1
|
Broseghini E, Venturi F, Veronesi G, Scotti B, Migliori M, Marini D, Ricci C, Casadei R, Ferracin M, Dika E. Exploring the Common Mutational Landscape in Cutaneous Melanoma and Pancreatic Cancer. Pigment Cell Melanoma Res 2025; 38:e13210. [PMID: 39609109 DOI: 10.1111/pcmr.13210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
Cutaneous melanoma (CM) and pancreatic cancer are aggressive tumors whose incidences are rapidly increasing in the last years. This review aims to provide a complete and update description about mutational landscape in CM and pancreatic cancer, focusing on similarities of these two apparently so different tumors in terms of site, type of cell involved, and embryonic origin. The familial forms of CM and pancreatic cancers are often characterized by a common mutated gene, namely CDKN2A. In fact, a germline mutation in CDKN2A gene can be responsible for the development of the familial atypical multiple mole and melanoma syndrome (FAMMM), which is characterized by melanomas and pancreatic cancer development. Sporadic melanoma and pancreatic cancer showed different key-driven genes. The open-access resource cBioPortal has been explored to deepen and investigate the common mutational landscape of these two tumors. We investigated the common mutated genes found in both melanoma and pancreatic cancer with a frequency of at least 5% of tested patients and copy number alterations with a frequency of at least of 3%. Data showed that 18 mutated genes and 3 copy number alterations are present in both melanoma and pancreatic cancers types. Since we found two patients that developed both melanoma and pancreatic cancer, we compared mutation landscape between the two tumors and identified a pathogenic variant in BRCA2 gene. This review gives valuable insights into the genetic underpinnings of melanoma and pancreatic cancer, urging the continued exploration and research of new genetic biomarkers able to identify patients and families at high risk of developing both cancers and to address to screening and to an effective clinical management of the patient.
Collapse
Affiliation(s)
| | - Federico Venturi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giulia Veronesi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Biagio Scotti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marina Migliori
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Internal Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Desy Marini
- Internal Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Claudio Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Pancreas and Endocrine Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Casadei
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Pancreas and Endocrine Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Manuela Ferracin
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Emi Dika
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Luo S, Li Z, Wang M, Liu Z, Wang D, Bai Y, Ge H, Yu Y, Yu Y, Chen W, Wang Y, Zhang C, Yu J, Song C, Lv C, Zhen Q, Han Y, Sun L. Genome wide association study and meta-analysis identified multiple new risk loci for freckles in 4813 Chinese individuals. Pigment Cell Melanoma Res 2024; 37:808-821. [PMID: 38970458 DOI: 10.1111/pcmr.13183] [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: 08/28/2023] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
Freckle is a prevalent pigmentary dermatosis with an obvious hereditary component. Dozens of freckles risk loci have been discovered through research on multiple traits or other diseases, rather than as an independent trait. To discover novel variants associated with freckles, we performed GWAS and meta-analysis in 4813 Chinese individuals. We conducted GWAS and meta-analysis of two cohorts: 197 patients and 1603 controls (Cohort I), and 336 patients and 2677 controls (Cohort II), both from China. Then we performed linkage disequilibrium (LD) analysis, eQTL study, and enrichment analysis with association results for functional implications. Finally, we discovered 59 new SNPs and 13 novel susceptibility genes associated with freckles (Pmeta <5 × 10-8), which has enriched the genetic research on freckles.
Collapse
Affiliation(s)
- Sihan Luo
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Zhuo Li
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Minhao Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Zhili Liu
- Dalian Dermatosis Hospital, Dalian, China
| | - Daiyue Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yuanming Bai
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Huiyao Ge
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yafen Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yanxia Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Weiwei Chen
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yirui Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Chang Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Jing Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Can Song
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | | | - Qi Zhen
- North China University of Science and Technology Affiliated Hospital Tangshan, Tangshan, China
| | - Yang Han
- North China University of Science and Technology Affiliated Hospital Tangshan, Tangshan, China
| | - Liangdan Sun
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
- North China University of Science and Technology Affiliated Hospital Tangshan, Tangshan, China
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
3
|
Saad MN, Hamed M. Transcriptome-Wide Association Study Reveals New Molecular Interactions Associated with Melanoma Pathogenesis. Cancers (Basel) 2024; 16:2517. [PMID: 39061157 PMCID: PMC11274789 DOI: 10.3390/cancers16142517] [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: 05/23/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
A transcriptome-wide association study (TWAS) was conducted on genome-wide association study (GWAS) summary statistics of malignant melanoma of skin (UK Biobank dataset) and The Cancer Genome Atlas-Skin Cutaneous Melanoma (TCGA-SKCM) gene expression weights to identify melanoma susceptibility genes. The GWAS included 2465 cases and 449,799 controls, while the gene expression testing was conducted on 103 cases. Afterward, a gene enrichment analysis was applied to identify significant TWAS associations. The melanoma's gene-microRNA (miRNA) regulatory network was constructed from the TWAS genes and their corresponding miRNAs. At last, a disease enrichment analysis was conducted on the corresponding miRNAs. The TWAS detected 27 genes associated with melanoma with p-values less than 0.05 (the top three genes are LOC389458 (RBAK), C16orf73 (MEIOB), and EIF3CL). After the joint/conditional test, one gene (AMIGO1) was dropped, resulting in 26 significant genes. The Gene Ontology (GO) biological process associated the extended gene set (76 genes) with protein K11-linked ubiquitination and regulation of cell cycle phase transition. K11-linked ubiquitin chains regulate cell division. Interestingly, the extended gene set was related to different skin cancer subtypes. Moreover, the enriched pathways were nsp1 from SARS-CoV-2 that inhibit translation initiation in the host cell, cell cycle, translation factors, and DNA repair pathways full network. The gene-miRNA regulatory network identified 10 hotspot genes with the top three: TP53, BRCA1, and MDM2; and four hotspot miRNAs: mir-16, mir-15a, mir-125b, and mir-146a. Melanoma was among the top ten diseases associated with the corresponding (106) miRNAs. Our results shed light on melanoma pathogenesis and biologically significant molecular interactions.
Collapse
Affiliation(s)
- Mohamed N. Saad
- Biomedical Engineering Department, Faculty of Engineering, Minia University, Minia 61519, Egypt
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, 18057 Rostock, Germany;
| | - Mohamed Hamed
- Institute for Biostatistics and Informatics in Medicine and Ageing Research (IBIMA), Rostock University Medical Center, 18057 Rostock, Germany;
- Faculty of Media Engineering and Technology, German University in Cairo, Cairo 11835, Egypt
| |
Collapse
|
4
|
Namikawa T, Tanaka T, Utsunomiya M, Yokota K, Munekage M, Maeda H, Kitagawa H, Kurioka Y, Satake H, Kobayashi M, Hanazaki K, Seo S. Gastric cancer with Fanconi anemia in adolescent and young adult patient diagnosed by comprehensive genome profiling using next-generation sequencing. Clin J Gastroenterol 2024; 17:12-17. [PMID: 37934348 DOI: 10.1007/s12328-023-01886-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023]
Abstract
Recently, the results of gastric cancer treatment have improved; however, its characteristics in adolescents and young adults are not well known. We report the case of a patient with advanced gastric cancer, Fanconi anemia (FA), and primary biliary cholangitis. A 26-year-old woman visited a local physician complaining of epigastralgia. Esophagogastroduodenoscopy revealed edematous changes with poor distension and circumferential thickened folds with erosions in the gastric body. Biopsy results of the lesion specimens revealed poorly differentiated adenocarcinoma. Abdominal contrast-enhanced computed tomography revealed gastric wall with irregular thickness, several nodules in the peritoneal cavity, and a mass lesion in the right ovary. We diagnosed the patient with T4N2M1 stage IV gastric cancer accompanied by peritoneal and ovarian metastases and initiated nivolumab with S-1 plus oxaliplatin as the first-line treatment regimen. Because of immune-related adverse events after one course of systemic treatment, the regimen was changed to ramucirumab combined with nab-paclitaxel chemotherapy as the second-line treatment. After three cycles of weekly nab-paclitaxel with ramucirumab, the decreased platelet count did not recover, and her general condition gradually deteriorated. Comprehensive genome profiling using next-generation sequencing was performed to determine the feasibility of genotype-matched therapies. Alterations in FA complementation group A (FANCA) F1263del (49.1%) and E484Q (12.3%), which encode a key component of the multiprotein FA complex, were identified. The patient died 10 months after treatment initiation. In conclusion, when treating malignancies in adolescent and young adult patients, the genomic background should be considered.
Collapse
Affiliation(s)
- Tsutomu Namikawa
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Tomoki Tanaka
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Masato Utsunomiya
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Keiichiro Yokota
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Masaya Munekage
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Hiromichi Maeda
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Hiroyuki Kitagawa
- Department of Operating Room Management, Kochi Medical School Hospital, Nankoku, Japan
| | - Yusuke Kurioka
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Michiya Kobayashi
- Department of Human Health and Medical Sciences, Kochi Medical School, Nankoku, Japan
| | - Kazuhiro Hanazaki
- Integrated Center for Advanced Medical Technologies, Kochi Medical School Hospital, Nankoku, Japan
| | - Satoru Seo
- Department of Surgery, Kochi Medical School, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
5
|
Jeremian R, Xie P, Fotovati M, Lefrançois P, Litvinov IV. Gene-Environment Analyses in a UK Biobank Skin Cancer Cohort Identifies Important SNPs in DNA Repair Genes That May Help Prognosticate Disease Risk. Cancer Epidemiol Biomarkers Prev 2023; 32:1599-1607. [PMID: 37642678 PMCID: PMC10840669 DOI: 10.1158/1055-9965.epi-23-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/12/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Despite well-established relationships between sun exposure and skin cancer pathogenesis/progression, specific gene-environment interactions in at-risk individuals remain poorly-understood. METHODS We leveraged a UK Biobank cohort of basal cell carcinoma (BCC, n = 17,221), cutaneous squamous cell carcinoma (cSCC, n = 2,331), melanoma in situ (M-is, n = 1,158), invasive melanoma (M-inv, n = 3,798), and healthy controls (n = 448,164) to quantify the synergistic involvement of genetic and environmental factors influencing disease risk. We surveyed 8,798 SNPs from 190 DNA repair genes, and 11 demographic/behavioral risk factors. RESULTS Clinical analysis identified darker skin (RR = 0.01-0.65) and hair (RR = 0.27-0.63) colors as protective factors. Eleven SNPs were significantly associated with BCC, three of which were also associated with M-inv. Gene-environment analysis yielded 201 SNP-environment interactions across 90 genes (FDR-adjusted q < 0.05). SNPs from the FANCA gene showed interactions with at least one clinical factor in all cancer groups, of which three (rs9926296, rs3743860, rs2376883) showed interaction with nearly every factor in BCC and M-inv. CONCLUSIONS We identified novel risk factors for keratinocyte carcinomas and melanoma, highlighted the prognostic value of several FANCA alleles among individuals with a history of sunlamp use and childhood sunburns, and demonstrated the importance of combining genetic and clinical data in disease risk stratification. IMPACT This study revealed genome-wide associations with important implications for understanding skin cancer risk in the context of the rapidly-evolving field of precision medicine. Major individual factors (including sex, hair and skin color, and sun protection use) were significant mediators for all skin cancers, interacting with >200 SNPs across four skin cancer types.
Collapse
Affiliation(s)
- Richie Jeremian
- Faculty of Medicine and Health Sciences, McGill University
- Department of Medicine, Division of Dermatology, Research Institute of the McGill University Health Centre (RI-MUHC) Montreal, Quebec
| | - Pingxing Xie
- Faculty of Medicine and Health Sciences, McGill University
- Department of Medicine, Division of Dermatology, Research Institute of the McGill University Health Centre (RI-MUHC) Montreal, Quebec
| | - Misha Fotovati
- Faculty of Medicine and Health Sciences, McGill University
- Department of Medicine, Division of Dermatology, Lady Davis Institute (LDI), Jewish General Hospital, Montreal, Quebec
| | - Philippe Lefrançois
- Faculty of Medicine and Health Sciences, McGill University
- Department of Medicine, Division of Dermatology, Lady Davis Institute (LDI), Jewish General Hospital, Montreal, Quebec
| | - Ivan V. Litvinov
- Faculty of Medicine and Health Sciences, McGill University
- Department of Medicine, Division of Dermatology, Research Institute of the McGill University Health Centre (RI-MUHC) Montreal, Quebec
| |
Collapse
|
6
|
Bokharaie H, Kolch W, Krstic A. Analysis of Alternative mRNA Splicing in Vemurafenib-Resistant Melanoma Cells. Biomolecules 2022; 12:993. [PMID: 35883549 PMCID: PMC9312936 DOI: 10.3390/biom12070993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 01/09/2023] Open
Abstract
Alternative mRNA splicing is common in cancers. In BRAF V600E-mutated malignant melanoma, a frequent mechanism of acquired resistance to BRAF inhibitors involves alternative splicing (AS) of BRAF. The resulting shortened BRAF protein constitutively dimerizes and conveys drug resistance. Here, we have analysed AS in SK-MEL-239 melanoma cells and a BRAF inhibitor (vemurafenib)-resistant derivative that expresses an AS, shortened BRAF V600E transcript. Transcriptome analysis showed differential expression of spliceosome components between the two cell lines. As there is no consensus approach to analysing AS events, we used and compared four common AS softwares based on different principles, DEXSeq, rMATS, ASpli, and LeafCutter. Two of them correctly identified the BRAF V600E AS in the vemurafenib-resistant cells. Only 12 AS events were identified by all four softwares. Testing the AS predictions experimentally showed that these overlapping predictions are highly accurate. Interestingly, they identified AS caused alterations in the expression of melanin synthesis and cell migration genes in the vemurafenib-resistant cells. This analysis shows that combining different AS analysis approaches produces reliable results and meaningful, biologically testable hypotheses.
Collapse
Affiliation(s)
- Honey Bokharaie
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland
| | - Aleksandar Krstic
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
| |
Collapse
|
7
|
Mo JL, Liu JS, Xiao Q, Hong WX, Yin JY, Chen J, Liu ZQ. Association of variations in the Fanconi anemia complementation group and prognosis in Non-small cell lung cancer patients with Platinum-based chemotherapy. Gene 2022; 825:146398. [PMID: 35306114 DOI: 10.1016/j.gene.2022.146398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/08/2022] [Accepted: 03/04/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE To explore the associations between FANC (FANCA, FANCC, FANCE, FANCF, and FANCJ) single nucleotide polymorphisms (SNPs) and prognosis of non-small cell lung cancer (NSCLC) patients with platinum-based chemotherapy. METHODS According to the inclusion criteria, we selected 395 DNA samples from NSCLC patients for genotyping and combined with clinical data for Cox regression analysis and stratification analyses to assess relationships between overall survival (OS) and progression free survival (PFS) with SNPs genotypes. RESULTS The results revealed that patients with FANCE rs6907678 TT genotype have a longer OS than TC and CC genotype (Additive model: P = 0.004, HR = 1.696, 95% CI = 1.186-2.425). In stratification analyses, Longer PFS is found in female, age ≤ 55 years old and non-smoking patients with FANCE rs6907678 TT genotype, and patients with TT genotypes were significantly had longer OS in male, age >55 years old, non-smoking, squamous cell carcinoma and stage IV stratification. CONCLUSION Our data demonstrates that patients with FANCE rs6907678 TT genotype are contributed to better prognosis. FANCE rs6907678 may be used as a clinical biomarker for predicting the prognosis of NSCLC patients with platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jun-Luan Mo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China
| | - Jia-Si Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Qi Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Wen-Xu Hong
- Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China; Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Juan Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China; Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China.
| |
Collapse
|
8
|
Chen W, Hou X, Hu Y, Huang G, Ye X, Nie S. A deep learning- and CT image-based prognostic model for the prediction of survival in non-small cell lung cancer. Med Phys 2021; 48:7946-7958. [PMID: 34661294 DOI: 10.1002/mp.15302] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/19/2021] [Accepted: 10/10/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To assist clinicians in arranging personalized treatment, planning follow-up programs and extending survival times for non-small cell lung cancer (NSCLC) patients, a method of deep learning combined with computed tomography (CT) imaging for survival prediction was designed. METHODS Data were collected from 484 patients from four research centers. The data from 344 patients were utilized to build the A_CNN survival prognosis model to classify 2-year overall survival time ranges (730 days cut-off). Data from 140 patients, including independent internal and external test sets, were utilized for model testing. First, a series of preprocessing techniques were used to process the original CT images and generate training and test data sets from the axial, coronal, and sagittal planes. Second, the structure of the A_CNN model was designed based on asymmetric convolution, bottleneck blocks, the uniform cross-entropy (UC) loss function, and other advanced techniques. After that, the A_CNN model was trained, and numerous comparative experiments were designed to obtain the best prognostic survival model. Last, the model performance was evaluated, and the predicted survival curves were analyzed. RESULTS The A_CNN survival prognosis model yielded a high patient-level accuracy of 88.8%, a patch-level accuracy of 82.9%, and an area under the receiver operating characteristic (ROC) curve (AUC) of 0.932. When tested on an external data set, the maximum patient-level accuracy was 80.0%. CONCLUSIONS The results suggest that using a deep learning method can improve prognosis in patients with NSCLC and has important application value in establishing individualized prognostic models.
Collapse
Affiliation(s)
- Wen Chen
- School of Medical Imaging, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Xuewen Hou
- School of Medical Imaging, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Ying Hu
- School of Medical Imaging, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Gang Huang
- Department of Radiology, Shanghai Chest Hospital, Shanghai, China
| | - Xiaodan Ye
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shengdong Nie
- School of Medical Imaging, Shanghai University of Medicine & Health Science, Shanghai, China
| |
Collapse
|
9
|
Podlipnik S, Potrony M, Puig S. Genetic markers for characterization and prediction of prognosis of melanoma subtypes: a 2021 update. Ital J Dermatol Venerol 2021; 156:322-330. [PMID: 33982545 DOI: 10.23736/s2784-8671.21.06957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this article we examined the most important genetic markers involved in melanoma susceptibility, initiation and progression, and their impact on the prognosis of the disease. Current knowledge in melanoma genetics identifies distinct pathways to the development of different melanoma subtypes characterized by specific clinico-pathological features and partially known genetic markers, modulated by high, low or absence of cumulative sun damage. The most prevalent somatic mutations are related to the activation of the MAPK pathway, which are classified into four major subtypes: BRAF mutant, NRAS mutant, NF1 mutant and triple wild type. Moreover, germinal mutations are also involved in the characterization and predictions of prognosis in melanoma. Currently, CDKN2A is seen as the main high-risk gene involved in melanoma susceptibility being mutated in around 20% of melanoma-prone families. Other high-risk susceptibility genes described include CDK4, POT1, BAP1, TERT promoter, ACD, and TERF2IP. Melanoma is one of the most genetically predisposed among all cancers in humans, and ultraviolet light from the sun is the main environmental factor. This genetic predisposition is starting to be understood, impacting not only on the risk of developing melanoma but also on the risk of developing other types of cancer, as well as on the prognosis of the disease.
Collapse
Affiliation(s)
- Sebastian Podlipnik
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain.,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Department of Biochemistry and Molecular Genetics, Hospital of Barcelona, Barcelona, Spain
| | - Susana Puig
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain - .,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
10
|
de Almeida LC, Calil FA, Machado-Neto JA, Costa-Lotufo LV. DNA damaging agents and DNA repair: From carcinogenesis to cancer therapy. Cancer Genet 2021; 252-253:6-24. [DOI: 10.1016/j.cancergen.2020.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 02/09/2023]
|
11
|
FANCA Polymorphism Is Associated with the Rate of Proliferation in Uterine Leiomyoma in Korea. J Pers Med 2020; 10:jpm10040228. [PMID: 33202820 PMCID: PMC7712130 DOI: 10.3390/jpm10040228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022] Open
Abstract
Uterine leiomyomas are the most common benign gynecologic tumors. This study was aimed to identify single nucleotide polymorphism of Fanconi anemia complementation group A (FANCA), associated with the rate of proliferation in uterine leiomyomas. In vitro study of patient-derived primary-cultured leiomyoma cells and direct sequencing of fresh frozen leiomyoma from each subject was conducted. Leiomyomas obtained from 44 patients who had underwent surgery were both primary-cultured and freshly frozen. Primary-cultured leiomyoma cells were divided into, according to the rate of proliferation, fast and slow groups. Single nucleotide polymorphism (SNP) of FANCA were determined from fresh frozen tissues of each patient using direct sequencing. Direct sequencing revealed a yet unidentified role of FANCA, a caretaker in the DNA damage-response pathway, as a possible biomarker molecule for the prediction of uterine leiomyoma proliferation. We identified that rs2239359 polymorphism, which causes a missense mutation in FANCA, is associated with the rate of proliferation in uterine leiomyomas. The frequency of C allele in the fast group was 35.29% while that in slow group was 11.11% (odds ratio (OR) 4.036 (1.176–13.855), p = 0.0266). We also found that the TC + CC genotype was more frequently observed in the fast group compared with that in the slow group (OR 6.44 (1.90–31.96), p = 0.0227). Taken together, the results in the current study suggested that a FANCA missense mutation may play an important regulatory role in the proliferation of uterine leiomyoma and thus may serve as a prognostic marker.
Collapse
|
12
|
Zhou B, Zhao YC, Liu H, Luo S, Amos CI, Lee JE, Li X, Nan H, Wei Q. Novel Genetic Variants of ALG6 and GALNTL4 of the Glycosylation Pathway Predict Cutaneous Melanoma-Specific Survival. Cancers (Basel) 2020; 12:E288. [PMID: 31991610 PMCID: PMC7072252 DOI: 10.3390/cancers12020288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 11/26/2022] Open
Abstract
Because aberrant glycosylation is known to play a role in the progression of melanoma, we hypothesize that genetic variants of glycosylation pathway genes are associated with the survival of cutaneous melanoma (CM) patients. To test this hypothesis, we used a Cox proportional hazards regression model in a single-locus analysis to evaluate associations between 34,096 genetic variants of 227 glycosylation pathway genes and CM disease-specific survival (CMSS) using genotyping data from two previously published genome-wide association studies. The discovery dataset included 858 CM patients with 95 deaths from The University of Texas MD Anderson Cancer Center, and the replication dataset included 409 CM patients with 48 deaths from Harvard University nurse/physician cohorts. In the multivariable Cox regression analysis, we found that two novel single-nucleotide polymorphisms (SNPs) (ALG6 rs10889417 G>A and GALNTL4 rs12270446 G>C) predicted CMSS, with an adjusted hazards ratios of 0.60 (95% confidence interval = 0.44-0.83 and p = 0.002) and 0.66 (0.52-0.84 and 0.004), respectively. Subsequent expression quantitative trait loci (eQTL) analysis revealed that ALG6 rs10889417 was associated with mRNA expression levels in the cultured skin fibroblasts and whole blood cells and that GALNTL4 rs12270446 was associated with mRNA expression levels in the skin tissues (all p < 0.05). Our findings suggest that, once validated by other large patient cohorts, these two novel SNPs in the glycosylation pathway genes may be useful prognostic biomarkers for CMSS, likely through modulating their gene expression.
Collapse
Affiliation(s)
- Bingrong Zhou
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China;
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; (Y.C.Z.); (H.L.)
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yu Chen Zhao
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; (Y.C.Z.); (H.L.)
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; (Y.C.Z.); (H.L.)
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Christopher I. Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeffrey E. Lee
- Department of Surgical Oncology, the University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Xin Li
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (X.L.); (H.N.)
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Hongmei Nan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (X.L.); (H.N.)
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA; (Y.C.Z.); (H.L.)
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
13
|
Yuan Y, Qin W, Buyyounouski M, Ibragimov B, Hancock S, Han B, Xing L. Prostate cancer classification with multiparametric MRI transfer learning model. Med Phys 2019; 46:756-765. [DOI: 10.1002/mp.13367] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yixuan Yuan
- Department of Electronic Engineering City University of Hong Kong Kowloon Tong Hong Kong
- Department of Radiation Oncology Stanford University Stanford 94305USA
| | - Wenjian Qin
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen 518055People's Republic of China
| | - Mark Buyyounouski
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen 518055People's Republic of China
| | - Bulat Ibragimov
- Department of Radiation Oncology Stanford University Stanford 94305USA
| | - Steve Hancock
- Department of Radiation Oncology Stanford University Stanford 94305USA
| | - Bin Han
- Department of Radiation Oncology Stanford University Stanford 94305USA
| | - Lei Xing
- Department of Radiation Oncology Stanford University Stanford 94305USA
| |
Collapse
|
14
|
Dębniak T, Scott RJ, Górski B, Masojć B, Kram A, Maleszka R, Cybulski C, Paszkowska-Szczur K, Kashyap A, Murawa D, Malińska K, Kiedrowicz M, Rogoża-Janiszewska E, Rudnicka H, Deptuła J, Domagała P, Kluźniak W, Lener MR, Lubiński J. BRCA1/2 mutations are not a common cause of malignant melanoma in the Polish population. PLoS One 2018; 13:e0204768. [PMID: 30286154 PMCID: PMC6171837 DOI: 10.1371/journal.pone.0204768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022] Open
Abstract
The association of BRCA1/2 mutations with melanoma is not completely determined; the interpretation of variants of unknown significance is also problematic. To evaluate these issues we explored the molecular basis of melanoma risk by performing whole-exome sequencing on a cohort of 96 unrelated Polish early-onset melanoma patients and targeted sequencing of BRCA1/2 genes on additional 30 melanoma patients with familial aggregation of breast and other cancers. Sequencing was performed on peripheral blood. We evaluated MutationTaster, Polyphen2, SIFT, PROVEAN algorithms, analyzed segregation with cancer disease (in both families with identified BRCA2 variants) and in one family performed LOH (based on 2 primary tumors). We found neither pathogenic mutations nor variants of unknown significance within BRCA1. We identified two BRCA2 variants of unknown significance: c.9334G>A and c.4534 C>T. Disease allele frequency was evaluated by genotyping of 1230 consecutive melanoma cases, 5000 breast cancer patients, 3500 prostate cancers and 9900 controls. Both variants were found to be absent among unselected cancer patients and healthy controls. The MutationTaster, Polyphen2 and SIFT algorithms indicate that c.9334G>A is a damaging variant. Due to lack of tumour tissue LOH analysis could not be performed for this variant. The variant segregated with the disease. The c.4534 C>T variant did not segregate with disease, there was no LOH of the variant. The c.9334G>A variant, classified as a rare variant of unknown significance, on current evidence may predisposes to cancers of the breast, prostate and melanoma. Functional studies to describe how the DNA change affects the protein function and a large multi-center study to evaluate its penetrance are required.
Collapse
Affiliation(s)
- Tadeusz Dębniak
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
- * E-mail:
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle and the Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Bohdan Górski
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Andrzej Kram
- West Pomeranian Oncology Center, Szczecin, Poland
| | | | - Cezary Cybulski
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Katarzyna Paszkowska-Szczur
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Aniruddh Kashyap
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dawid Murawa
- I Department of Oncological and General Surgery, Greater Poland Cancer Center, Poznań, Poland
| | - Karolina Malińska
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Emilia Rogoża-Janiszewska
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Helena Rudnicka
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jakub Deptuła
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Paweł Domagała
- Department of Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Wojciech Kluźniak
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Marcin R. Lener
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jan Lubiński
- Department of Genetics and Pathomorphology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
15
|
Visconti A, Duffy DL, Liu F, Zhu G, Wu W, Chen Y, Hysi PG, Zeng C, Sanna M, Iles MM, Kanetsky PA, Demenais F, Hamer MA, Uitterlinden AG, Ikram MA, Nijsten T, Martin NG, Kayser M, Spector TD, Han J, Bataille V, Falchi M. Genome-wide association study in 176,678 Europeans reveals genetic loci for tanning response to sun exposure. Nat Commun 2018; 9:1684. [PMID: 29739929 PMCID: PMC5940788 DOI: 10.1038/s41467-018-04086-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/03/2018] [Indexed: 12/03/2022] Open
Abstract
The skin’s tendency to sunburn rather than tan is a major risk factor for skin cancer. Here we report a large genome-wide association study of ease of skin tanning in 176,678 subjects of European ancestry. We identify significant association with tanning ability at 20 loci. We confirm previously identified associations at six of these loci, and report 14 novel loci, of which ten have never been associated with pigmentation-related phenotypes. Our results also suggest that variants at the AHR/AGR3 locus, previously associated with cutaneous malignant melanoma the underlying mechanism of which is poorly understood, might act on disease risk through modulation of tanning ability. The skin’s tanning response to sun exposure shows great interindividual variability. Here, Visconti et al. perform a genome-wide association study for ease of skin tanning and identify 20 genetic loci, ten of which had not previously been associated with pigmentation-related traits.
Collapse
Affiliation(s)
- Alessia Visconti
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
| | - Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Gu Zhu
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
| | - Wenting Wu
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin & Bren Simon Cancer Center, Indiana University, Indianapolis, 46202, IN, USA
| | - Yan Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pirro G Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Changqing Zeng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Marianna Sanna
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Mark M Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, UK
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, 33612, FL, USA
| | - Florence Demenais
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, 75010, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, 75010, France
| | - Merel A Hamer
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands.,Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, 4029, Australia
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, 3000 CA, The Netherlands
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin & Bren Simon Cancer Center, Indiana University, Indianapolis, 46202, IN, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Department of Dermatology, West Herts NHS Trust, Herts, HP2 4AD, UK
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.
