1
|
Joshi S, De Angelis PM, Zucknick M, Schjølberg AR, Andersen SN, Clausen OPF. Role of the Wnt signaling pathway in keratoacanthoma. Cancer Rep (Hoboken) 2019; 3:e1219. [PMID: 32672002 DOI: 10.1002/cnr2.1219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Keratoacanthoma (KA) has a unique life cycle of rapid growth and spontaneous regression that shows similarities to the hair follicle cycle, which involves an active Wnt signaling during physiological regeneration. We analyzed the expression of the Wnt signaling proteins β-catenin, Lef1, Sox9, and Cyclin D1 in young and old human KAs to investigate a possible role for Wnt signaling in KAs. AIM To investigate the role of the Wnt/β-catenin signaling pathway in human KAs. METHODS AND RESULTS Formalin-fixed, paraffin-embedded tissue samples of 67 KAs were analyzed for protein expression using immunohistochemistry. The majority of KAs were positive for Sox9 and Cyclin D1 but not for nuclear-localized β-catenin or Lef-1. No significant differences in protein expressions were seen between young and old KAs. However, we found a significant association between Ki67 and Cyclin D1 proteins (P= .008). CONCLUSIONS The Wnt signaling pathway does not appear to play a significant role in the biogenesis of human KA. Sox9 overexpression may be indicative of inhibition of Wnt signaling. Sox-9 and Cyclin D1 are proliferation markers that are most likely transactivated by alternate signaling pathways.
Collapse
Affiliation(s)
- Sarita Joshi
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Paula M De Angelis
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aasa R Schjølberg
- Department of Pathology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Solveig Norheim Andersen
- Institute of Clinical Medicine, Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | | |
Collapse
|
2
|
Zhan Y, Feng J, Lu J, Xu L, Wang W, Fan S. Expression of LEF1 and TCF1 (TCF7) proteins associates with clinical progression of nasopharyngeal carcinoma. J Clin Pathol 2019; 72:425-430. [DOI: 10.1136/jclinpath-2019-205698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/24/2022]
Abstract
AimsOur previous study has demonstrated that β-catenin pathway was abnormally activated in nasopharyngeal carcinoma (NPC). The purposes of the present study are to investigate whether the alterations of LEF1 and TCF1 (TCF7) proteins, the important components of the canonical Wnt/β-catenin pathway, are associated with clinicopathological features and prognostic implications.MethodsWe collected 391 cases of NPC, 53 non-cancerous control nasopharyngeal mucosa and 28 pairs of NPC and their matched metastases, detected expression of LEF1 and TCF1 (TCF7) proteins in these tissues by immunohistochemistry. ResultsResults showed that there were significantly increased expression of both LEF1 and TCF1 (TCF7) proteins and coexpression of LEF1 and TCF1 (TCF7) in NPC than these in non-cancerous nasopharyngeal mucosa (all p<0.001), as well as LEF1 and coexpression of LEF1 and TCF1 (TCF7) in matched metastasis NPCs than these in the primary NPCs (p=0.003 and p=0.014, respectively). In addition, expression of LEF1 and the coexpression of LEF1 and TCF1 (TCF7) proteins were positively correlated with lymph node metastasis (p=0.001 and p=0.020, respectively), advanced clinical stage (p<0.003 and p=0.027, respectively) and poor survival status of patients with NPC (p<0.001 and p=0.004, respectively). Moreover, multivariate Cox regression analysis identified that the positive expression of LEF1 was the independent poor prognostic factor for overall survival of patients with NPC (p<0.001).ConclusionsThe expression of LEF1 associated positively with TCF1 (TCF7) and clinical progression of NPC, and positive expression of LEF1 protein may act as valuable independent biomarker to predict poor prognosis for patients with NPC.
Collapse
|
3
|
Mormile R. Metformin and Colorectal Cancer Risk—Letter. Cancer Epidemiol Biomarkers Prev 2018; 27:1385. [DOI: 10.1158/1055-9965.epi-18-0559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/06/2018] [Accepted: 08/15/2018] [Indexed: 11/16/2022] Open
Affiliation(s)
- Raffaella Mormile
- Division of Pediatrics and Neonatology, Moscati Hospital, Aversa, Italy
| |
Collapse
|
4
|
Comparison of β-Catenin and LEF1 Immunohistochemical Stains in Desmoid-type Fibromatosis and its Selected Mimickers, With Unexpected Finding of LEF1 Positivity in Scars. Appl Immunohistochem Mol Morphol 2018; 26:648-653. [DOI: 10.1097/pai.0000000000000487] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
5
|
Przybyl J, Kidzinski L, Hastie T, Debiec-Rychter M, Nusse R, van de Rijn M. Gene expression profiling of low-grade endometrial stromal sarcoma indicates fusion protein-mediated activation of the Wnt signaling pathway. Gynecol Oncol 2018; 149:388-393. [PMID: 29544705 DOI: 10.1016/j.ygyno.2018.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/03/2018] [Accepted: 03/07/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Low-grade endometrial stromal sarcomas (LGESS) harbor chromosomal translocations that affect proteins associated with chromatin remodeling Polycomb Repressive Complex 2 (PRC2), including SUZ12, PHF1 and EPC1. Roughly half of LGESS also demonstrate nuclear accumulation of β-catenin, which is a hallmark of Wnt signaling activation. However, the targets affected by the fusion proteins and the role of Wnt signaling in the pathogenesis of these tumors remain largely unknown. METHODS Here we report the results of a meta-analysis of three independent gene expression profiling studies on LGESS and immunohistochemical evaluation of nuclear expression of β-catenin and Lef1 in 112 uterine sarcoma specimens obtained from 20 LGESS and 89 LMS patients. RESULTS Our results demonstrate that 143 out of 310 genes overexpressed in LGESS are known to be directly regulated by SUZ12. In addition, our gene expression meta-analysis shows activation of multiple genes implicated in Wnt signaling. We further emphasize the role of the Wnt signaling pathway by demonstrating concordant nuclear expression of β-catenin and Lef1 in 7/16 LGESS. CONCLUSIONS Based on our findings, we suggest that LGESS-specific fusion proteins disrupt the repressive function of the PRC2 complex similar to the mechanism seen in synovial sarcoma, where the SS18-SSX fusion proteins disrupt the mSWI/SNF (BAF) chromatin remodeling complex. We propose that these fusion proteins in LGESS contribute to overexpression of Wnt ligands with subsequent activation of Wnt signaling pathway and formation of an active β-catenin/Lef1 transcriptional complex. These observations could lead to novel therapeutic approaches that focus on the Wnt pathway in LGESS.
Collapse
Affiliation(s)
- Joanna Przybyl
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, 94305, CA, USA.
| | - Lukasz Kidzinski
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, 94305, CA, USA
| | - Trevor Hastie
- Department of Statistics, Stanford University, 390 Serra Mall, Stanford, 94305, CA, USA
| | - Maria Debiec-Rychter
- Department of Human Genetics, KU Leuven and University Hospitals Leuven, UZ, Herestraat 49, 3000 Leuven, Belgium
| | - Roel Nusse
- Department of Developmental Biology, Stanford University School of Medicine, 265 Campus Drive, Stanford, 94305, CA, USA
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, 94305, CA, USA
| |
Collapse
|
6
|
Tumminello K, Hosler GA. CDX2 and LEF-1 expression in pilomatrical tumors and their utility in the diagnosis of pilomatrical carcinoma. J Cutan Pathol 2018; 45:318-324. [PMID: 29369390 DOI: 10.1111/cup.13113] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/19/2017] [Accepted: 01/21/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND The Wnt signaling pathway has been implicated in the pathogenesis of pilomatrical tumors. Lymphoid enhancer-binding factor 1 (LEF-1) is a downstream component of this pathway, and Caudal-related homeobox transcription factor 2 (CDX2) has been postulated to regulate it, but little is known about expression of these transcription factors in pilomatrical tumors. METHODS Immunohistochemistry for CDX2, β-catenin, LEF-1, CK19, CK5, Special AT-rich sequence- binding protein 2 (SATB2), cadherin 17 and androgen receptor was performed on pilomatricomas (PMs) (N = 12), pilomatrical carcinomas (PMCAs) (N = 12) and non-pilomatrical cutaneous tumors (N = 18). RESULTS PMs and PMCAs were positive for CDX2 (9/12 PMs, sensitivity = 75%, specificity = 100%; 11/12 PMCAs, sensitivity = 92%, specificity = 100%; P < 0.01), β-catenin (12/12 PMs, sensitivity = 100%, specificity = 94%; 10/12 PMCAs, sensitivity = 83%, specificity = 94%; P < 0.01) and LEF-1 (12/12 PMs, sensitivity = 100%, specificity = 56%; 12/12 PMCAs, sensitivity = 100%, specificity = 56%; P < 0.01). CDX2 expression was commonly focal, within a discrete subpopulation of squamoid cells. The LEF-1 expression pattern was different and discernable between pilomatrical tumors (strong, diffuse) and non-pilomatrical tumors (weak, patchy). CONCLUSION This study reaffirms the importance of the Wnt signaling pathway in the tumorigenesis of pilomatrical tumors, and this introduces CDX2 as a possible regulator and marker of pilomatrical tumorigenesis. LEF-1 and CDX2 performed at least as well as β-catenin, if not better when taking into account expression pattern, as a diagnostic marker for PMCA, and should be considered in the workup of ambiguous primitive-appearing cutaneous tumors.
Collapse
Affiliation(s)
- Katherine Tumminello
- University of Mississippi Medical Center, Department of Pathology, Jackson, Mississippi.,ProPath, Division of Dermatopathology, Dallas, Texas
| | - Gregory A Hosler
- ProPath, Division of Dermatopathology, Dallas, Texas.,University of Texas Southwestern Medical Center, Department of Dermatology, Dallas, Texas.,University of Texas Southwestern Medical Center, Department and Pathology, Dallas, Texas
| |
Collapse
|
7
|
Mohindra S, Sakr H, Sturgis C, Chute DJ. LEF-1 is a Sensitive Marker of Cribriform Morular Variant of Papillary Thyroid Carcinoma. Head Neck Pathol 2017; 12:455-462. [PMID: 29243023 PMCID: PMC6232206 DOI: 10.1007/s12105-017-0873-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
Abstract
Cribriform morular variant of PTC (CMV-PTC) frequently shows activation of the CTNNB1/Wnt pathway with nuclear accumulation of beta catenin. The utility of LEF-1, also in the CTNNB1/WNT pathway, in the diagnosis of CMV-PTC has not been previously studied. LEF-1 immunohistochemistry was performed on seven CMV-PTC, 52 benign cases and 101 malignant thyroid neoplasms. LEF-1 was scored by stain intensity (0 = no nuclear stain, 1 = weak nuclear stain, less than lymphocyte and 2 = strong nuclear stain, intense as lymphocyte) and percentage of positive cells at each intensity, for a maximum total score of 200. Sensitivity and specificity of LEF-1 stain for all cases and to differentiate between regular PTC and CMV-PTC was also calculated. Six of the seven CMV-PTCs showed ≥ 30% strong (2+) nuclear LEF-1 staining and a total score over 100. Beta catenin also showed strong and diffuse nuclear staining in these cases. One CMV-PTC was negative for both LEF-1 and beta catenin and did not have a history of FAP. All control PTC cases uniformly lacked LEF-1 staining at 2+ intensity. LEF-1 had a sensitivity of 86% and specificity of 98% for the diagnosis of CMV-PTC. LEF-1 is highly sensitive and specific marker for CMV-PTC, especially when used in the setting of a PTC neoplasm. The pattern of staining is important with ≥ 30% of cells showing strong 2+ nuclear staining having the highest combined sensitivity and specificity.
Collapse
Affiliation(s)
- Shalini Mohindra
- 0000 0001 0675 4725grid.239578.2Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Ave, L25, Cleveland, OH 44195 USA
| | - Hany Sakr
- 0000 0001 0675 4725grid.239578.2Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Ave, L25, Cleveland, OH 44195 USA
| | - Charles Sturgis
- 0000 0001 0675 4725grid.239578.2Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Ave, L25, Cleveland, OH 44195 USA
| | - Deborah J. Chute
- 0000 0001 0675 4725grid.239578.2Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Ave, L25, Cleveland, OH 44195 USA
| |
Collapse
|
8
|
Huang LX, Hu CY, Jing L, Wang MC, Xu M, Wang J, Wang Y, Nan KJ, Wang SH. microRNA-219-5p inhibits epithelial-mesenchymal transition and metastasis of colorectal cancer by targeting lymphoid enhancer-binding factor 1. Cancer Sci 2017; 108:1985-1995. [PMID: 28771881 PMCID: PMC5623737 DOI: 10.1111/cas.13338] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/13/2017] [Accepted: 07/30/2017] [Indexed: 12/24/2022] Open
Abstract
Aberrant expression of microRNAs (miRs) has been shown to play a critical role in the pathogenesis and progression of tumors. microRNA‐219‐5p (miR‐219‐5p) has been reported to be abnormally expressed in some types of human tumors. However, the mechanism between miR‐219‐5p and colorectal cancer (CRC) metastasis remains unclear. In the present study, miR‐219‐5p was found to be downregulated in CRC tissue compared with matched normal tissue. Through luciferase reporter assay, we demonstrated lymphoid enhancer‐binding factor 1 (LEF1) as a direct target of miR‐219‐5p. Overexpression of miR‐219‐5p could inhibit motility, migration and invasion of CRC cells, and inhibit epithelial‐mesenchymal transition (EMT). Furthermore, silencing LEF1 phenocopied this metastasis‐suppressive function. The recovery experiment showed that re‐expression of LEF1 rescued this suppressive effect on tumor metastasis and reversed the expression of EMT markers caused by miR‐219‐5p. Additionally, we demonstrated that miR‐219‐5p exerted this tumor‐suppressive function by blocking activation of the AKT and ERK pathways. Finally, a nude mice experiment showed that miR‐219‐5p reduced the lung metastasis ability of CRC cells. Taken together, our findings indicate that miR‐219‐5p inhibits metastasis and EMT of CRC by targeting LEF1 and suppressing the AKT and ERK pathways, which may provide a new antitumor strategy to delay CRC metastasis.
