Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
Add more filters

Publication year range
1.
Gastroenterology ; 165(1): 88-103, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36921674

ABSTRACT

BACKGROUND & AIMS: Gastric cancer (GC) is a major cancer type characterized by high heterogeneity in both tumor cells and the tumor immune microenvironment (TIME). One intractable GC subtype is gastric signet-ring cell carcinoma (GSRCC), which is associated with poor prognosis. However, it remains unclear what the GSRCC TIME characteristics are and how these characteristics may contribute to clinical outcomes. METHODS: We enrolled 32 patients with advanced GC of diverse subtypes and profiled their TIME using an immune-targeted single-cell profiling strategy, including (1) immune-targeted single-cell RNA sequencing (n = 20 patients) and (2) protein expression profiling by a targeted antibody panel for mass cytometry (n = 12 patients). We also generated matched V(D)J (variable, diversity, and joining gene segments) sequencing of T and B cells along CD45+ immunocytes. RESULTS: We found that compared to non-GSRCC, the GSRCC TIME appears to be quiescent, where both CD4+ and CD8+ T cells are difficult to be mobilized, which further impairs the proper functions of B cells. CXCL13, mainly produced by follicular helper T cells, T helper type 17, and exhausted CD8+ T cells, is a central coordinator of this transformation. We show that CXCL13 expression can predict the response to immune checkpoint blockade in GC patients, which may be related to its effects on tertiary lymphoid structures. CONCLUSIONS: Our study provides a comprehensive molecular portrait of immune cell compositions and cell states in advanced GC patients, highlighting adaptive immune irresponsiveness in GSRCC and a mediator role of CXCL13 in TIME. Our targeted single-cell transcriptomic and proteomic profiling represents a powerful approach for TIME-oriented translational research.


Subject(s)
Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , CD8-Positive T-Lymphocytes , Proteomics , Carcinoma, Signet Ring Cell/genetics , Tumor Microenvironment
2.
Lancet Oncol ; 24(1): 107-116, 2023 01.
Article in English | MEDLINE | ID: mdl-36509094

ABSTRACT

BACKGROUND: Hereditary diffuse gastric cancer, generally caused by germline pathogenic variants in CDH1, presents with early-onset signet ring cell carcinoma. Prophylactic total gastrectomy is the definitive treatment. Endoscopic surveillance can inform the timing of prophylactic total gastrectomy through detection of microscopic signet ring cell carcinoma foci. However, evidence is scarce about the optimal endoscopic sampling technique and characterisation of signet ring cell carcinoma foci in hereditary diffuse gastric cancer. We aimed to formally assess the diagnostic yield of different sampling strategies and to identify criteria for the characterisation of endoscopic lesions. METHODS: For this prospective longitudinal cohort study, we included individuals aged 18 years or older at the Cambridge University Hospitals National Health Service (NHS) Foundation Trust who fulfilled testing criteria for hereditary diffuse gastric cancer between June 1, 2005, and July 31, 2021. The primary outcome was detection of intramucosal signet ring cell carcinoma foci. We assessed the detection rate and anatomical location of signet ring cell carcinoma in random biopsy samples taken according to a systematic protocol compared with biopsies targeted to endoscopic findings. Endoscopic lesions were examined with white-light and narrow band imaging with magnification to assess the likelihood of cancerous foci. FINDINGS: 145 individuals were included, of whom 68 (47%) were male and 92 (63%) carried the CDH1 pathogenic variant. 58 (40%) patients were diagnosed with invasive signet ring cell carcinoma over a median follow-up time of 51 months (IQR 18-80). The first diagnosis of signet ring cell carcinoma was most commonly made from random biopsies (29 [50%] of 58 patients), rather than targeted biopsies (15 [26%] patients). The anatomical distribution of signet ring cell carcinoma foci detected by random biopsies more accurately reflected those identified in prophylactic total gastrectomy specimens than did targeted biopsies. Omitting random biopsies in our cohort would have led to an under-diagnosis rate of 42%. Using a novel panel of endoscopic criteria, gastric lesions containing signet ring cell carcinoma were predicted with a sensitivity of 67·3% and a specificity of 90·2%. INTERPRETATION: Random biopsies enhance the early detection of signet ring cell carcinoma and are complementary to targeted biopsies in surveillance of hereditary diffuse gastric cancer. This sampling method should be the standard of care when performing all surveillance endoscopies for individuals with hereditary diffuse gastric cancer. FUNDING: UK Medical Research Council.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Male , Female , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/surgery , Longitudinal Studies , Prospective Studies , State Medicine , Early Detection of Cancer , Biopsy , Carcinoma, Signet Ring Cell/diagnosis , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/surgery , Gastrectomy , Germ-Line Mutation
3.
Lancet Oncol ; 24(4): 383-391, 2023 04.
Article in English | MEDLINE | ID: mdl-36990610

ABSTRACT

BACKGROUND: Loss of function variants in CDH1 are the most frequent cause of hereditary diffuse gastric cancer. Endoscopy is regarded as insufficient for early detection due to the infiltrative phenotype of diffuse-type cancers. Microscopic foci of invasive signet ring cells are pathognomonic of CDH1 and precede development of diffuse gastric cancer. We aimed to assess the safety and effectiveness of endoscopy for cancer interception in individuals with germline CDH1 variants, particularly in those who declined prophylactic total gastrectomy. METHODS: In this prospective cohort study, we included asymptomatic patients aged 2 years or older with pathogenic or likely pathogenic germline CDH1 variants who underwent endoscopic screening and surveillance at the National Institutes of Health (Bethesda, MD, USA) as part of a natural history study of hereditary gastric cancers (NCT03030404). Endoscopy was done with non-targeted biopsies and one or more targeted biopsy and assessment of focal lesions. Endoscopy findings, pathological data, personal and family cancer history, and demographics were recorded. Procedural morbidity, gastric cancer detection by endoscopy and gastrectomy, and cancer-specific events were assessed. Screening was defined as the initial endoscopy and all subsequent endoscopies were considered surveillance; follow-up endoscopy was at 6 to 12 months. The primary aim was to determine effectiveness of endoscopic surveillance for detection of gastric signet ring cell carcinoma. FINDINGS: Between Jan 25, 2017, and Dec 12, 2021, 270 patients (median age 46·6 years [IQR 36·5-59·8], 173 [64%] female participants, 97 [36%] male participants; 250 [93%] were non-Hispanic White, eight [3%] were multiracial, four [2%] were non-Hispanic Black, three [1%] were Hispanic, two [1%] were Asian, and one [<1%] was American Indian or Alaskan Native) with germline CDH1 variants were screened, in whom 467 endoscopies were done as of data cutoff (April 30, 2022). 213 (79%) of 270 patients had a family history of gastric cancer, and 176 (65%) reported a family history of breast cancer. Median follow-up was 31·1 months (IQR 17·1-42·1). 38 803 total gastric biopsy samples were obtained, of which 1163 (3%) were positive for invasive signet ring cell carcinoma. Signet ring cell carcinoma was detected in 76 (63%) of 120 patients who had two or more surveillance endoscopies, of whom 74 had occult cancer detected; the remaining two individuals developed focal ulcerations each corresponding to pT3N0 stage carcinoma. 98 (36%) of 270 patients proceeded to prophylactic total gastrectomy. Among patients who had a prophylactic total gastrectomy after an endoscopy with biopsy samples negative for cancer (42 [43%] of 98), multifocal stage IA gastric carcinoma was detected in 39 (93%). Two (1%) participants died during follow-up, one due to metastatic lobular breast cancer and the other due to underlying cerebrovascular disease, and no participants were diagnosed with advanced stage (III or IV) cancer during follow-up. INTERPRETATION: In our cohort, endoscopic cancer surveillance was an acceptable alternative to surgery in individuals with CDH1 variants who declined total gastrectomy. The low rate of incident tumours (>T1a) suggests that surveillance might be a rational alternative to surgery in individuals with CDH1 variants. FUNDING: Intramural Research Program, National Institutes of Health.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Stomach Neoplasms , Male , Humans , Female , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology , Stomach Neoplasms/genetics , Prospective Studies , Gastrectomy/adverse effects , Adenocarcinoma/surgery , Carcinoma, Signet Ring Cell/diagnosis , Carcinoma, Signet Ring Cell/epidemiology , Carcinoma, Signet Ring Cell/genetics , Germ-Line Mutation , Genetic Predisposition to Disease
4.
Mod Pathol ; 36(9): 100251, 2023 09.
Article in English | MEDLINE | ID: mdl-37355152

ABSTRACT

Signet-ring cell (SRC)/poorly cohesive cell carcinoma is an aggressive variant of pancreatic ductal adenocarcinoma (PDAC). This study aimed to clarify its clinicopathologic and molecular profiles based on a multi-institutional cohort of 20 cases. The molecular profiles were investigated using DNA and RNA sequencing. The clinicopathologic parameters and molecular alterations were analyzed based on survival indices and using a validation/comparative cohort of 480 conventional PDAC patients. The primary findings were as follows: (1) clinicopathologic features: SRC carcinomas are highly aggressive neoplasms with poor prognosis, and the lungs are elective metastatic sites; (2) survival analysis: a higher SRC component was indicative of poorer prognosis. In particular, the most clinically significant threshold of SRC was 80%, showing statistically significant differences in both disease-specific and disease-free survival; (3) genomic profiles: SRC carcinomas are similar to conventional PDAC with the most common alterations affecting the classic PDAC drivers KRAS (70% of cases), TP53 (55%), SMAD4 (25%), and CDKN2A (20%). EGFR alterations, RET::CCDC6 fusion gene, and microsatellite instability (3 different cases, 1 alteration per case) represent novel targets for precision oncology. The occurrence of SMAD4 mutations was associated with poorer prognosis; (4) pancreatic SRC carcinomas are genetically different from gastric SRC carcinomas: CDH1, the classic driver gene of gastric SRC carcinoma, is not altered in pancreatic SRC carcinoma; (5) transcriptome analysis: the cases clustered into 2 groups, one classical/exocrine-like, and the other squamous-like; and (6) SRC carcinoma-derived organoids can be successfully generated, and their cultures preserve the histologic and molecular features of parental SRC carcinoma. Although pancreatic SRC carcinoma shares similarities with conventional PDAC regarding the most important genetic drivers, it also exhibits important differences. A personalized approach for patients with this tumor type should consider the clinical relevance of histologic determination of the SRC component and the presence of potentially actionable molecular targets.


