1
|
Gao M, Wang M, Zhou S, Hou J, He W, Shu Y, Wang X. Machine learning-based prognostic model of lactylation-related genes for predicting prognosis and immune infiltration in patients with lung adenocarcinoma. Cancer Cell Int 2024; 24:400. [PMID: 39696439 DOI: 10.1186/s12935-024-03592-y] [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: 06/12/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Histone lactylation is a novel epigenetic modification that is involved in a variety of critical biological regulations. However, the role of lactylation-related genes in lung adenocarcinoma has yet to be investigated. METHODS RNA-seq data and clinical information of LUAD were downloaded from TCGA and GEO datasets. Unsupervised consistent cluster analysis was performed to identify differentially expressed genes (DEGs) between the two clusters, and risk prediction models were constructed by Cox regression analysis and LASSO analysis. Kaplan-Meier (KM) survival analysis, ROC curves and nomograms were used to validate the accuracy of the models. We also explored the differences in risk scores in terms of immune cell infiltration, immune cell function, TMB, TIDE, and anticancer drug sensitivity. In addition, single-cell clustering and trajectory analysis were performed to further understand the significance of lactylation-related genes. We further analyzed lactate content and glucose uptake in lung adenocarcinoma cells and tissues. Changes in LUAD cell function after knockdown of lactate dehydrogenase (LDHA) by CCK-8, colony formation and transwell assays. Finally, we analyzed the expression of KRT81 in LUAD tissues and cell lines using qRT-PCR, WB, and IHC. Changes in KRT81 function in LUAD cells were detected by CCK-8, colony formation, wound healing, transwell, and flow cytometry. A nude mouse xenograft model and a KrasLSL-G12D in situ lung adenocarcinoma mouse model were used to elucidate the role of KRT81 in LUAD. RESULTS After identifying 26 lactylation-associated DEGs, we constructed 10 lactylation-associated lung adenocarcinoma prognostic models with prognostic value for LUAD patients. A high score indicates a poor prognosis. There were significant differences between the high-risk and low-risk groups in the phenotypes of immune cell infiltration rate, immune cell function, gene mutation frequency, and anticancer drug sensitivity. TMB and TIDE scores were higher in high-risk score patients than in low-risk score patients. MS4A1 was predominantly expressed in B-cell clusters and was identified to play a key role in B-cell differentiation. We further found that lactate content was abnormally elevated in lung adenocarcinoma cells and cancer tissues, and glucose uptake by lung adenocarcinoma cells was significantly increased. Down-regulation of LDHA inhibits tumor cell proliferation, migration and invasion. Finally, we verified that the model gene KRT81 is highly expressed in LUAD tissues and cell lines. Knockdown of KRT81 inhibited cell proliferation, migration, and invasion, leading to cell cycle arrest in the G0/G1 phase and increased apoptosis. KRT81 may play a tumorigenic role in LUAD through the EMT and PI3K/AKT pathways. In vivo, KRT81 knockdown inhibited tumor growth. CONCLUSION We successfully constructed a new prognostic model for lactylation-related genes. Lactate content and glucose uptake are significantly higher in lung adenocarcinoma cells and cancer tissues. In addition, KRT81 was validated at cellular and animal levels as a possible new target for the treatment of LUAD, and this study provides a new perspective for the individualized treatment of LUAD.
Collapse
Affiliation(s)
- Mingjun Gao
- Dalian Medical University, Dalian, 116000, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, China
| | - Mengmeng Wang
- Dalian Medical University, Dalian, 116000, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, China
| | - Siding Zhou
- Department of Emergency, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jiaqi Hou
- Dalian Medical University, Dalian, 116000, China
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, China
| | - Wenbo He
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yusheng Shu
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, China.
- Clinical Medical College, Yangzhou University, Yangzhou, China.
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affliated to Yangzhou University, No. 98 Nantong West Road, Yangzhou, 225001, Jiangsu, China.
| | - Xiaolin Wang
- Yangzhou Clinical Medical College, Dalian Medical University, Yangzhou, 225001, China.
- Clinical Medical College, Yangzhou University, Yangzhou, China.
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affliated to Yangzhou University, No. 98 Nantong West Road, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
2
|
Marhaba, Anjum S, Mandal P, Agrawal S, Ansari KM. Zearalenone promotes endometrial cancer cell migration and invasion via activation of estrogen receptor-mediated Rho/ROCK/PMLC signaling pathway. Food Chem Toxicol 2024; 193:115017. [PMID: 39306225 DOI: 10.1016/j.fct.2024.115017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Zearalenone (ZEA), has emerged as a potential endocrine-disrupting chemical (EDC). Previous results show ZEA effects on endometrial stromal cell apoptosis, migration, and growth of endometriosis. Despite the reported presence of ZEA in Endometrial Cancer (EC) patient's blood and tissues, ZEA-induced EC promotion and its mechanism/s remain elusive. In this study, Ishikawa cells were used to investigate the ZEA effects on Ishikawa cell migration, invasion, and the underlying mechanism involved in these events. Ishikawa cells were exposed to low concentrations of ZEA (5, 25, and 125 nM) for 48 h, and morphological alterations, migration, invasion, markers associated with epithelial-mesenchymal transition (EMT), E-cadherin, Vimentin, RhoA/ROCK/PMLC pathway activation were analyzed. ZEA (25 nM) exposure caused morphological alterations like stress fiber, filopodia formation, loss of cell adhesion, and a significant increase in migration and invasive potential in extracellular matrix-coated porous membranes. Moreover, ZEA exposure also increases the Rho-GTPase activity and expression of pathway mediators, GEFH1, RhoA, ROCK1+2, CDC42, and PMLC/MLC. Furthermore, pre-treatment with specific pharmacological inhibitors for Estrogen receptor-alpha (ER-α) and ROCK attenuate the ZEA-induced stress fiber formation and altered expression of E-cadherin, Vimentin, and Rho/ROCK/PMLC pathway mediators. These findings suggest that Rho/ROCK/PMLC signaling pathways are involved in ZEA-induced Ishikawa cell migration and invasion.
Collapse
Affiliation(s)
- Marhaba
- Food Toxicology Laboratory, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR) Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India
| | - Saria Anjum
- Food Toxicology Laboratory, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR) Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India
| | - Payal Mandal
- Food Toxicology Laboratory, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Smriti Agrawal
- Department of Obstetrics & Gynaecology, Dr. Ram Manohar Lohia Institute of Medical Science, Lucknow, Uttar Pradesh, India
| | - Kausar Mahmood Ansari
- Food Toxicology Laboratory, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR) Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
3
|
Balasundaram A, C Doss GP. Deciphering the Impact of Rare Missense Variants in EGFR-TKI-Resistant Non-Small-Cell Lung Cancer through Whole Exome Sequencing: A Computational Approach. ACS OMEGA 2024; 9:16288-16302. [PMID: 38617633 PMCID: PMC11007825 DOI: 10.1021/acsomega.3c10229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024]
Abstract
Targeted therapy revolutionizes the treatment of non-small-cell lung cancer (NSCLC), harboring molecular change. Epidermal growth factor receptor(EGFR) mutations play a crucial role in the development of NSCLC, serving as a pivotal factor in its pathogenesis. We elucidated the mechanisms of resistance and potential therapeutic strategies in NSCLC resistant to the EGFR-tyrosine kinase inhibitor (EGFR-TKI). This is achieved by identifying rare missense variants through whole exome sequencing (WES). The goal is to enhance our understanding, identify biomarkers, and lay the groundwork for targeted interventions, thereby offering hope for an improved NSCLC treatment landscape. We conducted WES analysis on 16 NSCLC samples with EGFR-TKI-resistant NSCLC obtained from SRA-NCBI (PRJEB50602) to reveal genomic profiles within the EGFR-TKI. Our findings showed that 48% of the variants were missense, and after filtering with the Ensembl variant effect predictor, 53 rare missense variants in 23 genes were identified as highly deleterious. Further examination using pathogenic tools like PredictSNP revealed 12 deleterious rare missense variants in 7 genes: ZNF717, PSPH, ESRRA, SEMA3G, PTPN7, CAVIN4, and MYBBP1A. Molecular dynamics simulation (MDS) suggested that the L385P variant alters the structural flexibility of ESRRA, potentially leading to unfolding of ERRα proteins. This could impact their function and alter ERRα expression. These insights from MDS enhance our understanding of the structural and dynamic consequences of the L385P ESRRA variant and provide valuable implications for subsequent therapeutic considerations and targeted interventions.
Collapse
Affiliation(s)
- Ambritha Balasundaram
- Laboratory of Integrative
Genomics, Department of Integrative Biology, School of BioSciences
and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - George Priya C Doss
- Laboratory of Integrative
Genomics, Department of Integrative Biology, School of BioSciences
and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
4
|
Gao M, Wang M, Chen Y, Wu J, Zhou S, He W, Shu Y, Wang X. Identification and validation of tryptophan metabolism-related lncRNAs in lung adenocarcinoma prognosis and immune response. J Cancer Res Clin Oncol 2024; 150:171. [PMID: 38558328 PMCID: PMC10984901 DOI: 10.1007/s00432-024-05665-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: 12/07/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tryptophan (Trp) is an essential amino acid. Increasing evidence suggests that tryptophan metabolism plays a complex role in immune escape from Lung adenocarcinoma (LUAD). However, the role of long non-coding RNAs (lncRNAs) in tryptophan metabolism remains to be investigated. METHODS This study uses The Cancer Genome Atlas (TCGA)-LUAD dataset as the training cohort, and several datasets from the Gene Expression Omnibus (GEO) database are merged into the validation cohort. Genes related to tryptophan metabolism were identified from the Molecular Signatures Database (MSigDB) database and further screened for lncRNAs with Trp-related expression. Subsequently, a prognostic signature of lncRNAs related to tryptophan metabolism was constructed using Cox regression analysis, (Least absolute shrinkage and selection operator regression) and LASSO analysis. The predictive performance of this risk score was validated by Kaplan-Meier (KM) survival analysis, (receiver operating characteristic) ROC curves, and nomograms. We also explored the differences in immune cell infiltration, immune cell function, tumor mutational load (TMB), tumor immune dysfunction and exclusion (TIDE), and anticancer drug sensitivity between high- and low-risk groups. Finally, we used real-time fluorescence quantitative PCR, CCK-8, colony formation, wound healing, transwell, flow cytometry, and nude mouse xenotransplantation models to elucidate the role of ZNF8-ERVK3-1 in LUAD. RESULTS We constructed 16 tryptophan metabolism-associated lncRNA prognostic models in LUAD patients. The risk score could be used as an independent prognostic indicator for the prognosis of LUAD patients. Kaplan-Meier survival analysis, ROC curves, and risk maps validated the prognostic value of the risk score. The high-risk and low-risk groups showed significant differences in phenotypes, such as the percentage of immune cell infiltration, immune cell function, gene mutation frequency, and anticancer drug sensitivity. In addition, patients with high-risk scores had higher TMB and TIDE scores compared to patients with low-risk scores. Finally, we found that ZNF8-ERVK3-1 was highly expressed in LUAD tissues and cell lines. A series of in vitro experiments showed that knockdown of ZNF8-ERVK3-1 inhibited cell proliferation, migration, and invasion, leading to cell cycle arrest in the G0/G1 phase and increased apoptosis. In vivo experiments with xenografts have shown that knocking down ZNF8-ERVK3-1 can significantly inhibit tumor size and tumor proliferation. CONCLUSION We constructed a new prognostic model for tryptophan metabolism-related lncRNA. The risk score was closely associated with common clinical features such as immune cell infiltration, immune-related function, TMB, and anticancer drug sensitivity. Knockdown of ZNF8-ERVK3-1 inhibited LUAD cell proliferation, migration, invasion, and G0/G1 phase blockade and promoted apoptosis.
Collapse
Affiliation(s)
- Mingjun Gao
- Dalian Medical University, Dalian, 116000, China
| | | | - Yong Chen
- Dalian Medical University, Dalian, 116000, China
| | - Jun Wu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Siding Zhou
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Wenbo He
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Yusheng Shu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, Jiangsu, China.
| | - Xiaolin Wang
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, No. 98 Nantong West Road, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
5
|
Lee JY, Bhandare RR, Boddu SHS, Shaik AB, Saktivel LP, Gupta G, Negi P, Barakat M, Singh SK, Dua K, Chellappan DK. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer. Biomed Pharmacother 2024; 173:116275. [PMID: 38394846 DOI: 10.1016/j.biopha.2024.116275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Tumour suppressor genes play a cardinal role in the development of a large array of human cancers, including lung cancer, which is one of the most frequently diagnosed cancers worldwide. Therefore, extensive studies have been committed to deciphering the underlying mechanisms of alterations of tumour suppressor genes in governing tumourigenesis, as well as resistance to cancer therapies. In spite of the encouraging clinical outcomes demonstrated by lung cancer patients on initial treatment, the subsequent unresponsiveness to first-line treatments manifested by virtually all the patients is inherently a contentious issue. In light of the aforementioned concerns, this review compiles the current knowledge on the molecular mechanisms of some of the tumour suppressor genes implicated in lung cancer that are either frequently mutated and/or are located on the chromosomal arms having high LOH rates (1p, 3p, 9p, 10q, 13q, and 17p). Our study identifies specific genomic loci prone to LOH, revealing a recurrent pattern in lung cancer cases. These loci, including 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), 17p13 (TP53), exhibit a higher susceptibility to LOH due to environmental factors such as exposure to DNA-damaging agents (carcinogens in cigarette smoke) and genetic factors such as chromosomal instability, genetic mutations, DNA replication errors, and genetic predisposition. Furthermore, this review summarizes the current treatment landscape and advancements for lung cancers, including the challenges and endeavours to overcome it. This review envisages inspired researchers to embark on a journey of discovery to add to the list of what was known in hopes of prompting the development of effective therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates.
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada, Chebrolu, Guntur, Andhra Pradesh 522212, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Lakshmana Prabu Saktivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli 620024, India
| | - Gaurav Gupta
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Muna Barakat
- Department of Clinical Pharmacy & Therapeutics, Applied Science Private University, Amman-11937, Jordan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara 144411, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
6
|
Meng GQ, Wang Y, Luo C, Tan YM, Li Y, Tan C, Tu C, Zhang QJ, Hu L, Zhang H, Meng LL, Liu CY, Deng L, Lu GX, Lin G, Du J, Tan YQ, Sha Y, Wang L, He WB. Bi-allelic variants in DNAH3 cause male infertility with asthenoteratozoospermia in humans and mice. Hum Reprod Open 2024; 2024:hoae003. [PMID: 38312775 PMCID: PMC10834362 DOI: 10.1093/hropen/hoae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/21/2023] [Indexed: 02/06/2024] Open
Abstract
STUDY QUESTION Are there other pathogenic genes for asthenoteratozoospermia (AT)? SUMMARY ANSWER DNAH3 is a novel candidate gene for AT in humans and mice. WHAT IS KNOWN ALREADY AT is a major cause of male infertility. Several genes underlying AT have been reported; however, the genetic aetiology remains unknown in a majority of affected men. STUDY DESIGN SIZE DURATION A total of 432 patients with AT were recruited in this study. DNAH3 mutations were identified by whole-exome sequencing (WES). Dnah3 knockout mice were generated using the genome editing tool. The morphology and motility of sperm from Dnah3 knockout mice were investigated. The entire study was conducted over 3 years. PARTICIPANTS/MATERIALS SETTING METHODS WES was performed on 432 infertile patients with AT. In addition, two lines of Dnah3 knockout mice were generated. Haematoxylin and eosin (H&E) staining, transmission electron microscopy (TEM), immunostaining, and computer-aided sperm analysis (CASA) were performed to investigate the morphology and motility of the spermatozoa. ICSI was used to overcome the infertility of one patient and of the Dnah3 knockout mice. MAIN RESULTS AND THE ROLE OF CHANCE DNAH3 biallelic variants were identified in three patients from three unrelated families. H&E staining revealed various morphological abnormalities in the flagella of sperm from the patients, and TEM and immunostaining further showed the loss of the central pair of microtubules, a dislocated mitochondrial sheath and fibrous sheath, as well as a partial absence of the inner dynein arms. In addition, the two Dnah3 knockout mouse lines demonstrated AT. One patient and the Dnah3 knockout mice showed good treatment outcomes after ICSI. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION This is a preliminary report suggesting that defects in DNAH3 can lead to asthenoteratozoospermia in humans and mice. The pathogenic mechanism needs to be further examined in a future study. WIDER IMPLICATIONS OF THE FINDINGS Our findings show that DNAH3 is a novel candidate gene for AT in humans and mice and provide crucial insights into the biological underpinnings of this disorder. The findings may also be beneficial for counselling affected individuals. STUDY FUNDING/COMPETING INTERESTS This work was supported by grants from National Natural Science Foundation of China (82201773, 82101961, 82171608, 32322017, 82071697, and 81971447), National Key Research and Development Program of China (2022YFC2702604), Scientific Research Foundation of the Health Committee of Hunan Province (B202301039323, B202301039518), Hunan Provincial Natural Science Foundation (2023JJ30716), the Medical Innovation Project of Fujian Province (2020-CXB-051), the Science and Technology Project of Fujian Province (2023D017), China Postdoctoral Science Foundation (2022M711119), and Guilin technology project for people's benefit (20180106-4-7). The authors declare no competing interests.
Collapse
Affiliation(s)
- Gui-Quan Meng
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Yaling Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Chen Luo
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Yu-Mei Tan
- GuangDong Provincial Fertility Hospital (GuangDong Provincial Reproductive Science Institute), Guangzhou, China
| | - Yong Li
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Chen Tan
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Chaofeng Tu
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Qian-Jun Zhang
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Liang Hu
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Huan Zhang
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Lan-Lan Meng
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Chun-Yu Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Leiyu Deng
- Reproductive Center of No.924 Hospital of PLA Joint Logistic Support Force, Guilin, China
| | - Guang-Xiu Lu
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Ge Lin
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Juan Du
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Yue-Qiu Tan
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| | - Yanwei Sha
- Department of Andrology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Lingbo Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen-Bin He
- Genetic Department, Hunan Guangxiu Hospital, Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cells & Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Genetic Department, Reproductive and Genetic Hospital of CITIC-Xiangya & Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan, China
| |
Collapse
|
7
|
He Q, Sun C, Pan Y. Whole‑exome sequencing reveals Lewis lung carcinoma is a hypermutated Kras/Nras-mutant cancer with extensive regional mutation clusters in its genome. Sci Rep 2024; 14:100. [PMID: 38167599 PMCID: PMC10762126 DOI: 10.1038/s41598-023-50703-2] [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: 02/08/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Lewis lung carcinoma (LLC), as a widely used preclinical cancer model, has still not been genetically and genomically characterized. Here, we performed a whole-exome sequencing analysis on the LLC cell line to elucidate its molecular characteristics and etiologies. Our data showed that LLC originated from a male mouse belonging to C57BL/6L (a transitional strain between C57BL/6J and C57BL/6N) and contains substantial somatic SNV and InDel mutations (> 20,000). Extensive regional mutation clusters are present in its genome, which were caused mainly by the mutational processes underlying the SBS1, SBS5, SBS15, SBS17a, and SBS21 signatures during frequent structural rearrangements. Thirty three deleterious mutations are present in 30 cancer genes including Kras, Nras, Trp53, Dcc, and Cacna1d. Cdkn2a and Cdkn2b are biallelically deleted from the genome. Five pathways (RTK/RAS, p53, cell cycle, TGFB, and Hippo) are oncogenically deregulated or affected. The major mutational processes in LLC include chromosomal instability, exposure to metabolic mutagens, spontaneous 5-methylcytosine deamination, defective DNA mismatch repair, and reactive oxygen species. Our data also suggest that LLC is a lung cancer similar to human lung adenocarcinoma. This study lays a molecular basis for the more targeted application of LLC in preclinical research.
Collapse
Affiliation(s)
- Quan He
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Cuirong Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Yuanjiang Pan
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
8
|
Wang L, Diao M, Zhang Z, Jiang M, Chen S, Zhao D, Liu Z, Zhou C. Comparison of the somatic genomic landscape between central- and peripheral-type non-small cell lung cancer. Lung Cancer 2024; 187:107439. [PMID: 38113653 DOI: 10.1016/j.lungcan.2023.107439] [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: 08/31/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE Lung cancer is classified into central and peripheral types based on the anatomic location. The present study aimed to explore the distinct patterns of genomic alterations between central- and peripheral-type non-small cell lung cancers (NSCLCs) with negative driver genes and identify potential driver genes and biomarkers to improve therapy strategies for NSCLC. METHODS Whole-exome sequencing (WES) was performed with 182 tumor/control pairs of samples from 145 Chinese NSCLC patients without EGFR, ALK, or ROS1 alterations. Significantly mutated genes (SMGs) and somatic copy number alterations (SCNAs) were identified. Subsequently, tumor mutation burden (TMB), weighted genome integrity index (wGII), copy number alteration (CNA) burden, Shannon diversity index (SDI), intratumor heterogeneity (ITH), neoantigen load (NAL), and clonal variations were evaluated in central- and peripheral-type NSCLCs. Furthermore, mutational signature analysis and survival analysis were performed. RESULTS TP53 was the most frequently mutated gene in NSCLC and more frequently mutated in central-type NSCLC. Higher wGII, ITH, and SDI were found in central-type lung adenocarcinoma (LUAD) than in peripheral-type LUAD. The NAL of central-type lung squamous cell carcinoma (LUSC) with stage III/IV was significantly higher than that of peripheral-type LUSC. Mutational signature analysis revealed that SBS10b, SBS24, and ID7 were significantly different in central- and peripheral-type NSCLCs. Furthermore, central-type NSCLC was found to evolve at a higher level with fewer clones and more subclones, particularly in central-type LUSC. Survival analysis revealed that TMB, CNA burden, NAL, subclonal driver mutations, and subclonal mutations were negatively related to the overall survival (OS) and the progression-free survival (PFS) of central-type LUAD. CONCLUSIONS Central-type NSCLC tended to evolve at a higher level and might suggest a favorable response to immunotherapy. Our study also identified several potential driver genes and promising biomarkers for the prognosis and prediction of chemotherapy responses in NSCLC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Meng Diao
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Zheng Zhang
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Minlin Jiang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Shifu Chen
- HaploX Biotechnology Co., Shenzhen, PR China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| | - Zhenguo Liu
- Department of Anesthesiology, Weifang People's Hospital, Weifang, Shandong Province, PR China.
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| |
Collapse
|
9
|
Bağcı Ö, Özdemir EM, Şanlıtürk B. Variant Analysis of miRNA Regulatory Genes in 35 Sporadic Lung Carcinoma Tumors. DOKL BIOCHEM BIOPHYS 2023; 513:S1-S7. [PMID: 38472669 DOI: 10.1134/s1607672924600052] [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: 10/11/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 03/14/2024]
Abstract
Lung cancer is one of the cancer types with the highest mortality worldwide. The most frequently mutated genes known to be clinically important in lung cancers are EGFR, BRAF, and KRAS genes. Therefore, the therapeutic agents developed are directed against variants that cause over-activation of the EGFR-KRAS-BRAF-BRAF-MEK/ERK signalling pathway. However, different responses of patients to Tyrosine Kinase Inhibitors (TKIs) suggest that new prognostic biomarkers should be defined and epigenetic mechanisms may be related to this situation. METHODS In this study, sequence analyses of AGO2, DICER, and DROSHA genes involved in miRNA biogenesis and EGFR, KRAS, and BRAF genes were performed in 35 patients with sporadic lung cancer. RESULTS We found variations in genes involved in miRNA biogenesis that have not been previously reported in the literature. In addition, we found 4 different variants in the EGFR gene that have been described in the literature. In addition, a statistically significant association was found between the presence of mutations in at least one of the genes involved in miRNA biogenesis and metastasis (p:0.02). CONCLUSIONS In conclusion, genomic dysregulation of key miRNA biogenesis genes may be one of the possible reasons for the differential response of patients to therapeutic agents and the development of metastasis in EGFR wild type tumours.
Collapse
Affiliation(s)
- Özkan Bağcı
- Department of Medical Genetics, Selcuk University, School of Medicine, Konya, Turkey.
| | | | - Batuhan Şanlıtürk
- Department of Medical Genetics, Selcuk University, School of Medicine, Konya, Turkey
| |
Collapse
|
10
|
Moreno CS, Winham CL, Alemozaffar M, Klein ER, Lawal IO, Abiodun-Ojo OA, Patil D, Barwick BG, Huang Y, Schuster DM, Sanda MG, Osunkoya AO. Integrated Genomic Analysis of Primary Prostate Tumor Foci and Corresponding Lymph Node Metastases Identifies Mutations and Pathways Associated with Metastasis. Cancers (Basel) 2023; 15:5671. [PMID: 38067373 PMCID: PMC10705102 DOI: 10.3390/cancers15235671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 02/12/2024] Open
Abstract
Prostate cancer is a highly heterogeneous disease and mortality is mainly due to metastases but the initial steps of metastasis have not been well characterized. We have performed integrative whole exome sequencing and transcriptome analysis of primary prostate tumor foci and corresponding lymph node metastases (LNM) from 43 patients enrolled in clinical trial. We present evidence that, while there are some cases of clonally independent primary tumor foci, 87% of primary tumor foci and metastases are descended from a common ancestor. We demonstrate that genes related to oxidative phosphorylation are upregulated in LNM and in African-American patients relative to White patients. We further show that mutations in TP53, FLT4, EYA1, NCOR2, CSMD3, and PCDH15 are enriched in prostate cancer metastases. These findings were validated in a meta-analysis of 3929 primary tumors and 2721 metastases and reveal a pattern of molecular alterations underlying the pathology of metastatic prostate cancer. We show that LNM contain multiple subclones that are already present in primary tumor foci. We observed enrichment of mutations in several genes including understudied genes such as EYA1, CSMD3, FLT4, NCOR2, and PCDH15 and found that mutations in EYA1 and CSMD3 are associated with a poor outcome in prostate cancer.
Collapse
Affiliation(s)
- Carlos S. Moreno
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA; (C.L.W.); (A.O.O.)
- Department of Biomedical Informatics, Emory University, Atlanta, GA 30322, USA
| | - Cynthia L. Winham
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA; (C.L.W.); (A.O.O.)
| | - Mehrdad Alemozaffar
- Department of Urology, Emory University, Atlanta, GA 30322, USA (D.P.); (M.G.S.)
| | - Emma R. Klein
- Emory College of Arts and Sciences, Atlanta, GA 30322, USA
| | - Ismaheel O. Lawal
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA (O.A.A.-O.); (D.M.S.)
| | - Olayinka A. Abiodun-Ojo
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA (O.A.A.-O.); (D.M.S.)
| | - Dattatraya Patil
- Department of Urology, Emory University, Atlanta, GA 30322, USA (D.P.); (M.G.S.)
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Yijian Huang
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, USA;
| | - David M. Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA (O.A.A.-O.); (D.M.S.)
| | - Martin G. Sanda
- Department of Urology, Emory University, Atlanta, GA 30322, USA (D.P.); (M.G.S.)
| | - Adeboye O. Osunkoya
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA; (C.L.W.); (A.O.O.)
- Department of Urology, Emory University, Atlanta, GA 30322, USA (D.P.); (M.G.S.)
| |
Collapse
|
11
|
Becchi T, Beltrame L, Mannarino L, Calura E, Marchini S, Romualdi C. A pan-cancer landscape of pathogenic somatic copy number variations. J Biomed Inform 2023; 147:104529. [PMID: 37858853 DOI: 10.1016/j.jbi.2023.104529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/13/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
OBJECTIVE Copy number variations (CNVs) play crucial roles in physiological and pathological processes, including cancer. However, the functional implications of somatic CNVs in tumor progression and evolution remain unclear. This study focuses on identifying CNV alterations with high pathogenic potential that drive and sustain tumorigenesis, distinguishing them from passenger alterations that accumulate during tumor growth. Our goal is to explore the variability of CNVs across different tumor types and infer their impact on tumor cell functions. METHODS Starting from 7352 copy number profiles across 33 different cancer types, we infer the pathogenicity of each CNV and perform both intra- and inter-tumor analyses to predict the functional impact of different genomic patterns. We evaluate the actionability of genes belonging to altered regions and we correlate the presence of pathogenic regions with genome instability patterns and patients' survival. RESULTS Our analysis uncovered large heterogeneity among different tumors suggesting in many cases distinct genetic drivers of tumorigenesis. Recurrent genomic alterations frequently coincide with dysfunctional homologous recombination pathways and negative regulation of the immune system. In certain tumors, the number of pathogenic CNVs emerged as a prognostic biomarker, highlighting their significance in cancer progression. CONCLUSION This study contributes to elucidate the functional impact of pathogenic CNVs in tumor progression and sheds light on their potential as prognostic markers in specific cancer types.
Collapse
Affiliation(s)
- Tommaso Becchi
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Luca Beltrame
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Laura Mannarino
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano - Milan, Italy; Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele - Milan, Italy
| | - Enrica Calura
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Sergio Marchini
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
12
|
Pezzuto F, Hofman V, Bontoux C, Fortarezza F, Lunardi F, Calabrese F, Hofman P. The significance of co-mutations in EGFR-mutated non-small cell lung cancer: Optimizing the efficacy of targeted therapies? Lung Cancer 2023; 181:107249. [PMID: 37244040 DOI: 10.1016/j.lungcan.2023.107249] [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: 02/27/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/29/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common cause of cancer death worldwide. In non-squamous NSCLC, the identification of oncogenic drivers and the development of target-specific molecules led to remarkable progress in therapeutic strategies and overall survival over the last decade. Nevertheless, responses are limited by systematically acquired mechanisms of resistance early on after starting a targeted therapy. Moreover, mounting evidence has demonstrated that each oncogenic-driven cluster is actually heterogeneous in terms of molecular features, clinical behaviour, and sensitivity to targeted therapy. In this review, we aimed to examine the prognostic and predictive significance of oncogene-driven co-mutations, focusing mainly on EGFR and TP53. A narrative review was performed by searching MEDLINE databases for English articles published over the last decade (from January 2012 until November 2022). The bibliographies of key references were manually reviewed to select those eligible for the topic. The genetic landscape of EGFR-mutated NSCLC is more complicated than what is known so far. In particular, the occurrence of TP53 co-mutations stratify patients carrying EGFR mutations in terms of treatment response. The study provides a deeper understanding of the mechanisms underlying the variability of the genetic landscape of EGFR-mutated NSCLC and summarizes notably the clinical importance of TP53 co-mutations for an open avenue to more properly addressing the clinical decision-making in the near future.
Collapse
Affiliation(s)
- Federica Pezzuto
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France; Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy.
| | - Véronique Hofman
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France
| | - Christophe Bontoux
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France
| | - Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Paul Hofman
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France.
| |
Collapse
|
13
|
Badoiu SC, Greabu M, Miricescu D, Stanescu-Spinu II, Ilinca R, Balan DG, Balcangiu-Stroescu AE, Mihai DA, Vacaroiu IA, Stefani C, Jinga V. PI3K/AKT/mTOR Dysregulation and Reprogramming Metabolic Pathways in Renal Cancer: Crosstalk with the VHL/HIF Axis. Int J Mol Sci 2023; 24:8391. [PMID: 37176098 PMCID: PMC10179314 DOI: 10.3390/ijms24098391] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Renal cell carcinoma (RCC) represents 85-95% of kidney cancers and is the most frequent type of renal cancer in adult patients. It accounts for 3% of all cancer cases and is in 7th place among the most frequent histological types of cancer. Clear cell renal cell carcinoma (ccRCC), accounts for 75% of RCCs and has the most kidney cancer-related deaths. One-third of the patients with ccRCC develop metastases. Renal cancer presents cellular alterations in sugars, lipids, amino acids, and nucleic acid metabolism. RCC is characterized by several metabolic dysregulations including oxygen sensing (VHL/HIF pathway), glucose transporters (GLUT 1 and GLUT 4) energy sensing, and energy nutrient sensing cascade. Metabolic reprogramming represents an important characteristic of the cancer cells to survive in nutrient and oxygen-deprived environments, to proliferate and metastasize in different body sites. The phosphoinositide 3-kinase-AKT-mammalian target of the rapamycin (PI3K/AKT/mTOR) signaling pathway is usually dysregulated in various cancer types including renal cancer. This molecular pathway is frequently correlated with tumor growth and survival. The main aim of this review is to present renal cancer types, dysregulation of PI3K/AKT/mTOR signaling pathway members, crosstalk with VHL/HIF axis, and carbohydrates, lipids, and amino acid alterations.
Collapse
Affiliation(s)
- Silviu Constantin Badoiu
- Department of Anatomy and Embryology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Iulia-Ioana Stanescu-Spinu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Radu Ilinca
- Department of Medical Informatics and Biostatistics, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Daniela Gabriela Balan
- Department of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (D.G.B.); (A.-E.B.-S.)
| | - Andra-Elena Balcangiu-Stroescu
- Department of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (D.G.B.); (A.-E.B.-S.)
| | - Doina-Andrada Mihai
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Constantin Stefani
- Department of Family Medicine and Clinical Base, Dr. Carol Davila Central Military Emergency University Hospital, 134 Calea Plevnei, 010825 Bucharest, Romania;
| | - Viorel Jinga
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050653 Bucharest, Romania
- “Prof. Dr. Theodor Burghele” Clinical Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania
- Medical Sciences Section, Academy of Romanian Scientists, 050085 Bucharest, Romania
| |
Collapse
|
14
|
Li M, Mei YX, Wen JH, Jiao YR, Pan QR, Kong XX, Li J. Hepatoid adenocarcinoma-Clinicopathological features and molecular characteristics. Cancer Lett 2023; 559:216104. [PMID: 36863507 DOI: 10.1016/j.canlet.2023.216104] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023]
Abstract
Hepatoid adenocarcinoma (HAC) is a rare, malignant, extrahepatic tumor with histologic features similar to those of hepatocellular carcinoma. HAC is most often associated with elevated alpha-fetoprotein (AFP). HAC can occur in multiple organs, including the stomach, esophagus, colon, pancreas, lungs, and ovaries. HAC differs greatly from typical adenocarcinoma in terms of its biological aggression, poor prognosis, and clinicopathological characteristics. However, the mechanisms underlying its development and invasive metastasis remain unclear. The purpose of this review was to summarize the clinicopathological features, molecular traits, and molecular mechanisms driving the malignant phenotype of HAC, in order to support the clinical diagnosis and treatment of HAC.
Collapse
Affiliation(s)
- Ming Li
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China
| | - Yan-Xia Mei
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China
| | - Ji-Hang Wen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China
| | - Yu-Rong Jiao
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China
| | - Qiang-Rong Pan
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China
| | - Xiang-Xing Kong
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China.
| | - Jun Li
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Clinical Research Center for Cancer, China; Cancer Center of Zhejiang University, China.
| |
Collapse
|
15
|
Hoshi D, Kita E, Maru Y, Kogashi H, Nakamura Y, Tatsumi Y, Shimozato O, Nakamura K, Sudo K, Tsujimoto A, Yokoyama R, Kato A, Ushiku T, Fukayama M, Itami M, Yamaguchi T, Hippo Y. Derivation of pancreatic acinar cell carcinoma cell line HS-1 as a patient-derived tumor organoid. Cancer Sci 2023; 114:1165-1179. [PMID: 36382538 PMCID: PMC9986095 DOI: 10.1111/cas.15656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
Acinar cell carcinoma (ACC) of the pancreas is a malignant tumor of the exocrine cell lineage with a poor prognosis. Due to its rare incidence and technical difficulties, few authentic human cell lines are currently available, hampering detailed investigations of ACC. Therefore, we applied the organoid culture technique to various types of specimens, such as bile, biopsy, and resected tumor, obtained from a single ACC patient. Despite the initial propagation, none of these organoids achieved long-term proliferation or tolerated cryopreservation, confirming the challenging nature of establishing ACC cell lines. Nevertheless, the biopsy-derived early passage organoid developed subcutaneous tumors in immunodeficient mice. The xenograft tumor histologically resembled the original tumor and gave rise to infinitely propagating organoids with solid features and high levels of trypsin secretion. Moreover, the organoid stained positive for carboxylic ester hydrolase, a specific ACC marker, but negative for the duct cell marker CD133 and the endocrine lineage marker synaptophysin. Hence, we concluded the derivation of a novel ACC cell line of the pure exocrine lineage, designated HS-1. Genomic analysis revealed extensive copy number alterations and mutations in EP400 in the original tumor, which were enriched in primary organoids. HS-1 displayed homozygous deletion of CDKN2A, which might underlie xenograft formation from organoids. Although resistant to standard cytotoxic agents, the cell line was highly sensitive to the proteasome inhibitor bortezomib, as revealed by an in vitro drug screen and in vivo validation. In summary, we document a novel ACC cell line, which could be useful for ACC studies in the future.
Collapse
Affiliation(s)
- Daisuke Hoshi
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Emiri Kita
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yoshiaki Maru
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Hiroyuki Kogashi
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yuki Nakamura
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yasutoshi Tatsumi
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Osamu Shimozato
- Division of Oncogenomics, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Akiko Tsujimoto
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Ryo Yokoyama
- Division of Pathological Diagnosis, Matsudo City General Hospital, Chiba, Japan
| | - Atsushi Kato
- Department of Hepatobiliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology, Asahi General Hospital, Chiba, Japan
| | - Makiko Itami
- Division of Surgical Pathology, Chiba Cancer Center, Chiba, Japan
| | - Taketo Yamaguchi
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan.,Department of Internal Medicine, Funabashi Central Hospital, Chiba, Japan
| | - Yoshitaka Hippo
- Department of Molecular Carcinogenesis, Chiba Cancer Center Research Institute, Chiba, Japan
| |
Collapse
|
16
|
Design, synthesis of novel benzimidazole derivatives as ENL inhibitors suppressing leukemia cells viability via downregulating the expression of MYC. Eur J Med Chem 2023; 248:115093. [PMID: 36645983 DOI: 10.1016/j.ejmech.2023.115093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
Eleven-Nineteen-Leukemia Protein (ENL) containing YEATS domain, a potential drug target, has emerged as a reader of lysine acetylation. SGC-iMLLT bearing with benzimidazole scaffold was identified as an effective ENL inhibitor, but with weak activity against mixed-lineage leukemia (MLL)-rearranged cells proliferation. In this study, a series of compounds were designed and synthesized by structural optimization on SGC-iMLLT. All the compounds have been evaluated for their ENL inhibitory activities. The results showed that compounds 13, 23 and 28 are the most potential ones with the IC50 values of 14.5 ± 3.0 nM, 10.7 ± 5.3 nM, and 15.4 ± 2.2 nM, respectively, similar with that of SGC-iMLLT. They could interact with ENL protein and strengthen its thermal stability in vitro. Among them, compound 28 with methyl phenanthridinone moiety replacement of indazole in SGC-iMLLT, exhibited significantly inhibitory activities towards MV4-11 and MOLM-13 cell lines with IC50 values of 4.8 μM and 8.3 μM, respectively, exhibiting ∼7 folds and ∼9 folds more potent inhibition of cell growth than SGC-iMLLT. It could also increase the ENL thermal stability while SGC-iMLLT had no obvious effect on leukemia cells. Moreover, compound 28 could downregulate the expression of target gene MYC either alone or in combination with JQ-1 in cells, which was more effective than SGC-iMLLT. Besides, in vivo pharmacokinetic studies showed that the PK properties for compound 28 was much improved over that of SGC-iMLLT. These observations suggested compound 28 was a potential ligand for ENL-related MLL chemotherapy.
Collapse
|
17
|
Álvarez-Prado ÁF, Maas RR, Soukup K, Klemm F, Kornete M, Krebs FS, Zoete V, Berezowska S, Brouland JP, Hottinger AF, Daniel RT, Hegi ME, Joyce JA. Immunogenomic analysis of human brain metastases reveals diverse immune landscapes across genetically distinct tumors. Cell Rep Med 2023; 4:100900. [PMID: 36652909 PMCID: PMC9873981 DOI: 10.1016/j.xcrm.2022.100900] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023]
Abstract
Brain metastases (BrMs) are the most common form of brain tumors in adults and frequently originate from lung and breast primary cancers. BrMs are associated with high mortality, emphasizing the need for more effective therapies. Genetic profiling of primary tumors is increasingly used as part of the effort to guide targeted therapies against BrMs, and immune-based strategies for the treatment of metastatic cancer are gaining momentum. However, the tumor immune microenvironment (TIME) of BrM is extremely heterogeneous, and whether specific genetic profiles are associated with distinct immune states remains unknown. Here, we perform an extensive characterization of the immunogenomic landscape of human BrMs by combining whole-exome/whole-genome sequencing, RNA sequencing of immune cell populations, flow cytometry, immunofluorescence staining, and tissue imaging analyses. This revealed unique TIME phenotypes in genetically distinct lung- and breast-BrMs, thereby enabling the development of personalized immunotherapies tailored by the genetic makeup of the tumors.
Collapse
Affiliation(s)
- Ángel F Álvarez-Prado
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Roeltje R Maas
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Klara Soukup
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Florian Klemm
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Mara Kornete
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Fanny S Krebs
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Vincent Zoete
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sabina Berezowska
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; Brain and Spine Tumor Center, Departments of Clinical Neurosciences and Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy T Daniel
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monika E Hegi
- L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1011 Lausanne, Switzerland; Agora Cancer Research Center, 1011 Lausanne, Switzerland; L. Lundin and Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland.
| |
Collapse
|
18
|
Wang X, Yu G, Yan Z, Wan L, Wang W, Cui L. Lung Cancer Subtype Diagnosis by Fusing Image-Genomics Data and Hybrid Deep Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:512-523. [PMID: 34855599 DOI: 10.1109/tcbb.2021.3132292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Accurate diagnosis of cancer subtypes is crucial for precise treatment, because different cancer subtypes are involved with different pathology and require different therapies. Although deep learning techniques have made great success in computer vision and other fields, they do not work well on Lung cancer subtype diagnosis, due to the distinction of slide images between different cancer subtypes is ambiguous. Furthermore, they often over-fit to high-dimensional genomics data with limited samples, and do not fuse the image and genomics data in a sensible way. In this paper, we propose a hybrid deep network based approach LungDIG for Lung cancer subtype Diagnosis by fusing Image-Genomics data. LungDIG first tiles the tissue slide image into small patches and extracts the patch-level features by fine-tuning an Inception-V3 model. Since the patches may contain some false positives in non-diagnostic regions, it further designs a patch-level feature combination strategy to integrate the extracted patch features and maintain the diversity between different cancer subtypes. At the same time, it extracts the genomics features from Copy Number Variation data by an attention based nonlinear extractor. Next, it fuses the image and genomics features by an attention based multilayer perceptron (MLP) to diagnose cancer subtype. Experiments on TCGA lung cancer data show that LungDIG can not only achieve higher accuracy for cancer subtype diagnosis than state-of-the-art methods, but also have a high authenticity and good interpretability.
Collapse
|
19
|
Lan Y, Liu W, Hou X, Wang S, Wang H, Deng M, Wang G, Ping Y, Zhang X. Revealing the functions of clonal driver gene mutations in patients based on evolutionary dependencies. Hum Mutat 2022; 43:2187-2204. [PMID: 36218010 DOI: 10.1002/humu.24484] [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: 07/09/2022] [Revised: 09/19/2022] [Accepted: 10/06/2022] [Indexed: 01/25/2023]
Abstract
The clonal mutations in driver genes enable cells to gradually acquire growth advantage in tumor development. Therefore, revealing the functions of clonal driver gene mutations is important. Here, we proposed the method FCMP that considered evolutionary dependencies to analyze the functions of clonal driver gene mutations in a single patient. Applying our method to five cancer types from The Cancer Genome Atlas, we identified specific functions and common functions of clonal driver gene mutations. We found that the clonal driver gene mutations in the same patient played multiple functions. We also found that clonal mutations in the same driver gene performed different functions in different patients. These findings suggested that the clonal driver gene mutations showed strong tumor heterogeneity. In the pan-cancer analysis, the immune-related functions for clonal driver gene mutations were shared by multiple cancer types. In addition, clonal mutations in some driver genes predicted the survival of patients in cancers.
Collapse
Affiliation(s)
- Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaobo Hou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuai Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Menglan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Guiyu Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
20
|
Comprehensive Exploration of M2 Macrophages and Its Related Genes for Predicting Clinical Outcomes and Drug Sensitivity in Lung Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:1163924. [PMID: 36157235 PMCID: PMC9492411 DOI: 10.1155/2022/1163924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 02/05/2023]
Abstract
Background M2 macrophages play an important role in cancers. However, the role of M2 macrophages has not been clarified in lung squamous cell carcinoma. Methods All the open-accessed data were downloaded from The Cancer Genome Atlas database. All the analysis was performed in the R software. The CIBERSORT algorithm was utilized to quantify the immune cell infiltration in the tumor microenvironment. LASSO regression and multivariate Cox regression analysis were carried out for the creation of the prognostic model. Pathway enrichment analysis was performed using the single sample Gene Set Enrichment Analysis (ssGSEA) and clueGO algorithm. Results In our study, we comprehensively explored the role of M2 macrophages and its related genes in LUSC patients. We found that the patients with high M2 macrophage infiltration tend to have a worse prognosis. Also, some oncogenetic pathways were activated in the patients with high M2 macrophage infiltration. Further, a prognosis model based on six M2 macrophage-related genes was established, including TRIM58, VIPR2, CTNNA3, KIAA0408, CLEC4G, and MATN4, which showed a good prognosis prediction efficiency in both training and validation cohort. Pathway enrichment analysis showed that the pathway of allograft rejection, bile acid metabolism, coagulation, inflammatory response, IL6/JAK/STAT3 signaling, hedgehog signaling, peroxisome, and myogenesis were significantly activated in the high-risk patients. Based on the results of an investigation of immune infiltration, risk score was found to have a positive correlation with M2 macrophages and resting CD4+ memory T cells, but a negative correlation with follicular helper T cells, M1 macrophages, and Tregs. In addition, we discovered that patients in high-risk groups may respond better to immunotherapy than individuals in lower-risk groups. However, low-risk patients might be more sensitive to cisplatin. Conclusions Our model is a powerful tool to predict LUSC patient prognosis and could indicate the sensitivity of immunotherapy and chemotherapy.
Collapse
|
21
|
Zhang Q, Liu X, Chen Z, Zhang S. Novel GIRlncRNA Signature for Predicting the Clinical Outcome and Therapeutic Response in NSCLC. Front Pharmacol 2022; 13:937531. [PMID: 35991889 PMCID: PMC9382191 DOI: 10.3389/fphar.2022.937531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Non–small cell lung cancer (NSCLC) is highly malignant with driver somatic mutations and genomic instability. Long non-coding RNAs (lncRNAs) play a vital role in regulating these two aspects. However, the identification of somatic mutation-derived, genomic instability-related lncRNAs (GIRlncRNAs) and their clinical significance in NSCLC remains largely unexplored. Methods: Clinical information, gene mutation, and lncRNA expression data were extracted from TCGA database. GIRlncRNAs were screened by a mutator hypothesis-derived computational frame. Co-expression, GO, and KEGG enrichment analyses were performed to investigate the biological functions. Cox and LASSO regression analyses were performed to create a prognostic risk model based on the GIRlncRNA signature (GIRlncSig). The prediction efficiency of the model was evaluated by using correlation analyses with mutation, driver gene, immune microenvironment contexture, and therapeutic response. The prognostic performance of the model was evaluated by external datasets. A nomogram was established and validated in the testing set and TCGA dataset. Results: A total of 1446 GIRlncRNAs were selected from the screen, and the established GIRlncSig was used to classify patients into high- and low-risk groups. Enrichment analyses showed that GIRlncRNAs were mainly associated with nucleic acid metabolism and DNA damage repair pathways. Cox analyses further identified 19 GIRlncRNAs to construct a GIRlncSig-based risk score model. According to Cox regression and stratification analyses, 14 risk lncRNAs (AC023824.3, AC013287.1, AP000829.1, LINC01611, AC097451.1, AC025419.1, AC079949.2, LINC01600, AC004862.1, AC021594.1, MYRF-AS1, LINC02434, LINC02412, and LINC00337) and five protective lncRNAs (LINC01067, AC012645.1, AL512604.3, AC008278.2, and AC089998.1) were considered powerful predictors. Analyses of the model showed that these GIRlncRNAs were correlated with somatic mutation pattern, immune microenvironment infiltration, immunotherapeutic response, drug sensitivity, and survival of NSCLC patients. The GIRlncSig risk score model demonstrated good predictive performance (AUCs of ROC for 10-year survival was 0.69) and prognostic value in different NSCLC datasets. The nomogram comprising GIRlncSig and tumor stage exhibited improved robustness and feasibility for predicting NSCLC prognosis. Conclusion: The newly identified GIRlncRNAs are powerful biomarkers for clinical outcome and prognosis of NSCLC. Our study highlights that the GIRlncSig-based score model may be a useful tool for risk stratification and management of NSCLC patients, which deserves further evaluation in future prospective studies.
Collapse
Affiliation(s)
- Qiangzhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Xicheng Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
- *Correspondence: Sihe Zhang, , https://orcid.org/0000-0002-8923-1993
| |
Collapse
|
22
|
Zhang X, Kong W, Gao M, Huang W, Peng C, Huang Z, Xie Z, Guo H. Robust prognostic model based on immune infiltration-related genes and clinical information in ovarian cancer. J Cell Mol Med 2022; 26:3659-3674. [PMID: 35735060 PMCID: PMC9258710 DOI: 10.1111/jcmm.17360] [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: 12/03/2021] [Revised: 03/25/2022] [Accepted: 04/17/2022] [Indexed: 11/30/2022] Open
Abstract
Immune infiltration of ovarian cancer (OV) is a critical factor in determining patient's prognosis. Using data from TCGA and GTEx database combined with WGCNA and ESTIMATE methods, 46 genes related to OV occurrence and immune infiltration were identified. Lasso and multivariate Cox regression were applied to define a prognostic score (IGCI score) based on 3 immune genes and 3 types of clinical information. The IGCI score has been verified by K‐M curves, ROC curves and C‐index on test set. In test set, IGCI score (C‐index = 0.630) is significantly better than AJCC stage (C‐index = 0.541, p < 0.05) and CIN25 (C‐index = 0.571, p < 0.05). In addition, we identified key mutations to analyse prognosis of patients and the process related to immunity. Chi‐squared tests revealed that 6 mutations are significantly (p < 0.05) related to immune infiltration: BRCA1, ZNF462, VWF, RBAK, RB1 and ADGRV1. According to mutation survival analysis, we found 5 key mutations significantly related to patient prognosis (p < 0.05): CSMD3, FLG2, HMCN1, TOP2A and TRRAP. RB1 and CSMD3 mutations had small p‐value (p < 0.1) in both chi‐squared tests and survival analysis. The drug sensitivity analysis of key mutation showed when RB1 mutation occurs, the efficacy of six anti‐tumour drugs has changed significantly (p < 0.05).
Collapse
Affiliation(s)
- Xi Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Weikaixin Kong
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Institute Sanqu Technology (Hangzhou) Co., Ltd., Hangzhou, China
| | - Miaomiao Gao
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiran Huang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Chao Peng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhuo Huang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhengwei Xie
- Peking University International Cancer Institute and Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
23
|
In silico mutational analysis of ACE2 to check the susceptibility of lung cancer patients towards COVID-19. Sci Rep 2022; 12:7798. [PMID: 35552474 PMCID: PMC9098448 DOI: 10.1038/s41598-022-11805-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/25/2022] [Indexed: 11/24/2022] Open
Abstract
Being the second major cause of death worldwide, lung cancer poses a significant threat to the health of patients. This worsened during the era of pandemic since lung cancer is found to be more prone to SARS-CoV-2 infection. Many recent studies imply a high frequency of COVID-19 infection associated severe outcome. However, molecular studies are still lacking in this respect. Hence the current study is designed to investigate the binding affinities of ACE2 lung cancer mutants with the viral spike protein to find the susceptibility of respective mutants carrying patients in catching the virus. Quite interestingly, our study found lesser binding affinities of all the selected mutants thus implying that these cancer patients might be less affected by the virus than others. These results are opposed to the recent studies’ propositions and open new avenues for more in-depth studies.
Collapse
|
24
|
Development of a 5-Gene Signature to Evaluate Lung Adenocarcinoma Prognosis Based on the Features of Cancer Stem Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4404406. [PMID: 35480140 PMCID: PMC9036162 DOI: 10.1155/2022/4404406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 11/23/2022]
Abstract
Cancer stem cells (CSCs) can induce recurrence and chemotherapy resistance of lung adenocarcinoma (LUAD). Reliable markers identified based on CSC characteristic of LUAD may improve patients' chemotherapy response and prognosis. OCLR was used to calculate mRNA expression-based stemness index (mRNAsi) of LUAD patients' data in TCGA. Association analysis of mRNAsi was performed with clinical features, somatic mutation, and tumor immunity. A prognostic prediction model was established with LASSO Cox regression. Kaplan-Meier Plotter (KM-plotter) and time-dependent ROC were applied to assess signature performance. For LUAD, univariate and multivariate Cox analysis was performed to identify independent prognostic factors. LUAD tissues showed a noticeably higher mRNAsi in than nontumor tissues, and it showed significant differences in T, N, M, AJCC stages, and smoking history. The most frequently mutated gene was TP53, with a higher mRNAsi relating to more frequent mutation of TP53. The mRNAsi was significantly negatively correlated with immune score, stromal score, and ESTIMATE score in LUAD. The blue module was associated with mRNAsi. The 5-gene signature was confirmed as an independent indicator of LUAD prognosis that could promote personalized treatment of LUAD and accurately predict overall survival (OS) of LUAD patients.
Collapse
|
25
|
Jiang H, Wang Y, Xu H, Lei W, Yu X, Tian H, Meng C, Wang X, Zhao Z, Jin X. Identifying Actionable Variants Using Capture-Based Targeted Sequencing in 563 Patients With Non-Small Cell Lung Carcinoma. Front Oncol 2022; 11:812433. [PMID: 35186718 PMCID: PMC8854177 DOI: 10.3389/fonc.2021.812433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022] Open
Abstract
Although the NSCLC diagnostic standards recommend the detection of driver gene mutation, comprehensive genomic profiling has not been used widely in clinical practice. As to the different mutation spectrum characteristics between populations, the research based on Chinese NSCLC cohort is very important for clinical practice. Therefore, we collected 563 surgical specimens from patients with non-small cell lung carcinoma and applied capture-based sequencing using eight-gene panel. We identified 556 variants, with 416 potentially actionable variants in 54.88% (309/563) patients. These single nucleotide variants, insertions and deletions were most commonly found in EGFR (55%), followed by ERBB2 (12%), KRAS (11%), PIK3CA (9%), MET (8%), BRAF (7%), DDR2 (2%), NRAS (0.3%). By using ten protein function prediction algorithms, we also identified 30 novel potentially pathogenic variants. Ninety-eight patients harbored EFGR exon 21 p.L858R mutation and the catalytic domain of the protein tyrosine kinase (PTKc) in EGFR is largely mutated. In addition, there were nine frequent pathogenic variants found in five or more patients. This data provides the potential molecular basis for directing the treatment of lung cancer.
