
| | - Claudia Manini
- Department of Pathology, University of Turin, Turin, Italy
| | - Rahul Mannan
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - María G Martos
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Pritinanda Mishra
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Holger Moch
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Rodolfo Montironi
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Manas R Baisakh
- Department of Pathology, Prolife Diagnostics, Bhubaneswar, India
| | - George J Netto
- Department of Pathology, University of Pennsylvania, Philadelphia, USA
| | - Lovelesh K Nigam
- Department of Pathology, Institute of Kidney Diseases and Research Center, Ahmedabad, India
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, USA
| | - Francesca Pagliuca
- Department of Pathology, Università degliStudidella Campania Luigi Vanvitelli, Caserta, Italy
| | - Gladell P Paner
- Department of Pathology, University of Chicago, Chicago, USA
| | - Angel Panizo
- Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Hines, USA
| | - Susan Prendeville
- Department of Pathology, University Health Network, University of Toronto, Toronto, Canada
| | | | - Suvendu Purkait
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Francisco J Queipo
- Department of Pathology, Hospital Universitario de A Coruna, A Coruna, Spain
| | - B Vishal Rao
- Department of Pathology, Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Sankalp Sancheti
- Department of Pathology, Homi Bhabha Cancer Hospital, Punjab, India
| | - Ankur R Sangoi
- Department of Pathology, Stanford University, Stanford, USA
| | - Rohan Sardana
- Department of Pathology, Sardana Laboratories, Jalandhar, India
| | - Swati Satturwar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Rajal B Shah
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, USA
| | - Shivani Sharma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Mallika Dixit
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Monica Verma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Sirohi
- Department of Pathology, University of California, San Francisco, USA
| | - Steven C Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Shailesh Soni
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | | | | - Kiril Trpkov
- Department of Pathology, University of Calgary, Calgary, Canada
| | | | - Ming Zhou
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology, Unit of Anatomical Pathology, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | - Liang Cheng
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Sambit K Mohanty
- Department of Pathology, CORE Diagnostics, Gurgaon, India
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| |
Collapse
|
45
|
van der Ham CG, Suurenbroek LC, Kleisman MM, Antić Ž, Lelieveld SH, Yeong M, Westera L, Sonneveld E, Hoogerbrugge PM, van der Velden VHJ, van Leeuwen FN, Kuiper RP. Mutational mechanisms in multiply relapsed pediatric acute lymphoblastic leukemia. Leukemia 2024; 38:2366-2375. [PMID: 39232206 PMCID: PMC11518985 DOI: 10.1038/s41375-024-02403-7] [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: 06/14/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Pediatric acute lymphoblastic leukemia (ALL) is marked by low mutational load at initial diagnosis, which increases at relapse. To determine which processes are active in (relapsed) ALL and how they behave during disease progression before and after therapy, we performed whole genome sequencing on 97 tumor samples of 29 multiply relapsed ALL patients. Mutational load increased upon relapse in 28 patients and upon every subsequent relapse in 22 patients. In addition to two clock-like mutational processes, we identified UV-like damage, APOBEC activity, reactive oxygen species, thiopurine-associated damage and an unknown therapy component as drivers of mutagenesis. Mutational processes often affected patients over longer time periods, but could also occur in isolated events, suggesting the requirement of additional triggers. Thiopurine exposure was the most prominent source of new mutations in relapse, affecting over half of the studied patients in first and/or later relapse and causing potential relapse-driving mutations in multiple patients. Our data demonstrate that multiple mutational processes frequently act in parallel as prominent secondary drivers with dynamic activity during ALL development and progression.
