1
|
Harrold EC, Stadler ZK. Upper Gastrointestinal Cancers and the Role of Genetic Testing. Hematol Oncol Clin North Am 2024; 38:677-691. [PMID: 38458854 DOI: 10.1016/j.hoc.2024.01.006] [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] [Indexed: 03/10/2024]
Abstract
Beyond the few established hereditary cancer syndromes with an upper gastrointestinal cancer component, there is increasing recognition of the contribution of novel pathogenic germline variants (gPVs) to upper gastrointestinal carcinogenesis. The detection of gPVs has potential implications for novel treatment approaches of the index cancer patient as well as long-term implications for surveillance and risk-reducing measures for cancer survivors and far-reaching implications for the patients' family. With widespread availability of multigene panel testing, new associations may be identified with germline-somatic integration being critical to determining true causality of novel gPVs. Comprehensive cancer care should incorporate both somatic and germline testing.
Collapse
Affiliation(s)
- Emily C Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland. https://twitter.com/EmilyHarrold6
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Hao S, Zhao X, Fan Y, Liu Z, Zhang X, Li W, Yuan H, Zhang J, Zhang Y, Ma T, Tao H. Prevalence and spectrum of cancer predisposition germline mutations in young patients with the common late-onset cancers. Cancer Med 2023; 12:18394-18404. [PMID: 37610374 PMCID: PMC10524041 DOI: 10.1002/cam4.6445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Pathogenic germline variants (PGVs) can play a vital role in the oncogenesis process in carriers. Previous studies have recognized that PGVs contribute to early onset of tumorigenesis in certain cancer types, for example, colorectal cancer and breast cancer. However, the reported prevalence data of cancer-associated PGVs were highly inconsistent due to nonuniform patient cohorts, sequencing methods, and prominent difficulties in pathogenicity interpretation of variants. In addition to the above difficulties, due to the rarity of cases, the prevalence of cancer PGV carriers in young cancer patients affected by late-onset cancer types has not been comprehensively evaluated to date. METHODS A total of 131 young cancer patients (1-29 years old at diagnosis) were enrolled in this study. The patients were affected by six common late-onset cancer types, namely, lung cancer, liver cancer, colorectal cancer, gastric cancer, renal cancer, and head-neck cancer. Cancer PGVs were identified and analyzed. based on NGS-based targeted sequencing followed by bioinformatic screening and strict further evaluations of variant pathogenicity. RESULTS Twenty-three cancer PGVs in 21 patients were identified, resulting in an overall PGV prevalence of 16.0% across the six included cancer types, which was approximately double the prevalence reported in a previous pancancer study. Nine of the 23 PGVs are novel, thus expanding the cancer PGV spectrum. Seven of the 23 (30.4%) PGVs are potential therapeutic targets of olaparib, with potential implications for clinical manipulation. Additionally, a small prevalence of somatic mutations of some classic cancer hallmark genes in young patients, in contrast to all-age patients, was revealed. CONCLUSION This study demonstrates the high prevalence of PGVs in young cancer patients with the common late-onset cancers and the potentially significant clinical implications of cancer PGVs, the findings highlight the value of PGV screening in young patients across lung cancer, liver cancer, colorectal cancer, gastric cancer, renal cancer, or head-neck cancer.
Collapse
Affiliation(s)
- Shaoyu Hao
- Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ximeng Zhao
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Yue Fan
- Department of Integrated Traditional Chinese Medicine and Western MedicineZhong Shan Hospital, Fudan UniversityShanghaiChina
| | - Zhengchuang Liu
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
| | - Xiang Zhang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Wei Li
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | | | - Jie Zhang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | | | - Tonghui Ma
- Jichenjunchuang Clinical LaboratoryHangzhouChina
| | - Houquan Tao
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
- Department of GastroenterologyZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeHangzhouChina
| |
Collapse
|
3
|
Guerra J, Pinto C, Pinto P, Pinheiro M, Santos C, Peixoto A, Escudeiro C, Barbosa A, Porto M, Francisco I, Lopes P, Isidoro AR, Cunha AL, Albuquerque C, Claro I, Oliveira C, Silva J, Teixeira MR. Frequency of CDH1, CTNNA1 and CTNND1 Germline Variants in Families with Diffuse and Mixed Gastric Cancer. Cancers (Basel) 2023; 15:4313. [PMID: 37686589 PMCID: PMC10486404 DOI: 10.3390/cancers15174313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The most well-characterized hereditary form of gastric cancer is hereditary diffuse gastric cancer (HDGC), an autosomal dominant syndrome characterized by an increased risk of diffuse gastric and lobular breast cancer. HDGC is predominantly caused by germline pathogenic variants in the CDH1 gene, and more rarely in the CTNNA1 gene. Furthermore, the International Gastric Cancer Linkage Consortium (IGCLC) guidelines do not clarify whether or not mixed gastric cancer (with a diffuse component) should be considered in the HDGC genetic testing criteria. We aimed to evaluate the contribution of CTNNA1 and CTNND1 germline variants to HDGC. Additionally, we also intended to compare the frequencies of CDH1 and CTNNA1 (and eventually CTNND1) germline variants between patients with diffuse and mixed gastric carcinomas to evaluate if genetic testing for these genes should or should not be considered in patients with the latter. We analyzed the CDH1 gene in 67 cases affected with early-onset/familial mixed gastric carcinomas and the CTNNA1 and CTNND1 genes in 208 cases with diffuse or mixed gastric cancer who had tested negative for CDH1 pathogenic germline variants. A deleterious CTNNA1 germline variant was found in 0.7% (1/141) of diffuse gastric cancer patients meeting the 2020 IGCLC criteria, as compared to the rate of 2.8% of CDH1 deleterious variants found by us in this setting. No deleterious variants were found in CTNND1, but six variants of uncertain significance were identified in this gene. We did not find any pathogenic CDH1, CTNNA1 or CTNND1 variant in index patients with early-onset/familial mixed gastric cancer, so there is no evidence that supports including this tumor type in the testing criteria for germline variants in these genes. The role of the CTNND1 gene in inherited gastric cancer predisposition is still unclear.
Collapse
Affiliation(s)
- Joana Guerra
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Doctoral Programme in Biomedical Sciences, School Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Carla Pinto
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
- Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto, 4200-072 Porto, Portugal
| | - Pedro Pinto
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
| | - Manuela Pinheiro
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
| | - Catarina Santos
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Ana Peixoto
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Carla Escudeiro
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Ana Barbosa
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Miguel Porto
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
| | - Inês Francisco
- Molecular Pathobiology Research Unit, Portuguese Oncology Institute of Lisbon, 1099-023 Lisbon, Portugal; (I.F.); (C.A.)
| | - Paula Lopes
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (P.L.); (A.R.I.); (A.L.C.)
| | - Ana Raquel Isidoro
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (P.L.); (A.R.I.); (A.L.C.)
| | - Ana Luísa Cunha
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (P.L.); (A.R.I.); (A.L.C.)
| | - Cristina Albuquerque
- Molecular Pathobiology Research Unit, Portuguese Oncology Institute of Lisbon, 1099-023 Lisbon, Portugal; (I.F.); (C.A.)
| | - Isabel Claro
- Gastroenterology Department, Portuguese Oncology Institute of Lisbon, 1099-023 Lisbon, Portugal;
- Familiar Cancer Risk Clinic, Portuguese Oncology Institute of Lisbon, 1099-023 Lisbon, Portugal
| | - Carla Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, 4200-135 Porto, Portugal;
- IPATIMUP-Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
- FMUP-Faculty of Medicine of the University of Porto, 4100-179 Porto, Portugal
| | - João Silva
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Medical Genetics Department, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Manuel R. Teixeira
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal; (J.G.); (C.P.); (P.P.); (M.P.); (C.S.); (A.P.); (C.E.); (A.B.); (M.P.); (J.S.)
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
4
|
Torabian P, Yousefi H, Fallah A, Moradi Z, Naderi T, Delavar MR, Ertas YN, Zarrabi A, Aref AR. Cancer stem cell-mediated drug resistance: A comprehensive gene expression profile analysis in breast cancer. Pathol Res Pract 2023; 246:154482. [PMID: 37196466 DOI: 10.1016/j.prp.2023.154482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/19/2023]
Abstract
Breast cancer is the most frequently diagnosed malignancy in women and a major public health concern. In the current report, differential expression of the breast cancer resistance promoting genes with a focus on breast cancer stem cell related elements as well as the correlation of their mRNAs with various clinicopathologic characteristics, including molecular subtypes, tumor grade/stage, and methylation status, have been investigated using METABRIC and TCGA datasets. To achieve this goal, we downloaded gene expression data of breast cancer patients from TCGA and METABRIC. Then, statistical analyses were used to assess the correlation between the expression levels of stem cell related drug resistant genes and methylation status, tumor grades, various molecular subtypes, and some cancer hallmark gene sets such as immune evasion, metastasis, and angiogenesis. According to the results of this study, a number of stem cell related drug resistant genes are deregulated in breast cancer patients. Furthermore, we observe negative correlations between methylation of resistance genes and mRNA expression. There is a significant difference in the expression of resistance-promoting genes between different molecular subtypes. As mRNA expression and DNA methylation are clearly related, DNA methylation might be a mechanism that regulates these genes in breast cancer cells. As indicated by the differential expression of resistance-promoting genes among various breast cancer molecular subtypes, these genes may function differently in different subtypes of breast cancer. In conclusion, significant deregulation of resistance-promoting factors indicates that these genes may play a significant role in the development of breast cancer.
Collapse
Affiliation(s)
- Pedram Torabian
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Aysan Fallah
- Department of hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Moradi
- Department of hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tohid Naderi
- Department of Laboratory Hematology and Blood Bank, School of Allied Medicine, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey; Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396 Istanbul, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Ornella MSC, Badrinath N, Kim KA, Kim JH, Cho E, Hwang TH, Kim JJ. Immunotherapy for Peritoneal Carcinomatosis: Challenges and Prospective Outcomes. Cancers (Basel) 2023; 15:cancers15082383. [PMID: 37190310 DOI: 10.3390/cancers15082383] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Peritoneal metastasis, also known as peritoneal carcinomatosis (PC), is a refractory cancer that is typically resistant to conventional therapies. The typical treatment for PC is a combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Recently, research in this area has seen significant advances, particularly in immunotherapy as an alternative therapy for PC, which is very encouraging. Catumaxomab is a trifunctional antibody intraperitoneal (IP) immunotherapy authorized in Europe that can be used to diminish malignant ascites by targeting EpCAM. Intraperitoneal (IP) immunotherapy breaks immunological tolerance to treat peritoneal illness. Increasing T-cell responses and vaccination against tumor-associated antigens are two methods of treatment. CAR-T cells, vaccine-based therapeutics, dendritic cells (DCs) in combination with pro-inflammatory cytokines and NKs, adoptive cell transfer, and immune checkpoint inhibitors are promising treatments for PC. Carcinoembryonic antigen-expressing tumors are suppressed by IP administration of CAR-T cells. This reaction was strengthened by anti-PD-L1 or anti-Gr1. When paired with CD137 co-stimulatory signaling, CAR-T cells for folate receptor cancers made it easier for T-cell tumors to find their way to and stay alive in the body.