Collapse
Affiliation(s)
- Haiping Jiang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hanlin Xu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Lei
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyun Yu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiying Tian
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cong Meng
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueying Wang
- Research and Development Department, Shenzhen Byoryn Technology Co., Ltd, Shenzhen, China
| | - Zicheng Zhao
- Research and Development Department, Shenzhen Byoryn Technology Co., Ltd, Shenzhen, China
- *Correspondence: Zicheng Zhao, ; Xiangfeng Jin,
| | - Xiangfeng Jin
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Zicheng Zhao, ; Xiangfeng Jin,
| |
Collapse
|
26
|
Dressler L, Bortolomeazzi M, Keddar MR, Misetic H, Sartini G, Acha-Sagredo A, Montorsi L, Wijewardhane N, Repana D, Nulsen J, Goldman J, Pollitt M, Davis P, Strange A, Ambrose K, Ciccarelli FD. Comparative assessment of genes driving cancer and somatic evolution in non-cancer tissues: an update of the Network of Cancer Genes (NCG) resource. Genome Biol 2022; 23:35. [PMID: 35078504 PMCID: PMC8790917 DOI: 10.1186/s13059-022-02607-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/10/2022] [Indexed: 12/30/2022] Open
Abstract
Background Genetic alterations of somatic cells can drive non-malignant clone formation and promote cancer initiation. However, the link between these processes remains unclear and hampers our understanding of tissue homeostasis and cancer development. Results Here, we collect a literature-based repertoire of 3355 well-known or predicted drivers of cancer and non-cancer somatic evolution in 122 cancer types and 12 non-cancer tissues. Mapping the alterations of these genes in 7953 pan-cancer samples reveals that, despite the large size, the known compendium of drivers is still incomplete and biased towards frequently occurring coding mutations. High overlap exists between drivers of cancer and non-cancer somatic evolution, although significant differences emerge in their recurrence. We confirm and expand the unique properties of drivers and identify a core of evolutionarily conserved and essential genes whose germline variation is strongly counter-selected. Somatic alteration in even one of these genes is sufficient to drive clonal expansion but not malignant transformation. Conclusions Our study offers a comprehensive overview of our current understanding of the genetic events initiating clone expansion and cancer revealing significant gaps and biases that still need to be addressed. The compendium of cancer and non-cancer somatic drivers, their literature support, and properties are accessible in the Network of Cancer Genes and Healthy Drivers resource at http://www.network-cancer-genes.org/. Supplementary Information The online version contains supplementary material available at 10.1186/s13059-022-02607-z.
Collapse
|
27
|
Zhang S, Zeng X, Lin S, Liang M, Huang H. Identification of seven-gene marker to predict the survival of patients with lung adenocarcinoma using integrated multi-omics data analysis. J Clin Lab Anal 2021; 36:e24190. [PMID: 34951053 PMCID: PMC8841135 DOI: 10.1002/jcla.24190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022] Open
Abstract
Background The mechanism of cancer occurrence and development could be understood with multi‐omics data analysis. Discovering genetic markers is highly necessary for predicting clinical outcome of lung adenocarcinoma (LUAD). Methods Clinical follow‐up information, copy number variation (CNV) data, single nucleotide polymorphism (SNP), and RNA‐Seq were acquired from The Cancer Genome Atlas (TCGA). To obtain robust biomarkers, prognostic‐related genes, genes with SNP variation, and copy number differential genes in the training set were selected and further subjected to feature selection using random forests. Finally, a gene‐based prediction model for LUAD was validated in validation datasets. Results The study filtered 2071 prognostic‐related genes and 230 genomic variants, 1878 copy deletions, and 438 significant mutations. 218 candidate genes were screened through integrating genomic variation genes and prognosis‐related genes. 7 characteristic genes (RHOV, CSMD3, FBN2, MAGEL2, SMIM4, BCKDHB, and GANC) were identified by random forest feature selection, and many genes were found to be tumor progression‐related. A 7‐gene signature constructed by Cox regression analysis was an independent prognostic factor for LUAD patients, and at the same time a risk factor in the test set, external validation set, and training set. Noticeably, the 5‐year AUC of survival in the validation set and training set was all ˃ 0.67. Similar results were obtained from multi‐omics validation datasets. Conclusions The study builds a novel 7‐gene signature as a prognostic marker for the survival prediction of patients with LUAD. The current findings provided a set of new prognostic and diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Surong Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China
| | - Xueni Zeng
- Department of Infectious Diseases, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China.,Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China
| | - Shaona Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China
| | - Minchao Liang
- Department of Medicine, Shenzhen Haplox Biotechnology Co., Ltd, Shenzhen City, China
| | - Huaxing Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China
| |
Collapse
|
28
|
Anwar N, Memon FA, Shahid S, Shakeel M, Irfan M, Arshad A, Naz A, Ujjan ID, Shamsi T. The Dawn of next generation DNA sequencing in myelodysplastic syndromes- experience from Pakistan. BMC Genomics 2021; 22:903. [PMID: 34915860 PMCID: PMC8679965 DOI: 10.1186/s12864-021-08221-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 11/24/2021] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Myelodysplastic syndromes (MDS) are clonal disorders of hematopoietic stem cells exhibiting ineffective hematopoiesis and tendency for transformation into acute myeloid leukemia (AML). The available karyotyping and fluorescent in situ hybridization provide limited information on molecular abnormalities for diagnosis/prognosis of MDS. Next generation DNA sequencing (NGS), providing deep insights into molecular mechanisms being involved in pathophysiology, was employed to study MDS in Pakistani cohort.
Patients and methods
It was a descriptive cross-sectional study carried out at National institute of blood diseases and bone marrow transplant from 2016 to 2019. Total of 22 cases of MDS were included. Complete blood counts, bone marrow assessment and cytogenetic analysis was done. Patients were classified according to revised WHO classification 2016 and IPSS score was applied for risk stratification. Baseline blood samples were subjected to analysis by NGS using a panel of 54 genes associated with myeloid malignancies.
Results
The median age of patients was 48.5 ± 9.19 years. The most common presenting complaint was weakness 10(45.45%). Cytogenetics analysis revealed abnormal karyotype in 10 (45.45%) patients. On NGS, 54 non-silent rare frequency somatic mutational events in 29 genes were observed (average of 3.82 (SD ± 2.08) mutations per patient), including mutations previously not observed in MDS or AML. Notably, two genes of cohesin complex, RAD21 and STAG2, and two tumor suppressor genes, CDKN2A and TP53, contained highest number of recurrent non-silent somatic mutations in the MDS. Strikingly, a missense somatic mutation p.M272Rof Rad21 was observed in 13 cases. Overall, non-silent somatic mutations in these four genes were observed in 21 of the 22 cases. The filtration with PharmGKB database highlighted a non-synonymous genetic variant rs1042522 [G > C] located in the TP53. Genotype GG and GC of this variant are associated with decreased response to cisplatin and paclitaxel chemotherapy. These two genotypes were found in 13 cases.
Conclusion
Sequencing studies suggest that numerous genetic variants are involved in the initiation of MDS and in the development of AML. In countries like Pakistan where financial reservation of patients makes the use of such analysis even more difficult when the availability of advanced techniques is already a prevailing issue, our study could be an initiating effort in adding important information to the local data. Further studies and large sample size are needed in future to enlighten molecular profiling and ultimately would be helpful to compare and contrast the molecular characteristics of Asian versus global population.
Collapse
|
29
|
Valencia K, Sainz C, Bértolo C, de Biurrun G, Agorreta J, Azpilikueta A, Larrayoz MJ, Bosco G, Zandueta C, Redrado M, Redín E, Exposito F, Serrano D, Echepare M, Ajona D, Melero I, Pio R, Thomas R, Calvo A, Montuenga LM. Two alternative cell line models for the study of multiorganic metastasis and immunotherapy in Lung Squamous Cell Carcinoma. Dis Model Mech 2021; 15:273637. [PMID: 34870316 PMCID: PMC8822220 DOI: 10.1242/dmm.049137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
There is a paucity of adequate mouse models and cell lines available to study lung squamous cell carcinoma (LUSC). We have generated and characterized two models of phenotypically different transplantable LUSC cell lines (UN-SCC679 and UN-SCC680) derived from an N-nitroso-tris-chloroethylurea (NTCU) chemically-induced mouse model in A/J mice. Furthermore, we genetically characterized and compared both LUSC cell lines by performing whole exome and RNA sequencing. These experiments revealed similar genetic and transcriptomic patterns that may correspond to the classical LUSC human subtype. In addition, we compared the immune landscape generated by both tumor cells lines in vivo and assessed their response to immune checkpoint inhibition. The differences between the two cell lines are a good model for the remarkable heterogeneity of human squamous cell carcinoma. Study of the metastatic potential of these models revealed that both cell lines represent the human LUSC organotropism to the brain, bones, liver and adrenal glands. In summary, we have generated a very valuable cell line tools for LUSC research that recapitulates the complexity of the human disease.
Collapse
Affiliation(s)
- Karmele Valencia
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Cristina Sainz
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Cristina Bértolo
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Gabriel de Biurrun
- Department of Environmental Biology School of Sciences, University of Navarra, Pamplona, Spain
| | - Jackeline Agorreta
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Health Sciences, Biochemistry Area, Public University of Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain
| | - Marta J Larrayoz
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Graziella Bosco
- Department of Translational Genomics, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Carolina Zandueta
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Miriam Redrado
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Esther Redín
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Francisco Exposito
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Diego Serrano
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Mirari Echepare
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Daniel Ajona
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ignacio Melero
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, CIMA-University of Navarra, Pamplona, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain.,Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Roman Thomas
- Department of Translational Genomics, Medical Faculty, University of Cologne, 50931 Cologne, Germany.,Department of Pathology, University Hospital Cologne, 50937 Cologne, Germany.,German Cancer Research Center, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - Luis M Montuenga
- Program in Solid Tumors, CIMA-University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Health Research Institute (IDISNA), Pamplona, Spain
| |
Collapse
|
30
|
Leng Y, Dang S, Yin F, Gao T, Xiao X, Zhang Y, Chen L, Qin C, Lai N, Zhan XY, Huang K, Luo C, Kang Y, Wang N, Li Y, Liang Y, Huang B. GDPLichi: a DNA Damage Repair-Related Gene Classifier for Predicting Lung Adenocarcinoma Immune Checkpoint Inhibitors Response. Front Oncol 2021; 11:733533. [PMID: 34970479 PMCID: PMC8713481 DOI: 10.3389/fonc.2021.733533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/27/2021] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is one of the most common and mortal malignancies, usually with a poor prognosis in its advanced or recurrent stages. Recently, immune checkpoint inhibitors (ICIs) immunotherapy has revolutionized the treatment of human cancers including lung adenocarcinoma (LUAD), and significantly improved patients' prognoses. However, the prognostic and predictive outcomes differ because of tumor heterogeneity. Here, we present an effective method, GDPLichi (Genes of DNA damage repair to predict LUAD immune checkpoint inhibitors response), as the signature to predict the LUAD patient's response to the ICIs. GDPLichi utilized only 7 maker genes from 8 DDR pathways to construct the predictive model and classified LUAD patients into two subgroups: low- and high-risk groups. The high-risk group was featured by worse prognosis and decreased B cells, CD8+ T cells, CD8+ central memory T cells, hematopoietic stem cells (HSC), myeloid dendritic cells (MDC), and immune scores as compared to the low-risk group. However, our research also suggests that the high-risk group was more sensitive to ICIs, which might be explained by increased TMB, neoantigen, immune checkpoint molecules, and immune suppression genes' expression, but lower TIDE score as compared to the low-risk group. This conclusion was verified in three other LUAD cohort datasets (GSE30219, GSE31210, GSE50081).
Collapse
Affiliation(s)
- Yang Leng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shiying Dang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Fei Yin
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Tianshun Gao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xing Xiao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yi Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lin Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Changfei Qin
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Nannan Lai
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Yong Zhan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ke Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chuanming Luo
- Center for Clinical Neuroscience, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yun Li
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuhong Liang
- School of Medicine, Southern University Of Science And Technology, Shenzhen, China
| | - Bihui Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
31
|
Zhou W, Dong M, Wu H, Li HL, Xie JL, Ma RY, Su WW, Dai JY. Common mechanism of Citrus Grandis Exocarpium in treatment of chronic obstructive pulmonary disease and lung cancer. CHINESE HERBAL MEDICINES 2021; 13:525-533. [PMID: 36119362 PMCID: PMC9476385 DOI: 10.1016/j.chmed.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/07/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022] Open
Abstract
Objective “Same treatment for different diseases” is a unique treatment strategy in traditional Chinese medicine. Two kinds of malignant respiratory diseases endanger human health-chronic obstructive pulmonary disease (COPD) and lung cancer. Citrus Grandis Exocarpium (Huajuhong in Chinese, HJH), a famous herbal, is always applied by Chinese medicine practitioners to dispersion the lung to resolve phlegm based on “syndrome differentiation and treatment” theory. However, the common mechanism for HJH’s treatment of COPD and lung cancer is not clear. Methods In this study, based on network pharmacology and molecular docking technology, the common mechanism of HJH in the treatment of COPD and lung cancer was studied. The active ingredients and related targets of HJH were integrated from TCMSP, BATMAN-TAM, STP, and Pubchem databases. The standard names of these targets were united by UniProt database. Targets of COPD and lung cancer were enriched through GeneCards, NCBI (Gene), Therapeutic Target Database, and DisGeNET (v7.0) databases. Then the intersection targets of HJH and diseases were obtained. The STRING network and the Cytoscape 3.7.2 were used to construct PPI network, the DAVID database was used to perform GO and KEGG analysis. Then Cytoscape 3.6.1 was used to build “ingredient-target-signal pathway” network. Finally, AutoDock 1.5.6 software was used to perform molecular docking of key proteins and molecules. Results Eleven active ingredients in HJH were obtained by searching the database, corresponding to 184 HJH-COPD-lung cancer targets intersection. The results of biological network analysis showed that naringenin, the active component in HJH, could mainly act on target proteins such as AKT1, EGFR. Then through positive regulation of vasoconstriction and other biological processes, naringenin could regulate estrogen signaling pathway, VEGF signaling pathway, HIF-1 signaling pathway, ErbB signaling pathway, PI3K-Akt signaling pathway to play an important role in the treatment of both COPD and lung cancer. Conclusion Network pharmacology was employed to systematically investigate the active ingredients and targets of HJH in treatment of COPD and lung cancer. And then, the common pharmacodynamic network of HJH for the two malignant respiratory diseases was firstly described. Furthermore, naringenin was proved to strongly bind with AKT1 and EGFR. It may provide the scientific basis for understanding the “Same treatment for different diseases” strategy in traditional Chinese medicine and inspirit subsequent drug discovery for COPD, lung cancer and other malignant lung diseases.
Collapse
Affiliation(s)
- Wei Zhou
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Min Dong
- Department of Pulmonology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Hui-lin Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jia-le Xie
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Ru-yun Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Wei-wei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-evaluation of Post-marketed TCM, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Corresponding authors.
| | - Jian-ye Dai
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
- Corresponding authors.
| |
Collapse
|
32
|
Gialeli C, Tuysuz EC, Staaf J, Guleed S, Paciorek V, Mörgelin M, Papadakos KS, Blom AM. Complement inhibitor CSMD1 modulates epidermal growth factor receptor oncogenic signaling and sensitizes breast cancer cells to chemotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:258. [PMID: 34404439 PMCID: PMC8371905 DOI: 10.1186/s13046-021-02042-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Human CUB and Sushi multiple domains 1 (CSMD1) is a large membrane-bound tumor suppressor in breast cancer. The current study aimed to elucidate the molecular mechanism underlying the effect of CSMD1 in highly invasive triple negative breast cancer (TNBC). METHODS We examined the antitumor action of CSMD1 in three TNBC cell lines overexpressing CSMD1, MDA-MB-231, BT-20 and MDA-MB-486, in vitro using scanning electron microscopy, proteome array, qRT-PCR, immunoblotting, proximity ligation assay, ELISA, co-immunoprecipitation, immunofluorescence, tumorsphere formation assays and flow cytometric analysis. The mRNA expression pattern and clinical relevance of CSMD1 were evaluated in 3520 breast cancers from a modern population-based cohort. RESULTS CSMD1-expressing cells had distinct morphology, with reduced deposition of extracellular matrix components. We found altered expression of several cancer-related molecules, as well as diminished expression of signaling receptors including Epidermal Growth Factor Receptor (EGFR), in CSMD1-expressing cells compared to control cells. A direct interaction of CSMD1 and EGFR was identified, with the EGF-EGFR induced signaling cascade impeded in the presence of CSMD1. Accordingly, we detected increased ubiquitination levels of EGFR upon activation in CSMD1-expressing cells, as well as increased degradation kinetics and chemosensitivity. Accordingly, CSMD1 expression rendered tumorspheres pretreated with gefitinib more sensitive to chemotherapy. In addition, higher mRNA levels of CSMD1 tend to be associated with better outcome of triple negative breast cancer patients treated with chemotherapy. CONCLUSIONS Our results indicate that CSMD1 cross-talks with the EGFR endosomal trafficking cascade in a way that renders highly invasive breast cancer cells sensitive to chemotherapy. Our study unravels one possible underlying molecular mechanism of CSMD1 tumor suppressor function and may provide novel avenues for design of better treatment.
Collapse
Affiliation(s)
- Chrysostomi Gialeli
- Department of Translational Medicine, Lund University, Malmö, Sweden.,Experimental Cardiovascular Research Group, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Emre Can Tuysuz
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Lund, Sweden
| | - Safia Guleed
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Veronika Paciorek
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | | | | | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden.
| |
Collapse
|
33
|
Genomic and transcriptomic profiling of hepatoid adenocarcinoma of the stomach. Oncogene 2021; 40:5705-5717. [PMID: 34326469 DOI: 10.1038/s41388-021-01976-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022]
Abstract
Hepatoid adenocarcinoma of the stomach (HAS), a rare subtype of gastric cancer (GC), has a low incidence but a high mortality rate. Little is known about the molecular features of HAS. Here we applied whole-exome sequencing (WES) on 58 tumours and the matched normal controls from 54 HAS patients, transcriptome sequencing on 30 HAS tumours, and single-cell RNA sequencing (scRNA-seq) on one HAS tumour. Our results reveal that the adenocarcinomatous component and hepatocellular-like component of the same HAS tumour originate monoclonally, and HAS is likely to initiate from pluripotent precursor cells. HAS has high stemness and high methionine cycle activity compared to classical GC. Two genes in the methionine cycle, MAT2A, and AHCY are potential targets for HAS treatments. We provide the first integrative genomic profiles of HAS, which may facilitate its diagnosis, prognosis, and treatment.
Collapse
|
34
|
van Riet J, van de Werken HJG, Cuppen E, Eskens FALM, Tesselaar M, van Veenendaal LM, Klümpen HJ, Dercksen MW, Valk GD, Lolkema MP, Sleijfer S, Mostert B. The genomic landscape of 85 advanced neuroendocrine neoplasms reveals subtype-heterogeneity and potential therapeutic targets. Nat Commun 2021; 12:4612. [PMID: 34326338 PMCID: PMC8322054 DOI: 10.1038/s41467-021-24812-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Metastatic and locally-advanced neuroendocrine neoplasms (aNEN) form clinically and genetically heterogeneous malignancies, characterized by distinct prognoses based upon primary tumor localization, functionality, grade, proliferation index and diverse outcomes to treatment. Here, we report the mutational landscape of 85 whole-genome sequenced aNEN. This landscape reveals distinct genomic subpopulations of aNEN based on primary localization and differentiation grade; we observe relatively high tumor mutational burdens (TMB) in neuroendocrine carcinoma (average 5.45 somatic mutations per megabase) with TP53, KRAS, RB1, CSMD3, APC, CSMD1, LRATD2, TRRAP and MYC as major drivers versus an overall low TMB in neuroendocrine tumors (1.09). Furthermore, we observe distinct drivers which are enriched in somatic aberrations in pancreatic (MEN1, ATRX, DAXX, DMD and CREBBP) and midgut-derived neuroendocrine tumors (CDKN1B). Finally, 49% of aNEN patients reveal potential therapeutic targets based upon actionable (and responsive) somatic aberrations within their genome; potentially directing improvements in aNEN treatment strategies.
Collapse
Affiliation(s)
- Job van Riet
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Harmen J G van de Werken
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, the Netherlands
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Ferry A L M Eskens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Margot Tesselaar
- Department of Medical Oncology, Cancer Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Linde M van Veenendaal
- Department of Medical Oncology, Cancer Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Amsterdam University Medical Centers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marcus W Dercksen
- Department of Internal Medicine, Maxima Medisch Centrum, Veldhoven, The Netherlands
| | - Gerlof D Valk
- Department of Endocrine Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, the Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
35
|
Lu J, Ding Y, Chen Y, Jiang J, Chen Y, Huang Y, Wu M, Li C, Kong M, Zhao W, Wang H, Zhang J, Li Z, Lu Y, Yu X, Jin K, Zhou D, Zhou T, Teng F, Zhang H, Zhou Z, Wang H, Teng L. Whole-exome sequencing of alpha-fetoprotein producing gastric carcinoma reveals genomic profile and therapeutic targets. Nat Commun 2021; 12:3946. [PMID: 34168152 PMCID: PMC8225795 DOI: 10.1038/s41467-021-24170-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 06/01/2021] [Indexed: 02/05/2023] Open
Abstract
Alpha-fetoprotein producing gastric carcinoma (AFPGC) is a rare and aggressive subtype of gastric cancer. However, little is known about the genomic features of this disease. We perform whole-exome sequencing analysis of AFPGC, and identify 34 significantly mutated genes. Somatic copy number alterations analysis reveals several significant focal amplifications (e.g. 19q12, 17q12) and focal deletions (e.g. 1p36.11, 9p21.3), and some of these negatively affect the patient prognosis. Comparative analyses reveal that AFPGC has distinct genomic features from gastric cancer of The Cancer Genome Atlas as well as four molecular subtypes. Several frequently altered genes with potential as therapeutic targets are identified in AFPGC. Further analysis reveals that AFPGC with amplification of CCNE1 at 19q12 and/or ERBB2 at 17q12 show poorer survival and more aggressive. Subsequently, based on our established patient-derived xenograft models for AFPGC, translational research is performed and the therapeutic value of targeting CCNE1 and ERBB2 is validated. In this work, we provide an understanding of genomic characteristics of AFPGC and propose a platform to explore and validate the genome-guided personalized treatment for this disease.
Collapse
Affiliation(s)
- Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junjie Jiang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiran Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingying Huang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mengjie Wu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengzhi Li
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mei Kong
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenyi Zhao
- Innovation Institute for Artificial Intelligence in Medicine and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China
| | - Haohao Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongqi Li
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yimin Lu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiongfei Yu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ketao Jin
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianhua Zhou
- Institute of Gastroenterology, Cancer center, Zhejiang University, Hangzhou, China
| | - Fei Teng
- Hangzhou Oncocare Co. Ltd, Hangzhou, China
| | - Haibin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhan Zhou
- Innovation Institute for Artificial Intelligence in Medicine and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China.
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
36
|
The MLL3/4 H3K4 methyltransferase complex in establishing an active enhancer landscape. Biochem Soc Trans 2021; 49:1041-1054. [PMID: 34156443 PMCID: PMC8286814 DOI: 10.1042/bst20191164] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
Enhancers are cis-regulatory elements that play essential roles in tissue-specific gene expression during development. Enhancer function in the expression of developmental genes requires precise regulation, while deregulation of enhancer function could be the main cause of tissue-specific cancer development. MLL3/KMT2C and MLL4/KMT2D are two paralogous histone modifiers that belong to the SET1/MLL (also named COMPASS) family of lysine methyltransferases and play critical roles in enhancer-regulated gene activation. Importantly, large-scale DNA sequencing studies have revealed that they are amongst the most frequently mutated genes associated with human cancers. MLL3 and MLL4 form identical multi-protein complexes for modifying mono-methylation of histone H3 lysine 4 (H3K4) at enhancers, which together with the p300/CBP-mediated H3K27 acetylation can generate an active enhancer landscape for long-range target gene activation. Recent studies have provided a better understanding of the possible mechanisms underlying the roles of MLL3/MLL4 complexes in enhancer regulation. Moreover, accumulating studies offer new insights into our knowledge of the potential role of MLL3/MLL4 in cancer development. In this review, we summarize recent evidence on the molecular mechanisms of MLL3/MLL4 in the regulation of active enhancer landscape and long-range gene expression, and discuss their clinical implications in human cancers.
Collapse
|
37
|
Chamberlin K, Chamberlin G, Saunders K, Khagi S. Next-generation sequencing reveals novel mutations in a collision tumor of glioblastoma and meningioma. CNS Oncol 2021; 10:CNS70. [PMID: 34015961 PMCID: PMC8162195 DOI: 10.2217/cns-2020-0029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Primary intracranial collision tumors are rare in patients without predisposing factors. We report such a case in a 42-year-old female who presented with headaches and altered mental status. Imaging revealed a single heterogeneous, rim-enhancing lesion in the left parieto-occipital periventricular region, involving the corpus callosum. Stereotactic biopsy demonstrated glioblastoma. Subsequent tumor resection showed histologic evidence of glioblastoma and meningioma. Next-generation sequencing was performed on both tumor components. The glioblastoma exhibited a CDKN2A homozygous deletion and novel missense mutations in TAF1L and CSMD3, while no definitive genetic alterations were identified in the meningioma. Next-generation sequencing may yield insight into molecular drivers of intracranial collision tumors and aid in identifying future therapeutic targets. Next-generation sequencing (NGS) reveals novel mutations in a collision tumor of GBM and meningioma. NGS has the potential to yield insight into molecular drivers of intracranial tumors and identify therapeutic targets.