Collapse
Affiliation(s)
- Lan-Xuan Huang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chun-Yan Hu
- Department of Gynecology, North-western Women's and Children's Hospital, Xi'an, Shaanxi Province, China
| | - Li Jing
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Min-Cong Wang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Meng Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jing Wang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yu Wang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ke-Jun Nan
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Shu-Hong Wang
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
9
|
Wu F, Yuan G, Chen J, Wang C. Network analysis based on TCGA reveals hub genes in colon cancer. Contemp Oncol (Pozn) 2017; 21:136-144. [PMID: 28947883 PMCID: PMC5611503 DOI: 10.5114/wo.2017.68622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/17/2017] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is the third most widespread cancer in the world. Although many advances have been made in molecular biology, novel approaches are still required to reveal molecular mechanisms for the diagnosis and therapy of colon cancer. In this study, we aimed to determine and analyse the hub genes of CRC. First, we explored the mRNA and microRNA (miRNA) expression profiles of colon carcinoma, then we screened target genes of differentially expressed miRNAs and obtained the intersection between differently expressed genes and target genes. Gene Ontology (GO) classification and KEGG pathway analysis of differently expressed genes were performed, and gene-miRNA and TF-gene-miRNA networks were constructed to identify hub genes, miRNAs, and TFs. In total, 3436 significant differentially expressed genes (1709 upregulated and 1727 downregulated) and 216 differentially expressed miRNAs (99 upregulated and 117 downregulated) were identified in colon cancer. These differentially expressed genes were significantly enriched in GO terms and KEGG pathways, such as cell proliferation, cell adhesion, and cytokine-cytokine receptor interaction signalling pathways. GCNT4, EDN2, and so on were located in the central hub of the co-expression network. MYC, WT1, mir-34a, and LEF1 were located in the central hub of the network of TF-gene-miRNA. These findings increase our understanding of the molecular mechanisms of colon cancer and will aid in identifying potential targets for diagnostic and therapeutic usage.
Collapse
Affiliation(s)
- Fenzan Wu
- Science and Education Division, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Guoping Yuan
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Junjie Chen
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Chengzu Wang
- Clinical Laboratory, Cixi Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
10
|
Menter T, Trivedi P, Ahmad R, Flora R, Dirnhofer S, Tzankov A, Naresh KN. Diagnostic Utility of Lymphoid Enhancer Binding Factor 1 Immunohistochemistry in Small B-Cell Lymphomas. Am J Clin Pathol 2017; 147:292-300. [PMID: 28395058 DOI: 10.1093/ajcp/aqw208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVES Recent studies have shown that lymphoid enhancer binding factor 1 (LEF1) is a useful marker for chronic lymphocytic B-cell leukemia (CLL)/small lymphocytic lymphoma. Yet, it is not still being widely used in a diagnostic setting. In this study, we document the experience with LEF1 immunohistochemistry during routine diagnostics. METHODS In total, 191 B-cell lymphoma cases from Hammersmith Hospital, Imperial College NHS Healthcare Trust (London, UK) were investigated by immunohistochemistry for LEF1 during routine diagnostic workup. These cases included both bone marrow trephines and lymph node biopsy specimens. The monoclonal antibody clone EPR2029Y was used. RESULTS LEF1 expression was strong and diffuse (>70% of cells) in most cases. Few CLL cases showed a staining in proliferation centers only. Seventy-seven of 80 CLL cases expressed LEF1. Other entities expressing LEF1 included one of 38 follicular lymphomas, two of 33 marginal zone lymphomas, and one diffuse large B-cell lymphoma with a background of follicular lymphoma grade 3B. Sensitivity for LEF1 for the diagnosis of CLL was 0.96, and specificity was 0.93. CONCLUSIONS In this study, we could demonstrate the diagnostic utility of LEF1. LEF1 is a sensitive and specific marker for CLL and is helpful in the diagnosis of diagnostically challenging small B-cell lymphomas.
Collapse
Affiliation(s)
- Thomas Menter
- From the Department of Cellular and Molecular Pathology, Hammersmith Hospital Campus, Imperial College Healthcare NHS Trust, London, UK; and
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Pritesh Trivedi
- From the Department of Cellular and Molecular Pathology, Hammersmith Hospital Campus, Imperial College Healthcare NHS Trust, London, UK; and
| | - Raida Ahmad
- From the Department of Cellular and Molecular Pathology, Hammersmith Hospital Campus, Imperial College Healthcare NHS Trust, London, UK; and
| | - Rashpal Flora
- From the Department of Cellular and Molecular Pathology, Hammersmith Hospital Campus, Imperial College Healthcare NHS Trust, London, UK; and
| | - Stephan Dirnhofer
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Kikkeri N Naresh
- From the Department of Cellular and Molecular Pathology, Hammersmith Hospital Campus, Imperial College Healthcare NHS Trust, London, UK; and
| |
Collapse
|
11
|
Smith AL, Bascuñana C, Hall A, Salman A, Andrei AZ, Volenik A, Rothenmund H, Ferland D, Lamoussenery D, Kamath AS, Amre R, Caglar D, Gao ZH, Haegert DG, Kanber Y, Michel RP, Omeroglu-Altinel G, Asselah J, Bouganim N, Kavan P, Arena G, Barkun J, Chaudhury P, Gallinger S, Foulkes WD, Omeroglu A, Metrakos P, Zogopoulos G. Establishing a clinic-based pancreatic cancer and periampullary tumour research registry in Quebec. ACTA ACUST UNITED AC 2015; 22:113-21. [PMID: 25908910 DOI: 10.3747/co.22.2300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Enrolling patients in studies of pancreatic ductal adenocarcinoma (pdac) is challenging because of the high fatality of the disease. We hypothesized that a prospective clinic-based study with rapid ascertainment would result in high participation rates. Using that strategy, we established the Quebec Pancreas Cancer Study (qpcs) to investigate the genetics and causes of pdac and other periampullary tumours (pats) that are also rare and underrepresented in research studies. METHODS Patients diagnosed with pdac or pat were introduced to the study at their initial clinical encounter, with a strategy to enrol participants within 2 weeks of diagnosis. Patient self-referrals and referrals of unaffected individuals with an increased risk of pdac were also accepted. Family histories, epidemiologic and clinical data, and biospecimens were collected. Additional relatives were enrolled in families at increased genetic risk. RESULTS The first 346 completed referrals led to 306 probands being enrolled, including 190 probands affected with pdac, who represent the population focus of the qpcs. Participation rates were 88.4% for all referrals and 89.2% for pdac referrals. Family history, epidemiologic and clinical data, and biospecimens were ascertained from 91.9%, 54.6%, and 97.5% respectively of patients with pdac. Although demographics and trends in risk factors in our patients were consistent with published statistics for patients with pdac, the qpcs is enriched for families with French-Canadian ancestry (37.4%), a population with recurrent germ-line mutations in hereditary diseases. CONCLUSIONS Using rapid ascertainment, a pdac and pat research registry with high participation rates can be established. The qpcs is a valuable research resource and its enrichment with patients of French-Canadian ancestry provides a unique opportunity for studies of heredity in these diseases.
Collapse
Affiliation(s)
- A L Smith
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - C Bascuñana
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Hall
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Salman
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - A Z Andrei
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Volenik
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - H Rothenmund
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - D Ferland
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - D Lamoussenery
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - A S Kamath
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - R Amre
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - D Caglar
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - Z H Gao
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - D G Haegert
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - Y Kanber
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - R P Michel
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | | | - J Asselah
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - N Bouganim
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - P Kavan
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - G Arena
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - J Barkun
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - P Chaudhury
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - S Gallinger
- The Research Institute of the McGill University Health Centre, Montreal, QC
| | - W D Foulkes
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - A Omeroglu
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - P Metrakos
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - G Zogopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| |
Collapse
|