Collapse
Affiliation(s)
| | | | | | - Željko Antić
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Marley Yeong
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Liset Westera
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | - Peter M Hoogerbrugge
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | | | | | - Roland P Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Genetics, Utrecht University Medical Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
46
|
Tomkova M, McClellan MJ, Crevel G, Shahid AM, Mozumdar N, Tomek J, Shepherd E, Cotterill S, Schuster-Böckler B, Kriaucionis S. Human DNA polymerase ε is a source of C>T mutations at CpG dinucleotides. Nat Genet 2024; 56:2506-2516. [PMID: 39390083 PMCID: PMC11549043 DOI: 10.1038/s41588-024-01945-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2024] [Indexed: 10/12/2024]
Abstract
C-to-T transitions in CpG dinucleotides are the most prevalent mutations in human cancers and genetic diseases. These mutations have been attributed to deamination of 5-methylcytosine (5mC), an epigenetic modification found on CpGs. We recently linked CpG>TpG mutations to replication and hypothesized that errors introduced by polymerase ε (Pol ε) may represent an alternative source of mutations. Here we present a new method called polymerase error rate sequencing (PER-seq) to measure the error spectrum of DNA polymerases in isolation. We find that the most common human cancer-associated Pol ε mutant (P286R) produces an excess of CpG>TpG errors, phenocopying the mutation spectrum of tumors carrying this mutation and deficiencies in mismatch repair. Notably, we also discover that wild-type Pol ε has a sevenfold higher error rate when replicating 5mCpG compared to C in other contexts. Together, our results from PER-seq and human cancers demonstrate that replication errors are a major contributor to CpG>TpG mutagenesis in replicating cells, fundamentally changing our understanding of this important disease-causing mutational mechanism.
Collapse
Affiliation(s)
- Marketa Tomkova
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK.
| | | | - Gilles Crevel
- Molecular and Cellular Sciences, St George's University London, London, UK
| | | | - Nandini Mozumdar
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Jakub Tomek
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Emelie Shepherd
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Sue Cotterill
- Molecular and Cellular Sciences, St George's University London, London, UK
| | | | | |
Collapse
|
47
|
Chang Y, Zhou X, Nie K, Liu J, Zhang S. Insights into the historical trajectory and research trends of immune checkpoint blockade in colorectal cancer: visualization and bibliometric analysis. Front Immunol 2024; 15:1478773. [PMID: 39544944 PMCID: PMC11560439 DOI: 10.3389/fimmu.2024.1478773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Background Colorectal cancer (CRC) is a malignant tumor that poses a significant threat to human health due to rising incidence and mortality rates. In recent years, immune checkpoint blockade (ICB) therapy, represented by Programmed death receptor 1 (PD-1), T-lymphocyte-associated protein 4 (CTLA-4), and others, has been widely applied in CRC and has achieved encouraging results in some patients and has become a hot topic in both clinical and basic research. Objective This study undertakes a comprehensive bibliometric analysis of ICB research in CRC, aiming to evaluate the current status, identify future trends, and provide scientific insights for researchers and decision-makers. Methods Utilizing the Web of Science Core Collection (WoSCC), articles focusing on ICB in CRC from 2000 to 2022 were retrieved. Knowledge mapping and bibliometric analysis were conducted using tools such as CiteSpace, VOSviewer, SCImago Graphicay, and the R package bibliometrix. Results 6,718 publications were analyzed from 24,846 institutions across 639 regions. Temporally, ICB research in CRC is rapidly advancing, led by the USA and China with extensive global collaborations. Sun Yat-sen University from China stands out as the institution with the highest number of publications. Professor Thierry Andre from Sorbonne University in France is identified as a prolific author in this field, engaging in extensive collaboration for clinical trials on a global scale. Publications related to this research topic were published in 1,142 academic journals, demonstrating a positive co-citation relationship. Key clustering and burst terms analysis indicate that current research on ICB in CRC has shifted from basic experiments to clinical trials and from universal healthcare to precision medicine. Conclusion ICB therapies have shown substantial progress in CRC, highlighting their therapeutic potential. Research trends emphasize deeper drug mechanisms, treatment efficacy prediction, managing immune-related adverse events, and exploring novel drug delivery methods. Collaboration across borders remains crucial for further advancements.
Collapse
Affiliation(s)
- Yonglong Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Metabolic Diseases The Second Xiangya Hospital, Central South University Changsha China, Changsha, Hunan, China
| | - Xuhui Zhou
- Department of Addiction Medicine, Hunan Institute of Mental Health, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province, Changsha, Hunan, China
| | - Kechao Nie
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinhui Liu
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Sifang Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Metabolic Diseases The Second Xiangya Hospital, Central South University Changsha China, Changsha, Hunan, China
| |
Collapse
|
48
|
Liang P, Chen Q, Chen X, Zhang X, Xiao Y, Liang G, Liu M, He J, Liang W, Liang Y, Chen B. Microbiota modulate immune repertories in lung adenocarcinoma via microbiota-immunity interactive network. Transl Lung Cancer Res 2024; 13:2683-2697. [PMID: 39507044 PMCID: PMC11535827 DOI: 10.21037/tlcr-24-393] [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: 05/02/2024] [Accepted: 09/14/2024] [Indexed: 11/08/2024]
Abstract
Background While the resident microbiome of tumors has been shown to be associated with the occurrence and progression of non-small cell lung cancer, there remains a significant knowledge gap in understanding the correlation between the microbial spectrum and immunity response to cancer therapy. In the case of lung adenocarcinoma (LUAD), the tumor microenvironment, encompassing a diverse array of microbes and immune cells, plays a crucial role in modulating therapeutic response. Towards comprehending the underlying mechanism, we present the microbe-immunity interactive networks to delineate the microbiota and immunity repertoires for two distinct molecular subtypes in LUAD. Methods We obtained multi-omics data of LUAD patients from the publicly available database. In this study, we conducted a systematic exploration of the microbial and immunological etiology of cancer prognosis, by integrating the microbiome, genome, transcriptome, and clinic data. The mutational signature analysis, transcriptome analysis, gene set enrichment analysis, and microbiota-immunity network analysis were performed. Results Based on the transcriptome repertories, we classified the patients into two molecular subtypes and observed that the overall survival of molecular subtype 2 (MS2) was notably shortened. We identified the microbial biomarkers in patients that distinguished between these molecular subtypes. The significant up-regulation of γδT and neutrophil in MS2, suggesting the inflammation augmentation and stimulation of γδT activation. What is more, the MS2 are characterized by a correlation network between microbiota biomarkers and γδT cell, which may contribute to suppression of anti-tumor immunity and poor overall survival. Conclusions Our findings not only display the repertoires of tumor microbiota and immune cells, but also elucidate the potential contribution of the microbiota-immunity correlation network to unfavorable overall survival and therapeutic resistance, thereby exerting profound implications on future LUAD therapy.
Collapse
Affiliation(s)
- Peng Liang
- Center for Medical Research, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Qianxi Chen
- Center for Medical Research, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Xiaoping Chen
- Center for Medical Research, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Xiaolin Zhang
- Center for Medical Research, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Yizhen Xiao
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Guangni Liang
- Department of Thoracic Surgery, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Ming Liu
- Center for Medical Research, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Yufeng Liang
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Bo Chen
- Department of Thoracic Surgery, The First People’s Hospital of Yulin, The Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| |
Collapse
|
49
|
Sheng H, Gu J, Huang Y, Kołat D, Shi G, Yan L, Ye D. Cuproptosis-related signature predicts prognosis and indicates tumor immune infiltration in bladder cancer. Transl Androl Urol 2024; 13:2280-2293. [PMID: 39507864 PMCID: PMC11535731 DOI: 10.21037/tau-24-456] [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: 08/28/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Background Cuproptosis is a newly identified form of cell death that is dependent on copper (Cu) ions, termed Cu-dependent cytotoxicity. This process is distinct from other forms of cell death such as apoptosis, necrosis, and ferroptosis. The accumulation of copper is known to play a significant role in various biological processes, including angiogenesis (the formation of new blood vessels) and metastasis (the spread of cancer cells to different parts of the body). These processes are crucial for tumor growth and progression, indicating that copper and the cuproptosis-related genes (CPRGs) might be indispensable in the context of cancer development and progression. Given this background, we aimed to explore the relationship between CPRGs and both prognostic predictions and tumor microenvironment (TME) infiltration in bladder cancer (BLCA). Methods For this study, we utilized