Collapse
Affiliation(s)
- Kelly Chamberlin
- Department of Neurosurgery, UNC Hospitals, Chapel Hill, NC 27514, USA
| | - Gregory Chamberlin
- Department of Pathology & Laboratory Medicine, UNC Hospitals, Chapel Hill, NC 27514, USA
| | - Katherine Saunders
- Department of Pathology & Laboratory Medicine, UNC Hospitals, Chapel Hill, NC 27514, USA
| | - Simon Khagi
- Department of Neurosurgery, UNC Hospitals, Chapel Hill, NC 27514, USA.,Department of Medicine, Division of Medical Oncology, UNC Hospitals, Chapel Hill, NC 27514, USA
| |
Collapse
|
38
|
Luo R, Ge C, Xiao X, Song J, Miao S, Tang Y, Lai J, Nian W, Song F, Ran L. Identification of genetic variations associated with drug resistance in non-small cell lung cancer patients undergoing systemic treatment. Brief Bioinform 2021; 22:6278152. [PMID: 34013324 PMCID: PMC8574960 DOI: 10.1093/bib/bbab187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/05/2021] [Accepted: 04/22/2021] [Indexed: 12/31/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by relatively rapid response to systemic treatments yet inevitable resistance and predisposed to distant metastasis. We thus aimed at performing sequencing analysis to determine genomic events and underlying mechanisms concerning drug resistance in NSCLC. We performed targeted sequencing of 40 medication-relevant genes on plasma samples from 98 NSCLC patients and analyzed impact of genetic alterations on clinical presentation as well as response to systemic treatments. Profiling of multi-omics data from 1024 NSCLC tissues in public datasets was carried out for comparison and validation of identified molecular events implicated in resistance. A genetic association of CYP2D6 deletion with drug resistance was identified through circulating tumor DNA (ctDNA) profiling and response assessment. FCGR3A amplification was potentially involved in resistance to EGFR inhibitors. We further verified our findings in tissue samples and focused on potential resistance mechanisms, which uncovered that depleted CYP2D6 affected a set of genes involved in EMT, oncogenic signaling as well as inflammatory pathways. Tumor microenvironment analysis revealed that NSCLC with CYP2D6 loss manifested increased levels of immunomodulatory gene expressions, PD-L1 expression, relatively high mutational burden and lymphocyte infiltration. DNA methylation alterations were also found to be correlated with mRNA expressions and copy numbers of CYP2D6. Finally, MEK inhibitors were identified by CMap as the prospective therapeutic drugs for CYP2D6 deletion. These analyses identified novel resistance mechanisms to systemic NSCLC treatments and had significant implications for the development of new treatment strategies.
Collapse
Affiliation(s)
- Ruihan Luo
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, China
| | - Chuang Ge
- Clinical Laboratory of Chongqing University Cancer Hospital, China
| | - Xiao Xiao
- Department of Surgery, The First Affiliated Hospital of Chongqing Medical University, China
| | - Jing Song
- Molecular and Tumor Research Center, Chongqing Medical University, China
| | - Shiqi Miao
- Molecular and Tumor Research Center, Chongqing Medical University, China
| | - Yongyao Tang
- Molecular and Tumor Research Center, Chongqing Medical University, China
| | - Jiayi Lai
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, China
| | - Weiqi Nian
- Phase 1 Clinical Trial Center of Chongqing University Cancer Hospital, China
| | - Fangzhou Song
- Molecular and Tumor Research Center, Chongqing Medical University, China
| | - Longke Ran
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, China
| |
Collapse
|
39
|
Volz C, Breid S, Selenz C, Zaplatina A, Golfmann K, Meder L, Dietlein F, Borchmann S, Chatterjee S, Siobal M, Schöttle J, Florin A, Koker M, Nill M, Ozretić L, Uhlenbrock N, Smith S, Büttner R, Miao H, Wang B, Reinhardt HC, Rauh D, Hallek M, Acker-Palmer A, Heukamp LC, Ullrich RT. Inhibition of Tumor VEGFR2 Induces Serine 897 EphA2-Dependent Tumor Cell Invasion and Metastasis in NSCLC. Cell Rep 2021; 31:107568. [PMID: 32348765 DOI: 10.1016/j.celrep.2020.107568] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 03/11/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023] Open
Abstract
Anti-angiogenic treatment targeting vascular endothelial growth factor (VEGF)-VEGFR2 signaling has shown limited efficacy in lung cancer patients. Here, we demonstrate that inhibition of VEGFR2 in tumor cells, expressed in ∼20% of non-squamous non-small cell lung cancer (NSCLC) patients, leads to a pro-invasive phenotype. Drug-induced inhibition of tumor VEGFR2 interferes with the formation of the EphA2/VEGFR2 heterocomplex, thereby allowing RSK to interact with Serine 897 of EphA2. Inhibition of RSK decreases phosphorylation of Serine 897 EphA2. Selective genetic modeling of Serine 897 of EphA2 or inhibition of EphA2 abrogates the formation of metastases in vivo upon VEGFR2 inhibition. In summary, these findings demonstrate that VEGFR2-targeted therapy conditions VEGFR2-positive NSCLC to Serine 897 EphA2-dependent aggressive tumor growth and metastasis. These data shed light on the molecular mechanisms explaining the limited efficacy of VEGFR2-targeted anti-angiogenic treatment in lung cancer patients.
Collapse
Affiliation(s)
- Caroline Volz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Sara Breid
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Carolin Selenz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Alina Zaplatina
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Kristina Golfmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Lydia Meder
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Felix Dietlein
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, US Institute for Pathology, Cambridge, MA, USA
| | - Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany; University of Cologne, Department I of Internal Medicine, German Hodgkin Study Group (GHSG), Cologne, Germany; University of Cologne, Department I of Internal Medicine, Else Kröner Forschungskolleg Clonal Evolution in Cancer, Cologne, Germany
| | - Sampurna Chatterjee
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Maike Siobal
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Jakob Schöttle
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Department of Translational Genomics, University of Cologne, Medical Faculty, Cologne, Germany
| | - Alexandra Florin
- Institute of Pathology, University Hospital Medical School, Cologne, Germany
| | - Mirjam Koker
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Marieke Nill
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Luka Ozretić
- Department of Cellular Pathology, Royal Free Hospital, London NW3 2QG, UK
| | - Niklas Uhlenbrock
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Steven Smith
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Medical School, Cologne, Germany
| | - Hui Miao
- Rammelkamp Center for Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pharmacology and Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Bingcheng Wang
- Rammelkamp Center for Research, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pharmacology and Oncology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - H Christian Reinhardt
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany
| | - Daniel Rauh
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Amparo Acker-Palmer
- Institute for Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany
| | | | - Roland T Ullrich
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany; Center for Molecular Medicine, Cologne, Germany.
| |
Collapse
|
40
|
Suzuki M, Kasajima R, Yokose T, Ito H, Shimizu E, Hatakeyama S, Yokoyama K, Yamaguchi R, Furukawa Y, Miyano S, Imoto S, Yoshioka E, Washimi K, Okubo Y, Kawachi K, Sato S, Miyagi Y. Comprehensive molecular analysis of genomic profiles and PD-L1 expression in lung adenocarcinoma with a high-grade fetal adenocarcinoma component. Transl Lung Cancer Res 2021; 10:1292-1304. [PMID: 33889510 PMCID: PMC8044470 DOI: 10.21037/tlcr-20-1158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Fetal adenocarcinoma of the lung is a rare variant of lung adenocarcinoma and is subcategorized into low-grade and high-grade (H-FLAC) fetal adenocarcinoma. We previously reported poor prognosis in pulmonary adenocarcinomas with an H-FLAC component; however, the genetic abnormalities involved in H-FLAC remain unclear. Therefore, this study aimed to elucidate molecular abnormalities as potential therapeutic targets for H-FLACs. Methods We performed immunohistochemical analysis and comprehensive genetic analyses using whole-exome sequencing in 16 lung cancer samples with an H-FLAC component. DNA was extracted from formalin-fixed paraffin-embedded tissues after macrodissection of the H-FLAC component. Results Cancer-related mutations were identified in TP53 (7/16 cases), KMT2C (6/16 cases), KRAS (4/16 cases), NF1 (3/16 cases), STK11 (3/16 cases), CTNNB1 (2/16 cases), and EGFR (1/16 cases). A high tumor mutation burden of ≥10 mutations per megabase was observed in 3/16 cases. A high microsatellite instability was not detected in any case. Based on the cosine similarity with the Catalogue of Somatic Mutations in Cancer mutational signatures, H-FLACs were hierarchically clustered into three types: common adenocarcinoma-like (five cases), surfactant-deficient (ten cases), and signatures 2 and 13-related (one case). All common adenocarcinoma-like cases presented thyroid transcription factor-1 (TTF-1) expression, whereas surfactant-deficient cases often presented loss of TTF-1 and surfactant protein expression and included cases with mutations in the surfactant system genes NKX2-1 and SFTPC. H-FLACs displayed low programmed death ligand-1 (PD-L1) expression (1–49% of tumor cells) in 5/16 cases, and no case displayed high PD-L1 expression (≥50% of tumor cells). Conclusions This study indicates that lung cancers with an H-FLAC component rarely harbor currently targetable driver gene mutations for lung cancer but display a high frequency of KMT2C mutations. The microsatellite instability, tumor mutation burden, and PD-L1 expression status suggest a poor response to immune checkpoint therapy.
Collapse
Affiliation(s)
- Masaki Suzuki
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan.,Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroyuki Ito
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Eigo Shimizu
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seira Hatakeyama
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Rui Yamaguchi
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Integrated Data Science, Medical and Dental Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Emi Yoshioka
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Kota Washimi
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yoichiro Okubo
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Kae Kawachi
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| |
Collapse
|
41
|
Zengin T, Önal-Süzek T. Comprehensive Profiling of Genomic and Transcriptomic Differences between Risk Groups of Lung Adenocarcinoma and Lung Squamous Cell Carcinoma. J Pers Med 2021; 11:154. [PMID: 33672117 PMCID: PMC7926392 DOI: 10.3390/jpm11020154] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the second most frequently diagnosed cancer type and responsible for the highest number of cancer deaths worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are subtypes of non-small-cell lung cancer which has the highest frequency of lung cancer cases. We aimed to analyze genomic and transcriptomic variations including simple nucleotide variations (SNVs), copy number variations (CNVs) and differential expressed genes (DEGs) in order to find key genes and pathways for diagnostic and prognostic prediction for lung adenocarcinoma and lung squamous cell carcinoma. We performed a univariate Cox model and then lasso-regularized Cox model with leave-one-out cross-validation using The Cancer Genome Atlas (TCGA) gene expression data in tumor samples. We generated 35- and 33-gene signatures for prognostic risk prediction based on the overall survival time of the patients with LUAD and LUSC, respectively. When we clustered patients into high- and low-risk groups, the survival analysis showed highly significant results with high prediction power for both training and test datasets. Then, we characterized the differences including significant SNVs, CNVs, DEGs, active subnetworks, and the pathways. We described the results for the risk groups and cancer subtypes separately to identify specific genomic alterations between both high-risk groups and cancer subtypes. Both LUAD and LUSC high-risk groups have more downregulated immune pathways and upregulated metabolic pathways. On the other hand, low-risk groups have both up- and downregulated genes on cancer-related pathways. Both LUAD and LUSC have important gene alterations such as CDKN2A and CDKN2B deletions with different frequencies. SOX2 amplification occurs in LUSC and PSMD4 amplification in LUAD. EGFR and KRAS mutations are mutually exclusive in LUAD samples. EGFR, MGA, SMARCA4, ATM, RBM10, and KDM5C genes are mutated only in LUAD but not in LUSC. CDKN2A, PTEN, and HRAS genes are mutated only in LUSC samples. The low-risk groups of both LUAD and LUSC tend to have a higher number of SNVs, CNVs, and DEGs. The signature genes and altered genes have the potential to be used as diagnostic and prognostic biomarkers for personalized oncology.
Collapse
Affiliation(s)
- Talip Zengin
- Department of Molecular Biology and Genetics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey;
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| | - Tuğba Önal-Süzek
- Department of Bioinformatics, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
- Department of Computer Engineering, Muğla Sıtkı Koçman University, 48000 Muğla, Turkey
| |
Collapse
|
42
|
Kulkarni AA, Naqash AR, Puri S, Dienstmann R. Is It Time to Implement Adjuvant Targeted Therapy in EGFR-Mutant Non-Small-Cell Lung Cancer? JCO Precis Oncol 2021; 5:PO.20.00460. [PMID: 34109281 DOI: 10.1200/po.20.00460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 01/16/2023] Open
Affiliation(s)
- Amit A Kulkarni
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN
| | - Abdul Rafeh Naqash
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sonam Puri
- Division of Oncology, Thoracic Clinical Trials Research Group, Huntsman Cancer Institute, Salt Lake City, UT
| | - Rodrigo Dienstmann
- Oncoclínicas Precision Medicine, Grupo Oncoclínicas, São Paulo, Brazil.,Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
43
|
Cai X, Chen Z, Deng M, Li Z, Wu Q, Wei J, Dai C, Wang G, Luo C. Unique genomic features and prognostic value of COSMIC mutational signature 4 in lung adenocarcinoma and lung squamous cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1176. [PMID: 33241025 PMCID: PMC7576056 DOI: 10.21037/atm-20-5952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Analysis of mutational signatures is becoming routine in cancer genomics, with implications for pathogenesis, classification, and prognosis. Among the signatures cataloged at COSMIC, mutational signature 4 has been linked to smoking. However, the distribution of signature 4 in Chinese lung cancer patients has not been evaluated, and its clinical value has not been evaluated. Here we survey mutational signatures in Chinese lung cancer patients and explore the relationship between signature 4 and other genomic features in the patients. Methods We extracted mutational signatures from whole-exome sequencing data of Chinese non-small cell lung cancer patients. The data included 401 lung adenocarcinoma (LUAD) and 92 squamous cell carcinoma (LUSC). We then performed statistical analysis to search for genomic and clinical features that can be linked to mutation signatures. Results We found signature 4 is the most frequent mutational signature in LUSC and the second most frequent in LUAD. Fifty-six LUAD and thirty-five LUSC patients were named with high signature 4 similarities (cosine similarity >0.7). These patients have shorter survival and higher tumor mutational burden comparing to those with low signature 4 similarities. Dozens of genes with single nucleotide variation, index mutations, and copy number variations were differentially enriched in the patients with high signature 4 similarities. Among these genes, CSMD3, LRP1B, TP53, SYNE1, SLIT2, FGF4, and FGF19 are common in both LUADs and LUSCs with high signature 4 similarities, showing that these genes are tightly associated with signature 4. Conclusions The present study is the first to report a comparison in Chinese NSCLC patients with or without COSMIC mutational signature 4. These results will help find the Signature 4 related mutational process in NSCLC.