| |
Collapse
|
16
|
Yu Y, Hu H, Chen JS, Hu F, Fowler J, Scheet P, Zhao H, Huff CD. Integrated case-control and somatic-germline interaction analyses of melanoma susceptibility genes. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2247-2254. [PMID: 29317335 DOI: 10.1016/j.bbadis.2018.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/20/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022]
Abstract
While a number of genes have been implicated in melanoma susceptibility, the role of protein-coding variation in melanoma development and progression remains underexplored. To better characterize the role of germline coding variation in melanoma, we conducted a whole-exome case-control and somatic-germline interaction study involving 322 skin cutaneous melanoma cases from The Cancer Genome Atlas and 3607 controls of European ancestry. We controlled for cross-platform technological stratification using XPAT and conducted gene-based association tests using VAAST 2. Four established melanoma susceptibility genes achieved nominal statistical significance, MC1R (p = .0014), MITF (p = .0165) BRCA2 (p = .0206), and MTAP (p = .0393). We also observed a suggestive association for FANCA (p = .002), a gene previously implicated in melanoma survival. The association signal for BRCA2 was driven primarily by likely gene disrupting (LGD) variants, with an Odds Ratio (OR) of 5.62 (95% Confidence Interval (CI) 1.03-30.1). In contrast, the association signals for MC1R and MITF were driven primarily by predicted pathogenic missense variants, with estimated ORs of 1.4 to 3.0 for MC1R and 4.1 for MITF. MTAP exhibited an excess of both LGD and predicted damaging missense variants among cases, with ORs of 5.62 and 3.72, respectively, although neither category was significant. For individuals with known or predicted damaging variants, age of disease onset was significantly lower for two of the four genes, MC1R (p = .005) and MTAP (p = .035). In an analysis of germline carrier status and overlapping copy number alterations, we observed no evidence to support a two-hit model of carcinogenesis in any of the four genes. Although MC1R carriers were represented proportionally among the four molecular tumor subtypes, these individuals accounted for 69% of ultraviolet (UV) radiation mutational signatures among triple-wild type tumors (p = .040), highlighting the increased sensitivity to UV exposure among individuals with loss-of-function variants in MC1R.
Collapse
Affiliation(s)
- Yao Yu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Hu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiun-Sheng Chen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Fulan Hu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jerry Fowler
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chad D Huff
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Morgese F, Soldato D, Pagliaretta S, Giampieri R, Brancorsini D, Torniai M, Rinaldi S, Savini A, Onofri A, Scarpelli M, Berardi R. Impact of phosphoinositide-3-kinase and vitamin D3 nuclear receptor single-nucleotide polymorphisms on the outcome of malignant melanoma patients. Oncotarget 2017; 8:75914-75923. [PMID: 29100280 PMCID: PMC5652674 DOI: 10.18632/oncotarget.18304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 04/27/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Several studies associating single nucleotide polymorphisms (SNPs) frequencies with tumors outcome have been conducted, nevertheless malignant melanoma literature data are inconclusive.Therefore we evaluate the impact of different genotypes for phosphoinositide-3-kinase (PI3K) and vitamin D3 nuclear receptor (VDR) SNPs on melanoma patients' outcome. MATERIALS AND METHODS Genomic DNA of 88 patients was extracted from blood and tumor samples. SNPs were determined by PCR using TaqMan assays. We selected polymorphisms of the regulatory and catalytic subunit of PI3K (PIK3R1 and PIK3CA genes, respectively), analyzing rs2699887C>T of PIK3CA and rs3730089G>A of PIK3R1 SNPs. Furthermore we considered the following VDR SNPs: rs2228570A>G (Fok1), rs731236A>G (Taq1) and rs1544410C>T (Bsm1).Progression free survival (PFS) and overall survival (OS) were estimated with the Kaplan-Meier method and with Mantel-Haenszel log-rank test. RESULTS The statistical analysis for Fok1 of VDR showed a significant difference in PFS after the first line therapy (median PFS= 21.2 months in the homozygous recessive genotype group vs. 3.3 months of homozygous dominant and heterozygous ones, p= 0.03). In particular, in homozygous recessive patients for Fok1 SNPs of VDR a high rate of histological regression and BRAF (B- Rapidly Accelerated Fibrosarcoma gene) mutation were observed. Furthermore, more efficacy of BRAF +/- MEK (MAPK-ERK-Kinase) inhibitors therapies in homozygous recessive patients vs. homozygous dominant and heterozygous ones was shown. CONCLUSIONS Our study showed a significant correlation between homozygous recessive genotype of Fok1 SNPs of VDR gene and an increased PFS in patients who underwent a first line therapy with BRAF inhibitors.
Collapse
Affiliation(s)
- Francesca Morgese
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Davide Soldato
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Silvia Pagliaretta
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Riccardo Giampieri
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Donatella Brancorsini
- Section of Pathological Anatomy and Histopathology, Deparment of Neuroscience, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Mariangela Torniai
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Silvia Rinaldi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Agnese Savini
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Azzurra Onofri
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy and Histopathology, Deparment of Neuroscience, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti “Umberto I°-G.M. Lancisi-G. Salesi”, Ancona, Italy
| |
Collapse
|
18
|
Liu S, Wang Y, Xue W, Liu H, Xu Y, Shi Q, Wu W, Zhu D, Amos CI, Fang S, Lee JE, Hyslop T, Li Y, Han J, Wei Q. Genetic variants in the genes encoding rho GTPases and related regulators predict cutaneous melanoma-specific survival. Int J Cancer 2017; 141:721-730. [PMID: 28510328 PMCID: PMC5512872 DOI: 10.1002/ijc.30785] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/24/2017] [Accepted: 04/21/2017] [Indexed: 01/12/2023]
Abstract
Rho GTPases control cell division, motility, adhesion, vesicular trafficking and phagocytosis, which may affect progression and/or prognosis of cancers. Here, we investigated associations between genetic variants of Rho GTPases-related genes and cutaneous melanoma-specific survival (CMSS) by re-analyzing a published melanoma genome-wide association study (GWAS) and validating the results in another melanoma GWAS. In the single-locus analysis of 36,018 SNPs in 129 Rho-related genes, 427 SNPs were significantly associated with CMSS (p < 0.050 and false-positive report probability <0.2) in the discovery dataset, and five SNPs were replicated in the validation dataset. Among these, four SNPs (i.e., RHOU rs10916352 G > C, ARHGAP22 rs3851552 T > C, ARHGAP44 rs72635537 C > T and ARHGEF10 rs7826362 A > T) were independently predictive of CMSS (a meta-analysis derived p = 9.04 × 10-4 , 9.58 × 10-4 , 1.21 × 10-4 and 8.47 × 10-4 , respectively). Additionally, patients with an increasing number of unfavorable genotypes (NUGs) of these loci had markedly reduced CMSS in both discovery dataset and validation dataset (ptrend =1.47 × 10-7 and 3.12 × 10-5 ). The model including the NUGs and clinical variables demonstrated a significant improvement in predicting the five-year CMSS. Moreover, rs10916352C and rs3851552C alleles were significantly associated with an increased mRNA expression levels of RHOU (p = 1.8 × 10-6 ) and ARHGAP22 (p = 5.0 × 10-6 ), respectively. These results may provide promising prognostic biomarkers for CM personalized management and treatment.