data from The Cancer Genome Atlas (TCGA) to identify CPRGs and subsequently divided BLCA patients into three distinct molecular clusters based on these genes. To assess the proportions of various immune cell types within the TME, we employed single-sample gene set enrichment analysis (ssGSEA) and the Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) method. These computational techniques allowed us to quantify the infiltration of different immune cells, providing insights into the immune landscape of the tumors. Furthermore, we developed a risk score model using CPRGs to predict the survival prospects of BLCA patients. Results Our analysis identified three molecular clusters of BLCA patients, each exhibiting unique clinical features and patterns of TME infiltration. Among these clusters, cluster 1 was associated with a poor prognosis. Interestingly, this cluster also showed significant infiltration of activated CD4+ (ssGSEA P<0.001) and CD8+ T (ssGSEA P<0.05) cells, which are crucial components of the immune response against tumors. This finding suggests a complex interaction between the immune system and the tumor, where a high presence of T cells does not necessarily correlate with better outcomes. Additionally, our risk score model revealed that the high-risk group, characterized by a specific expression pattern of CPRGs, also had enhanced infiltration of CD4+ and CD8+ T cells. This indicates that the cuproptosis-based risk model has a robust ability to predict patient prognosis and can guide immunotherapy decisions. Conclusions Our study sheds light on the biological functions of CPRGs within the TME of BLCA and their correlations with clinical parameters and patient prognosis. The identification of distinct molecular clusters with varying prognoses and immune cell infiltrations highlights the heterogeneity of BLCA and underscores the potential of CPRGs as biomarkers for prognosis and therapeutic targets. These findings offer new perspectives for the development of immunotherapeutic strategies in the treatment of BLCA patients, potentially leading to more personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Haoyue Sheng
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiani Gu
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yongqiang Huang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Damian Kołat
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz, Poland
- Department of Functional Genomics, Medical University of Lodz, Lodz, Poland
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lihua Yan
- Department of Nursing, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Li AY, Bu J, Xiao HN, Zhao ZY, Zhang JL, Yu B, Li H, Li JP, Xiao T. Two-step consensus clustering approach to immune cell infiltration: An integrated exploration and validation of prognostic and immune implications in sarcomas. Heliyon 2024; 10:e38253. [PMID: 39492897 PMCID: PMC11531637 DOI: 10.1016/j.heliyon.2024.e38253] [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: 03/29/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 11/05/2024] Open
Abstract
To conduct a comprehensive investigation of the sarcoma immune cell infiltration (ImmCI) patterns and tumoral microenvironment (TME). We utilized transcriptomic, clinical, and mutation data of sarcoma patients (training cohort) obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) server. Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithms were applied to decipher the immune cell infiltration landscape and TME profiles of sarcomas. An unsupervised clustering method was utilized for classifying ImmCI clusters (initial clustering) and ImmCI-based differentially expressed gene-driven clusters (secondary clustering). Mortality rates and immune checkpoint gene levels was analyzed among the identified clusters. We calculated the ImmCI score through principal component analysis. The tumor immune dysfunction evaluation (TIDE) score was also employed to quantify immunotherapy efficacy between two ImmCI score groups. We further validated the biomarkers for ImmCI and gene-driven clusters via experimental verification and the accuracy of the ImmCI score in predicting survival outcomes and immunotherapy efficacy by external validation cohorts (testing cohort). We demonstrated that ImmCI cluster A and gene-driven cluster A, were beneficial prognostic biomarkers and indicators of immune checkpoint blockade response in sarcomas via in-silico and laboratory experiments. Additionally, the ImmCI score exhibited independent prognostic significance and was predictive of immunotherapy response. Our research underscores the clinical significance of ImmCI scores in identifying sarcoma patients likely to respond to immunotherapy.
Collapse
Affiliation(s)
- Ao-Yu Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jie Bu
- Department of Orthopedics, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui-Ni Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zi-Yue Zhao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jia-Lin Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Bin Yu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| | - Jin-Ping Li
- Department of Orthopedics, Changsha Central Hospital, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha, China
| |
Collapse
|