Collapse
Affiliation(s)
- Xiuyu Cai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhenghe Chen
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Meiling Deng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianchao Wu
- GenomiCare Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Jinwang Wei
- GenomiCare Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Chun Dai
- GenomiCare Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Guan Wang
- GenomiCare Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Chun Luo
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Deshpande RP, Sharma S, Watabe K. The Confounders of Cancer Immunotherapy: Roles of Lifestyle, Metabolic Disorders and Sociological Factors. Cancers (Basel) 2020; 12:E2983. [PMID: 33076303 PMCID: PMC7602474 DOI: 10.3390/cancers12102983] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Checkpoint blockade immunotherapy (CPI) is an effective treatment option for many types of cancers. Irrespective of its wide clinical implications, the overall efficacy remains unpredictable and even poor in certain pathologies such as breast cancer. Thus, it is imperative to understand the role of factors affecting its responsiveness. In this review, we provide an overview on the involvement of sociological factors, lifestyles and metabolic disorders in modulating the CPI response in patients from multiple malignancies. Lifestyle habits including exercise, and diet promoted therapeutic responsiveness while alcohol consumption mitigated the CPI effect by decreasing mutational burden and hampering antigen presentation by dendritic cells. Metabolic disorder such as obesity was recognized to enhance the PD-1 expression while diabetes and hypertension were consequences of CPI therapy rather than causes. Among the sociologic factors, sex and race positively influenced the CPI effectiveness on account of increased effector T cell activity and increased PD-1 expression while ageing impaired CPI responsiveness by decreasing functional T cell and increased toxicity. The combined effect of these factors was observed for obesity and gender, in which obese males had the most significant effect of CPI. Therefore these variables should be carefully considered before treating patients with CPI for optimal treatment outcome.
Collapse
Affiliation(s)
| | | | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (R.P.D.); (S.S.)
| |
Collapse
|
45
|
Marima R, Hull R, Dlamini Z, Penny C. Efavirenz and Lopinavir/Ritonavir Alter Cell Cycle Regulation in Lung Cancer. Front Oncol 2020; 10:1693. [PMID: 32984047 PMCID: PMC7484481 DOI: 10.3389/fonc.2020.01693] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022] Open
Abstract
Highly active anti-retroviral treatment (HAART) is currently the most effective treatment for HIV/AIDS. Additionally, HIV positive patients receiving HAART have a better health-related quality of life (HRQoL). Cancers previously associated with HIV/AIDS also known as the AIDS defining cancers (ADCs), such as Kaposi's sarcoma and non-Hodgkin's lymphoma have been on the decline since the introduction of HAART. However, non-AIDS defining cancers (NADCs), in particular, lung cancers have been documented to be on the rise. The association between the use of HAART components and lung carcinogenesis is poorly understood. This study aimed at elucidating the effects of two HAART components [efavirenz (EFV), and lopinavir/ritonavir (LPV/r)] on lung cancer. This was achieved through the use of in vitro cell biological approaches to assess cell health, including cell viability, Real Time Cell Analysis (RTCA) growth monitoring, evaluation of the cell cycle, and progression to apoptosis, following on drug treatments. At plasma level concentrations, both EFV and LPV/r induced S-phase arrest, while at lower concentrations both drugs promoted the progression of cells into G2/M phase following cell cycle FACS analysis. At higher concentrations although cell viability assays reflected anti-proliferative effects of the drugs, this was not statistically significant. RTCA showed a significant decline in cell viability in response to the highest dose of LPV/r. Dual staining by Annexin V-FITC and PI confirmed significant pro-apoptotic effects were promoted by LPV/r. Both EFV and LPV/r exert double-edged oncogenic effects on MRC-5 and A549 lung cells, acting to either promote cell proliferation or to enhance apoptosis. This is affected by EFV and LPV/r altering cell cycle progression, with a significant S-phase arrest, this being an indication of cellular stress, cytotoxicity, and DNA damage within the cell.
Collapse
Affiliation(s)
- Rahaba Marima
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| | - Rodney Hull
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa
| | - Zodwa Dlamini
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute, University of Pretoria, Pretoria, South Africa.,Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Parktown, South Africa
| |
Collapse
|
46
|
Liu Z, Li X, Ma J, Li D, Ju H, Liu Y, Chen Y, He X, Zhu Y. Integrative Analysis of the IQ Motif-Containing GTPase-Activating Protein Family Indicates That the IQGAP3-PIK3C2B Axis Promotes Invasion in Colon Cancer. Onco Targets Ther 2020; 13:8299-8311. [PMID: 32903879 PMCID: PMC7445521 DOI: 10.2147/ott.s257729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background Colon cancer (CRC) is a common type of tumour, and IQGAP family proteins play an important role in many tumours. However, their roles in CRC remain unclear. Methods First, we searched many public databases to comprehensively analyze expression of IQGAPs in CRC. Next, real-time PCR, immunohistochemical (IHC), transwell, siRNA transfection and Western blot assays were used to evaluate relationships among IQGAP3 expression, clinical pathological parameters and CRC prognosis, and the underlying molecular mechanism was investigated. Results IQGAP3 was elevated in CRC tissues, whereas there was no significant change in expression of IQGAP1 or IQGAP2. Additionally, IQGAP3 expression in CRC tissues was associated with tumour progression, invasion and poor prognosis. In mechanistic studies, we found that IQGAP3 was positively coexpressed with PIK3C2B. In an in vitro assay, the PIK3C2B expression level was increased after exogenous overexpression of IQGAP3, resulting in the promotion of cell invasion, which was blocked by pretransfecting cells with PIK3C2B siRNA. Furthermore, we found that high expression of IQGAP3 and PIK3C2B correlated with tumour stage and vessel invasion in human CRC, whereby patients with high expression of both in tumours had a worse prognosis compared with patients with single-positive or double-negative tumours. Conclusion The results of our current study and corresponding previous studies provide evidence that IQGAP3 is elevated in CRC and promotes colon cancer growth and metastasis by regulating PIK3C2B activation.
Collapse
Affiliation(s)
- Zhuo Liu
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| | - Xiao Li
- The 2nd Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China.,Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Dechuan Li
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| | - Haixing Ju
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| | - Yong Liu
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| | - Yinbo Chen
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| | - Xujun He
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Yuping Zhu
- Department of Colorectal Cancer, Institute of Cancer Research & Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Zhejiang 310022, People's Republic of China
| |
Collapse
|
47
|
Meitern R, Fort J, Giraudeau M, Rattiste K, Sild E, Sepp T. Age-dependent expression of cancer-related genes in a long-lived seabird. Evol Appl 2020; 13:1708-1718. [PMID: 32821278 PMCID: PMC7428815 DOI: 10.1111/eva.13024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/21/2020] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
Studies of model animals like mice and rats have led to great advances in our understanding of the process of tumorigenesis, but this line of study has less to offer for understanding the mechanisms of cancer resistance. Increasing the diversity of nonmodel species from the perspective of molecular mechanisms of natural cancer resistance can lead to new insights into the evolution of protective mechanisms against neoplastic processes and to a wider understanding of natural cancer defense mechanisms. Such knowledge could then eventually be harnessed for the development of human cancer therapies. We suggest here that seabirds are promising, albeit currently completely ignored candidates for studying cancer defense mechanisms, as they have a longer maximum life span than expected from their body size and rates of energy metabolism and may have thus evolved mechanisms to limit neoplasia progression, especially at older ages. We here apply a novel, intraspecific approach of comparing old and young seabirds for improving our understanding of aging and neoplastic processes in natural settings. We used the long-lived common gulls (Larus canus) for studying the age-related pattern of expression of cancer-related genes, based on transcriptome analysis and databases of orthologues of human cancer genes. The analysis of differently expressed cancer-related genes between young and old gulls indicated that similarly to humans, age is potentially affecting cancer risk in this species. Out of eleven differentially expressed cancer-related genes between the groups, three were likely artifactually linked to cancer. The remaining eight were downregulated in old gulls compared to young ones. The downregulation of five of them could be interpreted as a mechanism suppressing neoplasia risk and three as increasing the risk. Based on these results, we suggest that old gulls differ from young ones both from the aspect of cancer susceptibility and tumor suppression at the genetic level.
Collapse
Affiliation(s)
- Richard Meitern
- Institute of Ecology and Earth SciencesUniversity of TartuTartuEstonia
| | - Jérôme Fort
- Littoral Environnement et Sociétés (LIENSs)UMR 7266 CNRS‐La Rochelle UniversitéLa RochelleFrance
| | | | - Kalev Rattiste
- Institute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
| | - Elin Sild
- Institute of Ecology and Earth SciencesUniversity of TartuTartuEstonia
| | - Tuul Sepp
- Institute of Ecology and Earth SciencesUniversity of TartuTartuEstonia
| |
Collapse
|
48
|
Shahid S, Shakeel M, Siddiqui S, Ahmed S, Sohail M, Khan IA, Abid A, Shamsi T. Novel Genetic Variations in Acute Myeloid Leukemia in Pakistani Population. Front Genet 2020; 11:560. [PMID: 32655615 PMCID: PMC7324646 DOI: 10.3389/fgene.2020.00560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by clonal expansion of blast cells that exhibit great genetic heterogeneity. In this study, we describe the mutational landscape and its clinico-pathological significance in 26 myeloid neoplasm patients from a South Asian population (Pakistan) by using ultra-deep targeted next-generation DNA sequencing of 54 genes (∼5000×) and its subsequent bioinformatics analysis. The data analysis indicated novel non-silent somatic mutational events previously not reported in AML, including nine non-synonymous and one stop-gain mutations. Notably, two recurrent somatic non-synonymous mutations, i.e., STAG2 (causing p.L526F) and BCORL1 (p.A400V), were observed in three unrelated cases each. The BCOR was found to have three independent non-synonymous somatic mutations in three cases. Further, the SRSF2 with a protein truncating somatic mutation (p.Q88X) was observed for the first time in AML in this study. The prioritization of germline mutations with ClinVar, SIFT, Polyphen2, and Combined Annotation Dependent Depletion (CADD) highlighted 18 predicted deleterious/pathogenic mutations, including two recurrent deleterious mutations, i.e., a novel heterozygous non-synonymous SNV in GATA2 (p.T358P) and a frameshift insertion in NPM1 (p.L258fs), found in two unrelated cases each. The WT1 was observed with three independent potential detrimental germline mutations in three different cases. Collectively, non-silent somatic and/or germline mutations were observed in 23 (88.46%) of the cases (0.92 mutation per case). Furthermore, the pharmGKB database exploration showed a missense SNV rs1042522 in TP53, exhibiting decreased response to anti-cancer drugs, in 19 (73%) of the cases. This genomic profiling of AML provides deep insight into the disease pathophysiology. Identification of pharmacogenomics markers will help to adopt personalized approach for the management of AML patients in Pakistan.
Collapse
Affiliation(s)
- Saba Shahid
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Muhammad Shakeel
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Saima Siddiqui
- Department of Hematology, National Institute of Blood Diseases and Bone Marrow Transplantation Karachi, Karachi, Pakistan
| | - Shariq Ahmed
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Misha Sohail
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Ishtiaq Ahmad Khan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Aiysha Abid
- Centre for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Tahir Shamsi
- Department of Hematology, National Institute of Blood Diseases and Bone Marrow Transplantation Karachi, Karachi, Pakistan
| |
Collapse
|
49
|
Liao H, Long Y, Han R, Wang W, Xu L, Liao M, Zhang Z, Wu Z, Shang X, Li X, Peng J, Yuan K, Zeng Y. Deep learning-based classification and mutation prediction from histopathological images of hepatocellular carcinoma. Clin Transl Med 2020; 10:e102. [PMID: 32536036 PMCID: PMC7403820 DOI: 10.1002/ctm2.102] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 02/05/2023] Open
Affiliation(s)
- Haotian Liao
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yuxi Long
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ruijiang Han
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Wei Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Lin Xu
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Mingheng Liao
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Zhen Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenru Wu
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, China
| | - Xuefeng Li
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, China
| | - Kefei Yuan
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yong Zeng
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
50
|
Peng Y, Yuan C, Tao X, Zhao Y, Yao X, Zhuge L, Huang J, Zheng Q, Zhang Y, Hong H, Chen H, Sun Y. Integrated analysis of optical mapping and whole-genome sequencing reveals intratumoral genetic heterogeneity in metastatic lung squamous cell carcinoma. Transl Lung Cancer Res 2020; 9:670-681. [PMID: 32676329 PMCID: PMC7354123 DOI: 10.21037/tlcr-19-401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Intratumoral heterogeneity is a crucial factor to the outcome of patients and resistance to therapies, in which structural variants play an indispensable but undiscovered role. Methods We performed an integrated analysis of optical mapping and whole-genome sequencing on a primary tumor (PT) and matched metastases including lymph node metastasis (LNM) and tumor thrombus in the pulmonary vein (TPV). Single nucleotide variants, indels and structural variants were analyzed to reveal intratumoral genetic heterogeneity among tumor cells in different sites. Results Our results demonstrated there were less nonsynonymous somatic variants shared with PT in LNM than in TPV, while there were more structural variants shared with PT in LNM than in TPV. More private variants and its affected genes associated with tumorigenesis and progression were identified in TPV than in LNM. It should be noticed that optical mapping detected an average of 77.1% (74.5-78.5%) large structural variants (>5,000 bp) not detected by whole-genome sequencing and identified several structural variants private to metastases. Conclusions Our study does demonstrate structural variants, especially large structural variants play a crucial role in intratumoral genetic heterogeneity and optical mapping could make up for the deficiency of whole-genome sequencing to identify structural variants.
Collapse
Affiliation(s)
- Yizhou Peng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chongze Yuan
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoting Tao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yue Zhao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xingxin Yao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lingdun Zhuge
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | - Qiang Zheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yue Zhang
- Berry Genomics Corporation, Beijing 100015, China
| | - Hui Hong
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|