Subject(s)
Carcinoma, Pancreatic Ductal , Carcinoma, Signet Ring Cell , Pancreatic Neoplasms , Humans , Precision Medicine , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Genomics , Prognosis , Pancreatic Neoplasms
5.
Gastrointest Endosc ; 98(3): 326-336.e3, 2023 09.
Article in English | MEDLINE | ID: mdl-37094689

ABSTRACT

BACKGROUND AND AIMS: Individuals with germline pathogenic CDH1 variants have a high risk of hereditary diffuse gastric cancer. The sensitivity of EGD in detecting signet ring cell carcinoma (SRCC) in this population is low. We aimed to identify endoscopic findings and biopsy practices associated with detection of SRCC. METHODS: This retrospective cohort included individuals with a germline pathogenic/likely pathogenic CDH1 variant undergoing at least 1 EGD at Memorial Sloan Kettering Cancer Center between January 1, 2006, and March 25, 2022. The primary outcome was detection of SRCC on EGD. Findings on gastrectomy were also assessed. The study included periods before and after implementation of the Cambridge protocol for endoscopic surveillance, allowing for assessment of a spectrum of biopsy practices. RESULTS: Ninety-eight CDH1 patients underwent at least 1 EGD at our institution. SRCC was detected in 20 (20%) individuals on EGD overall and in 50 (86%) of the 58 patients undergoing gastrectomy. Most SRCC foci were detected in the gastric cardia/fundus (EGD, 50%; gastrectomy, 62%) and body/transition zone (EGD, 60%; gastrectomy, 62%). Biopsy results of gastric pale mucosal areas were associated with detection of SRCC (P < .01). The total number of biopsy samples taken on EGD was associated with increased detection of SRCC (P = .01), with 43% detected when ≥40 samples were taken. CONCLUSIONS: Targeted biopsy sampling of gastric pale mucosal areas and increasing number of biopsy samples taken on EGD were associated with detection of SRCC. SRCC foci were mostly detected in the proximal stomach, supporting updated endoscopic surveillance guidelines. Further studies are needed to refine endoscopic protocols to improve SRCC detection in this high-risk population.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Antigens, CD , Cadherins/genetics , Carcinoma, Signet Ring Cell/diagnostic imaging , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Gastrectomy , Gastroscopy/methods , Germ-Line Mutation , Retrospective Studies , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
6.
Gastric Cancer ; 26(4): 553-564, 2023 07.
Article in English | MEDLINE | ID: mdl-37036539

ABSTRACT

BACKGROUND: Patients with poorly cohesive gastric carcinoma (PCC) are known to have poor survival. However, detailed molecular biology of PCC has not been elucidated, except for mutations in CDH1 and RHOA. Additionally, the molecular profiles of signet-ring cell carcinoma (SRC) have not been fully investigated. We aimed to investigate the association between molecular profiles and survival in PCC and PCC subtypes. METHODS: The present study included 455 patients with gastric adenocarcinoma underwent radical gastrectomy. Whole-exome sequencing and gene expression profiling were conducted. Patients were classified according to the WHO classification as PCC or non-PCC, with PCC being further classified into SRC, combined, and PCC not-otherwise-specified (NOS). Clinicopathological factors and survival were compared with molecular profiles. RESULTS: Of the patients, 159 were classified with PCC, while 296 were classified with non-PCC. Among PCC, 44 were classified with SRC, 64 with combined, and 51 with PCC-NOS. Mutations in CDH1 and RHOA were remarkably more frequent in PCC than in non-PCC. PCC had worse overall survival (OS) and disease-specific survival (DSS) compared to non-PCC. For PCC, the SRC group had good OS and DSS, whereas PCC-NOS classification with CDH1 mutations was associated with extremely poor survival. In the PCC-NOS and combined groups, patients with mutations in the extracellular domain 1 of CDH1 had poor survival. CONCLUSIONS: Our findings suggest that PCC has poorer survival than non-PCC. Accumulation of CDH1 and RHOA mutations are unique profiles in PCC. Among PCC, CDH1 mutations may play a crucial role in the survival of non-SRC PCC.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Adenocarcinoma/pathology , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/surgery , Mutation , Gastrectomy
7.
Am J Dermatopathol ; 45(9): 626-630, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37506279

ABSTRACT

ABSTRACT: Signet-ring cell/histiocytoid carcinoma (SRCHC) is a rare appendageal tumor, mainly considering eyelids, more rarely axillae. This article describes 2 novel SRCHC cases of 71- and 66-year-old men and systematically reviews the literature on SRCHC. Of all cases reported in the literature, 73 (91.2%) were men and 7 (8.8%) were women. The median age at diagnosis was 71 years. Skin changes were located in the eyelids (68%) and axillae (32%). In all tested cases, SRCHC cells expressed CK7, CKAE1/AE3, EMA, CAM5.2, and AR and PIK3CA mutations. Future research should determine whether AR/PIK3CA-targeted therapies influence patients' survival.


