Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
2.
Mol Diagn Ther ; 27(4): 537-550, 2023 07.
Article in English | MEDLINE | ID: mdl-37099071

ABSTRACT

BACKGROUND: Circulating tumour DNA (ctDNA) analysis promises to improve the clinical care of people with cancer, address health inequities and guide translational research. This observational cohort study used ctDNA to follow 29 patients with advanced-stage cutaneous melanoma through multiple cycles of immunotherapy. METHOD: A melanoma-specific ctDNA next-generation sequencing (NGS) panel, droplet digital polymerase chain reaction (ddPCR) and mass spectrometry analysis were used to identify ctDNA mutations in longitudinal blood plasma samples from Aotearoa New Zealand (NZ) patients receiving immunotherapy for melanoma. These technologies were used in conjunction to identify the breadth and complexity of tumour genomic information that ctDNA analysis can reliably report. RESULTS: During the course of immunotherapy treatment, a high level of dynamic mutational complexity was identified in blood plasma, including multiple BRAF mutations in the same patient, clinically relevant BRAF mutations emerging through therapy and co-occurring sub-clonal BRAF and NRAS mutations. The technical validity of this ctDNA analysis was supported by high sample analysis-reanalysis concordance, as well as concordance between different ctDNA measurement technologies. In addition, we observed > 90% concordance in the detection of ctDNA when using cell-stabilising collection tubes followed by 7-day delayed processing, compared with standard EDTA blood collection protocols with rapid processing. We also found that the undetectability of ctDNA at a proportion of treatment cycles was associated with durable clinical benefit (DCB). CONCLUSION: We found that multiple ctDNA processing and analysis methods consistently identified complex longitudinal patterns of clinically relevant mutations, adding support for expanded clinical trials of this technology in a variety of oncology settings.


Subject(s)
Circulating Tumor DNA , Melanoma , Skin Neoplasms , Humans , Melanoma/genetics , Melanoma/therapy , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Circulating Tumor DNA/genetics , Proto-Oncogene Proteins B-raf/genetics , DNA, Neoplasm , Mutation , Immunotherapy , Melanoma, Cutaneous Malignant
3.
Gastroenterology ; 164(5): 719-735, 2023 04.
Article in English | MEDLINE | ID: mdl-36740198

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) is a dominantly inherited cancer syndrome characterized by a high incidence of diffuse gastric cancer (DGC) and lobular breast cancer (LBC). HDGC is caused by germline mutations in 2 genes involved in the epithelial adherens junction complex, CDH1 and CTNNA1. We discuss the genetics of HDGC and the variability of its clinical phenotype, in particular the variable penetrance of advanced DGC and LBC, both within and between families. We review the pathology of the disease, the mechanism of tumor initiation, and its natural history. Finally, we describe current best practice for the clinical management of HDGC, including emerging genetic testing criteria for the identification of new families, methods for endoscopic surveillance, the complications associated with prophylactic surgery, postoperative quality of life, and the emerging field of HDGC chemoprevention.


Subject(s)
Breast Neoplasms , Carcinoma, Lobular , Stomach Neoplasms , Humans , Female , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/prevention & control , Quality of Life , Genetic Testing/methods , Germ-Line Mutation , Cadherins/genetics , Breast Neoplasms/genetics , Genetic Predisposition to Disease
4.
Cancers (Basel) ; 14(7)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35406381

ABSTRACT

The CDH1 gene, encoding the cell adhesion protein E-cadherin, is one of the most frequently mutated genes in gastric cancer and inactivating germline CDH1 mutations are responsible for the cancer syndrome hereditary diffuse gastric cancer (HDGC). CDH1-deficient gastric cancers exhibit high AKT serine/threonine kinase 3 (AKT3) expression, but specific drugs against this AKT isoform are not available. We therefore used two publicly available datasets to identify AKT3-associated genes which could be used to indirectly target AKT3. Reactome analysis identified an enrichment of extracellular matrix remodelling genes in AKT3-high gastric cancers. Of the 51 genes that were significantly correlated with AKT3 (but not AKT1), discoidin domain receptor tyrosine kinase 2 (DDR2) showed the strongest positive association. Treatment of isogenic human cells and mouse gastric and mammary organoids with dasatinib, a small molecule inhibitor of multiple kinases including SRC, BCR-ABL and DDR2, preferentially slowed the growth and induced apoptosis of E-cadherin-deficient cells. Dasatinib treatment also preferentially slowed the growth of gastric and mammary organoids harbouring both Cdh1 and Tp53 mutations. In organoid models, dasatinib treatment was associated with decreased phosphorylation of total AKT, with a stronger effect seen in Cdh1-deficient organoids. Treatment with combinations of dasatinib and an inhibitor of AKT, MK2206, enhanced the effect of dasatinib in breast MCF10A cells. In conclusion, targeting the DDR2-SRC-AKT3 axis with dasatinib represents a promising approach for the chemoprevention and chemotherapy of gastric and breast cancers lacking E-cadherin.

5.
J Med Chem ; 64(24): 18114-18142, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34878770

ABSTRACT

Diffuse gastric cancer and lobular breast cancer are aggressive malignancies that are frequently associated with inactivating mutations in the tumor suppressor gene CDH1. Synthetic lethal (SL) vulnerabilities arising from CDH1 dysfunction represent attractive targets for drug development. Recently, SLEC-11 (1) emerged as a SL lead in E-cadherin-deficient cells. Here, we describe our efforts to optimize 1. Overall, 63 analogues were synthesized and tested for their SL activity toward isogenic mammary epithelial CDH1-deficient cells (MCF10A-CDH1-/-). Among the 26 compounds with greater cytotoxicity, AL-GDa62 (3) was four-times more potent and more selective than 1 with an EC50 ratio of 1.6. Furthermore, 3 preferentially induced apoptosis in CDH1-/- cells, and Cdh1-/- mammary and gastric organoids were significantly more sensitive to 3 at low micromolar concentrations. Thermal proteome profiling of treated MCF10A-CDH1-/- cell protein lysates revealed that 3 specifically inhibits TCOF1, ARPC5, and UBC9. In vitro, 3 inhibited SUMOylation at low micromolar concentrations.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Discovery , Stomach Neoplasms/drug therapy , Antigens, CD/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cadherins/genetics , Cell Line, Tumor , Humans , Mutation , Stomach Neoplasms/pathology
6.
J Pathol ; 254(3): 254-264, 2021 07.
Article in English | MEDLINE | ID: mdl-33797756

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) is a cancer syndrome caused by germline variants in CDH1, the gene encoding the cell-cell adhesion molecule E-cadherin. Loss of E-cadherin in cancer is associated with cellular dedifferentiation and poor prognosis, but the mechanisms through which CDH1 loss initiates HDGC are not known. Using single-cell RNA sequencing, we explored the transcriptional landscape of a murine organoid model of HDGC to characterize the impact of CDH1 loss in early tumourigenesis. Progenitor populations of stratified squamous and simple columnar epithelium, characteristic of the mouse stomach, showed lineage-specific transcriptional programs. Cdh1 inactivation resulted in shifts along the squamous differentiation trajectory associated with aberrant expression of genes central to gastrointestinal epithelial differentiation. Cytokeratin 7 (CK7), encoded by the differentiation-dependent gene Krt7, was a specific marker for early neoplastic lesions in CDH1 carriers. Our findings suggest that deregulation of developmental transcriptional programs may precede malignancy in HDGC. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cadherins/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Stomach Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Organoids , Single-Cell Analysis , Stomach Neoplasms/pathology , Transcriptome
7.
Cancers (Basel) ; 14(1)2021 Dec 26.
Article in English | MEDLINE | ID: mdl-35008266

ABSTRACT

Germline inactivating variants of CDH1 are causative of hereditary diffuse gastric cancer (HDGC), a cancer syndrome characterized by an increased risk of both diffuse gastric cancer and lobular breast cancer. Because loss of function mutations are difficult to target therapeutically, we have taken a synthetic lethal approach to identify targetable vulnerabilities in CDH1-null cells. We have previously observed that CDH1-null MCF10A cells exhibit a reduced rate of endocytosis relative to wildtype MCF10A cells. To determine whether this deficiency is associated with wider vulnerabilities in vesicle trafficking, we screened isogenic MCF10A cell lines with known inhibitors of autophagy, endocytosis, and sphingolipid metabolism. Relative to wildtype MCF10A cells, CDH1-/- MCF10A cells showed significantly greater sensitivity to several drugs targeting these processes, including the autophagy inhibitor chloroquine, the endocytosis inhibitors chlorpromazine and PP1, and the sphingosine kinase 1 inhibitor PF-543. Synthetic lethality was confirmed in both gastric and mammary organoid models of CDH1 loss, derived from CD44-Cre/Cdh1fl/fl/tdTomato mice. Collectively, these results suggest that both sphingolipid metabolism and vesicle trafficking represent previously unrecognised druggable vulnerabilities in CDH1-null cells and may lead to the development of new therapies for HDGC.

8.
Cancers (Basel) ; 14(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35008338

ABSTRACT

Inactivating germline mutations in the CDH1 gene (encoding the E-cadherin protein) are the genetic hallmark of hereditary diffuse gastric cancer (HDGC), and somatic CDH1 mutations are an early event in the development of sporadic diffuse gastric cancer (DGC) and lobular breast cancer (LBC). In this study, histone deacetylase (HDAC) inhibitors were tested for their ability to preferentially inhibit the growth of human cell lines (MCF10A and NCI-N87) and murine organoids lacking CDH1 expression. CDH1-/- breast and gastric cells were more sensitive to the pan-HDAC inhibitors entinostat, pracinostat, mocetinostat and vorinostat than wild-type cells, with an elevated growth inhibition that was, in part, attributable to increased apoptosis. CDH1-null cells were also sensitive to more class-specific HDAC inhibitors, but compared to the pan-inhibitors, these effects were less robust to genetic background. Increased sensitivity to entinostat was also observed in gastric organoids with both Cdh1 and Tp53 deletions. However, the deletion of Tp53 largely abrogated the sensitivity of the Cdh1-null organoids to pracinostat and mocetinostat. Finally, entinostat enhanced Cdh1 expression in heterozygous Cdh1+/- murine organoids. In conclusion, entinostat is a promising drug for the chemoprevention and/or treatment of HDGC and may also be beneficial for the treatment of sporadic CDH1-deficient cancers.

9.
Elife ; 92020 12 07.
Article in English | MEDLINE | ID: mdl-33284104

ABSTRACT

Mutations in genes encoding subunits of the cohesin complex are common in several cancers, but may also expose druggable vulnerabilities. We generated isogenic MCF10A cell lines with deletion mutations of genes encoding cohesin subunits SMC3, RAD21, and STAG2 and screened for synthetic lethality with 3009 FDA-approved compounds. The screen identified several compounds that interfere with transcription, DNA damage repair and the cell cycle. Unexpectedly, one of the top 'hits' was a GSK3 inhibitor, an agonist of Wnt signaling. We show that sensitivity to GSK3 inhibition is likely due to stabilization of ß-catenin in cohesin-mutant cells, and that Wnt-responsive gene expression is highly sensitized in STAG2-mutant CMK leukemia cells. Moreover, Wnt activity is enhanced in zebrafish mutant for cohesin subunits stag2b and rad21. Our results suggest that cohesin mutations could progress oncogenesis by enhancing Wnt signaling, and that targeting the Wnt pathway may represent a novel therapeutic strategy for cohesin-mutant cancers.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Synthetic Lethal Mutations/genetics , Wnt Signaling Pathway/physiology , Animals , Cell Division , Cell Line , Humans , Zebrafish , Cohesins
10.
Carcinogenesis ; 41(11): 1507-1517, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32955091

ABSTRACT

Accurate assessment of chemotherapy response provides the means to terminate ineffective treatment, trial alternative drug regimens or schedules and reduce dose to minimize toxicity. Here, we have compared circulating tumor DNA (ctDNA) with carcinoembryonic antigen (CEA) for the cycle by cycle assessment of chemotherapy response in 30 patients with metastatic colorectal cancer. CtDNA (quantified using individualized digital droplet PCR (ddPCR) assays) and CEA levels were determined immediately prior to each chemotherapy cycle over time periods ranging from 42-548 days (average of 10 time points/patient). Twenty-nine/thirty (97%) patients had detectable ctDNA compared with 83% whose tumors were CEA-positive (>5 ng/ml) during the monitoring course. Over the course of treatment, 20 disease progression events were detected by computed tomography; ctDNA predicted significantly more of these events than CEA (16 (80%) versus 6 (30%), respectively; P-value = 0.004). When progression was detected by both ctDNA and CEA, the rise in ctDNA occurred significantly earlier than CEA (P-value = 0.046). Partial responses to chemotherapy were also detected more frequently by ctDNA, although this was not significant (P-value = 0.07). In addition, another 28 colorectal cancer patients who underwent potentially curative surgery and showed no evidence of residual disease were monitored with ctDNA for up to 2 years. Clinical relapse was observed in 6/28 (21%) patients. Four out of 6 of these patients showed a significant increase in ctDNA at or prior to relapse. Overall, ctDNA analyses were able to be performed in a clinically relevant timeline and were a more sensitive and responsive measure of tumor burden than CEA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , Biomarkers, Tumor/analysis , Circulating Tumor DNA/analysis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA, Neoplasm/analysis , Follow-Up Studies , Humans , Prognosis , Prospective Studies , Tumor Burden
11.
N Z Med J ; 133(1514): 49-62, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32379739

ABSTRACT

AIMS: Increases in cancer survival may increase cancer prevalence and demand for healthcare. We aimed to estimate cancer prevalence in the New Zealand population. METHODS: We used national linked health, social and census datasets from the Stats NZ Integrated Data Infrastructure to identify the number of New Zealand residents who had at least one cancer diagnosis in New Zealand. We included all primary cancers recorded on the New Zealand Cancer Registry from January 1995 to June 2013, and used the 2013 census for demographic and socioeconomic data. RESULTS: On 30 June 2013, 140,600 of 4,438,900 (3.2%) New Zealand residents had been diagnosed with cancer in the last 18.5 years. In ≥15 year olds, the age-standardised prevalence of cancer diagnosed 0 to ≤1 year, and >1 to ≤5 years, prior to 30 June 2013 was 0.4% and 1.1% in men and 0.3% and 0.9% in women, respectively. Over the 18.5-year period prevalence was greatest in the oldest ages, European/Other, highest qualified, highest income, least deprived, ex-smokers, and Canterbury, Bay of Plenty and Nelson/Marlborough District Health Boards (age-standardised). CONCLUSIONS: Groups with the highest survival and the greatest access to healthcare had the highest cancer prevalences.


Subject(s)
Neoplasms/epidemiology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Big Data , Child , Child, Preschool , Ex-Smokers/statistics & numerical data , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neoplasms/ethnology , Neoplasms/mortality , New Zealand/epidemiology , Prevalence , Registries , Sex Factors , Socioeconomic Factors , Survival Rate , Time Factors , White People/statistics & numerical data , Young Adult
12.
Int J Mol Sci ; 20(20)2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31600923

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.


Subject(s)
Alleles , Antigens, CD/genetics , Cadherins/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Neoplastic Syndromes, Hereditary/genetics , Stomach Neoplasms/genetics , Adult , Female , Gastrectomy/methods , Genetic Association Studies , Humans , Male , Middle Aged , Neoplasm Staging , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/prevention & control , Pedigree , Prophylactic Surgical Procedures , Stomach Neoplasms/diagnosis , Stomach Neoplasms/prevention & control , Young Adult
13.
Cancers (Basel) ; 11(9)2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31540244

ABSTRACT

The CDH1 gene, encoding the cell adhesion protein E-cadherin, is one of the most frequently mutated genes in gastric cancer and inactivating germline CDH1 mutations are responsible for hereditary diffuse gastric cancer syndrome (HDGC). Using cell viability assays, we identified that breast (MCF10A) and gastric (NCI-N87) cells lacking CDH1 expression are more sensitive to allosteric AKT inhibitors than their CDH1-expressing isogenic counterparts. Apoptosis priming and total apoptosis assays in the isogenic MCF10A cells confirmed the enhanced sensitivity of E-cadherin-null cells to the AKT inhibitors. In addition, two of these inhibitors, ARQ-092 and MK2206, preferentially targeted mouse-derived gastric Cdh1-/- organoids for growth arrest. AKT protein expression and activation (as measured by phosphorylation of serine 473) were differentially regulated in E-cadherin-null MCF10A and NCI-N87 cells, with downregulation in the normal breast cells, but upregulation in the gastric cancer cells. Bioinformatic analysis of the TCGA STAD dataset revealed that AKT3, but not AKT1 or AKT2, is upregulated in the majority of E-cadherin-deficient gastric cancers. In conclusion, allosteric AKT inhibitors represent a promising class of drugs for chemoprevention and chemotherapy of cancers with E-cadherin loss.

14.
Sci Rep ; 9(1): 12511, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467357

ABSTRACT

The cell-cell adhesion protein E-cadherin (CDH1) is a tumor suppressor that is required to maintain cell adhesion, cell polarity and cell survival signalling. Somatic mutations in CDH1 are common in diffuse gastric cancer (DGC) and lobular breast cancer (LBC). In addition, germline mutations in CDH1 predispose to the autosomal dominant cancer syndrome Hereditary Diffuse Gastric Cancer (HDGC). One approach to target cells with mutations in specific tumor suppressor genes is synthetic lethality. To identify novel synthetic lethal compounds for the treatment of cancers associated with E-cadherin loss, we have undertaken a high-throughput screening campaign of ~114,000 lead-like compounds on an isogenic pair of human mammary epithelial cell lines - with and without CDH1 expression. This unbiased approach identified 12 novel compounds that preferentially harmed E-cadherin-deficient cells. Validation of these compounds using both real-time and end-point viability assays identified two novel compounds with significant synthetic lethal activity, thereby demonstrating that E-cadherin loss creates druggable vulnerabilities within tumor cells. In summary, we have identified novel synthetic lethal compounds that may provide a new strategy for the prevention and treatment of both sporadic and hereditary LBC and DGC.


Subject(s)
Antigens, CD/genetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Cadherins/genetics , Stomach Neoplasms/genetics , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Cadherins/deficiency , Cell Line, Tumor , Drug Screening Assays, Antitumor , Germ-Line Mutation , Humans , Stomach Neoplasms/metabolism
15.
Pract Lab Med ; 16: e00125, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31289732

ABSTRACT

OBJECTIVES: To compare the Roche Cell-Free DNA Collection Tubes® against the Streck Cell-Free DNA BCT®s for sample stability using Cell Free DNA (cfDNA) from healthy volunteers (n = 20). DESIGN & METHODS: Whole blood was drawn into five Roche and five Streck tubes per volunteer, stored at room temperature and processed at five different time points (Days 0, 4, 7, 10 and 14). One volunteer had blood drawn into ×10 K3EDTA tubes to observe the effect of no preservation buffer on White Blood Cell (WBC) lysis. DNA was extracted from the plasma and the concentration (ng/µL) measured using the Qubit Fluorometer® at each time point. The eluted DNA was further analysed by capillary electrophoresis to determine the proportion of cfDNA and gDNA contamination in the samples over the 14 days. RESULTS: There was no difference in individual (p = 0.097) and median paired (p = 0.26) DNA concentration across the five time points between the two tubes. However, a difference was observed for samples in the Roche tubes for pair days 0-7 (p = 0.01), 0 to 10 (p = 0.046) and 0 to 14 (p = 0.0016) in contrast to the Streck tubes after adjustment for multiple testing. CONCLUSION: The findings of this study indicate that the Roche Cell-Free DNA Collection Tubes® are a suitable alternative for sample collection and storage at room temperature, albeit for a duration of less than 7 days.

16.
Fam Cancer ; 18(3): 363-367, 2019 07.
Article in English | MEDLINE | ID: mdl-30989426

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.


Subject(s)
Adenocarcinoma/genetics , Antigens, CD/genetics , Cadherins/genetics , Polyps/genetics , Stomach Neoplasms/genetics , Family Health , Gene Silencing , Germ-Line Mutation , Humans , Stomach Neoplasms/pathology , Syndrome , alpha Catenin/genetics
17.
J Med Genet ; 56(4): 199-208, 2019 04.
Article in English | MEDLINE | ID: mdl-30661051

ABSTRACT

CDH1 encodes E-cadherin, a key protein in adherens junctions. Given that E-cadherin is involved in major cellular processes such as embryogenesis and maintenance of tissue architecture, it is no surprise that deleterious effects arise from its loss of function. E-cadherin is recognised as a tumour suppressor gene, and it is well established that CDH1 genetic alterations cause diffuse gastric cancer and lobular breast cancer-the foremost manifestations of the hereditary diffuse gastric cancer syndrome. However, in the last decade, evidence has emerged demonstrating that CDH1 mutations can be associated with lobular breast cancer and/or several congenital abnormalities, without any personal or family history of diffuse gastric cancer. To date, no genotype-phenotype correlations have been observed. Remarkably, there are reports of mutations affecting the same nucleotide but inducing distinct clinical outcomes. In this review, we bring together a comprehensive analysis of CDH1-associated disorders and germline alterations found in each trait, providing important insights into the biological mechanisms underlying E-cadherin's pleiotropic effects. Ultimately, this knowledge will impact genetic counselling and will be relevant to the assessment of risk of cancer development or congenital malformations in CDH1 mutation carriers.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Cell Differentiation/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Germ-Line Mutation , Alleles , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Cleft Lip/genetics , Cleft Palate/genetics , Ectropion/genetics , Female , Genetic Association Studies/methods , Genotype , Humans , Male , Stomach Neoplasms/genetics , Tooth Abnormalities/genetics
18.
Fam Cancer ; 18(1): 83-90, 2019 01.
Article in English | MEDLINE | ID: mdl-29589180

ABSTRACT

New Zealand Maori have a considerably higher incidence of gastric cancer compared to non-Maori, and are one of the few populations worldwide with a higher prevalence of diffuse-type disease. Pathogenic germline CDH1 mutations are causative of hereditary diffuse gastric cancer, a cancer predisposition syndrome primarily characterised by an extreme lifetime risk of developing diffuse gastric cancer. Pathogenic CDH1 mutations are well described in Maori families in New Zealand. However, the contribution of these mutations to the high incidence of gastric cancer is unknown. We have used next-generation sequencing, Sanger sequencing, and Multiplex Ligation-dependent Probe Amplification to examine germline CDH1 in an unselected series of 94 Maori gastric cancer patients and 200 healthy matched controls. Overall, 18% of all cases, 34% of cases diagnosed with diffuse-type gastric cancer, and 67% of cases diagnosed aged less than 45 years carried pathogenic CDH1 mutations. After adjusting for the effect of screening known HDGC families, we estimate that 6% of all advanced gastric cancers and 13% of all advanced diffuse-type gastric cancers would carry germline CDH1 mutations. Our results demonstrate that germline CDH1 mutations are a significant contributor to the high frequency of diffuse gastric cancer in New Zealand Maori.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Genetic Predisposition to Disease , Stomach Neoplasms/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , DNA Mutational Analysis , Female , Genetic Testing/methods , Genetic Testing/statistics & numerical data , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Incidence , Male , Middle Aged , New Zealand/epidemiology , Stomach Neoplasms/ethnology , Stomach Neoplasms/pathology , Young Adult
19.
Gastric Cancer ; 22(2): 273-286, 2019 03.
Article in English | MEDLINE | ID: mdl-30066183

ABSTRACT

BACKGROUND: The E-cadherin gene (CDH1) is frequently mutated in diffuse gastric cancer and lobular breast cancer, and germline mutations predispose to the cancer syndrome Hereditary Diffuse Gastric Cancer. We are taking a synthetic lethal approach to identify druggable vulnerabilities in CDH1-mutant cancers. METHODS: Density distributions of cell viability data from a genome-wide RNAi screen of isogenic MCF10A and MCF10A-CDH1-/- cells were used to identify protein classes affected by CDH1 mutation. The synthetic lethal relationship between selected protein classes and E-cadherin was characterised by drug sensitivity assays in both the isogenic breast MCF10A cells and CDH1-isogenic gastric NCI-N87. Endocytosis efficiency was quantified using cholera toxin B uptake. Pathway metagene expression of 415 TCGA gastric tumours was statistically correlated with CDH1 expression. RESULTS: MCF10A-CDH1-/- cells showed significantly altered sensitivity to RNAi inhibition of groups of genes including the PI3K/AKT pathway, GPCRs, ion channels, proteosomal subunit proteins and ubiquitinylation enzymes. Both MCF10A-CDH1-/- and NCI-N87-CDH1-/- cells were more sensitive than wild-type cells to compounds that disrupted plasma membrane composition and trafficking, but showed contrasting sensitivities to inhibitors of actin polymerisation and the chloride channel inhibitor NS3728. The MCF10A-CDH1-/- cell lines showed reduced capacity to endocytose cholera toxin B. Pathway metagene analysis identified 20 Reactome pathways that were potentially synthetic lethal in tumours. Genes involved in GPCR signalling, vesicle transport and the metabolism of PI3K and membrane lipids were strongly represented amongst the candidate synthetic lethal genes. CONCLUSIONS: E-cadherin loss leads to disturbances in receptor signalling and plasma membrane trafficking and organisation, creating druggable vulnerabilities.


Subject(s)
Cadherins/deficiency , Cell Membrane/metabolism , Cell Membrane/pathology , Antigens, CD/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Female , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Protein Transport/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
20.
Mol Cancer ; 17(1): 112, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068367

ABSTRACT

The aim of this study was to uncover the pathogenic relevance and the underlying molecular mechanism of a novel CDH1 variant found in a Hereditary Diffuse Gastric Cancer family (p.L13_L15del), which affects the signal peptide of E-cadherin without changing the remaining predicted sequence. We verified that p.L13_L15del cells yield low levels of E-cadherin, decreased cell adhesion and enhanced cell invasion. Further, we demonstrated that the disruption of the highly conserved hydrophobic core of the signal peptide hampers the binding of cellular components crucial for E-cadherin translation and translocation into the endoplasmic reticulum, constituting a new molecular basis for the loss of a tumour suppressor gene causative of hereditary cancer.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Protein Sorting Signals , Stomach Neoplasms/genetics , Adult , Cell Adhesion , Endoplasmic Reticulum/metabolism , Female , Genetic Variation , Humans , Male , Protein Transport , Sequence Analysis, DNA , Stomach Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...