Collapse
Affiliation(s)
- Mefotse Saha Cyrelle Ornella
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Narayanasamy Badrinath
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Kyeong-Ae Kim
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Jung Hee Kim
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Euna Cho
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Tae-Ho Hwang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Jae-Joon Kim
- Division of Hematology & Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
6
|
Huang J, Wang H, Xu Y, Li C, Lv X, Han X, Chen X, Chen Y, Yu Z. The Role of CTNNA1 in Malignancies: An Updated Review. J Cancer 2023; 14:219-230. [PMID: 36741258 PMCID: PMC9891874 DOI: 10.7150/jca.79236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
Catenin alpha 1 (CTNNA1), encoding α-catenin, is involved in several physiological activities, such as adherens junction synthesis and signal transduction. Recent studies have suggested additional functions for CTNNA1 malignancies. This review systematically summarizes the varying functions of CTNNA1 in different tumors and briefly describes the diverse pathways and mechanisms involved in different types of tumors. CTNNA1 is abnormally expressed in leukemia and solid tumor such as cancers of digestive system, genitourinary system and breast, and it's related to the occurrence, development, and prognosis of tumors. In addition, the possible physiological processes involving CTNNA1, such as methylation, miRNA interference, or regulatory axes, similar to those of CDH1, SETD2, and hsa-miR-30d-5p/GJA1 are also summarized here. The precise mechanism of CTNNA1 in most cancers remains uncertain; hence, additional pre-clinical studies of CTNNA1 are warranted for potential early tumor diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Jinhua Huang
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China.,College of Medicine, Shantou University, Shantou, 515041, Guangdong, China
| | - Huihui Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230023, China
| | - Yuting Xu
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China.,College of Medicine, Shantou University, Shantou, 515041, Guangdong, China
| | - Chunhua Li
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China
| | - Xinyue Lv
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China
| | - Xintong Han
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China
| | - Xiaochun Chen
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China
| | - Yu Chen
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China
| | - Zhiying Yu
- Department of Gynecology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, Guangdong, China.,✉ Corresponding author: Zhiying Yu, Shenzhen Second People's Hospital, 3002 Sungang West Road, Shenzhen, Guangdong, China, 518035. Tel: 0755-83366388; Fax: +86 83366388-3048; E-mail:
| |
Collapse
|
7
|
Coudert M, Drouet Y, Delhomelle H, Svrcek M, Benusiglio PR, Coulet F, Clark DF, Katona BW, van Hest LP, van der Kolk LE, Cats A, van Dieren JM, Nehoray B, Slavin T, Spier I, Hüneburg R, Lobo S, Oliveira C, Boussemart L, Masson L, Chiesa J, Schwartz M, Buecher B, Golmard L, Bouvier AM, Bonadona V, Stoppa-Lyonnet D, Lasset C, Colas C. First estimates of diffuse gastric cancer risks for carriers of CTNNA1 germline pathogenic variants. J Med Genet 2022; 59:1189-1195. [PMID: 36038258 DOI: 10.1136/jmg-2022-108740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/02/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Pathogenic variants (PV) of CTNNA1 are found in families fulfilling criteria for hereditary diffuse gastric cancer (HDGC) but no risk estimates were available until now. The aim of this study is to evaluate diffuse gastric cancer (DGC) risks for carriers of germline CTNNA1 PV. METHODS Data from published CTNNA1 families were updated and new families were identified through international collaborations. The cumulative risk of DGC by age for PV carriers was estimated with the genotype restricted likelihood (GRL) method, taking into account non-genotyped individuals and conditioning on all observed phenotypes and genotypes of the index case to obtain unbiased estimates. A non-parametric (NP) and the Weibull functions were used to model the shape of penetrance function with the GRL. Kaplan-Meier incidence curve and standardised incidence ratios were also computed. A 'leave-one-out' strategy was used to evaluate estimate uncertainty. RESULTS Thirteen families with 46 carriers of PV were included. The cumulative risks of DGC at 80 years for carriers of CTNNA1 PV are 49% and 57%, respectively with the Weibull GRL and NP GRL methods. Risk ratios to population incidence reach particularly high values at early ages and decrease with age. At 40 years, they are equal to 65 and 833, respectively with the Weibull GRL and NP GRL. CONCLUSION This is the largest series of CTNNA1 families that provides the first risk estimates of GC. These data will help to improve management and surveillance for these patients and support inclusion of CTNNA1 in germline testing panels.
Collapse
Affiliation(s)
- Marie Coudert
- Département de Génétique, Institut Curie, Paris, France
| | - Youenn Drouet
- CNRS UMR 5558 LBBE, Université de Lyon, Villeurbanne, France.,Département Prévention et Santé Publique, Centre Léon Bérard, Lyon, France
| | | | - Magali Svrcek
- AP-HP, Saint-Antoine Hospital, Department of Pathology, Sorbonne Université, Paris, France
| | - Patrick R Benusiglio
- Département de Génétique Médicale, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Florence Coulet
- Département de Génétique Médicale, AP-HP, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Dana Farengo Clark
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Liselotte P van Hest
- Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Annemieke Cats
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jolanda M van Dieren
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bita Nehoray
- Division of Clinical Cancer Genomics, City of Hope, Duarte, California, USA
| | - Thomas Slavin
- Departments of Medical Oncology and Population Sciences, City of Hope, Duarte, California, USA
| | - Isabel Spier
- Institute of Human Genetics/National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Robert Hüneburg
- Department of Internal Medicine/National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Silvana Lobo
- IPATIMUP-Institut of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,i3s, Universidade do Porto Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Carla Oliveira
- Instituto de Investigação e Inovação em Saúde & Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | | | | | - Jean Chiesa
- Génétique, Hopital Universitaire Caremeau, Nimes, France
| | | | - Bruno Buecher
- Département de Génétique, Institut Curie, Paris, France
| | - Lisa Golmard
- Département de Génétique, Institut Curie, Paris, France
| | - Anne-Marie Bouvier
- Digestive Cancer Registry of Burgundy, UMR 1231, Réseau FRANCIM (réseau Français des registres du cancer), Burgundy Franche-Comté University, Dijon, France
| | - Valérie Bonadona
- CNRS UMR 5558 LBBE, Université de Lyon, Villeurbanne, France.,Unité Clinique d'Oncologie génétique, Centre Leon Berard, Lyon, France
| | - Dominique Stoppa-Lyonnet
- Département de Génétique, Institut Curie, Paris, France.,INSERM U830, Université de Paris, Paris, France
| | - Christine Lasset
- CNRS UMR 5558 LBBE, Université de Lyon, Villeurbanne, France.,Département Prévention et Santé Publique, Centre Léon Bérard, Lyon, France
| | - Chrystelle Colas
- Département de Génétique, Institut Curie, Paris, France .,INSERM U830, Université de Paris, Paris, France
| |
Collapse
|
8
|
Castillo D, Yuan TA, Nehoray B, Cervantes A, Tsang KK, Yang K, Sand SR, Mokhnatkin J, Herzog J, Slavin TP, Hyman S, Schwartz A, Ebert BL, Amos CI, Garber JE, Weitzel JN. Clonal Hematopoiesis and Mosaicism Revealed by a Multi-Tissue Analysis of Constitutional TP53 Status. Cancer Epidemiol Biomarkers Prev 2022; 31:1621-1629. [PMID: 35654360 PMCID: PMC9866580 DOI: 10.1158/1055-9965.epi-21-1296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/02/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Though germline TP53 pathogenic/likely pathogenic variants (PV) are associated with Li-Fraumeni syndrome, many detected by multigene panels represent aberrant clonal expansion (ACE), most due to clonal hematopoiesis (CH). Discerning ACE/CH from germline variants and postzygotic mosaicism (PZM) is critically needed for risk assessment and management. METHODS Participants in the Li-Fraumeni & TP53 Understanding & Progress (LiFT UP) study with a TP53 PV were eligible. Demographics, personal/family cancer history, and clinical laboratory test reports were obtained. DNA from multiple tissues was analyzed using a custom QIAseq assay (ACE panel) that included TP53 and other CH-associated genes; the ACE panel and eyebrow follicles were assessed in a workflow to discern TP53 PV clinical categories. RESULTS Among 134 participants there was a significant difference for the age at diagnosis (P < 0.001), component cancers (P = 0.007), and clinical testing criteria (P < 0.001), comparing germline with PZM or ACE. ACE panel analysis of DNA from 55 sets of eyebrow follicles (mean 1.4 ug) and 36 formalin-fixed, paraffin imbedded tissues demonstrated low variance (SE, 3%; P = 0.993) for TP53 variant allele fraction, with no significant difference (P = 0.965) between tissue types, and detected CH gene PVs. Of 55 multi-tissue cases, germline status was confirmed for 20, PZM in seven, ACE for 25, and three were indeterminate. Additional CH variants were detected in six ACE and two germline cases. CONCLUSIONS We demonstrated an effective approach and tools for discerning germline TP53 status. IMPACT Discernment of PZM and TP53-driven CH increases diagnostic accuracy and enables risk-appropriate care.
Collapse
Affiliation(s)
| | - Tze-An Yuan
- City of Hope Cancer Center, Duarte, CA 91010, USA
| | - Bita Nehoray
- City of Hope Cancer Center, Duarte, CA 91010, USA
| | | | | | - Kai Yang
- City of Hope Cancer Center, Duarte, CA 91010, USA
| | | | | | - Josef Herzog
- City of Hope Cancer Center, Duarte, CA 91010, USA
| | | | - Sophie Hyman
- Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | - Jeffrey N. Weitzel
- City of Hope Cancer Center, Duarte, CA 91010, USA
- Latin American School of Oncology, Los Angeles, CA, USA
| |
Collapse
|
9
|
Identification of a Five-Gene Panel to Assess Prognosis for Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5593619. [PMID: 35187167 PMCID: PMC8850031 DOI: 10.1155/2022/5593619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
Methods Two datasets were used as training and validation cohorts to establish the predictive model. We used three types of screening criteria: background analysis, pathway analysis, and functional analysis provided by the cBioportal website. Fisher's exact test and multivariable logistic regression were performed to screen out related genes. Furthermore, we performed receiver operating characteristic (ROC) and Kaplan–Meier curve analyses to evaluate the correlation between the selected genes and overall survival. Result We screened five genes (KNL1, NRXN1, C6, CCDC169-SOHLH2, and TTN) that were highly related to recurrence of GC. The area under the receiver operating characteristic (ROC) curve was 0.813, which was much higher than that of the baseline model (AUC = 0.699). This result suggested that the mutation of five selected genes had a significant effect on the prediction of recurrence compared with other factors (age, stages, history, etc.). Furthermore, the Kaplan-Meier estimator also revealed that the mutation of five genes positively correlated with patient survival. Conclusions The patients who have mutations in these five genes may experience longer survival than those who do not have mutations. This five-gene panel will likely be a practical tool for prognostic evaluation and will provide another possible way for clinicians to determine therapy.
Collapse
|
10
|
Garcia-Pelaez J, Barbosa-Matos R, São José C, Sousa S, Gullo I, Hoogerbrugge N, Carneiro F, Oliveira C. Gastric cancer genetic predisposition and clinical presentations: Established heritable causes and potential candidate genes. Eur J Med Genet 2021; 65:104401. [PMID: 34871783 DOI: 10.1016/j.ejmg.2021.104401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 11/10/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Tumour risk syndromes (TRS) are characterized by an increased risk of early-onset cancers in a familial context. High cancer risk is mostly driven by loss-of-function variants in a single cancer-associated gene. Presently, predisposition to diffuse gastric cancer (DGC) is explained by CDH1 and CTNNA1 pathogenic and likely pathogenic variants (P/LP), causing Hereditary Diffuse Gastric Cancer (HDGC); while APC promoter 1B single nucleotide variants predispose to Gastric Adenocarcinoma and Proximal Polyposis of the Stomach (GAPPS). Familial Intestinal Gastric Cancer (FIGC), recognized as a GC-predisposing disease, remains understudied and genetically unsolved. GC can also occur in the spectrum of other TRS. Identification of heritable causes allows defining diagnostic testing criteria, helps to clinically classify GC families into the appropriate TRS, and allows performing pre-symptomatic testing identifying at-risk individuals for downstream surveillance, risk reduction and/or treatment. However, most of HDGC, some GAPPS, and most FIGC patients/families remain unsolved, expecting a heritable factor to be discovered. The missing heritability in GC-associated tumour risk syndromes (GC-TRS) is likely explained not by a single major gene, but by a diversity of genes, some, predisposing to other TRS. This would gain support if GC-enriched small families or apparently isolated early-onset GC cases were hiding a family history compatible with another TRS. Herein, we revisited current knowledge on GC-TRS, and searched in the literature for individuals/families bearing P/LP variants predisposing for other TRS, but whose probands display a clinical presentation and/or family history also fitting GC-TRS criteria. We found 27 families with family history compatible with HDGC or FIGC, harbouring 28 P/LP variants in 16 TRS-associated genes, mainly associated with DNA repair. PALB2 or BRCA2 were the most frequently mutated candidate genes in individuals with family history compatible with HDGC and FIGC, respectively. Consolidation of PALB2 and BRCA2 as HDGC- or FIGC-associated genes, respectively, holds promise and worth additional research. This analysis further highlighted the influence, that proband's choice and small or unreported family history have, for a correct TRS diagnosis, genetic screening, and disease management. In this review, we provide a rational for identification of particularly relevant candidate genes in GC-TRS.
Collapse
Affiliation(s)
- José Garcia-Pelaez
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Barbosa-Matos
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; International Doctoral Programme in Molecular and Cellular Biotechnology Applied to Health Sciences from Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Celina São José
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sónia Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Irene Gullo
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal; Centro Hospitalar e Universitário S. João, Porto, Portugal
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal; Centro Hospitalar e Universitário S. João, Porto, Portugal
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal.
| |
Collapse
|
11
|
Ruiz Hispán E, Pedregal M, Cristobal I, García-Foncillas J, Caramés C. Immunotherapy for Peritoneal Metastases from Gastric Cancer: Rationale, Current Practice and Ongoing Trials. J Clin Med 2021; 10:4649. [PMID: 34682772 PMCID: PMC8539177 DOI: 10.3390/jcm10204649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
Peritoneal metastases from gastric cancer play a key role in the fatal prognosis of the disease. The lack of efficacy of actual therapeutic approaches together with the outcomes achieved with checkpoint inhibitors in gastric cancer compel us to address the current state-of-the-art immunotherapy treatment of peritoneal dissemination. The immunogenicity of the peritoneum has been described to be particularly active at omentum and peritoneal lymph nodes. Also, both innate and acquired immunity seems to be involved at different molecular levels. Recent works show PDL1 expression being less present at the peritoneal level; however, some clinical trials have begun to yield results. For example, the ATTRACTION-2 trial has demonstrated the activity of Nivolumab in heavily pretreated patients even though peritoneal metastases were diagnosed in a 30% of them. Despite positive results in the metastatic setting, peritoneal responses to systemic checkpoint inhibitors remains unclear, therefore, new strategies for intraperitoneal immunotherapy are being proposed for different ongoing clinical trials.
Collapse
Affiliation(s)
- Eva Ruiz Hispán
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Manuel Pedregal
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Ion Cristobal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM Madrid, 28040 Madrid, Spain;
| | - Jesús García-Foncillas
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Cristina Caramés
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| |
Collapse
|
12
|
Herzog JS, Chavarri-Guerra Y, Castillo D, Abugattas J, Villarreal-Garza C, Sand S, Clague-Dehart J, Alvarez-Gómez RM, Wegman-Ostrosky T, Mohar A, Mora P, Del Toro-Valero A, Daneri-Navarro A, Rodriguez Y, Cruz-Correa M, Ashton-Prolla P, Alemar B, Mejia R, Gallardo L, Shaw R, Yang K, Cervantes A, Tsang K, Nehoray B, Barrera Saldana H, Neuhausen S, Weitzel JN. Genetic epidemiology of BRCA1- and BRCA2-associated cancer across Latin America. NPJ Breast Cancer 2021; 7:107. [PMID: 34413315 PMCID: PMC8377150 DOI: 10.1038/s41523-021-00317-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
The prevalence and contribution of BRCA1/2 (BRCA) pathogenic variants (PVs) to the cancer burden in Latin America are not well understood. This study aims to address this disparity. BRCA analyses were performed on prospectively enrolled Latin American Clinical Cancer Genomics Community Research Network participants via a combination of methods: a Hispanic Mutation Panel (HISPANEL) on MassARRAY; semiconductor sequencing; and copy number variant (CNV) detection. BRCA PV probability was calculated using BRCAPRO. Among 1,627 participants (95.2% with cancer), we detected 236 (14.5%) BRCA PVs; 160 BRCA1 (31% CNVs); 76 BRCA2 PV frequency varied by country: 26% Brazil, 9% Colombia, 13% Peru, and 17% Mexico. Recurrent PVs (seen ≥3 times), some region-specific, represented 42.8% (101/236) of PVs. There was no ClinVar entry for 14% (17/125) of unique PVs, and 57% (111/196) of unique VUS. The area under the ROC curve for BRCAPRO was 0.76. In summary, we implemented a low-cost BRCA testing strategy and documented a significant burden of non-ClinVar reported BRCA PVs among Latin Americans. There are recurrent, population-specific PVs and CNVs, and we note that the BRCAPRO mutation probability model performs adequately. This study helps address the gap in our understanding of BRCA-associated cancer in Latin America.
Collapse
Affiliation(s)
| | - Yanin Chavarri-Guerra
- Instituto Nacional de Ciencias Medicas y Nutrición, Salvador Zubiran, Mexico City, Mexico
| | | | | | - Cynthia Villarreal-Garza
- Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Monterrey, Mexico
- Instituto Nacional de Cancerología, México City, México
| | | | - Jessica Clague-Dehart
- City of Hope, Duarte, CA, USA
- School of Community & Global Health, Claremont Graduate University, Claremont, CA, USA
| | | | | | - Alejandro Mohar
- Instituto Nacional de Cancerología, México City, México
- Instituto de Investigaciones Biomédicas, Mexico City, Mexico
| | - Pamela Mora
- Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Azucena Del Toro-Valero
- Instituto Jalisciense de Cancerología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, México City, México
| | - Adrian Daneri-Navarro
- Instituto Jalisciense de Cancerología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, México City, México
| | | | - Marcia Cruz-Correa
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre and Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bárbara Alemar
- Hospital de Clínicas de Porto Alegre and Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Roche Pharmaceutical, Basel, Switzerland
| | | | | | - Robin Shaw
- Instituto Nacional de Cancerología, México City, México
| | | | | | | | | | | | | | - Jeffrey N Weitzel
- Latin American School of Oncology (Escuela Latinoamericana de Oncología), Tuxla Gutiérrez, Chiapas, Mexico.
| |
Collapse
|
13
|
Reckamp KL, Behrendt CE, Slavin TP, Gray SW, Castillo DK, Koczywas M, Cristea MC, Babski KM, Stearns D, Marcum CA, Rodriguez YP, Hass AJ, Vecchio MM, Mora P, Cervantes AE, Sand SR, Mejia RM, Tsou TC, Salgia R, Weitzel JN. Germline mutations and age at onset of lung adenocarcinoma. Cancer 2021; 127:2801-2806. [PMID: 33858029 PMCID: PMC8794435 DOI: 10.1002/cncr.33573] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND To identify additional at-risk groups for lung cancer screening, which targets persons with a long history of smoking and thereby misses younger or nonsmoking cases, the authors evaluated germline pathogenic variants (PVs) in patients with lung adenocarcinoma for an association with an accelerated onset. METHODS The authors assembled a retrospective cohort (1999-2018) of oncogenetic clinic patients with lung adenocarcinoma. Eligibility required a family history of cancer, data on smoking, and a germline biospecimen to screen via a multigene panel. Germline PVs (TP53/EGFR, BRCA2, other Fanconi anemia [FA] pathway genes, and non-FA DNA repair genes) were interrogated for associations with the age at diagnosis via an accelerated failure time model. RESULTS Subjects (n = 187; age, 28-89 years; female, 72.7%; Hispanic, 11.8%) included smokers (minimum of 5 pack-years; n = 65) and nonsmokers (lighter ever smokers [n = 18] and never smokers [n = 104]). Overall, 26.7% of the subjects carried 1 to 2 germline PVs: TP53 (n = 5), EGFR (n = 2), BRCA2 (n = 6), another FA gene (n = 11), or another DNA repair gene (n = 28). After adjustment for smoking, sex, and ethnicity, the diagnosis of lung adenocarcinoma was accelerated 12.2 years (95% confidence interval [CI], 2.5-20.6 years) by BRCA2 PVs, 9.0 years (95% CI, 0.5-16.5 years) by TP53/EGFR PVs, and 6.1 years (95% CI, -1.0 to 12.6 years) by PVs in other FA genes. PVs in other DNA repair genes showed no association. Germline associations did not vary by smoking. CONCLUSIONS Among lung adenocarcinoma cases, germline PVs (TP53, EGFR, BRCA2, and possibly other FA genes) may be associated with an earlier onset. With further study, the criteria for lung cancer screening may need to include carriers of high-risk PVs, and findings could influence precision therapy and reduce lung cancer mortality by earlier stage diagnosis.
Collapse
Affiliation(s)
- Karen L Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, California
| | | | - Thomas P Slavin
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Stacy W Gray
- City of Hope Comprehensive Cancer Center, Duarte, California
| | | | | | | | | | | | | | | | | | | | - Pamela Mora
- Instituto Nacional de Enfermedades Neoplasicas, Lima, Peru
| | | | - Sharon R Sand
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Rosa M Mejia
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Terrence C Tsou
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Ravi Salgia
- City of Hope Comprehensive Cancer Center, Duarte, California
| | | |
Collapse
|
14
|
Zhang YJ, Yang Y, Wei Q, Xu T, Zhang XT, Gao J, Tan SY, Liu BR, Zhang JD, Chen XB, Wang ZJ, Qiu M, Wang X, Shen L, Wang XC. A multicenter study assessing the prevalence of germline genetic alterations in Chinese gastric-cancer patients. Gastroenterol Rep (Oxf) 2021; 9:339-349. [PMID: 34567566 PMCID: PMC8460096 DOI: 10.1093/gastro/goab020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/10/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Approximately 10% of patients with gastric cancer (GC) have a genetic predisposition toward the disease. However, there is scant knowledge regarding germline mutations in predisposing genes in the Chinese GC population. This study aimed to determine the spectrum and distribution of predisposing gene mutations among Chinese GC patients known to have hereditary high-risk factors for cancer. METHODS A total of 40 GC patients from 40 families were recruited from seven medical institutions in China. Next-generation sequencing was performed on 171 genes associated with cancer predisposition. For probands carrying pathogenic/likely pathogenic germline variants, Sanger sequencing was applied to validate the variants in the probands as well as their relatives. RESULTS According to sequencing results, 25.0% (10/40) of the patients carried a combined total of 10 pathogenic or likely pathogenic germline variants involving nine different genes: CDH1 (n = 1), MLH1 (n = 1), MSH2 (n = 1), CHEK2 (n = 1), BLM (n = 1), EXT2 (n = 1), PALB2 (n = 1), ERCC2 (n = 1), and SPINK1 (n = 2). In addition, 129 variants of uncertain significance were identified in 27 patients. CONCLUSIONS This study indicates that approximately one in every four Chinese GC patients with hereditary high risk factors may harbor pathogenic/likely pathogenic germline alterations in cancer-susceptibility genes. The results further indicate a unique genetic background for GC among Chinese patients.
Collapse
Affiliation(s)
- Yin-Jie Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P. R. China
| | - Yang Yang
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Qing Wei
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, P. R. China
| | - Ting Xu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Xiao-Tian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Jing Gao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Si-Yi Tan
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Bao-Rui Liu
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Jing-Dong Zhang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, P. R. China
| | - Xiao-Bing Chen
- Department of Gastroenterology and Medical Oncology, Henan Cancer Hospital (Affiliated Cancer Hospital of Zhengzhou University), Zhengzhou, Henan, P. R. China
| | - Zhao-Jie Wang
- Department of Oncology, Henan Provincial People’s Hospital, Zhengzhou, Henan, P. R. China
| | - Meng Qiu
- Department of Medical Oncology, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Xin Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Disease, Fourth Military Medical University, Xi’an, Shaanxi, P. R. China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Xi-Cheng Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, P. R. China
| |
Collapse
|
15
|
Lam-Chung CE, Rodríguez LL, Vázquez JA, Chávarri-Guerra Y, Arízaga-Ramírez R, Antonio OF, De Anda González J, López-Hernández MA, Weitzel JN, Castillo D, Gómez-Pérez FJ, Cuevas-Ramos D. A Novel, Likely Pathogenic MAX Germline Variant in a Patient With Unilateral Pheochromocytoma. J Endocr Soc 2021; 5:bvab085. [PMID: 34169220 PMCID: PMC8218934 DOI: 10.1210/jendso/bvab085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Indexed: 12/14/2022] Open
Abstract
Context Inherited MYC-associated factor X (MAX) gene pathogenic variants (PVs) increase risk for pheochromocytomas (PCCs) and/or paragangliomas (PGLs) in adults and children. There is little clinical experience with such mutations. Objective This report highlights an important approach. Methods Clinical assessment, including blood chemistry, imaging studies, and genetic testing were performed. Results A 38-year-old Hispanic woman was diagnosed with PCC in 2015, treated with adrenalectomy, and referred to endocrinology clinic. Notably, she presented to her primary care physician 3 years earlier complaining of left flank pain, intermittent diaphoresis, and holocranial severe headache. We confirmed severe hypertension (180/100 mm Hg) over multiple antihypertensive regimens. Biochemical and radiological studies workup revealed high plasma metanephrine of 255 pg/mL (normal range, < 65 pg/mL) and plasma normetanephrine of 240 pg/mL (normal range, < 196 pg/mL). A noncontrast computed tomography scan of the abdomen revealed a 4.2 × 4.3 × 4.9-cm, round-shaped and heterogenous contrast enhancement of the left adrenal gland, and a 2-mm nonobstructive left kidney stone. A presumptive diagnosis of secondary hypertension was made. After pharmacological therapy, laparoscopic left adrenalectomy was performed and confirmed the diagnosis of pheochromocytoma. Based on her age, family history, and a high suspicion for genetic etiology, genetic testing was performed that revealed the presence of a novel likely pathogenic variant involving a splice consensus sequence in the MAX gene, designated c0.64-2A > G. Conclusion The phenotype of MAX PV-related disease and paraganglioma are highlighted. The novel c0.64-2A > G mutation is reported here and should be considered in the diagnostic workup of similar cases.
Collapse
Affiliation(s)
- César Ernesto Lam-Chung
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Larissa López Rodríguez
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Jazmín Arteaga Vázquez
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Yanin Chávarri-Guerra
- Department of Hemato-Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Rebeca Arízaga-Ramírez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico.,Department of Radiology and Imaging, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Orlando Falcon Antonio
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Jazmín De Anda González
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - María Aurelia López-Hernández
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | | | | | - Francisco Javier Gómez-Pérez
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan 14000, Mexico City, Mexico
| |
Collapse
|
16
|
Carli ALE, Afshar-Sterle S, Rai A, Fang H, O'Keefe R, Tse J, Ferguson FM, Gray NS, Ernst M, Greening DW, Buchert M. Cancer stem cell marker DCLK1 reprograms small extracellular vesicles toward migratory phenotype in gastric cancer cells. Proteomics 2021; 21:e2000098. [PMID: 33991177 DOI: 10.1002/pmic.202000098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/15/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a putative cancer stem cell marker, a promising diagnostic and prognostic maker for malignant tumors and a proposed driver gene for gastric cancer (GC). DCLK1 overexpression in a majority of solid cancers correlates with lymph node metastases, advanced disease and overall poor-prognosis. In cancer cells, DCLK1 expression has been shown to promote epithelial-to-mesenchymal transition (EMT), driving disruption of cell-cell adhesion, cell migration and invasion. Here, we report that DCLK1 influences small extracellular vesicle (sEV/exosome) biogenesis in a kinase-dependent manner. sEVs isolated from DCLK1 overexpressing human GC cell line MKN1 (MKN1OE -sEVs), promote the migration of parental (non-transfected) MKN1 cells (MKN1PAR ). Quantitative proteome analysis of MKN1OE -sEVs revealed enrichment in migratory and adhesion regulators (STRAP, CORO1B, BCAM, COL3A, CCN1) in comparison to MKN1PAR -sEVs. Moreover, using DCLK1-IN-1, a specific small molecule inhibitor of DCLK1, we reversed the increase in sEV size and concentration in contrast to other EV subtypes, as well as kinase-dependent cargo selection of proteins involved in EV biogenesis (KTN1, CHMP1A, MYO1G) and migration and adhesion processes (STRAP, CCN1). Our findings highlight a specific role of DCLK1-kinase dependent cargo selection for sEVs and shed new light on its role as a regulator of signaling in gastric tumorigenesis.
Collapse
Affiliation(s)
- Annalisa L E Carli
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Shoukat Afshar-Sterle
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia
| | - Ryan O'Keefe
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Janson Tse
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Ernst
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Buchert
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
17
|
Machlowska J, Kapusta P, Szlendak M, Bogdali A, Morsink F, Wołkow P, Maciejewski R, Offerhaus GJA, Sitarz R. Status of CHEK2 and p53 in patients with early-onset and conventional gastric cancer. Oncol Lett 2021; 21:348. [PMID: 33747205 PMCID: PMC7967923 DOI: 10.3892/ol.2021.12609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/08/2021] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common cause of cancer-associated death. Based on the age at diagnosis, GC is divided into early-onset GC (EOGC; ≤45 years) and conventional GC (CGC; >45 years). Mutations in the cell cycle checkpoint kinase 2 (CHEK2) and TP53 genes are associated with several types of cancer; however, their genetic defects in GC remain poorly understood. The aim of the present study was to determine the subcellular distribution of the CHEK2 protein and its redistribution following DNA damage, to improve the understanding of the DNA damage response. Genetic alterations and patterns of expression of CHEK2 and p53 proteins were investigated to identify potential biological markers and indicators of GC development. Additionally, the affected signaling pathways and their clinical importance in GC development and associated syndromes were investigated. A total of 196 GC samples (89 CGC and 107 EOGC samples) were used in the present study. DNA from 53 samples (18 CGC and 35 EOGC samples) was sequenced using targeted next-generation sequencing technology to identify and compare common and rare mutations associated with GC. Subsequently, the cytoplasmic and nuclear expression levels of CHEK2, phosphorylated (p)-CHEK2 at threonine 68 and p53 in GC tissues were determined via immunohistochemistry. Sequencing resulted in the identification of 63 single nucleotide polymorphisms (SNPs) in the CHEK2 gene amongst 5 different variants, and the intron variant c.319+379A>G was the most common SNP. In the TP53 gene, 57 different alterations were detected amongst 9 variant types, and the missense variant c.215C>G was the most common. Nuclear CHEK2 expression was high in both the EOGC and CGC subtypes. However, the prevalence of cytoplasmic CHEK2 expression (P<0.001) and nuclear p-CHEK2 expression (P=0.011) was significantly higher in CGC compared with in EOGC tissues. There was a statistically significant difference between high and low cytoplasmic CHEK2 expression in patients with p53-positive EOGC compared with in patients with p53-positive CGC (P=0.002). The present study was designed to determine the association between CHEK2 and p53 expression patterns in patients with EOGC and CGC, as well as genetic alterations in the CHEK2 and TP53 genes.
Collapse
Affiliation(s)
- Julita Machlowska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Kraków, Poland
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Przemysław Kapusta
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Kraków, Poland
| | - Małgorzata Szlendak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
- Department of Surgical Oncology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Anna Bogdali
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Kraków, Poland
| | - Folkert Morsink
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Paweł Wołkow
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Kraków, Poland
| | - Ryszard Maciejewski
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - G. Johan A. Offerhaus
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Robert Sitarz
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Department of Surgery, Center of Oncology of The Lublin Region St. Jana z Dukli, 20-090 Lublin, Poland
| |
Collapse
|
18
|
Herrera-Pariente C, Capó-García R, Díaz-Gay M, Carballal S, Muñoz J, Llach J, Sánchez A, Bonjoch L, Arnau-Collell C, Soares de Lima Y, Golubicki M, Jung G, Lozano JJ, Castells A, Balaguer F, Bujanda L, Castellví-Bel S, Moreira L. Identification of New Genes Involved in Germline Predisposition to Early-Onset Gastric Cancer. Int J Mol Sci 2021; 22:1310. [PMID: 33525650 PMCID: PMC7866206 DOI: 10.3390/ijms22031310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/24/2022] Open
Abstract
The genetic cause for several families with gastric cancer (GC) aggregation is unclear, with marked relevance in early-onset patients. We aimed to identify new candidate genes involved in GC germline predisposition. Whole-exome sequencing (WES) of germline samples was performed in 20 early-onset GC patients without previous germline mutation identified. WES was also performed in nine tumor samples to analyze the somatic profile using SigProfilerExtractor tool. Sequencing germline data were filtered to select those variants with plausible pathogenicity, rare frequency and previously involved in cancer. Then, a manual filtering was performed to prioritize genes according to current knowledge and function. These genetic variants were prevalidated with Integrative Genomics Viewer 2.8.2 (IGV). Subsequently, a further selection step was carried out according to function and information obtained from tumor samples. After IGV and selection step, 58 genetic variants in 52 different candidate genes were validated by Sanger sequencing. Among them, APC, FAT4, CTNND1 and TLR2 seem to be the most promising genes because of their role in hereditary cancer syndromes, tumor suppression, cell adhesion and Helicobacter pylori recognition, respectively. These encouraging results represent the open door to the identification of new genes involved in GC germline predisposition.
Collapse
Affiliation(s)
- Cristina Herrera-Pariente
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Roser Capó-García
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Marcos Díaz-Gay
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sabela Carballal
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Jenifer Muñoz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Joan Llach
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Ariadna Sánchez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Laia Bonjoch
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Coral Arnau-Collell
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Yasmin Soares de Lima
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Mariano Golubicki
- Oncology Section, Hospital of Gastroenterology “Dr. C. B. Udaondo”, C1264 Buenos Aires, Argentina;
- Molecular Biology Laboratory, Hospital of Gastroenterology “Dr. C. B. Udaondo”, C1264 Buenos Aires, Argentina
| | - Gerhard Jung
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Juan José Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, 08036 Barcelona, Spain;
| | - Antoni Castells
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Francesc Balaguer
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Biodonostia Health Research Institute, Basque Country University (UPV/EHU), 20014 San Sebastián, Spain;
| | - Sergi Castellví-Bel
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| | - Leticia Moreira
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (R.C.-G.); (M.D.-G.); (S.C.); (J.M.); (J.L.); (A.S.); (L.B.); (C.A.-C.); (Y.S.d.L.); (G.J.); (A.C.); (F.B.)
| |
Collapse
|
19
|
Zhu B, Cui H, Xu W. Hydrogen inhibits the proliferation and migration of gastric cancer cells by modulating lncRNA MALAT1/miR-124-3p/EZH2 axis. Cancer Cell Int 2021; 21:70. [PMID: 33482814 PMCID: PMC7821405 DOI: 10.1186/s12935-020-01743-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Background Gastric cancer is one of the most prevalent and deadly malignancies without efficient treatment option. This study aimed to investigate the effect of hydrogen gas on the behavior of gastric cancer cells. Methods Gastric cancer cell lines MGC-803 and BGC-823 were treated with or without H2 /O2 gas mixture (66.7%:33.3% v/v). Proliferation and migration were assessed by MTT and scratch wound healing assays respectively. The expression of lncRNA MALAT1, miR-124-3p, and EZH2 was analyzed by real-time quantitative PCR and/or western blot. Tumor growth was estimated using xenograft mouse model. Results H2 gas significantly inhibited gastric tumor growth in vivo and the proliferation, migration, and lncRNA MALAT1 and EZH2 expression of gastric cancer cells while upregulated miR-124-3p expression. LncRNA MALAT1 overexpression abolished all the aforementioned effects of H2. LncRNA MALAT1 and miR-124-3p reciprocally inhibited the expression of each other. MiR-124-3p mimics abrogated lncRNA MALAT1 promoted EZH2 expression and gastric cancer cell proliferation and migration. Conclusions These data demonstrated that H2 might be developed as a therapeutics of gastric cancer and lncRNA MALAT1/miR-124-3p/EZH2 axis could be a target for intervention.
Collapse
Affiliation(s)
- Baocheng Zhu
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Hengguan Cui
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Weiqiang Xu
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China. .,Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park East Road, Qingpu District, Shanghai, 201700, China.
| |
Collapse
|
20
|
Establish a Scoring Model for High-Risk Population of Gastric Cancer and Study on the Pattern of Opportunistic Screening. Gastroenterol Res Pract 2020; 2020:5609623. [PMID: 33061960 PMCID: PMC7545415 DOI: 10.1155/2020/5609623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/08/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
Objective To investigate and study the related risk factors of gastric cancer (GC) patients, to establish a high-risk scoring model of GC by multiple logistic regression analysis, and to explore the establishment of a GC screening mode with clinical opportunistic screening as the main method, and by using the pattern of opportunistic screening to establish the screening of high-risk GC patients and the choice of screening methods in the clinical outpatient work. Methods Collected the epidemiological questionnaire of 99 GC cases and 284 non-GC patients (other chronic gastric diseases and normal) diagnosed by the General Hospital of Ningxia Medical University from October 2017 to March 2019. Serum pepsinogen (PG) levels were measured by enzyme-linked immunosorbent assay (ELISA) and confirmed Helicobacter pylori (Hp) infection in gastric mucosa tissues by Giemsa staining. Determined the high-risk factors and established a scoring model through unconditional logistic regression model analysis, and the ROC curve determined the cut-off value. Then, we followed up 26 patients of nongastric cancer patients constituted a validation group, which validated the model. Results The high-risk factors of GC included age ≥ 55, male, drinking cellar or well water, family history of GC, Hp infection, PGI ≤ 43.6 μg/L, and PGI/PGII ≤ 2.1. Established the high-risk model: Y = A × age + 30 × gender + 30 × drinking water + 30 × Hp infection + 50 × family history of GC + B × PG level. The ROC curve determined that the cut-off value for high-risk GC population was ≥155, and the area under the curve (AUC) was 0.875, the sensitivity and specificity were 87.9% and 71.5%. Conclusions According to the risk factors of GC, using statistical methods can establish a high-risk scoring model of GC, and the score ≥ 155 is divided into the screening cut-off value for high-risk GC population. Using this model for clinical outpatient GC screening is cost-effective and has high sensitivity and specificity.
Collapse
|
21
|
Yao X, Ajani JA, Song S. Molecular biology and immunology of gastric cancer peritoneal metastasis. Transl Gastroenterol Hepatol 2020; 5:57. [PMID: 33073052 DOI: 10.21037/tgh.2020.02.08] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022] Open
Abstract
Peritoneal metastases occur in 55-60% of patients with gastric cancer (GC) and are associated with a 2% 5-year overall survival rate. There are limited treatment options for these patients, and no targeted therapy or immunotherapy is available. Rational therapeutic targets remain to be found. In this review, we present the published literature and our own recent experience in molecular biology to identify important molecules and signaling pathways as well as cellular immunity involved in the peritoneal metastasis of GC. We also suggest potential novel strategies for improving the outcomes of GC patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Wang A, Li Z, Wang M, Jia S, Chen J, Ji K, Ji X, Zong X, Wu X, Zhang J, Li Z, Zhang L, Hu Y, Bu Z, Zheng Q, Ji J. Molecular characteristics of synchronous multiple gastric cancer. Theranostics 2020; 10:5489-5500. [PMID: 32373223 PMCID: PMC7196298 DOI: 10.7150/thno.42814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/15/2020] [Indexed: 12/24/2022] Open
Abstract
Rationale: Multiple gastric cancer (MGC) is characterized by the presence of more than two different tumors in the stomach. However, the clonal relationship and carcinogenesis of MGC remain unclear. We investigated the clonal relationship and role of germline mutations in the carcinogenesis of MGC. Methods: We gathered 16 multiple gastric cancer patients. Thirty-three tumor samples and sixteen normal gastric tissue or blood samples were obtained from January 2016 to December 2017. We also conducted analyses for 208 gastric cancer and 49 esophagogastric junction cancer (GC-EGJ) tumors from TCGA. DNA extraction from our samples was conducted for whole-exome sequencing (WES). Results: Tumor mutation burden (TMB) was not statistically significant within database and our data in the GC-EGJ (P=0.0591) and GC groups (P=0.3113). The mutation spectrum and signatures also showed uniform distributions in GC and GC-EGJ groups within our data and TCGA database. Among sixteen patients, four were identified as monoclonal, in which 11, 10, 26 and 6 somatic mutations were shared within different tumors of P7, P8, P9 and P16, respectively. However, no common mutation between different tumors of the same patient was found among the other 12 patients. After identifying predisposing genes, we found that germline MSH2 and NCOR2 mutations were significantly dominant in 8/12 and 10/12 of genetic MGC patients. Additionally, all patients were identified with MSH2 mutations in cancer samples of those genetic MGC patients. Taking genetic MGCs as a whole, we identified that TP53 were significantly mutated in 14 of 25 tumor samples. Main conclusions: WES analyses are suggestive of monoclonal and polyclonal origin of MGC, which may promote the classification of MGC into genetic and metastatic MGC. For patients with genetic MGC, germline MSH2 X314_splice variants may contribute to carcinogenesis, thus prompting the consideration of more radical surgery and/or anti-PD-1/PD-L1 therapy.
Collapse
|
23
|
CDH1 Mutation Distribution and Type Suggests Genetic Differences between the Etiology of Orofacial Clefting and Gastric Cancer. Genes (Basel) 2020; 11:genes11040391. [PMID: 32260281 PMCID: PMC7231129 DOI: 10.3390/genes11040391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 01/16/2023] Open
Abstract
Pathogenic variants in CDH1, encoding epithelial cadherin (E-cadherin), have been implicated in hereditary diffuse gastric cancer (HDGC), lobular breast cancer, and both syndromic and non-syndromic cleft lip/palate (CL/P). Despite the large number of CDH1 mutations described, the nature of the phenotypic consequence of such mutations is currently not able to be predicted, creating significant challenges for genetic counselling. This study collates the phenotype and molecular data for available CDH1 variants that have been classified, using the American College of Medical Genetics and Genomics criteria, as at least ‘likely pathogenic’, and correlates their molecular and structural characteristics to phenotype. We demonstrate that CDH1 variant type and location differ between HDGC and CL/P, and that there is clustering of CL/P variants within linker regions between the extracellular domains of the cadherin protein. While these differences do not provide for exact prediction of the phenotype for a given mutation, they may contribute to more accurate assessments of risk for HDGC or CL/P for individuals with specific CDH1 variants.
Collapse
|
24
|
van der Post RS, Oliveira C, Guilford P, Carneiro F. Hereditary gastric cancer: what's new? Update 2013-2018. Fam Cancer 2019; 18:363-367. [PMID: 30989426 DOI: 10.1007/s10689-019-00127-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Around 10-20% of gastric cancer patients have relatives with a diagnosis of GC and in 1-3% of patients a genetic cause can be confirmed. Histopathologically, GC is classified into intestinal-type, with glandular growth, and diffuse-type with poorly cohesive growth pattern often with signet ring cells. Familial or hereditary GC is classified into hereditary diffuse GC (HDGC), familial intestinal GC (FIGC) and polyposis forms. This review focuses on recent research findings and new concepts of hereditary GC.
Collapse
Affiliation(s)
- Rachel S van der Post
- Department of Pathology, Radboud university medical centre, Nijmegen, The Netherlands
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Parry Guilford
- Cancer Genetics Laboratory, Department of Biochemistry, Centre for Translational Cancer Research, Te Aho Matatū, University of Otago, Dunedin, Aotearoa, New Zealand
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal. .,Ipatimup - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal. .,Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
25
|
Identification of c.1531C>T Pathogenic Variant in the CDH1 Gene as a Novel Germline Mutation of Hereditary Diffuse Gastric Cancer. Int J Mol Sci 2019; 20:ijms20204980. [PMID: 31600923 PMCID: PMC6829381 DOI: 10.3390/ijms20204980] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
Germline pathogenic variants in the CDH1 gene are a well-established cause of hereditary diffuse gastric cancer (HDGC) syndrome. The aim of this study was to characterize CDH1 mutations associated with HDGC from Chile, a country with one of the highest incidence and mortality rates in the world for gastric cancer (GC). Here, we prospectively include probands with family history/early onset of diffuse-type of GC. The whole coding sequence of the CDH1 gene was sequenced from genomic DNA in all patients, and a multidisciplinary team managed each family member with a pathogenic sequence variant. Thirty-six cases were included (median age 44 years/male 50%). Twenty-seven (75%) patients had diffuse-type GC at ≤50 years of age and 19 (53%) had first or second-degree family members with a history of HDGC. Two cases (5.5%) carried a non-synonymous germline sequence variant in the CDH1 gene: (a) The c.88C>A missense variant was found in a family with three diffuse-type GC cases; and (b) c.1531C>T a nonsense pathogenic variant was identified in a 22-year-old proband with no previous family history of HDGC. Of note, six family members carry the same nonsense pathogenic variant. Prophylactic gastrectomy in the proband's sister revealed stage I signet-ring cell carcinoma. The finding of 1531C>T pathogenic variant in the CDH1 in proband with no previous family history of HDGC warrants further study to uncover familial clustering of disease in CDH1 negative patients. This finding may be particularly relevant in high incidence countries, such as the case in this report.
Collapse
|
26
|
Multigene Panel Testing Increases the Number of Loci Associated with Gastric Cancer Predisposition. Cancers (Basel) 2019; 11:cancers11091340. [PMID: 31514334 PMCID: PMC6769562 DOI: 10.3390/cancers11091340] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/02/2019] [Accepted: 09/08/2019] [Indexed: 12/24/2022] Open
Abstract
The main gene involved in gastric cancer (GC) predisposition is CDH1, the pathogenic variants of which are associated with diffuse-type gastric cancer (DGC) and lobular breast cancer (LBC). CDH1 only explains a fraction (10–50%) of patients suspected of DGC/LBC genetic predisposition. To identify novel susceptibility genes, thus improving the management of families at risk, we performed a multigene panel testing on selected patients. We searched for germline pathogenic variants in 94 cancer-related genes in 96 GC or LBC Italian patients with early-onset and/or family history of GC. We found CDH1 pathogenic variants in 10.4% of patients. In 11.5% of cases, we identified loss-of-function variants in BRCA1, BRCA2, PALB2, and ATM breast/ovarian cancer susceptibility genes, as well as in MSH2, PMS2, BMPR1A, PRF1, and BLM genes. In 78.1% of patients, we did not find any variants with clear-cut clinical significance; however, 37.3% of these cases harbored rare missense variants predicted to be damaging by bioinformatics tools. Multigene panel testing decreased the number of patients that would have otherwise remained genetically unexplained. Besides CDH1, our results demonstrated that GC pathogenic variants are distributed across a number of susceptibility genes and reinforced the emerging link between gastric and breast cancer predisposition.
Collapse
|
27
|
Slavin TP, Weitzel JN, Neuhausen SL, Schrader KA, Oliveira C, Karam R. Genetics of gastric cancer: what do we know about the genetic risks? Transl Gastroenterol Hepatol 2019; 4:55. [PMID: 31463414 DOI: 10.21037/tgh.2019.07.02] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/11/2019] [Indexed: 01/11/2023] Open
Abstract
An appreciable number of patients with gastric cancer have an underlying hereditary cancer susceptibility syndrome as the cause of their gastric cancer, particularly those with early onset gastric cancer or a family history of gastric or other cancers. Pathogenic germline variants in specific genes account for the known gastric cancer predisposition syndromes. Germline genetic testing can identify individuals and their family members who carry inherited pathogenic gene variants, and thus have increased risk of developing gastric or other cancers. Ideally, germline pathogenic variants can be identified in family members before the onset of disease, when early detection or prevention strategies can be implemented most effectively to decrease gastric cancer- related morbidity and mortality. This article reviews some of the currently known gastric cancer predisposition syndromes and their associated cancer risks. We also discuss current research and advances in the field of genetic gastric cancer susceptibility.
Collapse
Affiliation(s)
- Thomas Paul Slavin
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA.,Department of Population Sciences, City of Hope, Duarte, CA, USA.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Jeffrey N Weitzel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA.,Department of Population Sciences, City of Hope, Duarte, CA, USA.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Susan L Neuhausen
- Department of Population Sciences, City of Hope, Duarte, CA, USA.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Kasmintan A Schrader
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Ambry Genetics Laboratories, Aliso Viejo, CA, USA
| | - Carla Oliveira
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,IPATIMUP, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Rachid Karam
- Ambry Genetics Laboratories, Aliso Viejo, CA, USA
| |
Collapse
|
28
|
Sokolova O, Naumann M. Crosstalk Between DNA Damage and Inflammation in the Multiple Steps of Gastric Carcinogenesis. Curr Top Microbiol Immunol 2019; 421:107-137. [PMID: 31123887 DOI: 10.1007/978-3-030-15138-6_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Over the last years, intensive investigations in molecular biology and cell physiology extended tremendously the knowledge about the association of inflammation and cancer. In frame of this paradigm, the human pathogen Helicobacter pylori triggers gastritis and gastric ulcer disease, and contributes to the development of gastric cancer. Mechanisms, by which the bacteria-induced inflammation in gastric mucosa leads to intestinal metaplasia and carcinoma, are represented in this review. An altered cell-signaling response and increased production of free radicals by epithelial and immune cells account for the accumulation of DNA damage in gastric mucosa, if infection stays untreated. Host genetics and environmental factors, especially diet, can accelerate the process, which offers the opportunity of intervention based on a balanced nutrition. It is supposed that inflammation might influence stem- or progenitor cells in gastric tissue predisposing for metaplasia or tumor relapse. Herein, DNA is strongly mutated and labile, which restricts therapy options. Thus, the understanding of the mechanisms that underlie gastric carcinogenesis will be of preeminent importance for the development of strategies for screening and early detection. As most gastric cancer patients face late-stage disease with a poor overall survival, the development of multi-targeted therapeutic intervention strategies is a major challenge for the future.
Collapse
Affiliation(s)
- Olga Sokolova
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| |
Collapse
|
29
|
Qiao H, Wang Y, Zhu B, Jiang L, Yuan W, Zhou Y, Guan Q. Enolase1 overexpression regulates the growth of gastric cancer cells and predicts poor survival. J Cell Biochem 2019; 120:18714-18723. [PMID: 31218757 DOI: 10.1002/jcb.29179] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 11/06/2022]
Abstract
Gastric cancer has become the third most common cancer around the world. In patients with gastric cancer, the 5-year survival rate is still low. However, the mechanism underlying gastric cancer remains largely unknown. As a glycolytic enzyme, enolase 1 (ENO1) is widely expressed in most tissues. The functions of ENO1 have been reported in various types of cancer. Here in this study, we identified that ENO1 promoted the growth of gastric cancer cells through diverse mechanisms. Our immunohistochemical, bioinformatic and Western blot data showed that ENO1 was significantly overexpressed in human gastric cancer cell lines and tissues. The survival analysis revealed that ENO1 overexpression predicted poor survival in the patients suffering gastric cancer. Knockdown of ENO1 expression repressed the rate of proliferation and capacity of colony formation in two human gastric cancer cell lines (MGC-803 and MKN-45). In addition, knockdown of the expression of ENO1 led to the arrest of the cell cycle at the G1 phase and promoted the apoptosis of MKN-45 and MGC-803 cells. The further microarray and bioinformatic analysis revealed that ENO1 regulated the expression of diverse genes, many of which are involved in the progress of cancer. Taken together, our data demonstrated that ENO1 was an oncogene-like factor and might serve as a promising target for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Hui Qiao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yufeng Wang
- Department of Oncology, Tumor Hospital of Gansu Province, Lanzhou, China
| | - Bingdong Zhu
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland.,Institute of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lei Jiang
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wenzhen Yuan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Disease of Gansu Province, Lanzhou, China
| | - Quanlin Guan
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Disease of Gansu Province, Lanzhou, China
| |
Collapse
|
30
|
Dominguez RL, Cherry CB, Estevez-Ordonez D, Mera R, Escamilla V, Pawlita M, Waterboer T, Wilson KT, Peek RM, Tavera G, Williams SM, Gulley ML, Emch M, Morgan DR. Geospatial analyses identify regional hot spots of diffuse gastric cancer in rural Central America. BMC Cancer 2019; 19:545. [PMID: 31174492 PMCID: PMC6554991 DOI: 10.1186/s12885-019-5726-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Geospatial technology has facilitated the discovery of disease distributions and etiology and helped target prevention programs. Globally, gastric cancer is the leading infection-associated cancer, and third leading cause of cancer mortality worldwide, with marked geographic variation. Central and South America have a significant burden, particularly in the mountainous regions. In the context of an ongoing population-based case-control study in Central America, our aim was to examine the spatial epidemiology of gastric cancer subtypes and H. pylori virulence factors. METHODS Patients diagnosed with gastric cancer from 2002 to 2013 in western Honduras were identified in the prospective gastric cancer registry at the principal district hospital. Diagnosis was based on endoscopy and confirmatory histopathology. Geospatial methods were applied using the ArcGIS v10.3.1 and SaTScan v9.4.2 platforms to examine regional distributions of the gastric cancer histologic subtypes (Lauren classification), and the H. pylori CagA virulence factor. Getis-Ord-Gi hot spot and Discrete Poisson SaTScan statistics, respectively, were used to explore spatial clustering at the village level (30-50 rural households), with standardization by each village's population. H. pylori and CagA serologic status was determined using the novel H. pylori multiplex assay (DKFZ, Germany). RESULTS Three hundred seventy-eight incident cases met the inclusion criteria (mean age 63.7, male 66.3%). Areas of higher gastric cancer incidence were identified. Significant spatial clustering of diffuse histology adenocarcinoma was revealed both by the Getis-Ord-GI* hot spot analysis (P-value < 0.0015; range 0.00003-0.0014; 99%CI), and by the SaTScan statistic (P-value < 0.006; range 0.0026-0.0054). The intestinal subtype was randomly distributed. H. pylori CagA had significant spatial clustering only in association with the diffuse histology cancer hot spot (Getis-Ord-Gi* P value ≤0.001; range 0.0001-0.0010; SaTScan statistic P value 0.0085). In the diffuse gastric cancer hot spot, the lowest age quartile range was 21-46 years, significantly lower than the intestinal cancers (P = 0.024). CONCLUSIONS Geospatial methods have identified a significant cluster of incident diffuse type adenocarcinoma cases in rural Central America, suggest of a germline genetic association. Further genomic and geospatial analyses to identify potential spatial patterns of genetic, bacterial, and environmental risk factors may be informative.
Collapse
Affiliation(s)
| | - Charlotte B Cherry
- Office of Public Health Informatics & Analytics, Tennessee Department of Public Health, Nashville, TN, USA
| | - Dago Estevez-Ordonez
- Vanderbilt Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, USA
| | - Robertino Mera
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Veronica Escamilla
- Carolina Population Center, University of North Carolina, Chapel Hill, USA
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Gloria Tavera
- Department of Population and Quantitative Health Sciences and Institute of Computational Biology, Case Western Reserve University, Cleveland, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences and Institute of Computational Biology, Case Western Reserve University, Cleveland, USA
| | - Margaret L Gulley
- Department of Pathology, University of North Carolina, Chapel Hill, USA
| | - Michael Emch
- Department of Geography, University of North Carolina, Chapel Hill, USA
| | - Douglas R Morgan
- Vanderbilt Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, USA.
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA.
- Division of Gastroenterology and Hepatology, The University of Alabama at Birmingham (UAB), 1808 7th Avenue South, BDB 373, Birmingham, AL, 35233, USA.
| |
Collapse
|
31
|
Yost S, Ruark E, Alexandrov LB, Rahman N. Insights into BRCA Cancer Predisposition from Integrated Germline and Somatic Analyses in 7632 Cancers. JNCI Cancer Spectr 2019; 3:pkz028. [PMID: 31360904 PMCID: PMC6649772 DOI: 10.1093/jncics/pkz028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/19/2019] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND It is often assumed any cancer in a germline BRCA1 or BRCA2 (collectively termed BRCA) mutation carrier was caused by that mutation. It is also often assumed the occurrence of breast or ovarian cancer in an individual with a variant of uncertain significance (VUS) suggests the VUS is pathogenic. These assumptions have profound management implications for cancer patients and healthy individuals. METHODS We compared the frequency of BRCA mutations, allele loss, and Signature 3 in 7632 individuals with 28 cancers and 1000 population controls. Because only increased frequency was the focus of the study, all statistical tests were one-sided. RESULTS Individuals with breast or ovarian cancer had increased germline BRCA pathogenic mutation frequencies compared to controls (P = 1.0x10-10 and P = 1.4x10-34, respectively). There was no increase in other cancer types. Wild-type allele loss and Signature 3 were statistically significantly higher in breast and ovarian cancers with BRCA mutations compared with other cancers with BRCA mutations (P = 5.1x10-10 and P = 3.7x10-9) and cancers without BRCA mutations (P = 2.8x10-53 and P = 1.0x10-134). There was no difference between non-breast and non-ovarian cancers with BRCA mutations and cancers without BRCA mutations. Allele loss and Signature 3 were statistically significantly higher in breast and ovarian cancers in individuals with BRCA pathogenic mutations compared to those with VUS (P = 3.8x10-17 and P = 1.6x10-8) or benign variants (P = 1.2x10-28 and P = 2.2x10-10). There was no difference between individuals with BRCA VUS and those with benign variants. CONCLUSIONS These data show that non-breast and non-ovarian cancers in individuals with germline BRCA pathogenic mutations are often not causally related to the mutation and that BRCA VUS are highly unlikely to be pathogenic. These results should reduce inappropriate management of germline BRCA information.
Collapse
Affiliation(s)
- Shawn Yost
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Elise Ruark
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine
- Department of Bioengineering, University of California, San Diego, La Jolla, CA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Nazneen Rahman
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK (NR)
| |
Collapse
|
32
|
Guio H, Poterico JA, Levano KS, Cornejo‐Olivas M, Mazzetti P, Manassero‐Morales G, Ugarte‐Gil MF, Acevedo‐Vásquez E, Dueñas‐Roque M, Piscoya A, Fujita R, Sanchez C, Casavilca‐Zambrano S, Jaramillo‐Valverde L, Sullcahuaman‐Allende Y, Iglesias‐Pedraz JM, Abarca‐Barriga H. Genetics and genomics in Peru: Clinical and research perspective. Mol Genet Genomic Med 2018; 6:873-886. [PMID: 30584990 PMCID: PMC6305655 DOI: 10.1002/mgg3.533] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/17/2022] Open
Abstract
Peruvians currently preserve in their DNA the history of 2.5 million years of human evolution and 150,000 years of migration from Africa to Peru or the Americas. The development of Genetics and Genomics in the clinical and academic field is shown in this review.
Collapse
Affiliation(s)
- Heinner Guio
- Instituto Nacional de Salud del PerúLimaPerú
- Universidad Científica del SurLimaPerú
| | - Julio A. Poterico
- Instituto Nacional de Salud del PerúLimaPerú
- Servicio de GenéticaInstituto Nacional de Salud del Niño San Borja (INSN‐SB)LimaPeru
| | | | - Mario Cornejo‐Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias NeurológicasLimaPerú
| | - Pilar Mazzetti
- Neurogenetics Research Center, Instituto Nacional de Ciencias NeurológicasLimaPerú
- School of MedicineUniversidad Nacional Mayor de San MarcosLimaPerú
| | | | - Manuel F. Ugarte‐Gil
- Universidad Científica del SurLimaPerú
- Rheumatology Department. Hospital Guillermo Almenara Irigoyen. EsSaludLimaPerú
| | - Eduardo Acevedo‐Vásquez
- School of MedicineUniversidad Nacional Mayor de San MarcosLimaPerú
- Clínica San FelipeLimaPerú
| | - Milagros Dueñas‐Roque
- Servicio de GenéticaHospital Nacional Edgardo Rebagliati MartinsLimaPerú
- Sociedad de Genética Médica del PerúLimaPeru
| | - Alejandro Piscoya
- Universidad San Ignacio de LoyolaLimaPerú
- Hospital Guillermo Kaelin de la FuenteLimaPerú
| | - Ricardo Fujita
- Centro de Genética y Biología Molecular, Universidad de San Martín de PorresLimaPerú
| | | | - Sandro Casavilca‐Zambrano
- Banco de Tejidos Tumorales, Instituto Nacional de Enfermedades NeoplásicasBanco de Tejidos TumoralesLimaPerú
- Instituto Nacional de Enfermedades NeoplásicasLimaPerú
| | | | | | - Juan M. Iglesias‐Pedraz
- Universidad Científica del SurLimaPerú
- Laboratorio de Genética Molecular y Bioquímica, Departamento de Investigación, Desarrollo e InnovaciónUniversidad Científica del SurLimaPerú
| | - Hugo Abarca‐Barriga
- Servicio de Genética & EIMInstituto Nacional de Salud del Niño Breña (INSN)LimaPeru
- Facultad de EstomatologíaUniversidad Científica del SurLimaPerú
- Facultad de Medicina HumanaUniversidad Ricardo PalmaLimaPerú
| |
Collapse
|
33
|
Rudloff U. Gastric adenocarcinoma and proximal polyposis of the stomach: diagnosis and clinical perspectives. Clin Exp Gastroenterol 2018; 11:447-459. [PMID: 30584346 PMCID: PMC6284852 DOI: 10.2147/ceg.s163227] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) is a recently described, rare gastric polyposis syndrome. It is characterized by extensive involvement of the fundus and body of the stomach with fundic gland polyps sparing the antrum and lesser curvature, an autosomal dominant inheritance pattern with incomplete penetrance, and a significant predisposition for the development of gastric adenocarcinoma. Due to the recent discovery of APC promotor IB mutations (c.-191T>C, c.-192A>G, and c.-195A>C), which reduce binding of the transcription factor Yin Yang 1 (YY1) and transcriptional activity of the promotor, as its underlying genetic perturbation, GAPPS has been added to the growing molecular class of APC-associated disorders. Recent reports on family members afflicted by gastric polyposis due to GAPPS have described the development of metastatic cancer or the presence of invasive gastric adenocarcinoma in total gastrectomy specimens after variable periods of endoscopic surveillance emphasizing the need for an improved understanding of the to-date poorly characterized natural history of the syndrome. There are, however, currently no guidelines on screening, timing of prophylactic gastrectomy, or endoscopic surveillance for GAPPS available. In this review, we summarize the clinical, pathological, and genetic aspects of GAPPS as well as management approaches to this rare cancer predisposition syndrome, highlighting the need for early recognition, a multidisciplinary approach, and the creation of prospective family registries and consensus guidelines in the near future.
Collapse
Affiliation(s)
- Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA,
| |
Collapse
|
34
|
Giryes A, Oweira H, Mannhart M, Decker M, Abdel-Rahman O. Exploring the differences between early-onset gastric cancer and traditional-onset gastric cancer. J Gastrointest Oncol 2018; 9:1157-1163. [PMID: 30603136 DOI: 10.21037/jgo.2018.06.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The current study sought to explore the potential clinical, epidemiological and genetic differences between early-onset gastric cancer (E-gastric cancer: defined as 20-39 years) and traditional-onset gastric cancer (T-gastric cancer: defined as ≥40 years). Methods Datasets from the following sources were searched: Surveillance, Epidemiology and End Results database [2000-2014], Behavioral Risk Factor Surveillance Survey and the cancer genome atlas (TCGA). Clinicopathological characteristics, trends, and genetic findings were compared between E-gastric cancer and T-gastric cancer. Moreover, correlations with relevant risk factors were sought after. Results A total of 95,323 gastric cancer patients were identified in the period from 2000 to 2014. While T-gastric cancer was decreasing during the study period (-1.4; P<0.05), E-gastric cancer was stable during the study period. E-gastric cancer is less prevalent in males (51.1% vs. 61.0%; P<0.0001), and white patients (68.9% vs. 71.4%; P<0.0001). E-gastric cancer patients usually present with poorly differentiated histology (55.3% vs. 48.0%; P<0.0001) as well as more aggressive histological subtypes (e.g., diffuse histology or linitis plastica). No difference can be detected with regards to risk factor correlations between E-gastric cancer and T-gastric cancer. Only four patients with E-gastric cancer were available in the provisional TCGA dataset at the time of the study. Conclusions E-gastric cancer is a potentially distinct disease entity with specific clinicopathological and trend patterns compared to conventional T-gastric cancer. Further studies are needed to explore the potential etiologic basis as well as to investigate the clinical consequences of this distinction. The impact of this distinction on minority populations requires further assessment as well.
Collapse
Affiliation(s)
| | - Hani Oweira
- Gastrointestinal Tumor Zentrum, Zürich, Switzerland
| | | | - Michael Decker
- Oncology Department, Zentrum für Integrative Onkologie, Zürich, Switzerland
| | - Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.,Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Lott PC, Carvajal-Carmona LG. Resolving gastric cancer aetiology: an update in genetic predisposition. Lancet Gastroenterol Hepatol 2018; 3:874-883. [PMID: 30507471 PMCID: PMC6500447 DOI: 10.1016/s2468-1253(18)30237-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/06/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022]
Abstract
Every year gastric cancer accounts for nearly 1 million new cases and more than 720 000 deaths worldwide. Prognosis is dismal because most patients are diagnosed with advanced disease; as such, gastric cancer outcomes will benefit from better methods for identification of at-risk individuals that can be targeted for early detection. One approach to targeting high-risk populations is to identify individuals who are genetically predisposed to gastric cancer, as up to 15% of all patients report family history of the disease. On the basis of clinical manifestations, three gastric cancer syndromes have been described, but the diagnosis of some of these syndromes is suboptimal and could benefit from genetic information. Over the past decade, genome-wide association and next-generation sequencing studies have identified several low penetrance variants and high-risk genes, considerably increasing our understanding of inherited gastric cancer predisposition. From these studies, PALB2 has emerged as a new familial gastric cancer gene. Furthermore, genetic analyses in patients with sporadic gastric cancer suggest that more than 10% of all cases have pathogenic mutations, a finding of great importance for cancer aetiology. In this Review, we summarise the role of genetics in gastric cancer aetiology and the implications of genetics findings for the prevention of this malignancy.
Collapse
Affiliation(s)
- Paul C Lott
- Genome Center, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Luis G Carvajal-Carmona
- Genome Center, School of Medicine, University of California at Davis, Davis, CA, USA; Population Sciences and Cancer Health Disparities Program, UC Davis Comprehensive Cancer Center, School of Medicine, University of California at Davis, Davis, CA, USA; Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Davis, CA, USA.
| |
Collapse
|
36
|
The effect of Ganoderma lucidum extract on immunological function and identify its anti-tumor immunostimulatory activity based on the biological network. Sci Rep 2018; 8:12680. [PMID: 30139984 PMCID: PMC6107651 DOI: 10.1038/s41598-018-30881-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 08/07/2018] [Indexed: 12/15/2022] Open
Abstract
Ganoderma lucidum extract (GLE) has shown positive effects for tumor treatment. However, the molecular mechanism of GLE treatment is unknown. In this study, a Hepa1-6-bearing C57 BL/6 mouse model was established to explore the anti-tumor and immunostimulatory activity of GLE treatment. The results showed that GLE effectively inhibited tumor growth without hepatic/renal toxicity and bone marrow suppression, and might enhancing immunological function. Based on the mRNA profiles of GLE treated and untreated mice, 302 differentially expressed (DE) mRNAs were identified and 6 kernel mRNAs were identified from the established protein-protein interaction (PPI) network. Quantitative RT-PCR and western-blot analysis indicated that 6 mRNAs have had statistically significant differences between the GLE treated and untreated mice. Furthermore, four kernel pathways were isolated from the KEGG-Target network, including the Jak-STAT signaling pathway, T cell receptor signaling pathway, PI3K-Akt signaling pathway, and cytokine-cytokine receptor interaction. Western-blot and cytokine detection results demonstrated that GLE suppressed growth and proliferation of tumors by the Jak-STAT signaling pathway, T cell receptor signaling pathway and PI3K-Akt signaling pathway, but also regulated the expression levels of serum immune cytokines and improved the anti-tumor immunostimulatory activity.
Collapse
|
37
|
Fewings E, Larionov A, Redman J, Goldgraben MA, Scarth J, Richardson S, Brewer C, Davidson R, Ellis I, Evans DG, Halliday D, Izatt L, Marks P, McConnell V, Verbist L, Mayes R, Clark GR, Hadfield J, Chin SF, Teixeira MR, Giger OT, Hardwick R, di Pietro M, O'Donovan M, Pharoah P, Caldas C, Fitzgerald RC, Tischkowitz M. Germline pathogenic variants in PALB2 and other cancer-predisposing genes in families with hereditary diffuse gastric cancer without CDH1 mutation: a whole-exome sequencing study. Lancet Gastroenterol Hepatol 2018; 3:489-498. [PMID: 29706558 PMCID: PMC5992580 DOI: 10.1016/s2468-1253(18)30079-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Germline pathogenic variants in the E-cadherin gene (CDH1) are strongly associated with the development of hereditary diffuse gastric cancer. There is a paucity of data to guide risk assessment and management of families with hereditary diffuse gastric cancer that do not carry a CDH1 pathogenic variant, making it difficult to make informed decisions about surveillance and risk-reducing surgery. We aimed to identify new candidate genes associated with predisposition to hereditary diffuse gastric cancer in affected families without pathogenic CDH1 variants. METHODS We did whole-exome sequencing on DNA extracted from the blood of 39 individuals (28 individuals diagnosed with hereditary diffuse gastric cancer and 11 unaffected first-degree relatives) in 22 families without pathogenic CDH1 variants. Genes with loss-of-function variants were prioritised using gene-interaction analysis to identify clusters of genes that could be involved in predisposition to hereditary diffuse gastric cancer. FINDINGS Protein-affecting germline variants were identified in probands from six families with hereditary diffuse gastric cancer; variants were found in genes known to predispose to cancer and in lesser-studied DNA repair genes. A frameshift deletion in PALB2 was found in one member of a family with a history of gastric and breast cancer. Two different MSH2 variants were identified in two unrelated affected individuals, including one frameshift insertion and one previously described start-codon loss. One family had a unique combination of variants in the DNA repair genes ATR and NBN. Two variants in the DNA repair gene RECQL5 were identified in two unrelated families: one missense variant and a splice-acceptor variant. INTERPRETATION The results of this study suggest a role for the known cancer predisposition gene PALB2 in families with hereditary diffuse gastric cancer and no detected pathogenic CDH1 variants. We also identified new candidate genes associated with disease risk in these families. FUNDING UK Medical Research Council (Sackler programme), European Research Council under the European Union's Seventh Framework Programme (2007-13), National Institute for Health Research Cambridge Biomedical Research Centre, Experimental Cancer Medicine Centres, and Cancer Research UK.
Collapse
Affiliation(s)
- Eleanor Fewings
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Alexey Larionov
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - James Redman
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Mae A Goldgraben
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - James Scarth
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Susan Richardson
- Familial Gastric Cancer Study, Department of Oncology, University of Cambridge, Cambridge, UK
| | | | | | - Ian Ellis
- Cheshire and Merseyside Regional Genetic Service, Liverpool, UK
| | | | - Dorothy Halliday
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Louise Izatt
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Peter Marks
- West Midlands Regional Genetics Service, Birmingham, UK
| | - Vivienne McConnell
- Northern Ireland Regional Genetics Centre, Belfast City Hospital, Belfast, UK
| | - Louis Verbist
- Department of Gastroenterology, ZNA Jan Palfijn, Antwerp, Belgium
| | - Rebecca Mayes
- Centre for Cancer Genetic Epidemiology, Strangeway's Research Laboratory, University of Cambridge, Cambridge, UK
| | - Graeme R Clark
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - James Hadfield
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Precision Medicine and Genomics, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute of Porto, Porto, Portugal; Institute of Biomedical Sciences, University of Porto, Porto, Portugal
| | - Olivier T Giger
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Richard Hardwick
- Department of Oesophago-Gastric Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Massimiliano di Pietro
- Medical Research Council (MRC) Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Strangeway's Research Laboratory, University of Cambridge, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Rebecca C Fitzgerald
- Medical Research Council (MRC) Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marc Tischkowitz
- Academic Laboratory of Medical Genetics, University of Cambridge, Cambridge, UK; National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|