Collapse
Affiliation(s)
- Shun Liu
- Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning, Guangxi 530021, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yanru Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - William Xue
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yinghui Xu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qiong Shi
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Dermatology, Xijing Hospital, Xi’an, Shanxi 710032, China
| | - Wenting Wu
- Department of Epidemiology, Fairbanks School of Public Health, and Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - Dakai Zhu
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Christopher I. Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Terry Hyslop
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27710, USA
| | - Yi Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, and Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Qingyi Wei
- Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning, Guangxi 530021, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
19
|
Li H, Wang Y, Liu H, Shi Q, Xu Y, Wu W, Zhu D, Amos CI, Fang S, Lee JE, Han J, Wei Q. Genetic variants in the integrin signaling pathway genes predict cutaneous melanoma survival. Int J Cancer 2016; 140:1270-1279. [PMID: 27914105 DOI: 10.1002/ijc.30545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/02/2016] [Indexed: 01/26/2023]
Abstract
To identify genetic variants involved in prognosis of cutaneous melanoma (CM), we investigated associations of single nucleotide polymorphisms (SNPs) of genes in the integrin signaling pathway with CM survival by re-analyzing a published genome-wide association study (GWAS) from The University of Texas M.D. Anderson Cancer Center (MDACC) and then validated significant SNPs in another GWAS from Harvard University. In the MDACC study, 1,148 SNPs were significantly associated with CM-specific survival (CMSS) (p ≤ 0.050 and false-positive report probability ≤ 0.20), and nine SNPs were validated in the Harvard study (p ≤ 0.050). Among these, three independent SNPs (i.e., DOCK1 rs11018104 T > A, rs35748949 C > T and PAK2 rs1718404 C > T) showed a predictive role in CMSS, with an effect-allele attributed adjusted hazards ratio [adjHR of 1.50 (95% confidence interval (CI) = 1.18-1.90, p = 7.46E-04), 1.53 (1.18-1.97, 1.18E-03) and 0.58 (0.45-0.76, 5.60E-05), respectively]. Haplotype analysis revealed that a haplotype carrying two risk alleles A-T in DOCK1 was associated with the poorest survival in both MDACC (adjHR = 1.73, 95% CI = 1.19-2.50, p = 0.004) and Harvard (adjHR = 1.95, 95% CI = 1.14-3.33, p = 0.010) studies. In addition, patients with an increasing number of unfavorable genotypes (NUGs) for these three SNPs had a poorer survival. Incorporating NUGs with clinical variables showed a significantly improved ability to classify CMSS (AUC increased from 86.8% to 88.6%, p = 0.031). Genetic variants in the integrin signaling pathway may independently or jointly modulate the survival of CM patients. Further large, prospective studies are needed to validate these findings.
Collapse
Affiliation(s)
- Hongyu Li
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC.,Department of Gastroenterology, Shenyang Northern Hospital, Shenyang, Liaoning, 110840, China
| | - Yanru Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Qiong Shi
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC.,Department of Dermatology, Xijing Hospital, Xi'an, Shanxi, 710032, China
| | - Yinghui Xu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC.,Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Wenting Wu
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN
| | - Dakai Zhu
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH
| | - Christopher I Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| |
Collapse
|
20
|
Zhang W, Liu H, Yin J, Wu W, Zhu D, Amos CI, Fang S, Lee JE, Li Y, Han J, Wei Q. Genetic variants in the PIWI-piRNA pathway gene DCP1A predict melanoma disease-specific survival. Int J Cancer 2016; 139:2730-2737. [PMID: 27578485 DOI: 10.1002/ijc.30409] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/12/2016] [Accepted: 08/18/2016] [Indexed: 12/30/2022]
Abstract
The Piwi-piRNA pathway is important for germ cell maintenance, genome integrity, DNA methylation and retrotransposon control and thus may be involved in cancer development. In this study, we comprehensively analyzed prognostic roles of 3,116 common SNPs in PIWI-piRNA pathway genes in melanoma disease-specific survival. A published genome-wide association study (GWAS) by The University of Texas M.D. Anderson Cancer Center was used to identify associated SNPs, which were later validated by another GWAS from the Harvard Nurses' Health Study and Health Professionals Follow-up Study. After multiple testing correction, we found that there were 27 common SNPs in two genes (PIWIL4 and DCP1A) with false discovery rate < 0.2 in the discovery dataset. Three tagSNPs (i.e., rs7933369 and rs508485 in PIWIL4; rs11551405 in DCP1A) were replicated. The rs11551405 A allele, located at the 3' UTR microRNA binding site of DCP1A, was associated with an increased risk of melanoma disease-specific death in both discovery dataset [adjusted Hazards ratio (HR) = 1.66, 95% confidence interval (CI) = 1.21-2.27, p =1.50 × 10-3 ] and validation dataset (HR = 1.55, 95% CI = 1.03-2.34, p = 0.038), compared with the C allele, and their meta-analysis showed an HR of 1.62 (95% CI, 1.26-2.08, p =1.55 × 10-4 ). Using RNA-seq data from the 1000 Genomes Project, we found that DCP1A mRNA expression levels increased significantly with the A allele number of rs11551405. Additional large, prospective studies are needed to validate these findings.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongliang Liu
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Jieyun Yin
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Wenting Wu
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Dakai Zhu
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Christopher I Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Yi Li
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, and Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana. .,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Qingyi Wei
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
21
|
Potrony M, Carreras E, Aranda F, Zimmer L, Puig-Butille JA, Tell-Martí G, Armiger N, Sucker A, Giménez-Xavier P, Martínez-Florensa M, Carrera C, Malvehy J, Schadendorf D, Puig S, Lozano F. Inherited functional variants of the lymphocyte receptor CD5 influence melanoma survival. Int J Cancer 2016; 139:1297-302. [PMID: 27169428 DOI: 10.1002/ijc.30184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/18/2016] [Accepted: 05/02/2016] [Indexed: 12/16/2022]
Abstract
Despite the recent progress in treatment options, malignant melanoma remains a deadly disease. Besides therapy, inherited factors might modulate clinical outcome, explaining in part widely varying survival rates. T-cell effector function regulators on antitumor immune responses could also influence survival. CD5, a T-cell receptor inhibitory molecule, contributes to the modulation of antimelanoma immune responses as deduced from genetically modified mouse models. The CD5 SNPs rs2241002 (NM_014207.3:c.671C > T, p.Pro224Leu) and rs2229177 (NM_014207.3:c.1412C > T, p.Ala471Val) constitute an ancestral haplotype (Pro224-Ala471) that confers T-cell hyper-responsiveness and worsens clinical autoimmune outcome. The assessment of these SNPs on survival impact from two melanoma patient cohorts (Barcelona, N = 493 and Essen, N = 215) reveals that p.Ala471 correlates with a better outcome (OR= 0.57, 95% CI = 0.33-0.99, Adj. p = 0.043, in Barcelona OR = 0.63, 95% CI = 0.40-1.01, Adj. p = 0.051, in Essen). While, p.Leu224 was associated with increased melanoma-associated mortality in both cohorts (OR = 1.87, 95% CI = 1.07-3.24, Adj. p = 0.030 in Barcelona and OR = 1.84, 95% CI = 1.04-3.26, Adj. p = 0.037, in Essen). Furthermore survival analyses showed that the Pro224-Ala471 haplotype in homozygosis improved melanoma survival in the entire set of patients (HR = 0.27, 95% CI 0.11-0.67, Adj. p = 0.005). These findings highlight the relevance of genetic variability in immune-related genes for clinical outcome in melanoma.
Collapse
Affiliation(s)
- Miriam Potrony
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain
| | - Esther Carreras
- Grup D'Immunoreceptors Del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Fernando Aranda
- Grup D'Immunoreceptors Del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium, Heidelberg, Germany
| | - Joan-Anton Puig-Butille
- Melanoma Unit, Molecular Biology and Genetics Department, Hospital Clínic De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Gemma Tell-Martí
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Noelia Armiger
- Grup D'Immunoreceptors Del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium, Heidelberg, Germany
| | - Pol Giménez-Xavier
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Mario Martínez-Florensa
- Grup D'Immunoreceptors Del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina Carrera
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Josep Malvehy
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany & German Cancer Consortium, Heidelberg, Germany
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clínic De Barcelona, IDIBAPS, Universitat De Barcelona, Barcelona, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Raras (CIBERER), Valencia, Spain
| | - Francisco Lozano
- Grup D'Immunoreceptors Del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Department of Immunology, Hospital Clínic De Barcelona, Barcelona, Spain.,Department of Cell Biology, Immunology and Neurosciences, School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Tuominen R, Engström PG, Helgadottir H, Eriksson H, Unneberg P, Kjellqvist S, Yang M, Lindén D, Edsgärd D, Hansson J, Höiom V. The role of germline alterations in the DNA damage response genes BRIP1 and BRCA2 in melanoma susceptibility. Genes Chromosomes Cancer 2016; 55:601-11. [PMID: 27074266 DOI: 10.1002/gcc.22363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 01/25/2023] Open
Abstract
We applied a targeted sequencing approach to identify germline mutations conferring a moderately to highly increased risk of cutaneous and uveal melanoma. Ninety-two high-risk melanoma patients were screened for inherited variation in 120 melanoma candidate genes. Observed gene variants were filtered based on frequency in reference populations, cosegregation with melanoma in families and predicted functional effect. Several novel or rare genetic variants in genes involved in DNA damage response, cell-cycle regulation and transcriptional control were identified in melanoma patients. Among identified genetic alterations was an extremely rare variant (minor allele frequency of 0.00008) in the BRIP1 gene that was found to cosegregate with the melanoma phenotype. We also found a rare nonsense variant in the BRCA2 gene (rs11571833), previously associated with cancer susceptibility but not with melanoma, which showed weak association with melanoma susceptibility in the Swedish population. Our results add to the growing knowledge about genetic factors associated with melanoma susceptibility and also emphasize the role of DNA damage response as an important factor in melanoma etiology. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rainer Tuominen
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pär G Engström
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Hanna Eriksson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Per Unneberg
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Sanela Kjellqvist
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Muyi Yang
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Diana Lindén
- Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Daniel Edsgärd
- Science for Life Laboratory, School of Biotechnology, Division of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Johan Hansson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Yin J, Liu H, Yi X, Wu W, Amos CI, Fang S, Lee JE, Han J, Wei Q. Genetic variants in the vitamin D pathway genes VDBP and RXRA modulate cutaneous melanoma disease-specific survival. Pigment Cell Melanoma Res 2016; 29:176-85. [PMID: 26575331 DOI: 10.1111/pcmr.12437] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/06/2015] [Accepted: 11/09/2015] [Indexed: 01/04/2023]
Abstract
Single nucleotide polymorphisms (SNPs) in the vitamin D pathway genes have been implicated in cutaneous melanoma (CM) risk, but their role in CM disease-specific survival (DSS) remains obscure. We comprehensively analyzed the prognostic roles of 2669 common SNPs in the vitamin D pathway genes using data from a published genome-wide association study (GWAS) at The University of Texas M.D. Anderson Cancer Center (MDACC) and then validated the SNPs of interest in another GWAS from the Nurses' Health Study and Health Professionals Follow-up Study. Among the 2669 SNPs, 203 were significantly associated with DSS in MDACC dataset (P < 0.05 and false-positive report probability < 0.2), of which 18 were the tag SNPs. In the replication, two of these 18 SNPs showed nominal significance: the VDBP rs12512631 T > C was associated with a better DSS [combined hazards ratio (HR) = 0.66]; and the same for RXRA rs7850212 C > A (combined HR = 0.38), which were further confirmed by the Fine and Gray competing-risks regression model. Further bioinformatics analyses indicated that these loci may modulate corresponding gene methylation status.
Collapse
Affiliation(s)
- Jieyun Yin
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Department of Epidemiology and Biostatistics and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Xiaohua Yi
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Wenting Wu
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Christopher I Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
24
|
He K, Li Y, Zhu J, Liu H, Lee JE, Amos CI, Hyslop T, Jin J, Lin H, Wei Q, Li Y. Component-wise gradient boosting and false discovery control in survival analysis with high-dimensional covariates. Bioinformatics 2016; 32:50-7. [PMID: 26382192 PMCID: PMC4757968 DOI: 10.1093/bioinformatics/btv517] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/07/2015] [Accepted: 08/25/2015] [Indexed: 12/22/2022] Open
Abstract
MOTIVATION Technological advances that allow routine identification of high-dimensional risk factors have led to high demand for statistical techniques that enable full utilization of these rich sources of information for genetics studies. Variable selection for censored outcome data as well as control of false discoveries (i.e. inclusion of irrelevant variables) in the presence of high-dimensional predictors present serious challenges. This article develops a computationally feasible method based on boosting and stability selection. Specifically, we modified the component-wise gradient boosting to improve the computational feasibility and introduced random permutation in stability selection for controlling false discoveries. RESULTS We have proposed a high-dimensional variable selection method by incorporating stability selection to control false discovery. Comparisons between the proposed method and the commonly used univariate and Lasso approaches for variable selection reveal that the proposed method yields fewer false discoveries. The proposed method is applied to study the associations of 2339 common single-nucleotide polymorphisms (SNPs) with overall survival among cutaneous melanoma (CM) patients. The results have confirmed that BRCA2 pathway SNPs are likely to be associated with overall survival, as reported by previous literature. Moreover, we have identified several new Fanconi anemia (FA) pathway SNPs that are likely to modulate survival of CM patients. AVAILABILITY AND IMPLEMENTATION The related source code and documents are freely available at https://sites.google.com/site/bestumich/issues. CONTACT yili@umich.edu.
Collapse
Affiliation(s)
| | | | - Ji Zhu
- Department of Statistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Hongliang Liu
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher I Amos
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03750, USA
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University and Duke Clinical Research Institute, Durham, NC 27710, USA
| | - Jiashun Jin
- Department of Statistics, Carnegie Mellon University, Pittsburgh, PA 15213, USA and
| | - Huazhen Lin
- Center of Statistical Research, School of Statistics, Southwestern University of Finance and Economics, Chengdu, Sichuan 611130, China
| | - Qinyi Wei
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Yi Li
- Department of Biostatistics and
| |
Collapse
|
25
|
Orlow I, Reiner AS, Thomas NE, Roy P, Kanetsky PA, Luo L, Paine S, Armstrong BK, Kricker A, Marrett LD, Rosso S, Zanetti R, Gruber SB, Anton-Culver H, Gallagher RP, Dwyer T, Busam K, Begg CB, Berwick M. Vitamin D receptor polymorphisms and survival in patients with cutaneous melanoma: a population-based study. Carcinogenesis 2015; 37:30-8. [PMID: 26521212 DOI: 10.1093/carcin/bgv157] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022] Open
Abstract
Factors known to affect melanoma survival include age at presentation, sex and tumor characteristics. Polymorphisms also appear to modulate survival following diagnosis. Result from other studies suggest that vitamin D receptor (VDR) polymorphisms (SNPs) impact survival in patients with glioma, renal cell carcinoma, lung, breast, prostate and other cancers; however, a comprehensive study of VDR polymorphisms and melanoma-specific survival is lacking. We aimed to investigate whether VDR genetic variation influences survival in patients with cutaneous melanoma. The analysis involved 3566 incident single and multiple primary melanoma cases enrolled in the international population-based Genes, Environment, and Melanoma Study. Melanoma-specific survival outcomes were calculated for each of 38 VDR SNPs using a competing risk analysis after adjustment for covariates. There were 254 (7.1%) deaths due to melanoma during the median 7.6 years follow-up period. VDR SNPs rs7299460, rs3782905, rs2239182, rs12370156, rs2238140, rs7305032, rs1544410 (BsmI) and rs731236 (TaqI) each had a statistically significant (trend P values < 0.05) association with melanoma-specific survival in multivariate analysis. One functional SNP (rs2239182) remained significant after adjustment for multiple testing using the Monte Carlo method. None of the SNPs associated with survival were significantly associated with Breslow thickness, ulceration or mitosis. These results suggest that the VDR gene may influence survival from melanoma, although the mechanism by which VDR exerts its effect does not seem driven by tumor aggressiveness. Further investigations are needed to confirm our results and to understand the relationship between VDR and survival in the combined context of tumor and host characteristics.
Collapse
Affiliation(s)
| | | | - Nancy E Thomas
- Department of Dermatology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | | | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Li Luo
- Department of Internal Medicine, Epidemiology and Cancer Prevention, University of New Mexico, Albuquerque, NM 87131, USA
| | - Susan Paine
- Department of Internal Medicine, Epidemiology and Cancer Prevention, University of New Mexico, Albuquerque, NM 87131, USA
| | - Bruce K Armstrong
- Sydney School of Public Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anne Kricker
- Sydney School of Public Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Loraine D Marrett
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, Ontario M5G 2L7, Canada
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin 10126, Italy
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin 10126, Italy
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Hoda Anton-Culver
- Department of Epidemiology, School of Medicine, University of California at Irvine, Irvine, CA 92617, USA
| | - Richard P Gallagher
- Cancer Control Research, British Columbia Cancer Research Centre, Vancouver, British Columbia V5Z 1L3, Canada
| | - Terence Dwyer
- The George Institute for Global Health, Oxford Martin School, University of Oxford, Oxford OX1 3BD, UK and
| | - Klaus Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Marianne Berwick
- Department of Internal Medicine, Epidemiology and Cancer Prevention, University of New Mexico, Albuquerque, NM 87131, USA
| | | |
Collapse
|
26
|
Abstract
Approximately 10% of melanoma cases report a relative affected with melanoma, and a positive family history is associated with an increased risk of developing melanoma. Although the majority of genetic alterations associated with melanoma development are somatic, the underlying presence of heritable melanoma risk genes is an important component of disease occurrence. Susceptibility for some families is due to mutation in one of the known high penetrance melanoma predisposition genes: CDKN2A, CDK4, BAP1, POT1, ACD, TERF2IP and TERT. However, despite such mutations being implicated in a combined total of approximately 50% of familial melanoma cases, the underlying genetic basis is unexplained for the remainder of high-density melanoma families. Aside from the possibility of extremely rare mutations in a few additional high penetrance genes yet to be discovered, this suggests a likely polygenic component to susceptibility, and a unique level of personal melanoma risk influenced by multiple low-risk alleles and genetic modifiers. In addition to conferring a risk of cutaneous melanoma, some 'melanoma' predisposition genes have been linked to other cancers, with cancer clustering observed in melanoma families at rates greater than expected by chance. The most extensively documented association is between CDKN2A germ line mutations and pancreatic cancer, and a cancer syndrome including cutaneous melanoma, uveal melanoma and mesothelioma has been proposed for BAP1 germ line mutations. Other medium to high penetrance melanoma predisposition genes have been associated with renal cell carcinoma (MITF, BAP1) and glioma (POT1). These associations between melanoma and other cancers hint at the possibility of common pathways for oncogenesis, and better knowledge of these pathways may improve understanding of the genetic basis underpinning familial melanoma. It is likely that 'melanoma' risk genes will impact on mutation screening and genetic counselling not only for melanoma but also a range of other cancers.
Collapse
Affiliation(s)
- Jazlyn Read
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia The University of Queensland, Brisbane, Queensland, Australia
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Zhang W, Liu H, Liu Z, Zhu D, Amos CI, Fang S, Lee JE, Wei Q. Functional Variants in Notch Pathway Genes NCOR2, NCSTN, and MAML2 Predict Survival of Patients with Cutaneous Melanoma. Cancer Epidemiol Biomarkers Prev 2015; 24:1101-10. [PMID: 25953768 PMCID: PMC4573541 DOI: 10.1158/1055-9965.epi-14-1380-t] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/05/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The Notch signaling pathway is constitutively activated in human cutaneous melanoma to promote growth and aggressive metastatic potential of primary melanoma cells. Therefore, genetic variants in Notch pathway genes may affect the prognosis of cutaneous melanoma patients. METHODS We identified 6,256 SNPs in 48 Notch genes in 858 cutaneous melanoma patients included in a previously published cutaneous melanoma genome-wide association study dataset. Multivariate and stepwise Cox proportional hazards regression and false-positive report probability corrections were performed to evaluate associations between putative functional SNPs and cutaneous melanoma disease-specific survival. Receiver operating characteristic curve was constructed, and area under the curve was used to assess the classification performance of the model. RESULTS Four putative functional SNPs of Notch pathway genes had independent and joint predictive roles in survival of cutaneous melanoma patients. The most significant variant was NCOR2 rs2342924 T>C (adjusted HR, 2.71; 95% confidence interval, 1.73-4.23; Ptrend = 9.62 × 10(-7)), followed by NCSTN rs1124379 G>A, NCOR2 rs10846684 G>A, and MAML2 rs7953425 G>A (Ptrend = 0.005, 0.005, and 0.013, respectively). The receiver operating characteristic analysis revealed that area under the curve was significantly increased after adding the combined unfavorable genotype score to the model containing the known clinicopathologic factors. CONCLUSIONS Our results suggest that SNPs in Notch pathway genes may be predictors of cutaneous melanoma disease-specific survival. IMPACT Our discovery offers a translational potential for using genetic variants in Notch pathway genes as a genotype score of biomarkers for developing an improved prognostic assessment and personalized management of cutaneous melanoma patients.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina. Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongliang Liu
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Zhensheng Liu
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Dakai Zhu
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Christopher I Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Qingyi Wei
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
28
|
van den Reek JM, Kievit W, Gniadecki R, Goeman JJ, Zweegers J, van de Kerkhof PC, Seyger MM, de Jong EM. Drug Survival Studies in Dermatology:Principles, Purposes, and Pitfalls. J Invest Dermatol 2015; 135:1-5. [DOI: 10.1038/jid.2015.171] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
29
|
Yuan H, Liu H, Liu Z, Zhu D, Amos CI, Fang S, Lee JE, Wei Q. Genetic variants in Hippo pathway genes YAP1, TEAD1 and TEAD4 are associated with melanoma-specific survival. Int J Cancer 2015; 137:638-45. [PMID: 25628125 DOI: 10.1002/ijc.29429] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/19/2014] [Accepted: 12/23/2014] [Indexed: 01/22/2023]
Abstract
Cutaneous melanoma (CM) is the most lethal form of skin cancers. The Hippo pathway controls cell migration, development and sizes of the organs in diverse species, and deregulation of this pathway may affect CM progression and prognosis. Therefore, we hypothesized that genetic variants of Hippo pathway genes might predict survival of CM patients. We used the genotyping data of 1,115 common single nucleotide polymorphisms (SNPs) in the 12 pathway core genes (i.e., MST1, MST2, SAV1, LATS1, LATS2, MOB1A, MOB1B, YAP1, TEAD1, TEAD2, TEAD3 and TEAD4) from the dataset of our previously published CM genome-wide association study and comprehensively analyzed their associations with CM-specific survival (CSS) in 858 CM patients by using the Kaplan-Meier analyses and Cox proportional hazards regression models. We found a predictive role of YAP1 rs11225163 CC, TEAD1 rs7944031 AG+GG and TEAD4 rs1990330 CA+AA in the prognosis of CM. In addition, patients with an increasing number of unfavorable genotypes (NUG) had a markedly increased risk of death. After incorporating NUG in the model with clinical variables, the new model showed a significantly improved discriminatory ability to classify CSS (AUC increased from 82.03% to 84.56%). Our findings suggest that genetic variants of Hippo pathway genes, particularly YAP1 rs11225163, TEAD1 rs7944031 and TEAD4 rs1990330, may independently or jointly modulate survival of CM patients. Additional large, prospective studies are needed to validate these findings.
Collapse
Affiliation(s)
- Hua Yuan
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC.,Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Zhensheng Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Dakai Zhu
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH
| | - Christopher I Amos
- Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Medicine, Duke University School of Medicine, Durham, NC
| |
Collapse
|