Subject(s)
Carcinoma, Signet Ring Cell , Eyelid Neoplasms , Skin Neoplasms , Male , Humans , Female , Aged , Immunohistochemistry , Carcinoma, Signet Ring Cell/epidemiology , Carcinoma, Signet Ring Cell/genetics , Skin Neoplasms/epidemiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Eyelid Neoplasms/pathology , Skin/pathology
8.
Br J Cancer ; 126(5): 797-803, 2022 03.
Article in English | MEDLINE | ID: mdl-34949788

ABSTRACT

BACKGROUND AND AIMS: CDH1 germline variants have been linked to heritability in diffuse gastric (DGC) and lobular breast cancer (LBC). Studies have not yet assessed whether CDH1 is a cancer-susceptibility gene in other cancers. Herein, we mapped the landscape of pathogenic and likely pathogenic (P/LP) germline variants in CDH1 across various cancers and ethnicities. METHODS: We evaluated CDH1 germline P/LP variants in 212,944 patients at one CLIA-certified laboratory (Invitae) and described their frequency in 7 cancer types. We screened for CDH1 variant enrichment in each cancer relative to a cancer-free population from The Genome Aggregation Database version 3 (gnomADv3). RESULTS: CDH1 P/LP variants were identified in 141 patients, most commonly in patients with DGC (27/408, 6.6%) followed by colorectal signet-ring cell cancer (CSRCC; 3/79, 3.8%), gastric cancer (56/2756, 2%), and LBC (22/6809, 0.3%). CDH1 P/LP variants were enriched in specific ethnic populations with breast cancer, gastric cancer, CRC, LBC, DGC, and CSRCC compared to matched ethnicities from gnomADv3. CONCLUSION: We report for the first time the prevalence of P/LP CDH1 variants across several cancers and show significant enrichment in CDH1 P/LP variants for patients with CSRCC, DGC, and LBC across various ethnicities. Future prospective studies are warranted to validate these findings.


Subject(s)
Antigens, CD/genetics , Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma, Lobular/genetics , Carcinoma, Signet Ring Cell/genetics , Colorectal Neoplasms/genetics , Germ-Line Mutation , Stomach Neoplasms/genetics , Adult , Aged , Breast Neoplasms/ethnology , Carcinoma, Lobular/ethnology , Carcinoma, Signet Ring Cell/ethnology , Colorectal Neoplasms/ethnology , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation Rate , Prevalence , Sequence Analysis, DNA , Stomach Neoplasms/ethnology , Young Adult
9.
Gastroenterology ; 160(3): 823-830, 2021 02.
Article in English | MEDLINE | ID: mdl-33217450

ABSTRACT

BACKGROUND AND AIMS: Diffuse-type gastric cancer (GC) is currently subdivided into signet-ring cell carcinoma (SRCC) and non-SRCC, referred to as poorly cohesive carcinoma not otherwise specified (PCC-NOS). Although these subtypes are considered to be independent, they often coexist in the same tumors, raising a question of whether they clonally differ or not. To tackle this question, we established an experimental platform for human diffuse GC that enables accurate modeling of histologic subtypes. METHODS: Seven patient-derived diffuse GC organoid lines were established, characterized by histopathologic analysis, in situ hybridization, and gene expression analysis. For genetic modeling of diffuse GC, we knocked out CDH1 and/or TP53 in human normal gastric organoids. Green fluorescent protein-labeled GC organoids were xenotransplanted into immune-deficient mice for in vivo assessment. RESULTS: PCC-NOS organoids transformed into SRCC-like structures on removal of Wnt and R-spondin from the culture medium. This morphologic change paralleled downregulation of Wnt-target and gastric stem cell genes, including LGR5, and elevation of differentiation markers, such as KRT20 and MUCs. The association between Wnt target gene expression and histologic subtypes was confirmed in 3 patient-derived GC tissues. In vivo, single clone-derived organoids formed tumors that comprised 2 distinct histologic compartments, each corresponding to SRCC and PCC-NOS. The transition from PCC-NOS to SRCC histology reflected the abundance of surrounding R-spondin-expressing fibroblasts. CONCLUSIONS: SRCC and PCC-NOS were clonally identical, and their morphology was regulated by extracellular Wnt and R-spondin expression. Our results decoded how genetic mutations and the tumor environment shape pathohistologic and biologic phenotypes in human diffuse GCs.


Subject(s)
Carcinoma, Signet Ring Cell/parasitology , Gastric Mucosa/pathology , Stomach Neoplasms/pathology , Wnt Signaling Pathway , Aged , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Signet Ring Cell/genetics , Female , Gastric Mucosa/cytology , Gene Knockout Techniques , Humans , Male , Mice , Middle Aged , Organoids/pathology , Primary Cell Culture , RNA-Seq , Stomach Neoplasms/genetics , Thrombospondins , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Exome Sequencing , Xenograft Model Antitumor Assays
10.
Gastric Cancer ; 25(5): 862-878, 2022 09.
Article in English | MEDLINE | ID: mdl-35661943

ABSTRACT

BACKGROUND: Loss of E-cadherin expression is frequently observed in signet ring carcinoma (SRCC). People with germline mutations in CDH1, which encodes E-cadherin, develop diffuse gastric cancer at a higher rate. Loss of E-cadherin expression is thus assumed to trigger oncogenic development. METHODS: To investigate novel therapeutic targets for gastric SRCC, we engineered an E-cadherin-deficient SRCC model in vitro using a human gastric organoid (hGO) with CDH1 knockout (KO). RESULTS: CDH1 KO hGO cells demonstrated distinctive morphological changes similar to SRCC and high cell motility. RNA-sequencing revealed up-regulation of matrix metalloproteinase (MMP) genes in CDH1 KO hGO cells compared to wild type. MMP inhibitors suppressed cell motility of CDH1 KO hGO cells and SRCC cell lines in vitro. Immunofluorescent analysis with 95 clinical gastric cancer tissues revealed that MMP-3 was specifically abundant in E-cadherin-aberrant SRCC. In addition, CXCR4 molecules translocated onto the cell membrane after CDH1 KO. Addition of CXCL12, a ligand of CXCR4, to the culture medium prolonged cell survival of CDH1 KO hGO cells and was abolished by the inhibitor, AMD3100. In clinical SRCC samples, CXCL12-secreting fibroblasts showed marked infiltration into the cancer area. CONCLUSIONS: E-cadherin deficient SRCCs might gain cell motility through upregulation of MMPs. CXCL12-positive cancer-associated fibroblasts could serve to maintain cancer-cell survival as a niche. MMPs and the CXCL12/CXCR4 axis represent promising candidates as novel therapeutic targets for E-cadherin-deficient SRCC.


Subject(s)
Carcinoma, Signet Ring Cell , Stomach Neoplasms , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Gene Expression Profiling , Germ-Line Mutation , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
11.
Langenbecks Arch Surg ; 407(1): 87-98, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34505199

ABSTRACT

PURPOSE: There has been an increase in the incidence of signet ring cell cancer (SRCC) of the stomach and gastro-esophageal junction (GEJ). The multistage carcinogenesis involving genetic and epigenetic aberrations may have a major role in the increasing incidence of SRCC. Although there are numerous studies on the prognostic value of SRCC, they are markedly inconsistent in their results, making it impossible to draw any meaningful conclusions. We aimed to examine the available evidences on molecular alterations and stage-stratified treatment approaches in SRCC of the stomach and GEJ. METHODS: A systematic search was carried out in PubMed. Studies available in English related to SRCC of stomach and gastro-esophageal junction were identified and evaluated. RESULTS: This study reviewed the current evidence and provided an insight into the molecular alterations, stage-stratified treatment approaches, and future challenges in the management of SRCC of the stomach and GEJ. Specific therapeutic strategies and personalized multimodal treatment have been recommended based on the tumor characteristics of SRCC. CONCLUSION: Multistage carcinogenesis involving genetic and epigenetic aberrations in SRCC is interlinked with stage-dependent prognosis. Specific therapeutic strategy and personalized multimodal treatment should be followed based on the tumor characteristics of SRCC. Endoscopic resection, radical surgery, and perioperative chemotherapy should be offered in carefully selected patients based on stage and prognostic stratification. Future studies in genetic and molecular analysis, histopathological classification, and options of multimodality treatment will improve the prognosis and oncological outcomes in SRCC of gastric and GEJ.


Subject(s)
Carcinoma, Signet Ring Cell , Stomach Neoplasms , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/therapy , Combined Modality Therapy , Esophagogastric Junction , Humans , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy
12.
Pathologe ; 43(1): 8-15, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34807275

ABSTRACT

The fifth edition of the World Health Organization (WHO) classification of digestive system tumours was published in 2019. The classification of invasive carcinoma is basically the same as in the fourth edition, but the description of each histological type has been updated, and some rare subtypes such as micropapillary carcinoma, gastric adenocarcinoma of the fundic gland type and undifferentiated carcinoma have been added and explained. Although this classification did not provide specific numerical criteria for the diagnosis of signet-ring cell carcinoma in poorly cohesive carcinoma, an additional study defined signet-ring cell carcinoma as having more than 90% signet-ring cells. The molecular classification of gastric cancer (Epstein-Barr virus-positive type, microsatellite instability type, genomically stable type, chromosomally unstable type) was additionally introduced. Many pages in the present classification have been devoted to precancerous lesions, and this article focuses on foveolar-type adenoma/dysplasia.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Epstein-Barr Virus Infections , Stomach Neoplasms , Adenocarcinoma/genetics , Carcinoma, Signet Ring Cell/genetics , Herpesvirus 4, Human , Humans , Stomach Neoplasms/genetics , World Health Organization
13.
Medicina (Kaunas) ; 58(7)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35888555

ABSTRACT

Background: Signet ring cell carcinoma (SC) accounts for 1% of total colorectal cancer (CRC) cases and is associated with aggressive behaviors, such as lymphatic invasion and distant metastases, resulting in poor prognosis. To date, there is still a lack of consensus on the genetic etiology underpinning this cancer subtype. This study aimed to clarify the molecular associations of SC by using meta-analysis and a systematic review. Methods: PubMed, Embase, and Cochrane Library were searched for studies evaluating the KRAS, BRAF, P53 statuses, and microsatellite instability (MSI) in CRC patients with different histological subtypes, including SC. The diagnosis of SC is defined as the signet ring cells comprising ≥50 percent of the tumor mass. By dividing the studies into subgroups based on the composition of control groups, such as classic adenocarcinoma (AC; no SC components) and non-SC (including those with SC components < 50%), the relative risk (RR) of molecular alterations for SC in each study were pooled using a random-effects model. Two reviewers identified trials for inclusion, assessed quality, and extracted data independently. Results: Data from 29 studies consisting of 9366 patients were included in this analysis. SC was associated positively with MSI (RR 1.78, 95% CI 1.34 to 2.37; 95% CI 0.77 to 4.15; p = 0.0005), BRAF mutation (RR 1.99, 95% CI 1.21 to 3.26; 95%CI 0.68 to 5.82; p = 0.0146), and negatively with KRAS mutation (RR 0.48, 95% CI 0.29 to 0.78; 95% CI 0.09 to 2.49; p = 0.0062). No association was found between SC and P53 expression (RR 0.92, 95% CI 0.76 to 1.13; 95%CI 0.61 to 1.39; p = 0.3790). Moreover, it was associated negatively with P53 gene mutations (RR 0.92, 95% CI 0.77 to 1.09; 95% CI 0.46 to 1.82; p = 0.1568), and P53 protein (RR 0.93, 95% CI 0.58 to 1.49; 95% CI 0.40 to 2.17; p = 0.6885). Conclusions: The molecular etiology of SC may be associated with the BRAF and MSI pathways. Its features, such as the high frequency of BRAF mutation, could partly explain its less favorable outcomes and limited effects of traditional chemotherapy.


Subject(s)
Carcinoma, Signet Ring Cell , Colorectal Neoplasms , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Microsatellite Instability , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
14.
Clin Gastroenterol Hepatol ; 19(1): 189-191, 2021 01.
Article in English | MEDLINE | ID: mdl-31678201

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) syndrome results from a germline CDH1 mutation, and microscopic foci of signet-ring carcinoma cells (SRCC) are present in nearly all gastrectomy specimens.1 The lifetime risk of invasive gastric cancer (GC) has been thought to be 70%,2 but recent data have suggested a lower risk of 37%.3 Prophylactic total gastrectomy is considered the standard of care, but many patients choose surveillance endoscopy instead. We sought to define the outcomes in CDH1-positive individuals who pursued endoscopic surveillance.


Subject(s)
Carcinoma, Signet Ring Cell , Neoplastic Syndromes, Hereditary , Stomach Neoplasms , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/surgery , Gastrectomy , Gastroscopy , Genetic Predisposition to Disease , Humans , Neoplastic Syndromes, Hereditary/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/surgery
15.
Gastric Cancer ; 24(5): 1102-1114, 2021 09.
Article in English | MEDLINE | ID: mdl-33961152

ABSTRACT

BACKGROUND: In Helicobacter pylori (Hp)-uninfected individuals, diffuse-type gastric cancer (DGC) was reported as the most common type of cancer. However, the carcinogenic mechanism of Hp-uninfected sporadic DGC is largely unknown. METHODS: We performed whole-exome sequencing of Hp-uninfected DGCs and Hp-uninfected normal gastric mucosa. For advanced DGCs, external datasets were also analyzed. RESULTS: Eighteen patients (aged 29-78 years) with DGCs and nine normal subjects (28-77 years) were examined. The mutation burden in intramucosal DGCs (10-66 mutations per exome) from individuals aged 29-73 years was not very different from that in the normal gastric glands, which showed a constant mutation accumulation rate (0.33 mutations/exome/year). Unbiased dN/dS analysis showed that CDH1 somatic mutation was a driver mutation for intramucosal DGC. CDH1 mutation was more frequent in intramucosal DGCs (67%) than in advanced DGCs (27%). In contrast, TP53 mutation was more frequent in advanced DGCs (52%) than in intramucosal DGCs (0%). This discrepancy in mutations suggests that CDH1-mutated intramucosal DGCs make a relatively small contribution to advanced DGC formation. Among the 16 intramucosal DGCs (median size, 6.5 mm), 15 DGCs were pure signet ring cell carcinoma (SRCC) with reduced E-cadherin expression and a low proliferative capacity (median Ki-67 index, 2.4%). Five SRCCs reviewed endoscopically over 2-5 years showed no progression. CONCLUSIONS: Impaired E-cadherin function due to CDH1 mutation was considered as an early carcinogenic event of Hp-uninfected intramucosal SRCC. Genetic and clinical analyses suggest that Hp-uninfected intramucosal SRCCs may be less likely to develop into advanced DGCs.


Subject(s)
Carcinoma, Signet Ring Cell , Helicobacter pylori , Stomach Neoplasms , Antigens, CD/genetics , Cadherins/genetics , Carcinoma, Signet Ring Cell/genetics , Helicobacter pylori/genetics , Humans , Mutation , Stomach Neoplasms/genetics
16.
J Cutan Pathol ; 48(1): 102-105, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32770562

ABSTRACT

Signet-ring cell/histiocytoid carcinoma (SRCHC) is a very rare skin appendage cancer, with an extremely rare occurrence in the axilla. This study describes the 11th case of SRCHC occurring in the axilla and reports the first gene alteration analysis performed for SRCHC. An 85-year-old Japanese male presented with a tumor in the left axilla. Biopsy of the axilla nodule demonstrated diffuse proliferation of histiocytoid neoplastic cells and signet-ring cells in the dermis and subcutis. Immunohistochemistry revealed loss of E-cadherin expression in these neoplastic cells. Accordingly, SRCHC of the axilla was diagnosed. Genetic analysis using next-generation sequencing demonstrated missense mutation of PIK3CA (c1633G>A, pGlu545Lys) and no CDH1 gene mutation.SRCHC of the axilla is considered equivalent to a histiocytoid variant of invasive lobular breast carcinoma. The present SRCHC case demonstrated a pathogenic PIK3CA mutation, which is observed in invasive lobular carcinoma. Additional large case studies are required to clarify the clinicopathological features and gene alterations in SRCHC of the axilla.


Subject(s)
Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Aged, 80 and over , Axilla , Carcinoma, Transitional Cell/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation, Missense , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Sequence Analysis, DNA , Urinary Bladder Neoplasms/pathology
17.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830394

ABSTRACT

Signet ring cell gastric carcinoma (SRCGC) is a lethal malignancy that has developed drug resistance to cisplatin therapies. The aim of this study was to characterize the acquisition of the cisplatin-resistance SRCGC cell line (KATO/DDP cells) and to understand the molecular mechanisms underlying cisplatin resistance. Transcriptomic and bioinformatic analyses were used to identify the candidate gene. This was confirmed by qPCR and Western blot. Aldoketoreductase1C1 and 1C3 (AKR1C1 and AKR1C3) were the most promising molecules in KATO/DDP cells. A specific inhibitor of AKR1C1 (5PBSA) and AKR1C3 (ASP9521) was used to enhance cisplatin-induced KATO/DPP cell death. Although cisplatin alone induced KATO/DDP apoptosis, a combination treatment of cisplatin and the AKR1C inhibitors had no influence on percent cell apoptosis. In conjunction with the autophagy inhibitor, 3MA, attenuated the effects of 5PBSA or ASP9521 to enhance cisplatin-induced cell death. These results indicated that AKR1C1 and 1C3 regulated cisplatin-induced KATO/DDP cell death via autophagy. Moreover, cisplatin in combination with AKR1C inhibitors and N-acetyl cysteine increased KATO/DDP cells' viability when compared with a combination treatment of cisplatin and the inhibitors. Taken together, our results suggested that AKR1C1 and 1C3 play a crucial role in cisplatin resistance of SRCGC by regulating redox-dependent autophagy.


Subject(s)
20-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3/genetics , Carcinoma, Signet Ring Cell/drug therapy , Stomach Neoplasms/drug therapy , Autophagic Cell Death/drug effects , Autophagic Cell Death/genetics , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptome/drug effects
18.
Gastroenterology ; 157(1): 87-96, 2019 07.
Article in English | MEDLINE | ID: mdl-30935944

ABSTRACT

BACKGROUND & AIMS: Pathogenic germline variants in CDH1 are associated with risk for diffuse gastric cancer (DGC) and lobular breast cancer. The reported high incidence of DGC and limited sensitivity of endoscopy in detection have prompted recommendation for total prophylactic gastrectomy for carriers of pathogenic or likely pathogenic (PLP) germline variants of CDH1. Multigene panel tests have identified increasing numbers of carriers of PLP variants in CDH1 who lack a family history of DGC. We evaluated outcomes of endoscopic surveillance for carriers of PLP variants of CDH1 with and without family history of DGC. METHODS: Individuals from 13 families with germline PLP variants of CDH1 were evaluated at the Michigan Medicine Cancer Genetics Clinic from January 1998 through May 2018. Outcomes of esophagogastroduodenoscopy examinations, histopathology analyses, and surgery were compared between individuals with and without a family history of DGC. RESULTS: We identified 20 carriers of germline CDH1 PLP variants; they underwent endoscopic examinations and/or gastrectomy. None had abnormal findings visible during endoscopy. Signet ring cell carcinoma (SRCC) was detected in 12 of 20 subjects. All but 1 of the carcinomas were tiny and confined to the lamina propria, and 1 was transmural. Seven of 12 subjects who had SRCC reported no diagnoses of DGC in first-degree relatives and did not meet established criteria for CDH1 analysis based on a 3-generation family pedigree. CONCLUSIONS: More than half of individuals with germlines variants of CDH1 that are PLP had histopathologic evidence for DGC on endoscopy and/or gastrectomy. Family history of DGC and endoscopic findings therefore do not appear to be reliable determinants of risk of SRCC in individuals with genetic predisposition to DGC.


Subject(s)
Carcinoma, Signet Ring Cell/diagnosis , Early Detection of Cancer/methods , Endoscopy, Digestive System , Neoplastic Syndromes, Hereditary/genetics , Stomach Neoplasms/diagnosis , Adult , Aged , Antigens, CD/genetics , Cadherins/genetics , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/prevention & control , Case-Control Studies , Female , Gastrectomy , Genetic Predisposition to Disease , Germ-Line Mutation , Heterozygote , Humans , Male , Middle Aged , Pedigree , Prophylactic Surgical Procedures , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control , Young Adult
19.
J Surg Res ; 250: 193-199, 2020 06.
Article in English | MEDLINE | ID: mdl-32078828

ABSTRACT

BACKGROUND: The signet-ring cell carcinoma (SRC), one of the histological subtypes of gastric cancer, often exhibits aggressive behavior in clinical practice and is therefore a commonly found subtype in advanced gastric cancer. However, SRC, especially at the early stages, has recently been reported to show a lower potential for malignancy than other pathological subtypes. Because the detailed molecular mechanisms underlying SRC pathology remain unclear, we focused on microRNAs (miRs) and aimed to elucidate the molecular mechanisms involved in endowing early SRC with its characteristic properties. MATERIALS AND METHODS: Formalin-fixed paraffin-embedded samples of patients diagnosed with various pathological subtypes of gastric cancer including SRC were used to purify and subsequently evaluate miRs. RESULTS: As expected, patients with SRC were observed to have significantly high miR-99a-5p expression. Further, high miR-99a-5p expression was found to closely correlate with less aggressive clinicopathological features, and functional studies probed by overexpression of miR-99a-5p resulted in inhibition of proliferation in two SRC cell lines. CONCLUSIONS: The present study reports that high levels of miR-99a-5p were observed in patients with SRC, particularly those with early stage SRC. In addition, miR-99a-5p expression was found to be related to cell proliferation. Therefore, miR-99a-5p could emerge as a diagnostic biomarker for early SRC and lymph node metastases or related adverse prognosis in patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Signet Ring Cell/diagnosis , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Stomach Neoplasms/diagnosis , Aged , Biomarkers, Tumor/analysis , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/mortality , Carcinoma, Signet Ring Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , Kaplan-Meier Estimate , Male , MicroRNAs/analysis , Prognosis , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
20.
Int J Gynecol Pathol ; 39(2): 193-198, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30676431

ABSTRACT

Signet-ring stromal tumor (SRST) is a rare benign stromal neoplasm of the ovary with only a handful of cases reported in the literature. To date, all but one reported cases have been unilateral, and the pathogenesis and underlying genetic abnormalities of this entity are not well characterized. Here we report a case of a 70-yr-old woman with bilateral ovarian SRST, clinically presenting with abdominal distention and rectal bleeding, and bilateral ovarian masses on imaging studies. Total hysterectomy and bilateral salpingo-oophorectomy were performed, revealing bilateral solid ovarian tumors with characteristic signet-ring cell morphology. The immunohistochemical profile-positive steroidogenic factor 1, calretinin, and smooth muscle actin, and negative epithelial membrane antigen-supported the diagnosis of SRST. CTNNB1 mutation and abnormal nuclear beta-catenin expression have recently been reported in 2 SRSTs. However, we did not identify any mutations in 54 oncogenes and tumor suppressor genes (including CTNNB1) by next-generation sequencing analysis, and PCR Sanger sequencing did not reveal FOXL2 C134W mutation, suggesting the possibility of heterogenous pathogenesis of these tumors.


Subject(s)
Carcinoma, Signet Ring Cell/pathology , Ovarian Neoplasms/pathology , Aged , Biomarkers, Tumor/analysis , Carcinoma, Signet Ring Cell/genetics , Female , Forkhead Box Protein L2/genetics , High-Throughput Nucleotide Sequencing , Humans , Ovarian Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL