1
|
Hereditary Diffuse Gastric Cancer: Molecular Genetics, Biological Mechanisms and Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms23147821. [PMID: 35887173 PMCID: PMC9319245 DOI: 10.3390/ijms23147821] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 12/14/2022] Open
Abstract
Hereditary diffuse gastric cancer is an autosomal dominant syndrome characterized by a high prevalence of diffuse gastric cancer and lobular breast cancer. It is caused by inactivating mutations in the tumor suppressor gene CDH1. Genetic testing technologies have become more efficient over the years, also enabling the discovery of other susceptibility genes for gastric cancer, such as CTNNA1 among the most important genes. The diagnosis of pathogenic variant carriers with an increased risk of developing gastric cancer is a selection process involving a multidisciplinary team. To achieve optimal long-term results, it requires shared decision-making in risk management. In this review, we present a synopsis of the molecular changes and current therapeutic approaches in HDGC based on the current literature.
Collapse
|
2
|
Pan Z, Fu Z, Luo C, Bao Y, Wang M, Cao W, Xu X. CDH1 germline mutations in a Chinese cohort with hereditary diffuse gastric cancer. J Cancer Res Clin Oncol 2021; 148:2145-2151. [PMID: 34537906 DOI: 10.1007/s00432-021-03775-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Germline mutations in CDH1 are associated with hereditary diffuse gastric cancer (HDGC) and have been identified in multiple ethnicities. However, CDH1 germline mutations have seldom been documented in Chinese patients with HDGC, and their frequency remains unclear. Here, we aimed to examine the frequency of CDH1 germline mutations in Chinese patients with HDGC. In total, 285 patients who met the International Gastric Cancer Linkage Consortium 2015 testing criteria of HDGC for CDH1 germline mutations were recruited. METHODS All 16 CDH1 exons, including neighboring intronic sequences, were amplified using polymerase chain reaction and screened using Sanger sequencing. Variants were analyzed using Mutation Surveyor V4.0, SIFT, and PolyPhen-2 software. RESULTS Three nonsense and nine missense CDH1 germline mutations were identified in 21 of 285 index cases (7.4%). Two CDH1 germline mutations, N405Y (Asn405Tyr) and W409X (Trp409Ter), were identified as new variants. In addition, up to 28.6% of CDH1 mutations in the 21 indicated patients were identified as c.1775G>C (E551Q). The frequency of CDH1 mutations was 6.5% (7/108) in HDGC and 7.9% (14/177) in early onset diffuse gastric cancer (EODGC). The mutation detection rates of CDH1 in males and females were 6.7% (4/60) and 8.5% (10/117) in EODGC and 4.6% (3/65) and 9.3% (4/43) in HDGC, respectively. CONCLUSION These data reveal, for the first time, the type and frequency of CDH1 germline mutations in Chinese HDGC and demonstrate that germline CDH1 mutations are a noteworthy contributor to the high frequency of HDGC in Chinese.
Collapse
Affiliation(s)
- Zhiwen Pan
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.,Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, Zhejiang, China
| | - Zhixuan Fu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Cong Luo
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yejiang Bao
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Mingli Wang
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenming Cao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiaohong Xu
- Clinical Laboratory Department, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Goud HK, Mehkari Z, Mohammed L, Javed M, Althwanay A, Ahsan F, Oliveri F, Rutkofsky IH. Significance of E-cadherin Gene Mutations in Patients With Hereditary Diffuse Gastric Cancer Syndrome: A Systematic Review. Cureus 2020; 12:e10406. [PMID: 33062523 PMCID: PMC7550226 DOI: 10.7759/cureus.10406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/12/2020] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer is the third-most fatal cancer in the world. Though over the years, we saw patients mostly with intestinal type accounting for the highest mortality rate, the recent rise of the diffuse form with germline E-cadherin (CDH1) mutations has added a whole new level of interest to study in detail about the association between CDH1 and diffuse gastric cancer (DGC). This introduced a set guideline formulated by Internal Gastric Cancer Linkage Consortium (IGCLC) for patients with family history of diffuse gastric cancer and invasive lobular breast cancer (ILBC). The analysis of this link was also important to set proper management protocol for patients who were CDH1 mutation carriers which now involves genetic counselling, endoscopic surveillance and screening and prophylactic total gastrectomy (PTG). The study was conducted in accordance to the 'PRISMA guidelines for reporting systematic review and meta-analysis'. Peer-reviewed studies were included from the PubMed database and relevant articles were selected to be included in the study. Appropriate inclusion/exclusion criteria with free full text English articles were applied while selecting the articles. A total of 10 studies on review with different study populations showed that of the 42 patients who were diagnosed with diffuse gastric cancer, 88% of them showed a positive germline E-cadherin gene mutation and 100% of the CDH1 mutation carriers showed microscopic changes of signet ring cell adenocarcinoma of the stomach. The beneficial effects of PTG with better survival rates and low mortality rates has outweighed other treatment modalities. Laparoscopic approach has proved to be more useful and a safer approach for gastrectomy surgeries with better post-operative management. The need for prophylactic mastectomy is also increased in the recent times and thus this requires a new set of guidelines for ILBC patients with hereditary diffuse gastric cancer (HDGC) syndrome.
Collapse
Affiliation(s)
- Harshit K Goud
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zainab Mehkari
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lubna Mohammed
- Obstetrics & Gynaecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Moiz Javed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aldanah Althwanay
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Farah Ahsan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Federico Oliveri
- Cardiology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ian H Rutkofsky
- Psychiatry, Neuroscience, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
4
|
Choi S, Jang J, Heo YJ, Kang SY, Kim ST, Lee J, Kang WK, Kim JW, Kim KM. CDH1 mutations in gastric cancers are not associated with family history. Pathol Res Pract 2020; 216:152941. [PMID: 32241597 DOI: 10.1016/j.prp.2020.152941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/03/2020] [Accepted: 03/21/2020] [Indexed: 02/07/2023]
Abstract
CDH1 mutation is the most frequent genetic alteration in hereditary diffuse gastric cancer (GC) and early onset diffuse GC patients. However, the incidence of CDH1 mutations in sporadic GC with or without family history has not been studied. This retrospective study includes a total of 993 Korean patients with primary advanced GC who underwent surgery and received palliative chemotherapy. Targeted deep sequencing was performed in all cases and family history of GC was searched with survival analysis. We found CDH1 alterations in 146 of 993 patients (14.7 %) and 8 were germline (0.8 %). Out of 146 patients with CDH1 mutations, 25 (17.1 %) had a family history of GC in one of their first relatives, and 12 patients (8.2 %) were diagnosed with familial GC (FGC). All cases with FGC were diffuse type by Lauren classification, and only one harbored a previously reported germline mutation of CDH1 (c.2638 G > A) and the remaining 11 harbored known somatic CDH1 mutations. Among all patients with CDH1 mutation, there was no significant survival difference between patients with family history or FGC. In the 847 patients without CDH1 mutation, 189 (22.3 %) had a family history of GC and 92 patients (10.9 %) were FGC. CDH1 mutations were more frequent in patients with early onset (<45 years) GC (45.5 %) compared with patients with late onset GC (10.9 %) (p = 0.001), but were not significantly associated with the family history of GC (p > 0.05). CDH1 mutations are mostly somatic and typically are not associated with family history.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jiryeon Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - You Jeong Heo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Tae Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeeyun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea; Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Won Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea; Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Donida BM, Tomasello G, Ghidini M, Buffoli F, Grassi M, Liguigli W, Maglietta G, Pergola L, Ratti M, Sabadini G, Toppo L, Ungari M, Passalacqua R. Epidemiological, clinical and pathological characteristics of gastric neoplasms in the province of Cremona: the experience of the first population-based specialized gastric cancer registry in Italy. BMC Cancer 2019; 19:212. [PMID: 30849945 PMCID: PMC6408835 DOI: 10.1186/s12885-019-5366-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The gastric cancer incidence rate differs widely across geographical areas. In Italy, in the province of Cremona the incidence is high, compared to the national situation. For this reason a specialized population-based registry was set up. METHODS The collection encompasses all gastric cancers diagnosed in the three districts of the province since January 1, 2010. The main data sources were the pathological and Hospital Discharge Records and patient clinical charts. Only diagnoses of primary gastric cancer were considered. For each case the following variables were registered: personal data, medical history and symptoms at diagnosis; imaging assessments performed, details on surgery and other treatments received; genetic background and biomolecular characteristics; social and environmental factors. RESULTS As of November 2017, 1087 cases were collected; of which 876, diagnosed up to December 2015, were analyzed. Male/female ratio was 1.4. The European Age-standardized Incidence Rate was 41.4 for males and 28.3 for females as compared to a national average of 33.3 and 17.0 respectively. Median age at diagnosis was 73 for male and 78 for female. Helicobacter Pylori infection was present in fewer than 20% of cases. HER-2 gene was amplified in about 25% of cases. Primary tumour location was the gastro-esophageal junction or cardia in 17.5% in males and 8.3% in females. The majority of cases (58.3%) were diagnosed at an advanced stage and overall only 41.2% underwent surgery. Median overall survival was 14.8 months for men and 18.5 for women. Age standardized 5-year relative survival was 31.4% for men and 40.5% for females. Neoadjuvant treatment was performed in fewer than 10% of patients who underwent surgery, and the rate of postoperative therapy adherence was low. DISCUSSION This study shows a high gastric cancer incidence in the province of Cremona, with a geographical spread across different districts. Moreover, a high percentage of gastric cancers were detected at an advanced stage of disease and a low rate of 5-year relative survival was registered. Based on these findings, effective preventive interventional health strategies and screening procedures need to be implemented to reduce the impact of this pathology in this geographical area.
Collapse
Affiliation(s)
- B M Donida
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy.
| | - G Tomasello
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| | - M Ghidini
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| | - F Buffoli
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| | - M Grassi
- ASST of Crema, Largo Ugo Dossena 2, 26013, Crema, CR, Italy
| | - W Liguigli
- Hospital of Suzzara, Via General Cantore 14/b, 46029, Suzzara, MN, Italy
| | - G Maglietta
- University Hospital of Parma, Via Gramsci 14, 43126, Parma, PR, Italy.,University of Florence, 50121, Parma, FI, Italy
| | - L Pergola
- ASST of Crema, Largo Ugo Dossena 2, 26013, Crema, CR, Italy
| | - M Ratti
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| | - G Sabadini
- Istituto Figlie San Camillo of Cremona, Via Fabio Filzi 56, 26100, Cremona, CR, Italy
| | - L Toppo
- Civil Hospital of Voghera, ASST of Pavia, Via Indipendenza 34, 27100, Pavia, PV, Italy
| | - M Ungari
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| | - R Passalacqua
- ASST of Cremona, Viale Concordia 1, 26100, Cremona, CR, Italy
| |
Collapse
|
6
|
Chen L, Lu Z, Yang Y, Du L, Zhou X, Chen Y. Effects of purified Omphalia lapidescens protein on metastasis, cell cycle, apoptosis and the JAK-STAT signaling pathway in SGC-7901 human gastric cells. Oncol Lett 2018. [PMID: 29541181 PMCID: PMC5835924 DOI: 10.3892/ol.2018.7830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer is one of the most common cancers globally with high rates of morbidity and mortality. Purified Omphalia lapidescens protein (pPeOp) is a protein extracted from the sclerotium of Omphalia lapidescens. The present study aimed to investigate the effects of pPeOp on the viability, migration, cell cycle progression and apoptosis of SGC-7901 cells. The expression of numerous proteins, namely matrix metallopeptidase (MMP)2, MMP9, p53, caspase-3, B-cell lymphoma (Bcl)-2, cyclin A2, cyclin B1, cyclin D1, cyclin dependent kinase (CDK)1, CDK2 and CDK4, were investigated using western blot analysis and reverse transcription-quantitative polymerase chain reaction. The results of the present study demonstrated that treating SGC-7901 cells with pPeOp markedly suppressed their migration, induced their apoptosis and arrested their progression in S phase. pPeOp also downregulated the expression of migration-associated proteins (MMP2 and MMP9) and cyclin-associated proteins (cyclin A2, cyclin B1, cyclin D1, CDK1, CDK2 and CDK4) in a dose-dependent manner. Cells treated with pPeOp significantly upregulated caspase-3 and p53 and downregulated Bcl-2. Finally, the impact of pPeOp on three key nodes of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway were investigated and it was revealed that expression levels of JAK1, JAK2 and STAT3 were significantly downregulated following treatment. Together, the results of the present study suggested that pPeOp suppresses metastasis, arrests cell cycle, induces apoptosis and inhibits the JAK-STAT signaling pathway in SGC-7901 cells. Therefore, pPeOp may serve as a novel therapeutic agent for patients with gastric cancer.
Collapse
Affiliation(s)
- Luchao Chen
- Microbiology and Immunology Laboratory, College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhongxia Lu
- Microbiology and Immunology Laboratory, College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yongle Yang
- Institute of Preventive and Veterinary Medicine and The Key Laboratory of Animal Virology of Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Lijun Du
- Microbiology and Immunology Laboratory, College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xiaofang Zhou
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yitao Chen
- Microbiology and Immunology Laboratory, College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
7
|
Zhang H, Feng M, Feng Y, Bu Z, Li Z, Jia S, Ji J. Germline mutations in hereditary diffuse gastric cancer. Chin J Cancer Res 2018; 30:122-130. [PMID: 29545726 DOI: 10.21147/j.issn.1000-9604.2018.01.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related deaths worldwide. Among which, about 1%-3% of gastric cancer patients were characterized by inherited gastric cancer predisposition syndromes, knowing as hereditary diffuse gastric cancer (HDGC). Studies reported that CDH1 germline mutations are the main cause of HDGC. With the help of rapid development of genetic testing technologies and data analysis tools, more and more researchers focus on seeking candidate susceptibility genes for hereditary cancer syndromes. In addition, National Comprehensive Cancer Network (NCCN) guidelines recommend that the patients of HDGC carrying CDH1 mutations should undergo prophylactic gastrectomy or routine endoscopic surveillances. Therefore, genetic counseling plays a key role in helping individuals with pathogenic mutations make appropriate risk management plans. Moreover, experienced and professional genetic counselors as well as a systematic multidisciplinary team (MDT) are also required to facilitate the development of genetic counseling and benefit pathogenic mutation carriers who are in need of regular and standardized risk management solutions. In this review, we provided an overview about the germline mutations of several genes identified in HDGC, suggesting that these genes may potentially act as susceptibility genes for this malignant cancer syndrome. Furthermore, we introduced information for prevention, diagnosis and risk management of HDGC. Investigations on key factors that may have effect on risk management decision-making and genetic data collection of more cancer syndrome family pedigrees are required for the development of HDGC therapeutic strategies.
Collapse
Affiliation(s)
- Hao Zhang
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yi Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhaode Bu
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Ziyu Li
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Shuqin Jia
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| |
Collapse
|
8
|
Yang YL, Gong WY, Chen FF, Chen LC, Chen YT. pPe Op from Omphalia lapidescens Schroet induces cell cycle arrest and inhibits the migration of MC-4 gastric tumor cells. Oncol Lett 2017; 14:533-540. [PMID: 28693202 PMCID: PMC5494755 DOI: 10.3892/ol.2017.6207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 01/19/2017] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to investigate the effect of purified Omphalia lapidescens protein (pPeOp) extracted by polyvinylpyrrolidone from the fungus Omphalia lapidescens Schroet on the proliferation and cell cycle progression of MC-4 human gastric tumor cells. Using polyvinylpyrrolidone, pPeOp was extracted from O. lapidescens Schroet. MC-4 cells were cultured with 30, 60 or 90 µg/ml pPeOp, with 5-fluorouracil used as a positive control. Survival rates of treated cells were significantly decreased compared with those of the untreated control group in a dose-dependent manner. Using flow cytometric analysis, cells treated with pPeOp were demonstrated to arrest in S phase and exhibit abnormal G0/G1 and G2/M phase cell cycle distribution. In addition, a wound healing assay demonstrated that pPeOp significantly inhibited the migration of MC-4 cells. The mRNA and protein expression levels of cyclin D1/cyclin-dependent kinase (CDK) 4, cyclin B/CDK1, cyclin A/CDK2, matrix metalloproteinase (MMP)-2 and MMP-9 were determined using reverse transcription-quantitative polymerase chain reaction analysis and western blotting. The mRNA expression level of CDK4 and cyclin A was significantly increased compared with the untreated control; however, cyclin D1, CDK1, CDK2, cyclin B, MMP-2, and MMP-9 exhibited a significantly decreased mRNA expression level, indicating that there is a negative association between concentration and cyclin D1 expression levels. The expression of the cycle arrest-associated proteins and migration-associated proteins examined were similar to the observed mRNA expression levels. In conclusion, pPeOp was identified to inhibit migration of and cause S phase cell cycle arrest in MC-4 cells.
Collapse
Affiliation(s)
- Yong-Le Yang
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Wei-Yao Gong
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Fei-Fei Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Lu-Chao Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yi-Tao Chen
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
9
|
López M, Cervera-Acedo C, Santibáñez P, Salazar R, Sola JJ, Domínguez-Garrido E. A novel mutation in the CDH1 gene in a Spanish family with hereditary diffuse gastric cancer. SPRINGERPLUS 2016; 5:1181. [PMID: 27512640 PMCID: PMC4961654 DOI: 10.1186/s40064-016-2852-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
Abstract
Hereditary diffuse gastric cancer (HDGC) is an inherited form of diffuse type gastric cancer. Germline CDH1 mutations have been identified in approximately 15-50 % of affected kindred that meet the clinical criteria for HDGC. If any of the criteria is met the individual is referred to genetic counseling and CDH1 testing is offered. In this report we present the case of a Spanish family with HDGC harboring a novel CDH1 mutation. A 47 year-old female with a diagnostic of gastric adenocarcinoma and some of her relatives were tested. Study of the entire CDH1 gene, including intron-exon boundaries, by PCR and sequencing and immunohistochemical determination of the expression of E-cadherin were performed. A novel heterozygous deletion in exon 9 of CDH1 gene (c.1220_1220delC, p.P407Qfs10), was found in the proband, one sister and a nephew. It generates a premature stop codon giving rise to a truncated protein that leads to a pathogenic variant. Expression of E-cadherin was absent or frankly reduced in the proband's tumor but normal in tumor cells of great-uncle. After these results, the sister underwent prophylactic total gastrectomy, and the nephew is under annual endoscopic surveillance. Personal or familial history of diffuse gastric cancer, above all at young age, should encourage CDH1 genetic testing. In this sense, the review of the criteria and the addition in the last guideline of the recommendation: "other families in which genetic testing may also be considered" broadens the number of individuals at risk detected. Since there are not reliable methods for early detection, DGC is usually diagnosed at an advanced stage and consequently associated with a poorer outcome. Thus, CDH1 mutations detection contributes to an improvement in diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- María López
- Molecular Diagnostics Laboratory, Fundación Rioja Salud, Logroño, La Rioja Spain
| | | | - Paula Santibáñez
- Molecular Diagnostics Laboratory, Fundación Rioja Salud, Logroño, La Rioja Spain
| | - Raquel Salazar
- Department of Medical Oncology, Hospital San Pedro, Logroño, La Rioja Spain
| | - Jesús-Javier Sola
- Department of Pathological Anatomy, Hospital San Pedro, Logroño, La Rioja Spain
| | | |
Collapse
|
10
|
van der Post RS, Vogelaar IP, Manders P, van der Kolk LE, Cats A, van Hest LP, Sijmons R, Aalfs CM, Ausems MGEM, Gómez García EB, Wagner A, Hes FJ, Arts N, Mensenkamp AR, van Krieken JH, Hoogerbrugge N, Ligtenberg MJL. Accuracy of Hereditary Diffuse Gastric Cancer Testing Criteria and Outcomes in Patients With a Germline Mutation in CDH1. Gastroenterology 2015; 149:897-906.e19. [PMID: 26072394 DOI: 10.1053/j.gastro.2015.06.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Germline mutations in the cadherin 1, type 1, E-cadherin gene (CDH1) cause a predisposition to gastric cancer. We evaluated the ability of the internationally accepted hereditary diffuse gastric cancer (HDGC) criteria to identify individuals with pathogenic mutations in CDH1, and assessed their outcomes. The criteria were as follows: families with 2 or more cases of gastric cancer, with at least 1 patient diagnosed with diffuse gastric cancer (DGC) before age 50; families with 3 or more cases of DGC; families with 1 DGC before the age of 40; and families with a history of DGC and lobular breast cancer, with 1 diagnosis before the age of 50. METHODS We collected results of a CDH1 mutation analysis of 578 individuals from 499 families tested in The Netherlands between 1999 and 2014 (118 families met the HDGC criteria for testing and 236 did not; there were 145 families with incomplete data and/or availability of only first-degree relatives). Data were linked with family histories and findings from clinical and pathology analyses. The Kaplan-Meier method and Cox regression analysis were used to evaluate the overall survival of patients with and without CDH1 mutations. RESULTS In a cohort study in The Netherlands, the HDGC criteria identified individuals with a germline CDH1 mutation with a positive predictive value of 14% and 89% sensitivity. There were 18 pathogenic CDH1 mutations in 499 families (4%); 16 of these mutations were detected in the 118 families who met the HDGC criteria for testing. One pathogenic CDH1 mutation was detected in the 236 families who did not meet HDGC criteria and 1 in the 145 families with incomplete data and/or availability of only first-degree relatives. No CDH1 mutations were found in the 67 families whose members developed intestinal-type gastric cancer, or in the 22 families whose families developed lobular breast cancer. Among patients who fulfilled the HDGC criteria and had pathogenic CDH1 mutations, 36% survived for 1 year and 4% survived for 5 years; among patients who fulfilled the HDGC criteria but did not carry pathogenic CDH1 mutations, 48% survived for 1 year and 13% survived for 5 years (P = .014 for comparative survival analysis between patients with and without a CDH1 mutation). CONCLUSIONS All individuals with a CDH1 mutation had a personal or family history of diffuse gastric cancer. Patients with gastric cancer and germline CDH1 mutations had shorter survival times than patients who met the HDGC criteria but did not have CDH1 mutations.
Collapse
Affiliation(s)
- Rachel S van der Post
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - Ingrid P Vogelaar
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Peggy Manders
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Lizet E van der Kolk
- Family Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Annemieke Cats
- Department of Gastroenterology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Liselotte P van Hest
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Rolf Sijmons
- Department of Genetics, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Cora M Aalfs
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Margreet G E M Ausems
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Encarna B Gómez García
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Frederik J Hes
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Neeltje Arts
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Arjen R Mensenkamp
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - J Han van Krieken
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Marjolijn J L Ligtenberg
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands; Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands.
| |
Collapse
|
11
|
Abstract
Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of cancer-related death worldwide. GC is a heterogeneous disease and the endpoint of a long multistep process largely influenced by Helicobacter pylori infection, genetic susceptibility, and environmental factors. In a subset of GC cases, infection with the Epstein-Barr virus (EBV) may also be involved. The development of GC is the consequence of the accumulation of multiple epi/genetic changes during the patient's lifetime that will result in oncogenic activation and/or tumor suppressor pathways' inactivation. This review will focus on the most recent updates on the characterization of the molecular phenotypes of sporadic and hereditary GC. This article will also update the most recent findings on the relationship between H. pylori infection and stem cells at the origin of GC. The understanding of the molecular genetics underlying gastric carcinogenesis is of paramount importance to identify novel potential targets for the development of screening and prognostic markers that can be clinically valuable for the management of GC patients and for the design of clinical trials.
Collapse
Affiliation(s)
- Ceu Figueiredo
- Department of Pathology and Oncology, Faculty of Medicine of the University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Susana Costa
- Department of Pathology and Oncology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Andreas Karameris
- Department of Pathology, Veterans Administration Hospital (NIMTS), Athens, Greece
| | - Jose Carlos Machado
- Department of Pathology and Oncology, Faculty of Medicine of the University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
van der Post RS, Vogelaar IP, Carneiro F, Guilford P, Huntsman D, Hoogerbrugge N, Caldas C, Schreiber KEC, Hardwick RH, Ausems MGEM, Bardram L, Benusiglio PR, Bisseling TM, Blair V, Bleiker E, Boussioutas A, Cats A, Coit D, DeGregorio L, Figueiredo J, Ford JM, Heijkoop E, Hermens R, Humar B, Kaurah P, Keller G, Lai J, Ligtenberg MJL, O'Donovan M, Oliveira C, Pinheiro H, Ragunath K, Rasenberg E, Richardson S, Roviello F, Schackert H, Seruca R, Taylor A, ter Huurne A, Tischkowitz M, Joe STA, van Dijck B, van Grieken NCT, van Hillegersberg R, van Sandick JW, Vehof R, van Krieken JH, Fitzgerald RC. Hereditary diffuse gastric cancer: updated clinical guidelines with an emphasis on germline CDH1 mutation carriers. J Med Genet 2015; 52:361-374. [PMID: 25979631 PMCID: PMC4453626 DOI: 10.1136/jmedgenet-2015-103094] [Citation(s) in RCA: 392] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/18/2015] [Indexed: 02/06/2023]
Abstract
Germline CDH1 mutations confer a high lifetime risk of developing diffuse gastric (DGC) and lobular breast cancer (LBC). A multidisciplinary workshop was organised to discuss genetic testing, surgery, surveillance strategies, pathology reporting and the patient's perspective on multiple aspects, including diet post gastrectomy. The updated guidelines include revised CDH1 testing criteria (taking into account first-degree and second-degree relatives): (1) families with two or more patients with gastric cancer at any age, one confirmed DGC; (2) individuals with DGC before the age of 40 and (3) families with diagnoses of both DGC and LBC (one diagnosis before the age of 50). Additionally, CDH1 testing could be considered in patients with bilateral or familial LBC before the age of 50, patients with DGC and cleft lip/palate, and those with precursor lesions for signet ring cell carcinoma. Given the high mortality associated with invasive disease, prophylactic total gastrectomy at a centre of expertise is advised for individuals with pathogenic CDH1 mutations. Breast cancer surveillance with annual breast MRI starting at age 30 for women with a CDH1 mutation is recommended. Standardised endoscopic surveillance in experienced centres is recommended for those opting not to have gastrectomy at the current time, those with CDH1 variants of uncertain significance and those that fulfil hereditary DGC criteria without germline CDH1 mutations. Expert histopathological confirmation of (early) signet ring cell carcinoma is recommended. The impact of gastrectomy and mastectomy should not be underestimated; these can have severe consequences on a psychological, physiological and metabolic level. Nutritional problems should be carefully monitored.
Collapse
Affiliation(s)
- Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ingrid P Vogelaar
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fátima Carneiro
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Department of Pathology and Oncology, Medical Faculty of the University of Porto, Porto, Portugal
- Centro Hospitalar São João, Porto, Portugal
| | - Parry Guilford
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - David Huntsman
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carlos Caldas
- Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Richard H Hardwick
- Department of Oesophago-Gastric Surgery, Addenbrooke's Hospital, Cambridge, UK
| | - Margreet G E M Ausems
- Department of Medical Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Linda Bardram
- Department of Surgical Gastroenterology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Tanya M Bisseling
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Eveline Bleiker
- Division of Psychosocial Research and Epidemiology/Family Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alex Boussioutas
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Annemieke Cats
- Department of Gastroenterology and Hepatology, Netherlands Cancer Institute/ Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Daniel Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Lynn DeGregorio
- The DeGregorio Family Foundation for Stomach and Esophageal Cancer Research, Pleasantville, New York, USA
| | - Joana Figueiredo
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - James M Ford
- Division of Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Esther Heijkoop
- Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Rosella Hermens
- Scientific Institute for Quality of Healthcare, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bostjan Humar
- Division of Surgical Research, University of Zurich, Zurich, Suisse
| | - Pardeep Kaurah
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gisella Keller
- Institute of Pathology, Technische Universität, Munich, Germany
| | - Jennifer Lai
- No Stomach For Cancer, Inc., Madison, Wisconsin, USA
| | - Marjolijn J L Ligtenberg
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Carla Oliveira
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Department of Pathology and Oncology, Medical Faculty of the University of Porto, Porto, Portugal
| | - Hugo Pinheiro
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Krish Ragunath
- NIHR Biomedical Research Unit, Nottingham Digestive Diseases Centre, Queens Medical Centre campus, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | | | - Susan Richardson
- Department of Oncology, Familial Gastric Cancer Registry, Cambridge University Hospital, Cambridge, UK
| | - Franco Roviello
- Department of General Surgery and Surgical Oncology, University of Siena, Siena, Italy
| | - Hans Schackert
- Department of Surgical Research, Technical University Dresden, Dresden, Germany
| | - Raquel Seruca
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Department of Pathology and Oncology, Medical Faculty of the University of Porto, Porto, Portugal
| | - Amy Taylor
- Cambridge University Hospital, Cambridge, UK
| | | | - Marc Tischkowitz
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Sheena Tjon A Joe
- Division of Oncology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | - Johanna W van Sandick
- Department of Surgery, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Rianne Vehof
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - J Han van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rebecca C Fitzgerald
- Cambridge NIHR Biomedical Research Centre, University of Cambridge NHS Foundation Trust
- MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge, UK
- Department Gastroenterology, Cambridge University Hospitals, UK
| |
Collapse
|