Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Raquel Seruca is active.

Publication


Featured researches published by Raquel Seruca.


Oncogene | 2003

BRAF mutations and RET/PTC rearrangements are alternative events in the etiopathogenesis of PTC.

Paula Soares; Vítor Trovisco; Ana Sofia Rocha; Jorge Lima; Patrícia Castro; Ana Preto; Valdemar Máximo; Tiago Botelho; Raquel Seruca; Manuel Sobrinho-Simões

Rearrangement of RET proto-oncogene is the major event in the etiopathogenesis of papillary thyroid carcinoma (PTC). We report a high prevalence of BRAFV599E mutation in sporadic PTC and in PTC-derived cell lines. The BRAFV599E mutation was detected in 23 of 50 PTC (46%) and in three of four PTC-derived cell lines. The prevalence of the BRAFV599E mutation in PTC is the highest reported to date in human carcinomas, being only exceeded by melanoma. PTC with RET/PTC rearrangement as well as the TPC-1 cell line (the only one harboring RET/PTC rearrangement) did not show the BRAFV599E mutation. BRAFV599E mutation was not detected in any of 23 nodular goiters, 51 follicular adenomas and 18 follicular carcinomas. A distinct mutation in BRAF (codon K600E) was detected in a follicular adenoma. Activating mutations in RAS genes were detected in 15% of FA, 33% of FTC and 7% of PTC. BRAFV599E mutation did not coexist with alterations in any of the RAS genes in any of the tumors. These results suggest that BRAFV599E mutation is frequent in the etiopathogenesis of PTC. The BRAFV599E mutation appears to be an alternative event to RET/PTC rearrangement rather than to RAS mutations, which are rare in PTC. BRAFV599E may represent an alternative pathway to oncogenic MAPK activation in PTCs without RET/PTC activation.


Gastroenterology | 2003

A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma

José Carlos Machado; Ceu Figueiredo; Paulo Canedo; Paul Pharoah; Ralph Carvalho; Sérgio Nabais; Catarina Castro Alves; Maria Luisa Campos; Leen-Jan van Doorn; Carlos Caldas; Raquel Seruca; Fátima Carneiro; Manuel Sobrinho-Simões

BACKGROUND & AIMS Pro-inflammatory polymorphisms within the genes interleukin (IL)-1B and IL-1RN are associated with risk for gastric carcinoma (GC) in Helicobacter pylori-infected individuals. We aimed to determine the association between variation of the tumor necrosis factor (TNF)-alpha gene and the risk for chronic atrophic gastritis (CAG) and GC. We also investigated the extent to which the combined effect of proinflammatory genetic polymorphisms (IL-1B, IL-1RN, and TNF-alpha), and the combined effect of TNF-alpha and bacterial genotypes each influence such a risk. METHODS In a case-control study including 306 controls, 221 individuals with chronic gastritis, and 287 GC patients, the TNF-alpha-308 and IL-1B-511 bi-allelic polymorphisms, the IL-1RN variable number of tandem repeats (VNTR), and the H. pylori genes vacA (s and m regions) and cagA were genotyped. RESULTS We found that carriers of the TNF-alpha-308*A allele are at increased risk for GC development with an odds ratio (OR) of 1.9 (95% confidence interval [CI], 1.3-2.7). For both CAG and GC, the odds of developing disease increased with the number of high-risk genotypes. Individuals carrying high-risk genotypes at the 3 loci are at increased risk for CAG and GC with an OR of 5.8 (95% CI, 1.1-31.0) and 9.7 (95% CI, 2.6-36.0), respectively. The risk for GC was not affected significantly by the combination of bacterial and TNF-alpha-308 genotypes. CONCLUSIONS These findings show that a proinflammatory polymorphism in the TNF-alpha gene is associated with increased risk for GC, and that it is possible to define a specific genetic profile associated with highest risk for CAG and GC.


Journal of Medical Genetics | 1999

Familial gastric cancer: overview and guidelines for management*

Carlos Caldas; Fátima Carneiro; Henry T. Lynch; Jun Yokota; Georgia L. Wiesner; Steven M. Powell; Frank R. Lewis; David Huntsman; Paul Pharoah; Janusz Jankowski; Patrick MacLeod; Holger Vogelsang; Gisela Keller; Ken G M Park; Frances M. Richards; Eamonn R. Maher; Simon A. Gayther; Carla Oliveira; Nicola Grehan; Derek Wight; Raquel Seruca; Franco Roviello; Bruce A.J. Ponder; Charles E. Jackson

Families with autosomal dominant inherited predisposition to gastric cancer have been described. More recently, germlineE-cadherin/CDH1mutations have been identified in hereditary diffuse gastric cancer kindred. The need to have protocols to manage and counsel these families in the clinic led a group of geneticists, gastroenterologists, surgeons, oncologists, pathologists, and molecular biologists to convene a workshop to produce consensus statements and guidelines for familial gastric cancer. Review of the available cancer pathology from people belonging to families with documented germlineE-cadherin/CDH1mutations confirmed that the gastric cancers were all of the diffuse type. Criteria to define the different types of familial gastric cancer syndromes were agreed. Foremost among these criteria was that review of histopathology should be part of the evaluation of any family with aggregation of gastric cancer cases. Guidelines for genetic testing and counselling in hereditary diffuse gastric cancer were produced. Finally, a proposed strategy for clinical management in families with high penetrance autosomal dominant predisposition to gastric cancer was defined.


The New England Journal of Medicine | 2001

Early Gastric Cancer in Young, Asymptomatic Carriers of Germ-Line E-Cadherin Mutations

David Huntsman; Fátima Carneiro; Frank R. Lewis; Patrick MacLeod; Allen Hayashi; Kristin G. Monaghan; Raymond Maung; Raquel Seruca; Charles E. Jackson; Carlos Caldas

BACKGROUND Germ-line truncating mutations in the E-cadherin (CDH1) gene have been found in families with hereditary diffuse gastric cancer. These families are characterized by a highly penetrant susceptibility to diffuse gastric cancer with an autosomal dominant pattern of inheritance, predominantly in young persons. We describe genetic screening, surgical management, and pathological findings in young persons with truncating mutations in CDH1 from two unrelated families with hereditary diffuse gastric cancer. METHODS Mutation-specific predictive genetic testing was performed by polymerase-chain-reaction amplification, followed by restriction-enzyme digestion and DNA sequencing in Family 1 and by heteroduplex analysis in Family 2. A total gastrectomy was performed prophylactically in five carriers of mutations who were between 22 and 40 years old. In each case, the entire mucosa of the stomach was extensively sampled for microscopical analysis. RESULTS Superficial infiltrates of malignant signet-ring cells were identified in the surgical samples from all five persons who underwent gastrectomy. These early diffuse gastric cancers were multifocal in three of the five cases, and in one person infiltrates of malignant signet-ring cells were present in 65 of the 140 tissue blocks analyzed, representing in aggregate less than 2 percent of the gastric mucosa. CONCLUSIONS We recommend genetic counseling and consideration of prophylactic gastrectomy in young, asymptomatic carriers of germ-line truncating CDH1 mutations who belong to families with highly penetrant hereditary diffuse gastric cancer.


Journal of Medical Genetics | 2004

BRAF screening as a low-cost effective strategy for simplifying HNPCC genetic testing

Enric Domingo; Päivi Laiho; Miina Ollikainen; Mafalda Pinto; Liang Wang; Amy J. French; Jantine L. Westra; Thierry Frebourg; E Espin; Manel Armengol; Richard Hamelin; Hiroyuki Yamamoto; Robert Hofstra; Raquel Seruca; Annika Lindblom; Päivi Peltomäki; Stephen N. Thibodeau; Lauri A. Aaltonen; Simó Schwartz

Background: According to the international criteria for hereditary non-polyposis colorectal cancer (HNPCC) diagnostics, cancer patients with a family history or early onset of colorectal tumours showing high microsatellite instability (MSI-H) should receive genetic counselling and be offered testing for germline mutations in DNA repair genes, mainly MLH1 and MSH2. Recently, an oncogenic V600E hotspot mutation within BRAF, a kinase encoding gene from the RAS/RAF/MAPK pathway, has been found to be associated with sporadic MSI-H colon cancer, but its association with HNPCC remains to be further clarified. Methods: BRAF-V600E mutations were analysed by automatic sequencing in colorectal cancers from 206 sporadic cases with MSI-H and 111 HNPCC cases with known germline mutations in MLH1 and MSH2. In addition, 45 HNPCC cases showing abnormal immunostaining for MSH2 were also analysed. Results: The BRAF-V600E hotspot mutation was found in 40% (82/206) of the sporadic MSI-H tumours analysed but in none of the 111 tested HNPCC tumours or in the 45 cases showing abnormal MSH2 immunostaining. Conclusions: Detection of the V600E mutation in a colorectal MSI-H tumour argues against the presence of a germline mutation in either the MLH1 or MSH2 gene. Therefore, screening of these mismatch repair (MMR) genes can be avoided in cases positive for V600E if no other significant evidence, such as fulfilment of the strict Amsterdam criteria, suggests MMR associated HNPCC. In this context, mutation analysis of the BRAF hotspot is a reliable, fast, and low cost strategy which simplifies genetic testing for HNPCC.


Journal of Medical Genetics | 2004

Germline E-cadherin mutations in hereditary diffuse gastric cancer: assessment of 42 new families and review of genetic screening criteria

Angela Brooks-Wilson; Pardeep Kaurah; Gianpaolo Suriano; Stephen Leach; Janine Senz; Nicola Grehan; Yaron S N Butterfield; J Jeyes; J Schinas; J Bacani; Megan M. Kelsey; Paulo A. Ferreira; B MacGillivray; Patrick MacLeod; M Micek; James M. Ford; William D. Foulkes; Karlene Australie; C. R. Greenberg; M LaPointe; Catherine Gilpin; S Nikkel; Dawna Gilchrist; R Hughes; Charles E. Jackson; Kristin G. Monaghan; Maria José Oliveira; Raquel Seruca; Steve Gallinger; Carlos Caldas

Background: Mutations in the E-cadherin (CDH1) gene are a well documented cause of hereditary diffuse gastric cancer (HDGC). Development of evidence based guidelines for CDH1 screening for HDGC have been complicated by its rarity, variable penetrance, and lack of founder mutations. Methods: Forty three new gastric cancer (GC) families were ascertained from multiple sources. In 42 of these families at least one gastric cancer was pathologically confirmed to be a diffuse gastric cancer (DGC); the other family had intestinal type gastric cancers. Screening of the entire coding region of the CDH1 gene and all intron/exon boundaries was performed by bi-directional sequencing. Results: Novel mutations were found in 13 of the 42 DGC families (31% overall). Twelve of these mutations occur among the 25 families with multiple cases of gastric cancer and with pathologic confirmation of diffuse gastric cancer phenotype in at least one individual under the age of 50 years. The mutations found include small insertions and deletions, splice site mutations, and three non-conservative amino acid substitutions (A298T, W409R, and R732Q). All three missense mutations conferred loss of E-cadherin function in in vitro assays. Multiple cases of breast cancers including pathologically confirmed lobular breast cancers were observed both in mutation positive and negative families. Conclusion: Germline truncating CDH1 mutations are found in 48% of families with multiple cases of gastric cancer and at least one documented case of DGC in an individual under 50 years of age. We recommend that these criteria be used for selecting families for CDH1 mutational analysis.


Virchows Archiv | 2008

KRAS mutation testing for predicting response to anti-EGFR therapy for colorectal carcinoma: proposal for an European quality assurance program.

J.H.J.M. van Krieken; Andreas Jung; Thomas Kirchner; Fátima Carneiro; Raquel Seruca; Fred T. Bosman; P. Quirke; J.F. Fléjou; T. Plato Hansen; G. De Hertogh; Pedro Jares; Cord Langner; Gerald Hoefler; M.J.L. Ligtenberg; Dina Tiniakos; Sabine Tejpar; Generoso Bevilacqua; Arzu Ensari

Novel therapeutic agents targeting the epidermal growth factor receptor (EGFR) have improved outcomes for patients with colorectal carcinoma. However, these therapies are effective only in a subset of patients. Activating mutations in the KRAS gene are found in 30–40% of colorectal tumors and are associated with poor response to anti-EGFR therapies. Thus, KRAS mutation status can predict which patient may or may not benefit from anti-EGFR therapy. Although many diagnostic tools have been developed for KRAS mutation analysis, validated methods and standardized testing procedures are lacking. This poses a challenge for the optimal use of anti-EGFR therapies in the management of colorectal carcinoma. Here we review the molecular basis of EGFR-targeted therapies and the resistance to treatment conferred by KRAS mutations. We also present guideline recommendations and a proposal for a European quality assurance program to help ensure accuracy and proficiency in KRAS mutation testing across the European Union.


Genes, Chromosomes and Cancer | 1998

Determination of the replication error phenotype in human tumors without the requirement for matching normal DNA by analysis of mononucleotide repeat microsatellites

Xiao-Ping Zhou; Jean-Marc Hoang; You-Jun Li; Raquel Seruca; Fátima Carneiro; Manuel Sobrinho-Simões; Ragnhild A. Lothe; Catherine M. Gleeson; S. E. Hilary Russell; Françoise Muzeau; Jean-François Fléjou; Khê Hoang-Xuan; Rosette Lidereau; Gilles Thomas; Richard Hamelin

Microsatellite instability (MI) characterizing tumors with replication errors (RER+ tumors) was first described in colorectal tumors from hereditary non‐polyposis colorectal cancer (HNPCC) patients as well as in sporadic cases. It has also been observed in subgroups of extracolonic sporadic tumors, but there is no consensus as to the number of microsatellite loci to examine, and the threshold percentage of unstable loci required to classify a tumor as RER+. We have recently shown that BAT‐26, a mononucleotide repeat microsatellite, was quasi‐monomorphic in DNA from normal individuals and from colorectal RER− samples, and showed important size variations in RER+ samples. In the present work, we analyzed BAT‐26 allelic profiles in tumors of the breast (n = 107), brain (n = 78), stomach (n = 59), prostate (n = 49), esophagus (n = 36), thyroid (n = 31), endometrium (n = 12), and cervix (n = 10) whose RER status was already known, thus extending BAT‐26 analysis to a total of 542 human solid tumors. BAT‐26 alleles were quasi‐monomorphic in RER‐ samples (475/481) and shortened in RER+ tumors (57/61), including four tumors shown to have been misclassified on the basis of dinucleotide repeat microsatellite analysis. In 3/481 RER− and 4/61 RER+ cases, BAT‐26 size variation was important enough to attract attention, but not sufficient to establish the RER status of the corresponding tumors. In these cases, the analysis of BAT‐25 and BAT‐34C4, two other mononucleotide repeat microsatellites, was necessary to resolve the ambiguity. There were only 3 false positive cases. In conclusion, BAT‐26 was able to identify the RER status of 539 out of 542 tumors from various origins (99.5% efficiency) in a single‐step experiment without the requirement for matching normal DNA. Genes Chromosomes Cancer 21:101–107, 1998.


Oncogene | 2001

E-cadherin gene (CDH1) promoter methylation as the second hit in sporadic diffuse gastric carcinoma.

Jc Machado; Carla Oliveira; Ralph Carvalho; Paula Soares; Geert Berx; Carolina Caldas; Raquel Seruca; Fátima Carneiro; Manuel Sobrinho-Simões

In diffuse gastric carcinoma, despite common E-cadherin gene (CDH1) mutations, tumors show absence of CDH1 loss of heterozigosity (LOH) in most cases. This observation challenges the classical two-hit model of tumor suppressor gene inactivation. In order to investigate whether or not CDH1 promoter methylation may function as the second hit we analysed a series of 23 sporadic gastric carcinomas for the presence of CDH1 mutations, CDH1 promoter methylation, LOH and E-cadherin expression. CDH1 mutations were detected in nine of the 16 (56.3%) diffuse gastric carcinomas and in none of the seven intestinal gastric carcinomas. In diffuse gastric carcinomas harboring CDH1 mutations, LOH was observed in a single case. Loss of plasma membrane E-cadherin expression was consistently found in all nine cases with CDH1 mutation, suggesting that tumors inactivated the remaining CDH1 allele via a different mechanism. CDH1 promoter methylation was observed in nine of the 16 (56.3%) diffuse-type gastric carcinoma cases, including six of the nine cases (66.7%) harboring CDH1 mutations. CDH1 promoter methylation was also seen in two (28.6%) intestinal-type cases. Our results show that CDH1 promoter methylation is the second hit in more than half of the sporadic diffuse gastric carcinoma cases harboring CDH1 mutations.


The Journal of Pathology | 2004

Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening

Fátima Carneiro; David Huntsman; Thomas C. Smyrk; David A. Owen; Raquel Seruca; Paul Pharoah; Carlos Caldas; Manuel Sobrinho-Simões

Hereditary diffuse gastric cancer (HDGC) is a familial cancer syndrome caused, in 30–40% of cases, by germline mutations of the E‐cadherin/CDH1 gene. The presence of clinically undetectable early gastric cancers has been previously reported in ten of ten prophylactic gastrectomies from germline E‐cadherin mutation carriers. In the present study, detailed maps of the distribution of invasive cancers in nine of these ten stomachs were produced and precursor lesions of HDGC searched for. The nine gastrectomy specimens contained from 1 to 161 foci of early diffuse gastric cancer, occupying 0.005–2.96% of the gastric mucosa. Seven specimens contained focal in situ signet ring carcinoma. Pagetoid spread of signet ring cells was observed beneath the epithelial lining of gastric foveolae/glands. Helicobacter pylori organisms and associated pathology were absent from all cases. Two‐dimensional maps of the gastrectomy specimens revealed lesions throughout the gastric mucosa without evidence of antral clustering. The distribution and size of the cancers in the gastrectomy specimens indicate that standard endoscopic screening with random or geographically targeted biopsies is unlikely to provide sufficiently sensitive clinical screening for at‐risk individuals. An in situ precursor of signet ring carcinoma was identified and a model for neoplastic progression in the setting of HDGC is proposed. Copyright

Collaboration


Dive into the Raquel Seruca's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge