1
|
Chen C, Fan G, Li P, Yang E, Jing S, Shi Y, Gong Y, Zhang L, Wang Z. Effect of smoking on the recurrence and progression of non-muscle-invasive bladder cancer. Clin Transl Oncol 2025; 27:1695-1711. [PMID: 39266874 DOI: 10.1007/s12094-024-03694-z] [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: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND It is well established that smoking is the most significant risk factor for bladder cancer, yet the impact of smoking on the recurrence and progression of non-muscle-invasive bladder cancer (NMIBC) remains a contentious issue. OBJECTIVE To review all relevant literature published to date, providing a comprehensive assessment of the effects of smoking on the recurrence and progression of NMIBC, thereby offering a basis for smoking cessation management in NMIBC patients. METHODS A search was conducted for all relevant literature published up to April 2024 in PubMed, Web of Science, and Embase databases. The existing literature results and deficiencies were analyzed, and the gaps in understanding between different studies were highlighted, with recommendations for future research. RESULTS A total of 24 studies were included in this work. Among them, 14 studies suggested that smoking promotes the recurrence and progression of NMIBC, while another 10 studies concluded that smoking has no effect on the recurrence and progression of NMIBC patients. CONCLUSIONS Our research indicates that smoking increases the risk of recurrence and progression in NMIBC patients, and quitting smoking can improve health-related quality of life. High-quality, large-sample prospective cohort studies (or randomized controlled studies) are still needed in the future.
Collapse
Affiliation(s)
- Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, Chengguan District, No.82 Linxia Road, Lanzhou, 730030, People's Republic of China.
- Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou, People's Republic of China.
| |
Collapse
|
2
|
Islam MA, Mubashshira S, Rahman MM, Kabir Y. Contribution of ERCC2 rs13181 (Lys751Gln) and rs1799793 (Asp312Asn) polymorphisms to the risk of bladder cancer in Bangladesh. Cancer Genet 2024; 288-289:126-132. [PMID: 39551027 DOI: 10.1016/j.cancergen.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Human Excision Repair Cross-Complementation Group 2 (ERCC2) proteins play a vital role in the nucleotide excision repair pathway through ATP-dependent helicase activity. Several studies found that polymorphisms in the ERCC2 gene are associated with susceptibility to different cancers, although the outcomes were confusing. OBJECTIVE As a result, in this retrospective study, we investigated the relationship between genetic polymorphisms of the ERCC2 gene at codons 312 (rs1799793) and 751 (rs13181) and bladder cancer susceptibility in Bangladesh, as well as the disease's aggressiveness. METHODS Genetic polymorphisms of ERCC2 were assayed by the polymerase chain reaction-based restriction fragment length polymorphism (PCR-RFLP) method with 121 bladder cancer patients and 130 healthy controls. RESULTS Patients who had the Gln/Gln polymorphism of ERCC2 at codon 751 (OR=3.27; 95% CI=1.19-8.67; p<0.05) and Asp/Asn at codon 312 (OR=2.14; 95% CI=1.03-4.29; p<0.05) were significantly associated with a higher risk of developing bladder cancer. Again, Gln/Gln polymorphisms in bladder cancer (p<0.05) were more likely to be present in individuals with cancer in the family. CONCLUSIONS This study reveals that susceptibility and bladder cancer aggressiveness are associated with polymorphisms at codon 751 and Asp/Asn at codon 312 of the ERCC2 gene.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh; Bangladesh Atomic Energy Commission, Dhaka, Bangladesh
| | - Saima Mubashshira
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | | | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
3
|
Dai X, Shi X, Luo M, Li P, Gao Y. Integrative analysis of transcriptomic and metabolomic profiles reveals enhanced arginine metabolism in androgen-independent prostate cancer cells. BMC Cancer 2023; 23:1241. [PMID: 38104097 PMCID: PMC10724921 DOI: 10.1186/s12885-023-11707-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/02/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Prostate cancer is a common solid tumor that affects a significant number of men worldwide. Conventional androgen deprivation therapy (ADT) increases the risk of developing castration-resistant prostate cancer (CRPC). Effective clinical management of patients with CRPC is challenging due to the limited understanding. METHODS In this study, transcriptomic and metabolomic profiles of androgen-dependent prostate cancer cell line LNCaP and the androgen-independent cells developed from LNCaP cells (LNCaP-ADR) were investigated using RNA-sequencing and LC-MS/MS, respectively. The differentially expressed genes and metabolites were analyzed, and integrative analysis of transcriptomic and metabolomic data was further conducted to obtain a comprehensive understanding of the metabolic characteristics in LNCaP-ADR cells. Quantitative real-time PCR (QPCR) was employed to ascertain the mRNA expression levels of the selected differentially expressed genes. RESULTS The arginine and proline metabolism pathway was identified as a commonly altered pathway at both the transcriptional and metabolic levels. In the LNCaP-ADR cells, significant upregulation was observed for metabolites including 5-Aminopentanoic acid, L-Arginine, L-Glutamic acid, N-Acetyl-L-alanine, and Pyrrole-2-carboxylic acid at the metabolic level. At the transcriptional level, MAOA, ALDH3A2, ALDH2, ARG1, CKMT2, and CNDP1 were found to be significantly upregulated in the LNCaP-ADR cells. Gene set enrichment analysis (GSEA) identified various enriched gene sets in the LNCaP-ADR cells, encompassing inflammatory response, 9plus2 motile cilium, motile cilium, ciliary plasm, cilium or flagellum-dependent cell motility, cilium movement, cilium, response to endoplasmic reticulum stress, PTEN DN.V1 DN, SRC UP.V1 UP, IL15 UP.V1 DN, RB DN.V1 DN, AKT UP MTOR DN.V1 UP, VEGF A UP.V1 UP, and KRAS.LUNG.BREAST UP.V1 UP. CONCLUSIONS These findings highlight the substantial association between the arginine and proline metabolism pathway and CRPC, emphasizing the need to prioritize strategies that target dysregulated metabolites and differentially expressed genes as essential interventions in the clinical management of CRPC.
Collapse
Affiliation(s)
- Xingchen Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Department of Nephrology, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xinyi Shi
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Ankang Central Hospital, Ankang, China
| | - Mingxiu Luo
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Pu Li
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
4
|
Wang D, Ning H, Wu H, Song Y, Chu Y, Liu F, Zhao Z, Wu F, Lyu J. Construction and evaluation of a novel prognostic risk model of aging-related genes in bladder cancer. Curr Urol 2023; 17:236-245. [PMID: 37994343 PMCID: PMC10662852 DOI: 10.1097/cu9.0000000000000218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 04/22/2023] [Indexed: 11/24/2023] Open
Abstract
Background Bladder cancer (BLCA) is the most common malignancy of the urinary system. Muscle-invasive bladder cancer (MIBC), which constitutes approximately 25% of all BLCA cases, is characterized by frequent recurrence and early onset of metastasis. Bladder cancer most commonly occurs in elderly patients and is significantly associated with aging. However, the prognostic value of age-related genes in BLCA, especially in MIBC, remains unclear. Materials and methods Training and testing sets were obtained from The Cancer Genome Atlas BLCA project. Differentially expressed genes between BLCA and normal samples intersected with human aging-related genes. Univariate Cox regression and least absolute shrinkage and selection operator regression analyses were used to identify prognostic aging-related signatures, followed by the construction of a risk score model and nomogram. Kaplan-Meier and receiver operating characteristic analyses were conducted to assess the predictive power. An independent BLCA cohort of 165 samples was included for external validation. The CIBERSORT algorithm was used to explore the characteristics of the immune microenvironment. Results Seven genes (IGF1, NGF, GCLM, PYCR1, EFEMP1, APOC3, and IFNB1) were identified by Cox and least absolute shrinkage and selection operator analyses. After combining the gene signature with the clinical parameters of patients with BLCA, a risk-prognosis model and nomogram were constructed and validated with the testing set. Bladder cancer cases with high 7-gene signature scores (high-risk group) and low scores (low-risk group) showed distinct prognoses. Furthermore, 7 types of immune cells were significantly altered between the low- and high-risk groups. Conclusions Collectively, our data provide a 7-gene signature that serves as a potential biomarker for BLCA, especially MIBC. Moreover, this 7-gene signature highlights the role of the tumor immune microenvironment in prognosis and thus might be related to the response to anti-programmed cell death protein 1-based immunotherapy.
Collapse
Affiliation(s)
- Delin Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Ning
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Haihu Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yufeng Song
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yaru Chu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Feifan Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhenlin Zhao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jiaju Lyu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Department of Urology, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
5
|
Jiang Y, Sun X, Song X, Li Z, Zhang P, Zhang W, Tang D. Patient-derived bladder cancer organoid model to predict sensitivity and feasibility of tailored precision therapy. Curr Urol 2023; 17:221-228. [PMID: 37994334 PMCID: PMC10662868 DOI: 10.1097/cu9.0000000000000219] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/12/2023] [Indexed: 11/24/2023] Open
Abstract
Background Bladder cancer is a common and highly heterogeneous malignant tumor with a relatively poor prognosis. Thus, personalized treatment strategies for bladder cancer are essential for improving patient outcomes. Materials and methods We developed an efficient 3-dimensional in vitro organoid culture system for bladder cancer organoids (BCOs), which maintains the homology with the original patient tumors and the heterogeneity between different individuals. In addition, we constructed chimeric antigen receptor (CAR)-T cells targeting B7H3 and evaluated the antitumor function of CAR-T cells by coculturing them with BCOs. Results The BCOs closely resembled the characteristics of human tumors and were used to test individual sensitivity to platinum-based drugs and olaparib therapy. Coculture with CAR-T cells demonstrated specific antigen recognition and immune activation, indicating their potential in immunotherapy. Conclusions Our study highlights the potential of BCOs to facilitate the development of personalized medicine for bladder cancer and improve the efficiency of drug discovery for bladder cancer therapy.
Collapse
Affiliation(s)
- Ying Jiang
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| | - Xun Sun
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaoyun Song
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| | - Zhen Li
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| | - Ping Zhang
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| | - Wen Zhang
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| | - Dongqi Tang
- Center for Gene and Immunotherapy, The Second Hospital of Shandong University, Cheeloo College of Medicine, Jinan, China
| |
Collapse
|
6
|
Gercek O, Keles I, Saritas TB, Koyuncu B, Topal K, Demirbas A. Effect of obturator nerve block during transurethral resection of bladder tumors on the disease recurrence, progression and surgery outcomes. Int Urol Nephrol 2023; 55:2765-2772. [PMID: 37531039 DOI: 10.1007/s11255-023-03727-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE In our study, we aimed to evaluate the effect of the obturator nerve block (ONB) on the operation time, duration of hospital stay, complete resection, presence of muscle tissue in the pathology, second resection, recurrence, and progression, when applied in addition to spinal anesthesia in patients with primary bladder lateral wall tumor and Transurethral Resection of Bladder Tumor (TURBT) was planned. MATERIALS AND METHODS Seventy patients with bladder lateral wall tumors were included in the study. In addition, ONB was applied to 35 of the patients who underwent spinal anesthesia. The two groups were compared in terms of obturator reflex development, perforation, complete resection, presence of muscle tissue in pathology samples, need for second resection, need for second resection due to inadequate muscle tissue, and 1 year recurrence and progression rates. RESULTS When the two groups were compared for obturator reflex and bladder perforation, both were found to be lower in the ONB group (p = 0.002, p = 0.198, respectively). The rate of complete resection and the presence of muscle tissue in the pathology samples were higher in the ONB group (p = 0.045, p = 0.034, respectively). The rates of second resection and second resection due to inadequate muscle tissue were found to be higher in the group without ONB (p = 0.015, p = 0.106, respectively). In the 1-year follow-up, the recurrence rate was significantly lower in the ONB group (p < 0.001), while there was no significant difference between the progression rates (p = 0.106). CONCLUSION In our study, we found out that ONB applied in addition to spinal anesthesia increases the rate of complete and muscle tissue resection by decreasing the obturator reflex, and causes a significant reduction in the need for second resection and tumor recurrence.
Collapse
Affiliation(s)
- O Gercek
- Department of Urology, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey.
| | - I Keles
- Department of Urology, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - T B Saritas
- Department of Anesthesiology and Reanimation, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - B Koyuncu
- Department of Anesthesiology and Reanimation, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - K Topal
- Department of Urology, Afyonkarahisar State Hospital, Afyonkarahisar, Turkey
| | - A Demirbas
- Department of Urology, Bursa Doruk Hospital, Bursa, Turkey
| |
Collapse
|
7
|
Karimi M, Mendez-Pineda S, Blanché H, Boland A, Besse C, Deleuze JF, Meng XY, Sirab N, Groussard K, Lebret T, Bonastre J, Allory Y, Radvanyi F, Benhamou S, Michiels S. A Case-Only Genome-Wide Interaction Study of Smoking and Bladder Cancer Risk: Results from the COBLAnCE Cohort. Cancers (Basel) 2023; 15:4218. [PMID: 37686494 PMCID: PMC10487226 DOI: 10.3390/cancers15174218] [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: 07/06/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Bladder cancer (BC) is the 6th most common cancer worldwide, with tobacco smoking considered as its main risk factor. Accumulating evidence has found associations between genetic variants and the risk of BC. Candidate gene-environment interaction studies have suggested interactions between cigarette smoking and NAT2/GSTM1 gene variants. Our objective was to perform a genome-wide association case-only study using the French national prospective COBLAnCE cohort (COhort to study BLAdder CancEr), focusing on smoking behavior. The COBLAnCE cohort comprises 1800 BC patients enrolled between 2012 and 2018. Peripheral blood samples collected at enrolment were genotyped using the Illumina Global Screening Array with a Multi-Disease drop-in panel. Genotyping data (9,719,614 single nucleotide polymorphisms (SNP)) of 1674, 1283, and 1342 patients were analyzed for smoking status, average tobacco consumption, and age at smoking initiation, respectively. A genome-wide association study (GWAS) was conducted adjusting for gender, age, and genetic principal components. The results suggest new candidate loci (4q22.1, 12p13.1, 16p13.3) interacting with smoking behavior for the risk of BC. Our results need to be validated in other case-control or cohort studies.
Collapse
Affiliation(s)
- Maryam Karimi
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
- Bureau de Biostatistique et d’Épidémiologie, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Sebastian Mendez-Pineda
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
| | - Hélène Blanché
- CEPH-Biobank, Fondation Jean Dausset-CEPH, 75010 Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), CEA, Université Paris-Saclay, 91057 Evry, France
| | - Céline Besse
- Centre National de Recherche en Génomique Humaine (CNRGH), CEA, Université Paris-Saclay, 91057 Evry, France
| | - Jean-François Deleuze
- CEPH-Biobank, Fondation Jean Dausset-CEPH, 75010 Paris, France
- Centre National de Recherche en Génomique Humaine (CNRGH), CEA, Université Paris-Saclay, 91057 Evry, France
| | | | - Nanor Sirab
- Curie Institute, CNRS, UMR144, Molecular Oncology Team, PSL Research University, 75005 Paris, France
| | - Karine Groussard
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
| | | | - Julia Bonastre
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
- Bureau de Biostatistique et d’Épidémiologie, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Yves Allory
- CNRS UMR144, Curie Institute, 75005 Paris, France
- UVSQ, Curie Institute, Department of Pathology, Université Paris-Saclay, 92210 Saint-Cloud, France
| | | | - Simone Benhamou
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
| | - Stefan Michiels
- Oncostat U1018 Inserm, Équipe Labellisée Ligue Contre le Cancer, Université Paris-Saclay, 94805 Villejuif, France
- Bureau de Biostatistique et d’Épidémiologie, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| |
Collapse
|
8
|
Li XG, Niu C, Lu P, Wan HW, Jin WD, Wang CX, Mao WY, Zhang ZP, Zhang WF, Li B. Screening and identification of hub-gene associated with brain metastasis in breast cancer. Medicine (Baltimore) 2023; 102:e32771. [PMID: 36800575 PMCID: PMC9935999 DOI: 10.1097/md.0000000000032771] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND The presence of breast cancer in the brain, also known as brain metastasis (BMS), is the primary reason for a bad prognosis in cases of breast cancer. Breast cancer is the most prevalent malignant tumor seen in women in developing nations. At present, there is no effective method to inhibit brain metastasis of breast cancer. Therefore, it is necessary to conduct a systematic study on BMS of breast cancer, which will not provide ideas and sites for follow-up studies on the treatment and inhibition of BMS. METHODS In this study, data set GSE43837 was screened from gene expression omnibus database, and then R language tool was used for differential analysis of its expression spectrum, The gene ontology functional enrichment and Kyoto encyclopedia of genes and genomes signal pathway enrichment analyses, as well as the interactive gene retrieval tool for hub-gene analysis, were performed. RESULTS According to the findings, the primary genes linked to breast cancer brain metastases are those that involve interactions between cytokines and their respective receptors and between neuroactive ligands and their respective receptors. The majority of the gene ontology enrichment took place in the extracellular structural tissues, the extracellular matrix tissues, and the second message-mediated signaling. We were able to identify 8 genes that are linked to breast cancer spreading to the brain. The gene score for matrix metallopeptidase1 (MMP-1) was the highest among them, and the genes MMP10, tumor necrosis factor alpha-inducible protein 8, collagen type I alpha 2 chain, vascular cell adhesion molecule 1, and TNF superfamily member 11 were all connected to 1 another in an interaction way. CONCLUSIONS There is a possibility that the 8 key genes that were identified in this research are connected to the progression of BMS in breast cancer. Among them, MMP1 is 1 that has the potential to have a role in the diagnosis and treatment of BMS in breast cancer.
Collapse
Affiliation(s)
- Xiao-Gang Li
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Chao Niu
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Ping Lu
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Hong-Wei Wan
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Wen-Di Jin
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Chun-Xiao Wang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Wen-Yuan Mao
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Zhi-Ping Zhang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Wan-Fu Zhang
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
| | - Bo Li
- Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming, China
- * Correspondence: Bo Li, Department of General Surgery, Affiliated Hospital of Yunnan University, Kunming 650021, China (e-mail: )
| |
Collapse
|
9
|
Future Strategies Involving Immune Checkpoint Inhibitors in Advanced Urothelial Carcinoma. Curr Treat Options Oncol 2020; 22:7. [PMID: 33269438 DOI: 10.1007/s11864-020-00799-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Immune checkpoint inhibitors have importantly improved the outcome of patients with urothelial carcinoma. Different immune checkpoint inhibitors are currently approved and used in first- and second-line setting. The multiple agents currently approved in these setting make the choice sometimes difficult for clinicians. Furthermore, only a minority of patients present drastic response and long-term benefit with current immunotherapy. In this review, we describe the current use of immunotherapy in urothelial carcinoma but we also highlight the new strategies of treatment involving immune checkpoint inhibitors; we describe the place of immunotherapy with chemotherapy, targeted agents, and anti-angiogenic agents, incorporating the recent results presented at ASCO 2020. This review explores also the different action mechanisms of immune checkpoint inhibitors and the molecular rational to evaluate these agents in other strategies, such as maintenance and salvage strategies. The new advances in biomarker development are also presented.
Collapse
|
10
|
Chiang CL, Huang HH, Huang TY, Shih YL, Hsieh TY, Lin HH. Nonalcoholic Fatty Liver Disease Associated With Bladder Cancer. Am J Med Sci 2020; 360:161-165. [DOI: 10.1016/j.amjms.2020.04.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/29/2020] [Accepted: 04/24/2020] [Indexed: 12/17/2022]
|
11
|
Lipunova N, Wesselius A, Cheng KK, van Schooten FJ, Cazier JB, Bryan RT, Zeegers MP. Systematic Review: Genetic Associations for Prognostic Factors of Urinary Bladder Cancer. BIOMARKERS IN CANCER 2019; 11:1179299X19897255. [PMID: 31908559 PMCID: PMC6937527 DOI: 10.1177/1179299x19897255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Many germline associations have been reported for urinary bladder cancer (UBC) outcomes and prognostic characteristics. It is unclear whether there are overlapping genetic patterns for various prognostic endpoints. We aimed to review contemporary literature on genetic associations with UBC prognostic outcomes and to identify potential overlap in reported genes. METHODS EMBASE, MEDLINE, and PubMed databases were queried for relevant articles in English language without date restrictions. The initial search identified 1346 articles. After exclusions, 112 studies have been summarized. Cumulatively, 316 single-nucleotide polymorphisms (SNPs) were reported across prognostic outcomes (recurrence, progression, death) and characteristics (tumor stage, grade, size, age, risk group). There were considerable differences between studied outcomes in the context of genetic associations. The most commonly reported SNPs were located in OGG1, TP53, and MDM2. For outcomes with the highest number of reported associations (ie, recurrence and death), functional enrichment annotation yields different terms, potentially indicating separate biological mechanisms. CONCLUSIONS Our study suggests that all UBC prognostic outcomes may have different biological origins with limited overlap. Further validation of these observations is essential to target a phenotype that could best predict patient outcome and advance current management practices.
Collapse
Affiliation(s)
- Nadezda Lipunova
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Complex Genetics, Maastricht University, Maastricht, The Netherlands
- Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - Anke Wesselius
- Department of Complex Genetics, Maastricht University, Maastricht, The Netherlands
| | - Kar K Cheng
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | | | - Jean-Baptiste Cazier
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - Richard T Bryan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Maurice P Zeegers
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Complex Genetics, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
12
|
Critselis E, Rava M, Marquez M, Lygirou V, Chatzicharalambous D, Liapi P, Lichtinghagen R, Brand K, Cecchini L, Vlahou A, Malats N, Zoidakis J. Diagnostic and Prognostic Performance of Secreted Protein Acidic and Rich in Cysteine (SPARC) Assay for Detecting Primary and Recurrent Urinary Bladder Cancer. Proteomics Clin Appl 2019; 13:e1800148. [DOI: 10.1002/prca.201800148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/03/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Elena Critselis
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Marta Rava
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Mirari Marquez
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Vasiliki Lygirou
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Despoina Chatzicharalambous
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Panagiota Liapi
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Ralph Lichtinghagen
- Institute of Clinical ChemistryHannover Medical School Hannover 30625 Germany
| | - Korbinian Brand
- Institute of Clinical ChemistryHannover Medical School Hannover 30625 Germany
| | | | - Antonia Vlahou
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| | - Nuria Malats
- Genetic and Molecular Epidemiology GroupSpanish National Cancer Research Centre (Centro Nacional de Investigaciones Oncologicas) Madrid and CIBERONC 28029 Spain
| | - Jerome Zoidakis
- Proteomics Laboratory, Biotechnology DivisionBiomedical Research Foundation of the Academy of Athens Athens 11527 Greece
| |
Collapse
|
13
|
Zoppino FCM, Guerrero-Gimenez ME, Castro GN, Ciocca DR. Comprehensive transcriptomic analysis of heat shock proteins in the molecular subtypes of human breast cancer. BMC Cancer 2018; 18:700. [PMID: 29954368 PMCID: PMC6022707 DOI: 10.1186/s12885-018-4621-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/20/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Heat Shock Proteins (HSPs), a family of genes with key roles in proteostasis, have been extensively associated with cancer behaviour. However, the HSP family is quite large and many of its members have not been investigated in breast cancer (BRCA), particularly in relation with the current molecular BRCA classification. In this work, we performed a comprehensive transcriptomic study of the HSP gene family in BRCA patients from both The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohorts discriminating the BRCA intrinsic molecular subtypes. METHODS We examined gene expression levels of 1097 BRCA tissue samples retrieved from TCGA and 1981 samples of METABRIC, focusing mainly on the HSP family (95 genes). Data were stratified according to the PAM50 gene expression (Luminal A, Luminal B, HER2, Basal, and Normal-like). Transcriptomic analyses include several statistical approaches: differential gene expression, hierarchical clustering and survival analysis. RESULTS Of the 20,531 analysed genes we found that in BRCA almost 30% presented deregulated expression (19% upregulated and 10% downregulated), while of the HSP family 25% appeared deregulated (14% upregulated and 11% downregulated) (|fold change| > 2 comparing BRCA with normal breast tissues). The study revealed the existence of shared HSP genes deregulated in all subtypes of BRCA while other HSPs were deregulated in specific subtypes. Many members of the Chaperonin subfamily were found upregulated while three members (BBS10, BBS12 and CCTB6) were found downregulated. HSPC subfamily had moderate increments of transcripts levels. Various genes of the HSP70 subfamily were upregulated; meanwhile, HSPA12A and HSPA12B appeared strongly downregulated. The strongest downregulation was observed in several HSPB members except for HSPB1. DNAJ members showed heterogeneous expression pattern. We found that 23 HSP genes correlated with overall survival and three HSP-based transcriptional profiles with impact on disease outcome were recognized. CONCLUSIONS We identified shared and specific HSP genes deregulated in BRCA subtypes. This study allowed the recognition of HSP genes not previously associated with BRCA and/or any cancer type, and the identification of three clinically relevant clusters based on HSPs expression patterns with influence on overall survival.
Collapse
Affiliation(s)
- Felipe C. M. Zoppino
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Martin E. Guerrero-Gimenez
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Gisela N. Castro
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| | - Daniel R. Ciocca
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500 Mendoza, Argentina
| |
Collapse
|
14
|
Esquinas C, Alonso J, Mateo E, Dotor A, Martín A, Dorado J, Arance I, Angulo J. Prospective study comparing laparoscopic and open radical cystectomy: Surgical and oncological results. Actas Urol Esp 2018. [PMID: 28624175 DOI: 10.1016/j.acuro.2017.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Laparoscopic radical cystectomy with lymphadenectomy and urinary diversion is an increasingly widespread operation. Studies are needed to support the oncological effectiveness and safety of this minimally invasive approach. PATIENTS AND METHODS A nonrandomised, comparative prospective study between open radical cystectomy (ORC) and laparoscopic radical cystectomy (LRC) was conducted in a university hospital. The main objective was to compare cancer-specific survival. The secondary objective was to compare the surgical results and complications according to the Clavien-Dindo scale. RESULTS We treated 156 patients with high-grade invasive bladder cancer with either ORC (n=70) or LRC (n=86). The mean follow-up was 33.5±23.8 (range 12-96) months. The mean age was 66.9+9.4 years, and the male to female ratio was 19:1. Both groups were equivalent in age, stage, positive lymph nodes, in situ carcinoma, preoperative obstructive uropathy, adjuvant chemotherapy and type of urinary diversion. There were no differences between the groups in terms of cancer-specific survival (log-rank; P=.71). The histopathology stage was the only independent variable that predicted the prognosis. The hospital stay (P=.01) and operative transfusion rates (P=.002) were less for LRC. The duration of the surgery was greater for LRC (P<.001). There were no differences in the total complications rate (p=.62) or major complications (P=.69). The risk of evisceration (P=.02), surgical wound infection (P=.005) and pneumonia (P=.017) was greater for ORC. The risk of rectal lesion (P=.017) and urethrorectal fistulae (P=.065) was greater for LRC. CONCLUSION LRC is an equivalent treatment to ORC in terms of oncological efficacy and is advantageous in terms of transfusion rates and hospital stays but not in terms of operating room time and overall safety. Studies are needed to better define the specific safety profile for each approach.
Collapse
|
15
|
Rava M, Czachorowski MJ, Silverman D, Márquez M, Kishore S, Tardón A, Serra C, García-Closas M, Garcia-Closas R, Carrato A, Rothman N, Real FX, Kogevinas M, Malats N. Asthma status is associated with decreased risk of aggressive urothelial bladder cancer. Int J Cancer 2018; 142:470-476. [PMID: 28940228 PMCID: PMC6982397 DOI: 10.1002/ijc.31066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/23/2017] [Accepted: 07/31/2017] [Indexed: 12/18/2022]
Abstract
Previous studies suggested an association between atopic conditions and specific cancers. The results on the association with urothelial bladder cancer (UBC) are scarce and inconsistent. To evaluate the association between asthma and risk of UBC, we considered 936 cases and 1,022 controls from the Spanish Bladder Cancer/EPICURO Study (86% males, mean age 65.4 years), a multicenter and hospital-based case-control study conducted during 1998-2001. Participants were asked whether they had asthma and detailed information about occupational exposures, smoking habits, dietary factors, medical conditions and history of medication was collected through face-to-face questionnaires performed by trained interviewers. Since asthma and UBC might share risk factors, association between patients' characteristics and asthma was studied in UBC controls. Association between UBC and asthma was assessed using logistic regression unadjusted and adjusted for potential confounders. The complex interrelationships, direct and mediating effect of asthma on UBC, were appraised using counterfactual mediation models. Asthma was associated with a reduced risk of UBC (odds ratio (OR) = 0.54, 95% confidence interval (CI) 0.37, 0.79) after adjusting for a wide range of confounders. No mediating effect was identified. The reduced risk associated with asthma was restricted to patients with high-risk non-muscle invasive (OR = 0.25, 95%CI 0.10, 0.62) and muscle invasive UBC (OR = 0.32, 95%CI 0.15, 0.69). Our results support that asthma is associated with a decreased risk of UBC, especially among aggressive tumors. Further work on the relationship between asthma and other atopic conditions and cancer risk should shed light on the relationship between immune response mechanisms and bladder carcinogenesis.
Collapse
Affiliation(s)
- Marta Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC, Spain
| | - Maciej J Czachorowski
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC, Spain
| | - Debra Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Mirari Márquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC, Spain
| | - Sirish Kishore
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC, Spain
| | - Adonina Tardón
- Department of Preventive Medicine, Universidad de Oviedo, and CIBERESP, Spain
| | - Consol Serra
- Center for Research in Occupational Health (CiSAL), Universitat Pompeu Fabra, IMIM-Hospital del Mar Medical Research Institut, Barcelona, and CIBERESP, Spain
| | - Montse García-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, USA
| | | | - Alfredo Carrato
- Servicio de Oncología, Hospital Universitario Ramon y Cajal, Madrid, Servicio de Oncología, Hospital Universitario de Elche, and CIBERONC, Spain
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, and CIBERONC, Spain
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL), Parc de Salut Mar, Barcelona, and CIBERESP, Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC, Spain
| |
Collapse
|
16
|
Risk Factors and Molecular Features Associated with Bladder Cancer Development. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-64769-2_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
17
|
López de Maturana E, Malats N. Genetic Testing, Genetic Variation, and Genetic Susceptibility. Bladder Cancer 2018. [DOI: 10.1016/b978-0-12-809939-1.00033-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat Rev Urol 2017; 15:92-111. [PMID: 29133939 DOI: 10.1038/nrurol.2017.179] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Effective management of advanced urothelial bladder cancer is challenging. New discoveries that improve our understanding of molecular bladder cancer subtypes have revealed numerous potentially targetable genomic alterations and demonstrated the efficacy of treatments that harness the immune system. These findings have begun to change paradigms of bladder cancer therapy. For example, DNA repair pathway mutations in genes such as ERCC2, FANCC, ATM, RB1, and others can predict responses to neoadjuvant platinum-based chemotherapies and to targeted therapies on the basis of mutation status. Furthermore, an increasing number of pan-cancer clinical trials (commonly referred to as basket or umbrella trials) are enrolling patients on the basis of molecular and genetic predictors of response. These studies promise to provide improved insight into the true utility of personalized medicine in the treatment of bladder cancer and many other cancer types. Finally, therapies that modulate immune responses have shown great benefit in many cancer types. Several immune checkpoint inhibitors that target programmed cell death protein 1 (PD1), its ligand PDL1, and cytotoxic T lymphocyte-associated protein 4 (CTLA4) have already been approved for use in bladder cancer, representing the most important change to the urological oncologist's tool-kit in over a decade. These advances also provide opportunities for personalization of bladder cancer therapy.
Collapse
|
19
|
Abstract
Bladder cancer is a highly prevalent disease and is associated with substantial morbidity, mortality and cost. Environmental or occupational exposures to carcinogens, especially tobacco, are the main risk factors for bladder cancer. Most bladder cancers are diagnosed after patients present with macroscopic haematuria, and cases are confirmed after transurethral resection of bladder tumour (TURBT), which also serves as the first stage of treatment. Bladder cancer develops via two distinct pathways, giving rise to non-muscle-invasive papillary tumours and non-papillary (solid) muscle-invasive tumours. The two subtypes have unique pathological features and different molecular characteristics. Indeed, The Cancer Genome Atlas project identified genetic drivers of muscle-invasive bladder cancer (MIBC) as well as subtypes of MIBC with distinct characteristics and therapeutic responses. For non-muscle-invasive bladder cancer (NMIBC), intravesical therapies (primarily Bacillus Calmette-Guérin (BCG)) with maintenance are the main treatments to prevent recurrence and progression after initial TURBT; additional therapies are needed for those who do not respond to BCG. For localized MIBC, optimizing care and reducing morbidity following cystectomy are important goals. In metastatic disease, advances in our genetic understanding of bladder cancer and in immunotherapy are being translated into new therapies.
Collapse
|
20
|
Bureau A, Croteau J. Polyunphased: an extension to polytomous outcomes of the Unphased package for family-based genetic association analysis. Stat Appl Genet Mol Biol 2017; 16:75-81. [PMID: 28186960 DOI: 10.1515/sagmb-2016-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Polytomous phenotypes arise when a disease has multiple subtypes or when two dichotomous phenotypes are analyzed simultaneously. Few software programs offer the option to analyze such phenotypes in family studies, and none implements conditional polytomous logistic regression for within-family analysis robust to population stratification. We introduce Polyunphased, an extension to polytomous phenotypes of the Unphased package, a flexible software tool for genetic association analysis in nuclear families. Like Unphased, Polyunphased is written in C++ and runs from the command line or from a Java graphical user interface. Most Unphased options remain available in Polyunphased, including those handling missing parental genotypes while preserving robustness to population stratification, and the modelling options. Simulation studies confirmed the expected statistical behaviour of the maximum likelihood estimates of the association parameters of the conditional logistic regression model when the corresponding association parameters in the parental term of the likelihood function are set to 0, but revealed convergence problems when estimating these parental association parameters separately. The former approach is thus recommended with polytomous phenotypes.
Collapse
|
21
|
Verma M. Genome-wide association studies and epigenome-wide association studies go together in cancer control. Future Oncol 2016; 12:1645-64. [PMID: 27079684 PMCID: PMC5551540 DOI: 10.2217/fon-2015-0035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/22/2016] [Indexed: 02/07/2023] Open
Abstract
Completion of the human genome a decade ago laid the foundation for: using genetic information in assessing risk to identify individuals and populations that are likely to develop cancer, and designing treatments based on a person's genetic profiling (precision medicine). Genome-wide association studies (GWAS) completed during the past few years have identified risk-associated single nucleotide polymorphisms that can be used as screening tools in epidemiologic studies of a variety of tumor types. This led to the conduct of epigenome-wide association studies (EWAS). This article discusses the current status, challenges and research opportunities in GWAS and EWAS. Information gained from GWAS and EWAS has potential applications in cancer control and treatment.
Collapse
Affiliation(s)
- Mukesh Verma
- Methods & Technologies Branch, Epidemiology & Genomics Research Program, Division of Cancer Control & Population Sciences, National Cancer Institute (NCI), NIH, 9609 Medical Center Drive, Suite 4E102, Rockville, MD 20850, USA
| |
Collapse
|
22
|
Wen Y, Lu Q. A Clustered Multiclass Likelihood-Ratio Ensemble Method for Family-Based Association Analysis Accounting for Phenotypic Heterogeneity. Genet Epidemiol 2016; 40:512-9. [PMID: 27321816 DOI: 10.1002/gepi.21987] [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: 12/03/2015] [Revised: 05/04/2016] [Accepted: 05/08/2016] [Indexed: 12/24/2022]
Abstract
Although compelling evidence suggests that the genetic etiology of complex diseases could be heterogeneous in subphenotype groups, little attention has been paid to phenotypic heterogeneity in genetic association analysis of complex diseases. Simply ignoring phenotypic heterogeneity in association analysis could result in attenuated estimates of genetic effects and low power of association tests if subphenotypes with similar clinical manifestations have heterogeneous underlying genetic etiologies. To facilitate the family-based association analysis allowing for phenotypic heterogeneity, we propose a clustered multiclass likelihood-ratio ensemble (CMLRE) method. The proposed method provides an alternative way to model the complex relationship between disease outcomes and genetic variants. It allows for heterogeneous genetic causes of disease subphenotypes and can be applied to various pedigree structures. Through simulations, we found CMLRE outperformed the commonly adopted strategies in a variety of underlying disease scenarios. We further applied CMLRE to a family-based dataset from the International Consortium to Identify Genes and Interactions Controlling Oral Clefts (ICOC) to investigate the genetic variants and interactions predisposing to subphenotypes of oral clefts. The analysis suggested that two subphenotypes, nonsyndromic cleft lip without palate (CL) and cleft lip with palate (CLP), shared similar genetic etiologies, while cleft palate only (CP) had its own genetic mechanism. The analysis further revealed that rs10863790 (IRF6), rs7017252 (8q24), and rs7078160 (VAX1) were jointly associated with CL/CLP, while rs7969932 (TBK1), rs227731 (17q22), and rs2141765 (TBK1) jointly contributed to CP.
Collapse
Affiliation(s)
- Yalu Wen
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Qing Lu
- Department of Epidemiology and Biostatics, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
23
|
Patient-derived bladder cancer xenografts in the preclinical development of novel targeted therapies. Oncotarget 2016; 6:21522-32. [PMID: 26041878 PMCID: PMC4673283 DOI: 10.18632/oncotarget.3974] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/15/2015] [Indexed: 01/03/2023] Open
Abstract
Optimal animal models of muscle invasive bladder cancer (MIBC) are necessary to overcome the current lack of novel targeted therapies for this malignancy. Here we report on the establishment and characterization of patient-derived primary xenografts (PDX). Patient tumors were grafted under the renal capsule of mice and subsequently transplanted over multiple generations. Patient tumor and PDX were processed for analysis of copy number variations by aCGH, gene expression by microarray, and expression of target pathways by immunohistochemistry (IHC). One PDX harbouring an FGFR3 mutation was treated with an inhibitory monoclonal antibody targeting FGFR3. Five PDX were successfully established. Tumor doubling time ranged from 5 to 11 days. Array CGH revealed shared chromosomal aberrations in the patient tumors and PDX. Gene expression microarray and IHC confirmed that PDXs maintain similar patterns to the parental tumors. Tumor growth in the PDX with an FGFR3 mutation was inhibited by the FGFR3 inhibitor. PDXs recapitulate the tumor biology of the patients' primary tumors from which they are derived. Investigations related to tumor biology and drug testing in these models are therefore more likely to be relevant to the disease state in patients. They represent a valuable tool for developing precision therapy in MIBC.
Collapse
|
24
|
Camarena V, Wang G. The epigenetic role of vitamin C in health and disease. Cell Mol Life Sci 2016; 73:1645-58. [PMID: 26846695 PMCID: PMC4805483 DOI: 10.1007/s00018-016-2145-x] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/28/2015] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
Abstract
Recent advances have uncovered a previously unknown function of vitamin C in epigenetic regulation. Vitamin C exists predominantly as an ascorbate anion under physiological pH conditions. Ascorbate was discovered as a cofactor for methylcytosine dioxygenases that are responsible for DNA demethylation, and also as a likely cofactor for some JmjC domain-containing histone demethylases that catalyze histone demethylation. Variation in ascorbate bioavailability thus can influence the demethylation of both DNA and histone, further leading to different phenotypic presentations. Ascorbate deficiency can be presented systematically, spatially and temporally in different tissues at the different stages of development and aging. Here, we review how ascorbate deficiency could potentially be involved in embryonic and postnatal development, and plays a role in various diseases such as neurodegeneration and cancer through epigenetic dysregulation.
Collapse
Affiliation(s)
- Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building, Rm. 608, 1501 NW 10th Ave, Miami, FL, 33136, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building, Rm. 608, 1501 NW 10th Ave, Miami, FL, 33136, USA.
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
25
|
Wu H, Wang X, Zhang L, Mo N, Lv Z. Association Between N-acetyltransferase 2 Polymorphism and Bladder Cancer Risk: Results From Studies of the Past Decade and a Meta-Analysis. Clin Genitourin Cancer 2016; 14:122-9. [DOI: 10.1016/j.clgc.2015.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/04/2015] [Accepted: 10/17/2015] [Indexed: 12/24/2022]
|
26
|
Vukovic V, Ianuale C, Leoncini E, Pastorino R, Gualano MR, Amore R, Boccia S. Lack of association between polymorphisms in the CYP1A2 gene and risk of cancer: evidence from meta-analyses. BMC Cancer 2016; 16:83. [PMID: 26865042 PMCID: PMC4750358 DOI: 10.1186/s12885-016-2096-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 01/28/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Polymorphisms in the CYP1A2 genes have the potential to affect the individual capacity to convert pre-carcinogens into carcinogens. With these comprehensive meta-analyses, we aimed to provide a quantitative assessment of the association between the published genetic association studies on CYP1A2 single nucleotide polymorphisms (SNPs) and the risk of cancer. METHODS We searched MEDLINE, ISI Web of Science and SCOPUS bibliographic online databases and databases of genome-wide association studies (GWAS). After data extraction, we calculated Odds Ratios (ORs) and 95% confidence intervals (CIs) for the association between the retrieved CYP1A2 SNPs and cancer. Random effect model was used to calculate the pooled ORs. Begg and Egger tests, one-way sensitivity analysis were performed, when appropriate. We conducted stratified analyses by study design, sample size, ethnicity and tumour site. RESULTS Seventy case-control studies and one GWA study detailing on six different SNPs were included. Among the 71 included studies, 42 were population-based case-control studies, 28 hospital-based case-control studies and one genome-wide association study, including total of 47,413 cancer cases and 58,546 controls. The meta-analysis of 62 studies on rs762551, reported an OR of 1.03 (95% CI, 0.96-1.12) for overall cancer (P for heterogeneity < 0.01; I(2) = 50.4%). When stratifying for tumour site, an OR of 0.84 (95% CI, 0.70-1.01; P for heterogeneity = 0.23, I(2) = 28.5%) was reported for bladder cancer for those homozygous mutant of rs762551. An OR of 0.79 (95% CI, 0.65-0.95; P for heterogeneity = 0.09, I(2) = 58.1%) was obtained for the bladder cancer from the hospital-based studies and on Caucasians. CONCLUSIONS This large meta-analysis suggests no significant effect of the investigated CYP1A2 SNPs on cancer overall risk under various genetic models. However, when stratifying according to the tumour site, our results showed a borderline not significant OR of 0.84 (95% CI, 0.70-1.01) for bladder cancer for those homozygous mutant of rs762551. Due to the limitations of our meta-analyses, the results should be interpreted with attention and need to be further confirmed by high-quality studies, for all the potential CYP1A2 SNPs.
Collapse
Affiliation(s)
- Vladimir Vukovic
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy.
| | - Carolina Ianuale
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| | - Emanuele Leoncini
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| | - Roberta Pastorino
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| | - Maria Rosaria Gualano
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| | - Rosarita Amore
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| | - Stefania Boccia
- Institute of Public Health- Section of Hygiene, Università Cattolica del Sacro Cuore, Largo F.Vito 1, 00168, Rome, Italy
| |
Collapse
|
27
|
Zhang K, Zhang L, Zhou J, Hao Z, Fan S, Yang C, Liang C. Association between interleukin-6 polymorphisms and urinary system cancer risk: evidence from a meta-analysis. Onco Targets Ther 2016; 9:567-77. [PMID: 26869801 PMCID: PMC4734788 DOI: 10.2147/ott.s94348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Interleukin-6 (IL-6) is a multifunctional proinflammatory cytokine involved in cancer initiation and progression. Numerous studies have investigated the associations between IL-6 polymorphisms (IL-6 −174G>C, −592G>C, −597G>A) and risk of urinary system cancers, including prostate cancer, bladder cancer, and renal cell cancer. However, conclusions from these studies were controversial. Thus, we conducted the current meta-analysis to obtain the comprehensive profile regarding the association between IL-6 polymorphisms and urinary system cancer risk. Methods According to inclusion and exclusion criteria, the associations of IL-6 polymorphisms with urinary system cancer were searched from database and analyzed using STATA 12.0 statistical software. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of the associations. Results A total of 20 previous publications consisting of 15,033 cases and 17,655 controls were involved in this meta-analysis. Significant association was observed in overall population regarding IL-6 −592G>C polymorphisms (G vs C: OR =0.1.30, 95% CI =1.13−2.52; GG vs CC: OR =1.81, 95% CI =1.31−2.52; GG vs GC + CC: OR =1.33, 95% CI =1.02−1.75; GG + GC vs CC: OR =1.41, 95% CI =1.09−1.83). In the stratified analyses by ethnicity, the significant associations were found among Asian (GG vs CC: OR =1.89, 95% CI =1.34−2.66; GG + GC vs CC: OR =1.43, 95% CI =1.09−1.87) and Black population (GC vs CC: OR =0.20, 95% CI =0.05−0.82) rather than Caucasian men. Likewise, there were noticeable associations in almost all the other subanalyses such as cancer types, control sources, genotyped methods, and sample sizes. However, no significant associations were identified between any of IL-6 −174G>C polymorphisms with urinary system cancer, except for Asian population (G vs C: OR =0.81, 95% CI =0.70−0.95; GG vs CC: OR =0.51, 95% CI =0.35−0.74; GC vs CC: OR =0.49, 95% CI =0.33−0.72; GG + GC vs CC: OR =0.50, 95% CI =0.35−0.72; respectively). In addition, no significant associations were detected between IL-6 −597G>A polymorphism and urinary system cancer, regardless of whole or subgroups. Conclusion This meta-analysis presents a relatively comprehensive view of the associations between IL-6 polymorphism and urinary system cancer risk to explore the carcinogenic mechanisms, which will help shed light on the clinical diagnosis and therapy for urinary system cancer. However, further detailed studies are needed to verify our conclusion.
Collapse
Affiliation(s)
- Kaiping Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Cheng Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
28
|
Angulo JC, García-Tello A, Mateo E, Gimbernat H, Redondo C, Andrés G. Two-Port Approach Compared to Standard Laparoscopic Radical Cystectomy. J Endourol 2015; 29:1030-7. [PMID: 26115458 DOI: 10.1089/end.2015.0161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Two-port laparoscopic radical cystectomy (LRC) using a multichannel platform through the umbilicus and one additional 10-mm in the right iliac fossa has been recently described. We compare the perioperative and early results of this technique with a cohort of patients simultaneously treated in our institution with four-port LRC and a 7 to 10 cm midline incision. MATERIALS AND METHODS A matched-pair study comparing perioperative outcomes, postoperative visual analog pain scale (VAPS), and morbidity of two-port (n=30) and four-port (n=30) LRC. Preoperative and perioperative data analyzed and compared included demographics, type of urinary diversion, operative time, blood loss, transfusion requirements, decrease in serum hemoglobin, operative complications, analgesic needs, length of stay, number of nodes removed, and other pathologic information. Complications in the first 3 months were evaluated according to the Clavien-Dindo classification. RESULTS There was no significant difference between the two groups regarding patient age, gender, body-mass index, American Society of Anesthesiologists (ASA) score, Charlson comorbidity index, tumor stage, percent of positive nodes, positive margin rate, number of nodes retrieved, proportion of neobladders, secondary carcinoma in situ, incidental prostate cancer, or need of systemic chemotherapy. Median operative time was 20 minutes higher in the two-port procedure, but this difference was not statistically significant (p=0.2). Estimated blood loss and differential hemoglobin change were lower in the two-port technique (each p=0.03), but no difference was detected regarding the intraoperative or postoperative transfusion rate. Length of hospital stay was also equivalent. Differences were not observed either in the number or severity of complications. VAPS at days 2, 3, and 5 was significantly lower for two-port LRC (p<0.01). CONCLUSIONS Two-port LRC performed through an umbilical platform and an accessory 10-mm port seems technically equivalent to standard four-port LRC. Perioperative outcomes regarding hospital stay and complications are tantamount. Diminished blood loss and, especially, less postoperative pain favor the reduced-port approach in this complex urologic minimally invasive procedure.
Collapse
Affiliation(s)
- Javier C Angulo
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| | - Ana García-Tello
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| | - Erika Mateo
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| | - Helena Gimbernat
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| | - Cristina Redondo
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| | - Guillermo Andrés
- Servicio de Urología, Fundación para la Investigación Biomédica del Hospital Universitario de Getafe , Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
29
|
Abstract
Emerging evidence suggests that ascorbate, the dominant form of vitamin C under physiological pH conditions, influences activity of the genome via regulating epigenomic processes. Ascorbate serves as a cofactor for Ten-eleven translocation (TET) dioxygenases that catalyze the oxidation of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC), and further to 5-formylcytosine (5fC) and to 5-carboxylcytosine (5caC), which are ultimately replaced by unmodified cytosine. The Jumonji C (JmjC)-domain-containing histone demethylases also require ascorbate as a cofactor for histone demethylation. Thus, by primarily participating in the demethylation of both DNA and histones, ascorbate appears to be a mediator of the interface between the genome and environment. Furthermore, redox status has a profound impact on the bioavailability of ascorbate in the nucleus. In order to bridge the gap between redox biology and genomics, we suggest an interdisciplinary research field that can be termed redox genomics to study dynamic redox processes in health and diseases. This review examines the evidence and potential molecular mechanism of ascorbate in the demethylation of the genome, and it highlights potential epigenetic roles of ascorbate in various diseases.
Collapse
|
30
|
Yang H, Li J, Tang R, Liu Y, Shao Y, Huang Q, Shi J. Telomere reverse transcriptase (TERT) rs2735940 increases cancer risk. Med Sci Monit 2015; 21:612-6. [PMID: 25716168 PMCID: PMC4354426 DOI: 10.12659/msm.893087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Telomerase reverse transcriptase (TERT) rs2735940 polymorphism was found to be associated with increased cancer risk. However, recent studies reported controversial results. The aim of our study was to detect its relationship with cancer risk. Material/Methods EMBASE and PubMed databases were searched for all publications until October 2014. ORs and 95% CIs were applied to investigate the association in the random-effects model. Results Thirteen case-control studies with 19385 cases and 17558 controls were included in this study. We found a significant association between cancer risk and TERT rs2735940 polymorphism (OR=1.06, 95% CI 1.02–1.11, P=0.005). In the subgroup analysis by ethnicity, a marginal association was found in Caucasians (OR=1.05, 95% CI 1.00–1.10, P=0.05), but not in Asians (OR=1.01, 95% CI 0.82–1.24, P=0.93). In the subgroup analysis by cancer site, this polymorphism was significantly associated with lung cancer risk (OR=1.08, 95% CI 1.02–1.13, P=0.004). Conclusions TERT rs2735940 polymorphism was significantly associated with cancer risk, especially lung cancer.
Collapse
Affiliation(s)
- Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Jing Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Ruihua Tang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Yaxiong Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Yan Shao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Qingsheng Huang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| | - Junling Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, China (mainland)
| |
Collapse
|
31
|
Angulo JC, Cáceres F, Cabrera PM, García-Tello A, Arance I, Romero I. Two-port Laparoscopic Radical Cystectomy With Reusable Umbilical System: A Feasibility Study. Urology 2014; 84:1088-93. [DOI: 10.1016/j.urology.2014.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/07/2014] [Accepted: 07/07/2014] [Indexed: 10/24/2022]
|
32
|
Fu YP, Kohaar I, Moore LE, Lenz P, Figueroa JD, Tang W, Porter-Gill P, Chatterjee N, Scott-Johnson A, Garcia-Closas M, Muchmore B, Baris D, Paquin A, Ylaya K, Schwenn M, Apolo AB, Karagas MR, Tarway M, Johnson A, Mumy A, Schned A, Guedez L, Jones MA, Kida M, Hosain GMM, Malats N, Kogevinas M, Tardon A, Serra C, Carrato A, Garcia-Closas R, Lloreta J, Wu X, Purdue M, Andriole GL, Grubb RL, Black A, Landi MT, Caporaso NE, Vineis P, Siddiq A, Bueno-de-Mesquita HB, Trichopoulos D, Ljungberg B, Severi G, Weiderpass E, Krogh V, Dorronsoro M, Travis RC, Tjønneland A, Brennan P, Chang-Claude J, Riboli E, Prescott J, Chen C, De Vivo I, Govannucci E, Hunter D, Kraft P, Lindstrom S, Gapstur SM, Jacobs EJ, Diver WR, Albanes D, Weinstein SJ, Virtamo J, Kooperberg C, Hohensee C, Rodabough RJ, Cortessis VK, Conti DV, Gago-Dominguez M, Stern MC, Pike MC, Van Den Berg D, Yuan JM, Haiman CA, Cussenot O, Cancel-Tassin G, Roupret M, Comperat E, Porru S, Carta A, Pavanello S, Arici C, Mastrangelo G, Grossman HB, Wang Z, Deng X, Chung CC, Hutchinson A, Burdette L, Wheeler W, Fraumeni J, Chanock SJ, Hewitt SM, Silverman DT, Rothman N, Prokunina-Olsson L. The 19q12 bladder cancer GWAS signal: association with cyclin E function and aggressive disease. Cancer Res 2014; 74:5808-18. [PMID: 25320178 PMCID: PMC4203382 DOI: 10.1158/0008-5472.can-14-1531] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A genome-wide association study (GWAS) of bladder cancer identified a genetic marker rs8102137 within the 19q12 region as a novel susceptibility variant. This marker is located upstream of the CCNE1 gene, which encodes cyclin E, a cell-cycle protein. We performed genetic fine-mapping analysis of the CCNE1 region using data from two bladder cancer GWAS (5,942 cases and 10,857 controls). We found that the original GWAS marker rs8102137 represents a group of 47 linked SNPs (with r(2) ≥ 0.7) associated with increased bladder cancer risk. From this group, we selected a functional promoter variant rs7257330, which showed strong allele-specific binding of nuclear proteins in several cell lines. In both GWASs, rs7257330 was associated only with aggressive bladder cancer, with a combined per-allele OR = 1.18 [95% confidence interval (CI), 1.09-1.27, P = 4.67 × 10(-5)] versus OR = 1.01 (95% CI, 0.93-1.10, P = 0.79) for nonaggressive disease, with P = 0.0015 for case-only analysis. Cyclin E protein expression analyzed in 265 bladder tumors was increased in aggressive tumors (P = 0.013) and, independently, with each rs7257330-A risk allele (P(trend) = 0.024). Overexpression of recombinant cyclin E in cell lines caused significant acceleration of cell cycle. In conclusion, we defined the 19q12 signal as the first GWAS signal specific for aggressive bladder cancer. Molecular mechanisms of this genetic association may be related to cyclin E overexpression and alteration of cell cycle in carriers of CCNE1 risk variants. In combination with established bladder cancer risk factors and other somatic and germline genetic markers, the CCNE1 variants could be useful for inclusion into bladder cancer risk prediction models.
Collapse
Affiliation(s)
- Yi-Ping Fu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Indu Kohaar
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lee E Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Petra Lenz
- Clinical Research Directorate/Clinical Monitoring Research Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Tang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Patricia Porter-Gill
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nilanjan Chatterjee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alexandra Scott-Johnson
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Brian Muchmore
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dalsu Baris
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ashley Paquin
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kris Ylaya
- Laboratory of Pathology, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Andrea B Apolo
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | - McAnthony Tarway
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Adam Mumy
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alan Schned
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Liliana Guedez
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael A Jones
- Department of Pathology and Laboratory Medicine, Maine Medical Center, Portland, Maine
| | - Masatoshi Kida
- Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont
| | | | - Nuria Malats
- Spanish National Cancer Research Centre, Madrid, Spain
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain. Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain. National School of Public Health, Athens, Greece. CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - Adonina Tardon
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain. Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
| | - Consol Serra
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain. Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Reina Garcia-Closas
- Unidad de Investigación, Hospital Universitario de Canarias, La Laguna, Spain
| | - Josep Lloreta
- Hospital del Mar-IMIM, Univesitat Pompeu Fabra, Barcelona, Spain
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Mark Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Gerald L Andriole
- Division of Urologic Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert L Grubb
- Division of Urologic Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria T Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Paolo Vineis
- School of Public Health, Imperial College London, London, United Kingdom. Human Genetics Foundation (HuGeF), Torino, Italy
| | - Afshan Siddiq
- School of Public Health, Imperial College London, London, United Kingdom
| | - H Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands. Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands. Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom. Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Dimitrios Trichopoulos
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts. Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece. Hellenic Health Foundation, Athens, Greece
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Gianluca Severi
- Human Genetics Foundation (HuGeF), Torino, Italy. Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Australia. Centre for Epidemiology and Biostatistics, University of Melbourne, Australia
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway. Department of Research, Cancer Registry of Norway, Oslo, Norway. Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden. Samfundet Folkhälsan, Helsinki, Finland
| | - Vittorio Krogh
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Miren Dorronsoro
- Public Health Division of Gipuzkoa, Basque Regional Health Department and Ciberesp-Biodonostia, San Sebastian, Spain
| | - Ruth C Travis
- Cancer Epidemiology Unit, University of Oxford, Oxford, United Kingdom
| | | | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elio Riboli
- School of Public Health, Imperial College London, London, United Kingdom
| | - Jennifer Prescott
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Constance Chen
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Edward Govannucci
- Department of Nutrition and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - David Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Sara Lindstrom
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - W Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jarmo Virtamo
- National Institute for Health and Welfare, Helsinki, Finland
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, Washington
| | - Chancellor Hohensee
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, Washington
| | - Rebecca J Rodabough
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, Washington
| | - Victoria K Cortessis
- Department of Obstetrics and Gynecology, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California. Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - David V Conti
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Manuela Gago-Dominguez
- Genomic Medicine Group, Galician Foundation of Genomic Medicine, Complejo Hospitalario Universitario de Santiago, Servicio Galego de Saude (SERGAS), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Mariana C Stern
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Malcolm C Pike
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - David Van Den Berg
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Jian-Min Yuan
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Olivier Cussenot
- AP-HP, Hopital Tenon, GHU-Est, Department of Urology, Paris, France. Centre de Recherche sur les Pathologies Prostatiques, Paris, France. UPMC Univ Paris 06, ONCOTYPE-URO, Paris, France
| | - Geraldine Cancel-Tassin
- Centre de Recherche sur les Pathologies Prostatiques, Paris, France. UPMC Univ Paris 06, ONCOTYPE-URO, Paris, France
| | - Morgan Roupret
- Centre de Recherche sur les Pathologies Prostatiques, Paris, France. UPMC Univ Paris 06, ONCOTYPE-URO, Paris, France. AP-HP, Hopital Pitie-Salpetriere, GHU-Est, Departments of Urology and Pathology, Paris, France
| | - Eva Comperat
- Centre de Recherche sur les Pathologies Prostatiques, Paris, France. UPMC Univ Paris 06, ONCOTYPE-URO, Paris, France. AP-HP, Hopital Pitie-Salpetriere, GHU-Est, Departments of Urology and Pathology, Paris, France
| | - Stefano Porru
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Angela Carta
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padua, Italy
| | - Cecilia Arici
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppe Mastrangelo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padua, Italy
| | - H Barton Grossman
- Department of Urology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Zhaoming Wang
- Cancer Genomics Research Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Xiang Deng
- Cancer Genomics Research Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Charles C Chung
- Cancer Genomics Research Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Amy Hutchinson
- Cancer Genomics Research Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Laurie Burdette
- Cancer Genomics Research Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | - Joseph Fraumeni
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen M Hewitt
- Laboratory of Pathology, Center of Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ludmila Prokunina-Olsson
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
33
|
Bureau A, Croteau J, Chagnon YC, Roy MA, Maziade M. Extension of the generalized disequilibrium test to polytomous phenotypes and two-locus models. Front Genet 2014; 5:258. [PMID: 25152751 PMCID: PMC4126369 DOI: 10.3389/fgene.2014.00258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/14/2014] [Indexed: 12/04/2022] Open
Abstract
We extend the usual logistic model between a dichotomous phenotype and an allele count in two ways: a polytomous phenotype with K > 2 levels, and modeling of allele counts at two unlinked marker loci. Inference is based on within-family information to guard against potential bias due to population genetic structure. Score tests of the model coefficients taking into account the correlation between relatives in entire pedigrees are derived as an extension of the Generalized Disequilibrium Test (GDT). Simulations confirm that the tests have the expected statistical properties, and that their power exceeds that of the GDT under a favorable scenario. The score tests are illustrated with candidate genetic markers, a major psychosis phenotype and a cognitive endophenotype in large kindreds from Eastern Quebec.
Collapse
Affiliation(s)
- Alexandre Bureau
- Département de Médecine Sociale et Préventive, Université Laval Québec, QC, Canada ; Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Québec, QC, Canada
| | - Jordie Croteau
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Québec, QC, Canada
| | - Yvon C Chagnon
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Québec, QC, Canada
| | - Marc-André Roy
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Québec, QC, Canada ; Département de Psychiatrie et Neurosciences, Université Laval Québec, QC, Canada
| | - Michel Maziade
- Centre de Recherche de L'Institut Universitaire en Santé Mentale de Québec Québec, QC, Canada ; Département de Psychiatrie et Neurosciences, Université Laval Québec, QC, Canada
| |
Collapse
|
34
|
Pineda S, Milne RL, Calle ML, Rothman N, López de Maturana E, Herranz J, Kogevinas M, Chanock SJ, Tardón A, Márquez M, Guey LT, García-Closas M, Lloreta J, Baum E, González-Neira A, Carrato A, Navarro A, Silverman DT, Real FX, Malats N. Genetic variation in the TP53 pathway and bladder cancer risk. a comprehensive analysis. PLoS One 2014; 9:e89952. [PMID: 24818791 PMCID: PMC4018346 DOI: 10.1371/journal.pone.0089952] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/24/2014] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Germline variants in TP63 have been consistently associated with several tumors, including bladder cancer, indicating the importance of TP53 pathway in cancer genetic susceptibility. However, variants in other related genes, including TP53 rs1042522 (Arg72Pro), still present controversial results. We carried out an in depth assessment of associations between common germline variants in the TP53 pathway and bladder cancer risk. MATERIAL AND METHODS We investigated 184 tagSNPs from 18 genes in 1,058 cases and 1,138 controls from the Spanish Bladder Cancer/EPICURO Study. Cases were newly-diagnosed bladder cancer patients during 1998-2001. Hospital controls were age-gender, and area matched to cases. SNPs were genotyped in blood DNA using Illumina Golden Gate and TaqMan assays. Cases were subphenotyped according to stage/grade and tumor p53 expression. We applied classical tests to assess individual SNP associations and the Least Absolute Shrinkage and Selection Operator (LASSO)-penalized logistic regression analysis to assess multiple SNPs simultaneously. RESULTS Based on classical analyses, SNPs in BAK1 (1), IGF1R (5), P53AIP1 (1), PMAIP1 (2), SERINPB5 (3), TP63 (3), and TP73 (1) showed significant associations at p-value≤0.05. However, no evidence of association, either with overall risk or with specific disease subtypes, was observed after correction for multiple testing (p-value≥0.8). LASSO selected the SNP rs6567355 in SERPINB5 with 83% of reproducibility. This SNP provided an OR = 1.21, 95%CI 1.05-1.38, p-value = 0.006, and a corrected p-value = 0.5 when controlling for over-estimation. DISCUSSION We found no strong evidence that common variants in the TP53 pathway are associated with bladder cancer susceptibility. Our study suggests that it is unlikely that TP53 Arg72Pro is implicated in the UCB in white Europeans. SERPINB5 and TP63 variation deserve further exploration in extended studies.
Collapse
Affiliation(s)
- Silvia Pineda
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Roger L. Milne
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - M. Luz Calle
- Systems Biology Department, University of Vic, Vic, Spain
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, United States of America
| | | | - Jesús Herranz
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Manolis Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Institut Municipal d'Investigació Mèdica – Hospital del Mar, Barcelona, Spain
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, United States of America
| | - Adonina Tardón
- Department of Preventive Medicine, Universidad de Oviedo, Oviedo, Spain
| | - Mirari Márquez
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Lin T. Guey
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Montserrat García-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, United States of America
| | - Josep Lloreta
- Institut Municipal d'Investigació Mèdica – Hospital del Mar, Barcelona, Spain
- Departament de Patologia, Hospital del Mar – IMAS, Barcelona, Spain
| | - Erin Baum
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Alfredo Carrato
- Servicio de Oncología, Hospital Universitario de Elche, Elche, Spain
- Servicio de Oncología, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Arcadi Navarro
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
- Institut de Biologia Evolutiva (UPF-CSIC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Instituto Nacional de Bioinformática, Barcelona, Spain
| | - Debra T. Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Department of Health and Human Services, Bethesda, Maryland, United States of America
| | - Francisco X. Real
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Núria Malats
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
- * E-mail:
| |
Collapse
|
35
|
de Maturana EL, Chanok SJ, Picornell AC, Rothman N, Herranz J, Calle ML, García-Closas M, Marenne G, Brand A, Tardón A, Carrato A, Silverman DT, Kogevinas M, Gianola D, Real FX, Malats N. Whole genome prediction of bladder cancer risk with the Bayesian LASSO. Genet Epidemiol 2014; 38:467-76. [PMID: 24796258 DOI: 10.1002/gepi.21809] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/05/2014] [Accepted: 03/20/2014] [Indexed: 11/11/2022]
Abstract
To build a predictive model for urothelial carcinoma of the bladder (UCB) risk combining both genomic and nongenomic data, 1,127 cases and 1,090 controls from the Spanish Bladder Cancer/EPICURO study were genotyped using the HumanHap 1M SNP array. After quality control filters, genotypes from 475,290 variants were available. Nongenomic information comprised age, gender, region, and smoking status. Three Bayesian threshold models were implemented including: (1) only genomic information, (2) only nongenomic data, and (3) both sources of information. The three models were applied to the whole population, to only nonsmokers, to male smokers, and to extreme phenotypes to potentiate the UCB genetic component. The area under the ROC curve allowed evaluating the predictive ability of each model in a 10-fold cross-validation scenario. Smoking status showed the highest predictive ability of UCB risk (AUCtest = 0.62). On the other hand, the AUC of all genetic variants was poorer (0.53). When the extreme phenotype approach was applied, the predictive ability of the genomic model improved 15%. This study represents a first attempt to build a predictive model for UCB risk combining both genomic and nongenomic data and applying state-of-the-art statistical approaches. However, the lack of genetic relatedness among individuals, the complexity of UCB etiology, as well as a relatively small statistical power, may explain the low predictive ability for UCB risk. The study confirms the difficulty of predicting complex diseases using genetic data, and suggests the limited translational potential of findings from this type of data into public health interventions.
Collapse
|
36
|
Nielsen ME, Smith AB, Meyer AM, Kuo TM, Tyree S, Kim WY, Milowsky MI, Pruthi RS, Millikan RC. Trends in stage-specific incidence rates for urothelial carcinoma of the bladder in the United States: 1988 to 2006. Cancer 2013; 120:86-95. [PMID: 24122346 DOI: 10.1002/cncr.28397] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/21/2013] [Accepted: 08/26/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND Bladder cancer is notable for a striking heterogeneity of disease-specific risks. Among the approximately 75% of incident cases found to be superficial to the muscularis propria at the time of presentation (non-muscle-invasive bladder cancer), the risk of progression to the lethal phenotype of muscle-invasive disease is strongly associated with stage and grade of disease. Given the suggestion of an increasing percentage of low-risk cases in hospital-based registry data in recent years, the authors hypothesized that population-based data may reveal changes in the stage distribution of early-stage cases. METHODS Surveillance, Epidemiology, and End Results (SEER) data were used to examine trends for the stage-specific incidence of bladder cancer between 1988 and 2006, adjusted for age, race, and sex, using Joinpoint and nonparametric tests. RESULTS The adjusted incidence rate of papillary noninvasive (Ta) predominantly low grade (77%) disease was found to increase from 5.52 to 9.09 per 100,000 population (P < .0001), with an average annual percentage change of +3.3. Over the same period, concomitant, albeit smaller, decreases were observed for flat in situ (Tis) and lamina propria-invasive (T1) disease (2.57 to 1.19 and 6.65 to 4.61 per 100,000 population [both P < .0001]; average annual percent change of -5.0 and -1.6, respectively). The trend was most dramatic among patients in the oldest age strata, suggesting a previously unappreciated cohort phenomenon. CONCLUSIONS The findings of the current study should motivate further epidemiological investigations of differential associations of genetic and environmental factors with different bladder cancer phenotypes as well as further scrutiny of clinical practice guideline recommendations for the growing subgroup of predominantly older patients with lower-risk disease.
Collapse
Affiliation(s)
- Matthew E Nielsen
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina; Department of Urology, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wen Y, Lu Q. A multiclass likelihood ratio approach for genetic risk prediction allowing for phenotypic heterogeneity. Genet Epidemiol 2013; 37:715-25. [PMID: 23934726 DOI: 10.1002/gepi.21751] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 06/09/2013] [Accepted: 07/03/2013] [Indexed: 01/04/2023]
Abstract
The translation of human genome discoveries into health practice is one of the major challenges in the coming decades. The use of emerging genetic knowledge for early disease prediction, prevention, and pharmacogenetics will advance genome medicine and lead to more effective prevention/treatment strategies. For this reason, studies to assess the combined role of genetic and environmental discoveries in early disease prediction represent high priority research projects, as manifested in the multiple risk prediction studies now underway. However, the risk prediction models formed to date lack sufficient accuracy for clinical use. Converging evidence suggests that diseases with the same or similar clinical manifestations could have different pathophysiological and etiological processes. When heterogeneous subphenotypes are treated as a single entity, the effect size of predictors can be reduced substantially, leading to a low-accuracy risk prediction model. The use of more refined subphenotypes facilitates the identification of new predictors and leads to improved risk prediction models. To account for the phenotypic heterogeneity, we have developed a multiclass likelihood-ratio approach, which simultaneously determines the optimum number of subphenotype groups and builds a risk prediction model for each group. Simulation results demonstrated that the new approach had more accurate and robust performance than existing approaches under various underlying disease models. The empirical study of type II diabetes (T2D) by using data from the Genes and Environment Initiatives suggested heterogeneous etiology underlying obese and nonobese T2D patients. Considering phenotypic heterogeneity in the analysis leads to improved risk prediction models for both obese and nonobese T2D subjects.
Collapse
Affiliation(s)
- Yalu Wen
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan
| | | |
Collapse
|
38
|
Balbás-Martínez C, Rodríguez-Pinilla M, Casanova A, Domínguez O, Pisano DG, Gómez G, Lloreta J, Lorente JA, Malats N, Real FX. ARID1A alterations are associated with FGFR3-wild type, poor-prognosis, urothelial bladder tumors. PLoS One 2013; 8:e62483. [PMID: 23650517 PMCID: PMC3641081 DOI: 10.1371/journal.pone.0062483] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/20/2013] [Indexed: 02/06/2023] Open
Abstract
Urothelial bladder cancer (UBC) is heterogeneous at the clinical, pathological, genetic, and epigenetic levels. Exome sequencing has identified ARID1A as a novel tumor suppressor gene coding for a chromatin remodeling protein that is mutated in UBC. Here, we assess ARID1A alterations in two series of patients with UBC. In the first tumor series, we analyze exons 2–20 in 52 primary UBC and find that all mutant tumors belong to the aggressive UBC phenotype (high grade non-muscle invasive and muscle invasive tumors) (P = 0.05). In a second series (n = 84), we assess ARID1A expression using immunohistochemistry, a surrogate for mutation analysis, and find that loss of expression increases with higher stage/grade, it is inversely associated with FGFR3 overexpression (P = 0.03) but it is not correlated with p53 overexpression (P = 0.30). We also analyzed the expression of cytokeratins in the same set of tumor and find, using unsupervised clustering, that tumors with ARID1A loss of expression are generally KRT5/6-low. In this patient series, loss of ARID1A expression is also associated with worse prognosis, likely reflecting the higher prevalence of losses found in tumors of higher stage and grade. The independent findings in these two sets of patients strongly support the notion that ARID1A inactivation is a key player in bladder carcinogenesis occurring predominantly in FGFR3 wild type tumors.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Base Sequence
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/mortality
- Carcinoma, Transitional Cell/pathology
- Cell Line, Tumor
- DNA Mutational Analysis
- DNA-Binding Proteins
- Female
- HEK293 Cells
- Humans
- Kaplan-Meier Estimate
- Male
- Middle Aged
- Mutation, Missense
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Prognosis
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/mortality
- Urinary Bladder Neoplasms/pathology
- Urothelium/pathology
Collapse
Affiliation(s)
- Cristina Balbás-Martínez
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - María Rodríguez-Pinilla
- Lymphoma Group, Molecular Pathology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Ariel Casanova
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Orlando Domínguez
- Genomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - David G. Pisano
- Bioinformatics Unit, Structural and Computational Biology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Gonzalo Gómez
- Bioinformatics Unit, Structural and Computational Biology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Josep Lloreta
- Department of Pathology, Hospital del Mar, Barcelona, Spain
- Departament de Ciències Experimentals de i la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Núria Malats
- Genetic and Molecular Epidemiology Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Molecular Pathology Programme, Spanish National Cancer Research Centre, Madrid, Spain
- Departament de Ciències Experimentals de i la Salut, Universitat Pompeu Fabra, Barcelona, Spain
- * E-mail:
| |
Collapse
|
39
|
Hsu I, Vitkus S, Da J, Yeh S. Role of oestrogen receptors in bladder cancer development. Nat Rev Urol 2013; 10:317-26. [PMID: 23588401 DOI: 10.1038/nrurol.2013.53] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Early studies documented the existence of sexual dimorphism in bladder cancer occurrence and progression, with a greater bladder cancer incidence in males than females. However, the progression of bladder cancer after diagnosis is much quicker in females than males. These findings can be explained by the effects of female hormones (predominantly oestrogens) and their binding receptors, including oestrogen receptor 1 (ESR1; also known as ERα), oestrogen receptor 2 (ESR2; also known as ERβ), and GPR30 protein on bladder cancer incidence and progression. Results from studies using various in vitro cell lines and in vivo mouse models demonstrate differential roles of oestrogen receptors in cancer initiation and progression. ERα suppresses bladder cancer initiation and invasion, whereas ERβ promotes bladder cancer initiation and progression. Mechanistic studies suggest that ERα and ERβ exert these effects via modulation of the AKT pathway and DNA replication complex, respectively. Targeting these signalling pathways--for example, with ERα agonists, ERβ antagonists, or selective oestrogen receptor modulators such as 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (also known as PHTPP)--could lead to the development of new therapeutic approaches for controlling bladder cancer progression.
Collapse
Affiliation(s)
- Iawen Hsu
- Department of Urology, George Whipple Lab for Cancer Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
40
|
Robles C, Viscidi R, Malats N, Silverman DT, Tardon A, Garcia-Closas R, Serra C, Carrato A, Herranz J, Lloreta J, Rothman N, Real FX, de Sanjose S, Kogevinas M. Bladder cancer and seroreactivity to BK, JC and Merkel cell polyomaviruses: the Spanish bladder cancer study. Int J Cancer 2013; 133:597-603. [PMID: 23355322 DOI: 10.1002/ijc.28053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 12/18/2012] [Indexed: 12/16/2022]
Abstract
An infectious etiology for bladder cancer has long been suspected. Merkel cell virus (MCV), BKV and JCV polyomaviruses are possible causative agents but data remain scarce. Therefore, we evaluated the seroresponse to these three polyomaviruses in association with bladder cancer risk. 1,135 incident bladder cancer subjects from five Spanish regions and 982 hospital controls matched by sex, age and region were included. 99% of cases were urothelial-cell carcinomas. Antibody response against MCV, BKV and JCV was measured by enzyme immunoassay using Virus-Like-Particles. Our results show a similar seroprevalence in cases and controls: 64/60% for BKV, 83/82% for MCV and 87/83% for JCV. However, among seropositive subjects, higher median seroreactivities were observed in cases compared to controls for BKV (0.84 vs. 0.70, p-value = 0.009) and MCV (1.81 vs. 0.65, p-value < 0.001). Increased bladder cancer risk was observed for BKV (OR = 1.4, 95%CI 1.04-1.8) and for MCV (OR = 1.5, 95%CI 1.2-1.9), when comparing highest to lowest seroreactivity tertiles. The associations of BKV and MCV with bladder cancer were independent of each other and neither smoking status nor disease stage and grade modified them. Furthermore, no association was observed between seroresponse to JCV and bladder cancer. Therefore, we conclude that BKV and MCV polyomavirus infection could be related to an increased bladder cancer risk.
Collapse
Affiliation(s)
- Claudia Robles
- Unit of Infections and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology, IDIBELL, 08908 L' Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Amasyali AS, Kucukgergin C, Erdem S, Sanli O, Seckin S, Nane I. Nitric oxide synthase (eNOS4a/b) gene polymorphism is associated with tumor recurrence and progression in superficial bladder cancer cases. J Urol 2012; 188:2398-403. [PMID: 23088972 DOI: 10.1016/j.juro.2012.07.096] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Indexed: 12/20/2022]
Abstract
PURPOSE We investigated the relationship between the distribution of the eNOS4a/b polymorphism and the clinical features of superficial bladder cancer. MATERIALS AND METHODS This study included 201 healthy controls with a mean ± SD age of 62.35 ± 7.96 years and 123 patients with a mean age of 64.03 ± 11.00 years diagnosed with histopathologically confirmed superficial bladder cancer. The eNOS4a/b polymorphism genotype (aa, bb or ab) was identified by polymerase chain reaction. Blood glutathione and plasma malondialdehyde levels were measured by spectrophotometry as an indicator of oxidative stress. We estimated total plasma levels of nitric oxide metabolites using a colorimetric assay kit. RESULTS There were no significant differences in age or body mass index between patients and controls. Malondialdehyde and nitric oxide metabolite levels were statistically significantly increased (p = 0.000 and 0.024, respectively) and glutathione levels were decreased (p = 0.000) in patients with superficial bladder cancer. The bb genotype of the eNOS4a/b polymorphism is the most frequent one in the Turkish population and the aa genotype was significantly more common in patients with superficial bladder cancer (p = 0.000). Also, the aa plus ab genotype was significantly more common in patients with high grade tumors (p = 0.013) and in those with more progression to muscle invasive disease (p = 0.000). This genotype was also a significant independent risk factor for recurrence after adjusting for smoking status, stage, grade and the presence of carcinoma in situ on logistic regression analyses (OR 3.095, 95% CI 1.21-7.86, p = 0.018). CONCLUSIONS The current study suggests that a genotype containing the a allele of the eNOS4a/b polymorphism may be a risk factor for bladder cancer. Additionally, patients harboring the aa plus ab genotype are more likely to experience tumor recurrence and progression.
Collapse
|
42
|
Liu MS, Liu CH, Wu G, Zhou Y. Antisense inhibition of secretory and cytosolic phospholipase A2 reduces the mortality in rats with sepsis*. Crit Care Med 2012; 40:2132-40. [PMID: 22564957 DOI: 10.1097/ccm.0b013e31824e1e20] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Phospholipase A(2) has been implicated to play a pivotal role in the pathogenesis of sepsis syndrome. The two major forms of phospholipase A(2) isoenzymes, secretory phospholipase A(2) and cytosolic phospholipase A(2), are overexpressed during sepsis. The objective of this study was to test the hypothesis that inhibition of the overexpressed secretory phospholipase A(2) and cytosolic phospholipase A(2) during sepsis benefits the disease's eventual outcome. DESIGN Short-chain antisense oligonucleotide molecules were designed with the aid of computer software programs, and their in vitro efficacies were assessed in cell culture systems based on inhibition of target protein expression. The in vivo efficacies were determined in intact sepsis rats using 35-day survival rate as a primary efficacy end point. SETTING Animal research laboratory at a university. SUBJECTS Male Sprague-Dawley rats (180-200 g). INTERVENTIONS Sepsis was induced by cecal ligation and puncture. Antibiotics were administered subcutaneously once daily at 12 mg/kg, for 20 days. Oligonucleotides (antisense or mismatch) were administered intravenously once daily at 2 mg/kg to 0.8 mg/kg in a decreasing order, for 20 days. MEASUREMENTS AND MAIN RESULTS In cell culture systems, 21 of the 105 antisense constructs were found to be efficacious in inhibiting secretory phospholipase A(2) IIa and cytosolic phospholipase A(2) IVa protein expression. In sepsis rats, antisense oligonucleotides were capable of reducing their target protein expression by 18%-61% in major organs such as liver, heart, and kidney. In animal experiments, sepsis without any treatment (Group 1) had a median survival time of 2 days and a zero (0) percent survival rate at day 14. Sepsis with antibiotic treatment (Group 2) had a median survival time of 6 days and a 35-day survival rate of 28%. Sepsis with cotreatment of antibiotics and antisense oligonucleotides (one against secretory phospholipase A2 IIa and the other against cytosolic phospholipase A(2) IVa) (Group 4) increased the median survival time from 6 to 35 days and the 35-day survival rate from 28% to 58.8% as compared with antibiotics alone (Group 4 vs. Group 2; p <.05). Sepsis with cotreatment of antibiotics and mismatch oligonucleotides (Group 3) did not affect the median survival time and the 35-day survival rate as compared to antibiotics alone (Group 3 vs. Group 2; p >.05). CONCLUSIONS The results demonstrate that antisense strategy against secretory phospholipase A(2) IIa and cytosolic phospholipase A(2) IVa can inhibit their target protein expression in major organs and greatly improve the clinical outcome, i.e., an absolute reduction in 35-day mortality of 30.8%, in rats with sepsis. Our studies, thus, provide an improved method for the treatment of sepsis by targeting multiple forms of phospholipase A(2) isoenzymes with DNA antisense oligomers.
Collapse
Affiliation(s)
- Maw-Shung Liu
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St Louis, MO, USA.
| | | | | | | |
Collapse
|
43
|
Burger M, Catto JWF, Dalbagni G, Grossman HB, Herr H, Karakiewicz P, Kassouf W, Kiemeney LA, La Vecchia C, Shariat S, Lotan Y. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol 2012; 63:234-41. [PMID: 22877502 DOI: 10.1016/j.eururo.2012.07.033] [Citation(s) in RCA: 1436] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/12/2012] [Indexed: 12/14/2022]
Abstract
CONTEXT Urothelial bladder cancer (UBC) is a disease of significant morbidity and mortality. It is important to understand the risk factors of this disease. OBJECTIVE To describe the incidence, prevalence, and mortality of UBC and to review and interpret the current evidence on and impact of the related risk factors. EVIDENCE ACQUISITION A literature search in English was performed using PubMed. Relevant papers on the epidemiology of UBC were selected. EVIDENCE SYNTHESIS UBC is the 7th most common cancer worldwide in men and the 17th most common cancer worldwide in women. Approximately 75% of newly diagnosed UBCs are noninvasive. Each year, approximately 110 500 men and 70 000 women are diagnosed with new cases and 38 200 patients in the European Union and 17 000 US patients die from UBC. Smoking is the most common risk factor and accounts for approximately half of all UBCs. Occupational exposure to aromatic amines and polycyclic aromatic hydrocarbons are other important risk factors. The impact of diet and environmental pollution is less evident. Increasing evidence suggests a significant influence of genetic predisposition on incidence. CONCLUSIONS UBC is a frequently occurring malignancy with a significant impact on public health and will remain so because of the high prevalence of smoking. The importance of primary prevention must be stressed, and smoking cessation programs need to be encouraged and supported.
Collapse
Affiliation(s)
- Maximilian Burger
- Department of Urology and Pediatric Urology, Julius-Maximilians-University Medical Center, Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mocellin S, Verdi D, Pooley KA, Landi MT, Egan KM, Baird DM, Prescott J, De Vivo I, Nitti D. Telomerase reverse transcriptase locus polymorphisms and cancer risk: a field synopsis and meta-analysis. J Natl Cancer Inst 2012; 104:840-54. [PMID: 22523397 DOI: 10.1093/jnci/djs222] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Several recent studies have provided evidence that polymorphisms in the telomerase reverse transcriptase (TERT) gene sequence are associated with cancer development, but a comprehensive synopsis is not available. We conducted a systematic review and meta-analysis of the available molecular epidemiology data regarding the association between TERT locus polymorphisms and predisposition to cancer. METHODS A systematic review of the English literature was conducted by searching PubMed, Embase, Cancerlit, Google Scholar, and ISI Web of Knowledge databases for studies on associations between TERT locus polymorphisms and cancer risk. Random-effects meta-analysis was performed to pool per-allele odds ratios for TERT locus polymorphisms and risk of cancer, and between-study heterogeneity and potential bias sources (eg, publication and chasing bias) were assessed. Because the TERT locus includes the cleft lip and palate transmembrane 1-like (CLPTM1L) gene, which is in linkage disequilibrium with TERT, CLPTM1L polymorphisms were also analyzed. Cumulative evidence for polymorphisms with statistically significant associations was graded as "strong," "moderate," and "weak" according to the Venice criteria. The joint population attributable risk was calculated for polymorphisms with strong evidence of association. RESULTS Eighty-five studies enrolling 490 901 subjects and reporting on 494 allelic contrasts were retrieved. Data were available on 67 TERT locus polymorphisms and 24 tumor types, for a total of 221 unique combinations of polymorphisms and cancer types. Upon meta-analysis, a statistically significant association with the risk of any cancer type was found for 22 polymorphisms. Strong, moderate, and weak cumulative evidence for association with at least one tumor type was demonstrated for 11, 9, and 14 polymorphisms, respectively. For lung cancer, which was the most studied tumor type, the estimated joint population attributable risk for three polymorphisms (TERT rs2736100, intergenic rs4635969, and CLPTM1L rs402710) was 41%. Strong evidence for lack of association was identified for five polymorphisms in three tumor types. CONCLUSIONS To our knowledge, this is the largest collection of data for associations between TERT locus polymorphisms and cancer risk. Our findings support the hypothesis that genetic variability in this genomic region can modulate cancer susceptibility in humans.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Oncological and Surgical Sciences, Meta-analysis Unit, University of Padova, Padova, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kotsinas A, Aggarwal V, Tan EJ, Levy B, Gorgoulis VG. PIG3: a novel link between oxidative stress and DNA damage response in cancer. Cancer Lett 2011; 327:97-102. [PMID: 22178897 DOI: 10.1016/j.canlet.2011.12.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 11/25/2022]
Abstract
Reactive oxygen species (ROS), the most prominent free radicals produced in cells, can have both beneficial and detrimental effects on them. Many genes are known to be involved in ROS regulation. P53 inducible gene 3 (PIG3 or TP53I3) was identified in an analysis of genes induced by p53 before the onset of apoptosis. It is a widely conserved gene between many species. Until now it has been shown to exert two disparate cellular roles. The first is that of ROS producer linked to p53 induced apoptosis. In this context, it exhibits a NADPH dependent reductase activity with orthoquinones. The second is that of a component of the DNA damage response pathway. While it is considered as a p53 dependent pro-apoptotic gene, it is rarely affected in cancer. This data does not support an anti-tumor activity. In the present review we present and discuss aspects on the regulation and function of this factor and how it is implicated in cancer. We conclude by proposing that PIG3 may possibly have a role in cancer cell survival.
Collapse
Affiliation(s)
- Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Greece
| | | | | | | | | |
Collapse
|
46
|
Porta N, Calle ML, Malats N, Gómez G. A dynamic model for the risk of bladder cancer progression. Stat Med 2011; 31:287-300. [PMID: 22161505 DOI: 10.1002/sim.4433] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 09/15/2011] [Indexed: 11/05/2022]
Abstract
We propose a multistate modeling approach to describe the observed evolution of patients diagnosed with non-muscle-invasive bladder cancer. On the basis of data from the Spanish Bladder Cancer/EPICURO study, we adjust a multistate model taking into account the disease-related events of interest (recurrence, progression, and disease-related deaths) as well as competing deaths due to other causes. We then develop a dynamic predictive process for bladder cancer progression, which allows the risk of a patient to be updated whenever new information of his or her evolution is available. By using specific measures of prospective accuracy in the presence of competing risks, the proposed dynamic model has shown to improve prediction accuracy and provides a more personalized management of bladder patients.
Collapse
Affiliation(s)
- Núria Porta
- Universitat Politècnica de Catalunya, Jordi Girona 1-3, 08034, Barcelona, Spain.
| | | | | | | |
Collapse
|
47
|
Garzón J, Rodríguez-Muñoz M, Vicente-Sánchez A, García-López MÁ, Martínez-Murillo R, Fischer T, Sánchez-Blázquez P. SUMO-SIM interactions regulate the activity of RGSZ2 proteins. PLoS One 2011; 6:e28557. [PMID: 22163035 PMCID: PMC3232247 DOI: 10.1371/journal.pone.0028557] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/10/2011] [Indexed: 11/18/2022] Open
Abstract
The RGSZ2 gene, a regulator of G protein signaling, has been implicated in cognition, Alzheimer's disease, panic disorder, schizophrenia and several human cancers. This 210 amino acid protein is a GTPase accelerating protein (GAP) on Gαi/o/z subunits, binds to the N terminal of neural nitric oxide synthase (nNOS) negatively regulating the production of nitric oxide, and binds to the histidine triad nucleotide-binding protein 1 at the C terminus of different G protein-coupled receptors (GPCRs). We now describe a novel regulatory mechanism of RGS GAP function through the covalent incorporation of Small Ubiquitin-like MOdifiers (SUMO) into RGSZ2 RGS box (RH) and the SUMO non covalent binding with SUMO-interacting motifs (SIM): one upstream of the RH and a second within this region. The covalent attachment of SUMO does not affect RGSZ2 binding to GPCR-activated GαGTP subunits but abolishes its GAP activity. By contrast, non-covalent binding of SUMO with RH SIM impedes RGSZ2 from interacting with GαGTP subunits. Binding of SUMO to the RGSZ2 SIM that lies outside the RH does not affect GαGTP binding or GAP activity, but it could lead to regulatory interactions with sumoylated proteins. Thus, sumoylation and SUMO-SIM interactions constitute a new regulatory mechanism of RGS GAP function and therefore of GPCR cell signaling as well.
Collapse
Affiliation(s)
- Javier Garzón
- Cajal Institute, CSIC, Madrid, Spain
- CIBER of Mental Health, ISCIII, Madrid, Spain
| | | | - Ana Vicente-Sánchez
- Cajal Institute, CSIC, Madrid, Spain
- CIBER of Mental Health, ISCIII, Madrid, Spain
| | | | | | - Thierry Fischer
- Department of Immunology and Oncology, National Centre of Biotechnology, CSIC, Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Cajal Institute, CSIC, Madrid, Spain
- CIBER of Mental Health, ISCIII, Madrid, Spain
- * E-mail:
| |
Collapse
|
48
|
Garcia-Closas M, Ye Y, Rothman N, Figueroa JD, Malats N, Dinney CP, Chatterjee N, Prokunina-Olsson L, Wang Z, Lin J, Real FX, Jacobs KB, Baris D, Thun M, De Vivo I, Albanes D, Purdue MP, Kogevinas M, Kamat AM, Lerner SP, Grossman HB, Gu J, Pu X, Hutchinson A, Fu YP, Burdett L, Yeager M, Tang W, Tardón A, Serra C, Carrato A, García-Closas R, Lloreta J, Johnson A, Schwenn M, Karagas MR, Schned A, Andriole G, Grubb R, Black A, Jacobs EJ, Diver WR, Gapstur SM, Weinstein SJ, Virtamo J, Hunter DJ, Caporaso N, Landi MT, Fraumeni JF, Silverman DT, Chanock SJ, Wu X. A genome-wide association study of bladder cancer identifies a new susceptibility locus within SLC14A1, a urea transporter gene on chromosome 18q12.3. Hum Mol Genet 2011; 20:4282-9. [PMID: 21824976 PMCID: PMC3188994 DOI: 10.1093/hmg/ddr342] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 08/01/2011] [Indexed: 11/13/2022] Open
Abstract
Genome-wide and candidate-gene association studies of bladder cancer have identified 10 susceptibility loci thus far. We conducted a meta-analysis of two previously published genome-wide scans (4501 cases and 6076 controls of European background) and followed up the most significant association signals [17 single nucleotide polymorphisms (SNPs) in 10 genomic regions] in 1382 cases and 2201 controls from four studies. A combined analysis adjusted for study center, age, sex, and smoking status identified a novel susceptibility locus that mapped to a region of 18q12.3, marked by rs7238033 (P = 8.7 × 10(-9); allelic odds ratio 1.20 with 95% CI: 1.13-1.28) and two highly correlated SNPs, rs10775480/rs10853535 (r(2)= 1.00; P = 8.9 × 10(-9); allelic odds ratio 1.16 with 95% CI: 1.10-1.22). The signal localizes to the solute carrier family 14 member 1 gene, SLC14A1, a urea transporter that regulates cellular osmotic pressure. In the kidney, SLC14A1 regulates urine volume and concentration whereas in erythrocytes it determines the Kidd blood groups. Our findings suggest that genetic variation in SLC14A1 could provide new etiological insights into bladder carcinogenesis.
Collapse
|
49
|
|
50
|
Seiler R, Thalmann GN, Fleischmann A. MMP-2 and MMP-9 in lymph-node-positive bladder cancer. J Clin Pathol 2011; 64:1078-82. [DOI: 10.1136/jclinpath-2011-200153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BackgroundMatrix metalloproteinases (MMP), particularly MMP-2 and MMP-9, participate in tumour progression and metastasis in various cancers. Their significance in urothelial cancer of the bladder (UCB) is unclear. Expression analysis of MMP-2 and MMP-9 in tissue microarrays (TMA) constructed of corresponding samples from histopathological normal urothelium, tumour centre and invasion front of primary tumours and lymph-node (LN) metastases might help to elucidate their relevance in UCB.MethodMMP-2 and MMP-9 expression was evaluated in TMA of 150 surgically treated LN-positive UCB patients. Biomarker expression was correlated with tumour characteristics (primary tumour and LN stage, number, total diameter and extracapsular extension of LN metastases) and overall survival.ResultsWhile there was a significant increase in MMP-9 expression from normal urothelium over primary tumours to metastases (median scores: 20, 50, 65; p<0.005), no such trend was observed in MMP-2 (median scores: 5, 22, 10; p<0.005). A comparison of expression in the tumour centre and the invasion front showed no difference in both MMP. No association between expression and histopathological tumour characteristics was identified. There was a non-significant trend for a more favourable outcome for patients with high MMP-2 expression in primary tumours.ConclusionIn LN-positive UCB, MMP-2 and MMP-9 expression was not increased at the invasion front, suggesting an infiltration strategy independent of MMP-2 and MMP-9 activity. Larger series are needed to detect a potential significant trend for favourable outcome in cancers with high MMP-2 expression.
Collapse
|