Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Clin Obstet Gynecol ; 63(1): 64-73, 2020 03.
Article in English | MEDLINE | ID: mdl-31764001

ABSTRACT

Eleven genes have been identified that increase the lifetime risk of developing ovarian cancer. The cumulative cancer risk of ovarian cancer varies with the mutation type and age. Ovarian cancer risk management options include surgical risk reduction with salpingo-oophorectomy and a newer step-wise approach with interval salpingectomy and delayed oophorectomy. Women should be counseled on the pros and cons of hysterectomy in the setting of reducing the risk of other cancers; eliminating the risk of endometrial cancer in Lynch Syndrome, potential risk of serous/serous-like endometrial cancer in BRCA1 carriers, and elimination of progestogen therapy that may increase breast cancer risk.


Subject(s)
Ovarian Neoplasms/prevention & control , Prophylactic Surgical Procedures/methods , Adult , Aged , Breast Neoplasms/genetics , Breast Neoplasms/prevention & control , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/prevention & control , Decision Making , Endometrial Neoplasms/genetics , Endometrial Neoplasms/prevention & control , Female , Genes, BRCA1 , Genes, BRCA2 , Humans , Lynch Syndrome II/complications , Lynch Syndrome II/genetics , Lynch Syndrome II/surgery , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Risk Assessment , Salpingo-oophorectomy
2.
Cancer ; 125(18): 3172-3183, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31150123

ABSTRACT

BACKGROUND: Universal tumor testing for defective DNA mismatch repair (MMR) is recommended for all women diagnosed with endometrial cancer to identify those with underlying Lynch syndrome. However, the effectiveness of these screening methods in identifying individuals with Lynch syndrome across the population has not been well studied. The aim of this study was to evaluate outcomes of MMR immunohistochemistry (IHC), mutL homolog 1 (MLH1) methylation, and microsatellite instability (MSI) analysis among patients with endometrial cancer. METHODS: A complete systematic search of online databases (PubMed, EMBASE, MEDLINE, and the Cochrane Library) for 1990-2018 was performed. A DerSimonian-Laird random effects model meta-analysis was used to estimate the weighted prevalence of Lynch syndrome diagnoses. RESULTS: The comprehensive search produced 4400 publications. Twenty-nine peer-reviewed studies met the inclusion criteria. Patients with endometrial cancer (n = 6649) were identified, and 206 (3%) were confirmed to have Lynch syndrome through germline genetic testing after positive universal tumor molecular screening. Among 5917 patients who underwent tumor IHC, 28% had abnormal staining. Among 3140 patients who underwent MSI analysis, 31% had MSI. Among patients with endometrial cancer, the weighted prevalence of Lynch syndrome germline mutations was 15% (95% confidence interval [CI], 11%-18%) with deficient IHC staining and 19% (95% CI, 13%-26%) with a positive MSI analysis. Among 1159 patients who exhibited a loss of MLH1 staining, 143 (13.7%) were found to be MLH1 methylation-negative among those who underwent methylation testing, and 32 demonstrated a germline MLH1 mutation (2.8% of all absent MLH1 staining cases and 22.4% of all MLH1 methylation-negative cases). Forty-three percent of patients with endometrial cancer who were diagnosed with Lynch syndrome via tumor typing would have been missed by family history-based screening alone. CONCLUSIONS: Despite the widespread implementation of universal tumor testing in endometrial cancer, data regarding testing results remain limited. This study provides predictive values that will help practitioners to evaluate abnormal results in the context of Lynch syndrome and aid them in patient counseling.


Subject(s)
Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , Lynch Syndrome II/diagnosis , Neoplasms, Cystic, Mucinous, and Serous/genetics , Carcinoma, Endometrioid/etiology , Carcinoma, Endometrioid/metabolism , DNA Methylation/genetics , DNA Mismatch Repair , DNA-Binding Proteins/genetics , Endometrial Neoplasms/etiology , Endometrial Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Lynch Syndrome II/complications , Lynch Syndrome II/genetics , Microsatellite Instability , Middle Aged , Mismatch Repair Endonuclease PMS2/genetics , Molecular Diagnostic Techniques , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/genetics , Neoplasms, Cystic, Mucinous, and Serous/etiology , Neoplasms, Cystic, Mucinous, and Serous/metabolism
3.
BMC Cancer ; 18(1): 576, 2018 May 21.
Article in English | MEDLINE | ID: mdl-29783979

ABSTRACT

BACKGROUND: Lynch syndrome is an autosomal dominant inherited disease caused by germline mutations in mismatch repair genes. Analysis for microsatellite instability (MSI) and immunohistochemistry (IHC) of protein expressions of disease-associated genes is used to screen for Lynch syndrome in endometrial cancer patients. When losses of both MLH1 and PMS2 proteins are observed by IHC, MLH1 promoter methylation analysis is conducted to distinguish Lynch syndrome-associated endometrial cancer from sporadic cancer. CASE PRESENTATION: Here we report a woman who developed endometrial cancer at the age of 49 years. She had a family history of colorectal cancer (first-degree relative aged 52 years) and stomach cancer (second-degree relative with the age of onset unknown). No other family history was present, and she failed to meet the Amsterdam II criteria for the diagnosis of Lynch syndrome. Losses of MLH1 and PMS2, but not MSH2 and MSH6, proteins were observed by IHC in endometrial cancer tissues. Because MLH1 promoter hypermethylation was detected in endometrial cancer tissue samples, the epigenetic silencing of MLH1 was suspected as the cause of the protein loss. However, because of the early onset of endometrial cancer and the positive family history, a diagnosis of Lynch syndrome was also suspected. Therefore, we provided her with genetic counseling. After obtaining her consent, MLH1 promoter methylation testing and genetic testing of peripheral blood were performed. MLH1 promoter methylation was not observed in peripheral blood. However, genetic testing revealed a large deletion of exon 5 in MLH1; thus, we diagnosed the presence of Lynch syndrome. CONCLUSIONS: Both MLH1 germline mutation and MLH1 promoter hypermethylation may be observed in endometrial cancer. Therefore, even if MLH1 promoter hypermethylation is detected, a diagnosis of Lynch syndrome cannot be excluded.


Subject(s)
DNA Methylation/genetics , Endometrial Neoplasms/genetics , Lynch Syndrome II/genetics , MutL Protein Homolog 1/genetics , Promoter Regions, Genetic/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Endometrium/pathology , Female , Genetic Counseling , Germ-Line Mutation , Humans , Hysterectomy , Lynch Syndrome II/diagnosis , Microsatellite Instability , Middle Aged , Salpingo-oophorectomy
4.
Clin Med (Lond) ; 17(6): 568-572, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29196360

ABSTRACT

Advances in sequencing technology have led to the introduction of panel testing in hereditary breast and ovarian cancer. While direct-to-consumer testing services have become widely available, the clinical validity of many of the genes on panel tests remains contentious and risk management guidelines are often lacking. This article gives an overview of advantages with panel testing as well as important challenges, including clinical translation of test results.


Subject(s)
Genetic Testing/methods , Hereditary Breast and Ovarian Cancer Syndrome/genetics , AMP-Activated Protein Kinase Kinases , Antigens, CD , Cadherins/genetics , DNA-Binding Proteins/genetics , Epithelial Cell Adhesion Molecule/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Predisposition to Disease , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , High-Throughput Nucleotide Sequencing , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Lynch Syndrome II/diagnosis , Lynch Syndrome II/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , PTEN Phosphohydrolase/genetics , Peutz-Jeghers Syndrome/diagnosis , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
5.
Gynecol Oncol ; 146(2): 217-224, 2017 08.
Article in English | MEDLINE | ID: mdl-28596016

ABSTRACT

OBJECTIVE: To assess current practice, advise minimum standards, and identify educational gaps relevant to genetic screening, counseling, and testing of women affected by gynecologic cancers. METHODS: The Society of Gynecologic Oncology (SGO) organized a multidisciplinary summit that included representatives from the American College of Obstetricians and Gynecologists (ACOG), the American Society Clinical Oncology (ASCO), the National Society of Genetic Counselors (NSGC), and patient advocacy groups, BrightPink and Facing our Risk of Cancer Empowered (FORCE). Three subject areas were discussed: care delivery models for genetic testing, barriers to genetic testing, and educational opportunities for providers of genetic testing. RESULTS: The group endorsed current SGO, National Comprehensive Cancer Network (NCCN), and NSGC genetic testing guidelines for women affected with ovarian, tubal, peritoneal cancers, or DNA mismatch repair deficient endometrial cancer. Three main areas of unmet need were identified: timely and universal genetic testing for women with ovarian, fallopian tube, and peritoneal cancers; education regarding minimum standards for genetic counseling and testing; and barriers to implementation of testing of both affected individuals as well as cascade testing of family members. Consensus building among all stakeholders resulted in an action plan to address gaps in education of gynecologic oncology providers and delivery of cancer genetics care.


Subject(s)
Genetic Services , Genital Neoplasms, Female/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Lynch Syndrome II/genetics , Congresses as Topic , Consensus Development Conferences as Topic , Female , Genetic Counseling/methods , Genetic Testing/methods , Gynecology , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Humans , Lynch Syndrome II/diagnosis , Patient Selection , Practice Guidelines as Topic , Societies, Medical , Surgical Oncology
6.
Am J Obstet Gynecol ; 217(5): 512-521, 2017 11.
Article in English | MEDLINE | ID: mdl-28411145

ABSTRACT

The world of hereditary cancers has seen exponential growth in recent years. While hereditary breast and ovarian cancer and Lynch syndrome account for the majority of mutations encountered by gynecologists, newly identified deleterious genetic mutations continue to be unearthed with their associated risks of malignancies. However, these advances in genetic cancer predispositions then force practitioners and their patients to confront the uncertainties of these less commonly identified mutations and the fact that there is limited evidence to guide them in expected cancer risk and appropriate risk-reduction strategies. Given the speed of information, it is imperative to involve cancer genetics experts when counseling these patients. In addition, coordination of screening and care in conjunction with specialty high-risk clinics, if available, allows for patients to have centralized management for multiple cancer risks under the guidance of physicians with experience counseling these patients. The objective of this review is to present the current literature regarding genetic mutations associated with gynecologic malignancies as well to propose screening and risk-reduction options for these high-risk patients.


Subject(s)
Genital Neoplasms, Female/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , AMP-Activated Protein Kinase Kinases , DNA Mismatch Repair/genetics , DNA Polymerase III/genetics , DNA-Binding Proteins/genetics , Epithelial Cell Adhesion Molecule/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Predisposition to Disease , Genetic Testing , Genital Neoplasms, Female/genetics , Hamartoma Syndrome, Multiple/diagnosis , Hamartoma Syndrome, Multiple/genetics , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Lynch Syndrome II/diagnosis , Lynch Syndrome II/genetics , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Neoplastic Syndromes, Hereditary/genetics , PTEN Phosphohydrolase/genetics , Peutz-Jeghers Syndrome/diagnosis , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , RNA Helicases/genetics , Tumor Suppressor Protein p53/genetics
7.
Cancer ; 123(10): 1721-1730, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28085182

ABSTRACT

BACKGROUND: As panel testing becomes more common in clinical practice, it is important to understand the prevalence and trends associated with the pathogenic variants (PVs) identified. This is especially true for genetically heterogeneous cancers, such as breast cancer (BC), in which PVs in different genes may be associated with various risks and cancer subtypes. The authors evaluated the outcomes of genetic testing among women who had a personal history of BC. METHODS: A total of 35,409 women with a single diagnosis of BC who underwent clinical genetic testing with a 25-gene panel were included in the current analysis. Women with multiple BCs and men with BC were excluded. The frequency and distribution of PVs were assessed for the overall cohort, among women with triple-negative BC (TNBC) (n = 4797), and by age at diagnosis. RESULTS: PVs were identified in 9.3% of women tested; 51.5% of PVs were identified in genes other than breast cancer 1 (BRCA1) and BRCA2, including checkpoint kinase 2 (CHEK2) (11.7%), ataxia telangiectasia mutated (ATM; ATM serine/threonine kinase) (9.7%), and partner and localizer of BRCA2 (PALB2) (9.3%). The prevalence of PVs in BRCA1, PALB2, BRCA1-associated RING domain 1 (BARD1), BRCA1-interacting protein C-terminal helicase 1 (BRIP1), and RAD51 paralog C (RAD51C) was statistically higher among women with TNBC. The PV rate was higher among women aged <40 years, lower among women aged >59 years, and relatively constant (8.5%-9.0%) among women who were diagnosed between ages 40 and 59 years. CONCLUSIONS: These results demonstrate that panel testing increased the number of women identified as carrying a PV in this cohort compared with BRCA testing alone. Furthermore, the proportion of women identified who carried a PV in this cohort did not decrease between ages 40 and 59 years. Cancer 2017;123:1721-1730. © 2017 American Cancer Society.


Subject(s)
Breast Neoplasms/genetics , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Lynch Syndrome II/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins/genetics , Checkpoint Kinase 2/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group N Protein , Fanconi Anemia Complementation Group Proteins , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Testing , Humans , Middle Aged , Neoplastic Syndromes, Hereditary/genetics , Nuclear Proteins/genetics , RNA Helicases/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Young Adult
8.
Int J Surg Pathol ; 25(3): 253-257, 2017 May.
Article in English | MEDLINE | ID: mdl-27903930

ABSTRACT

Lynch syndrome is a hereditary disease with germline mutation in a DNA mismatch repair gene, most often presenting with colorectal and/or endometrial carcinomas; however, the spectrum of Lynch syndrome-associated tumors is expanding. In this article, we report a case of a primary peritoneal epithelioid mesothelioma that developed in a Lynch syndrome patient 10 months after diagnosis of uterine endometrioid adenocarcinoma. To our knowledge, this is the first reported case of a Lynch syndrome patient with metachronous uterine endometrioid adenocarcinoma and primary peritoneal mesothelioma.


Subject(s)
Carcinoma, Endometrioid/pathology , Lynch Syndrome II/pathology , Neoplasms, Mesothelial/pathology , Peritoneal Neoplasms/pathology , Uterine Neoplasms/pathology , Biomarkers, Tumor/analysis , Carcinoma, Endometrioid/etiology , DNA-Binding Proteins/genetics , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Lynch Syndrome II/complications , Lynch Syndrome II/genetics , Middle Aged , Neoplasms, Mesothelial/etiology , Peritoneal Neoplasms/etiology , Uterine Neoplasms/etiology
9.
Gynecol Oncol ; 143(3): 453-459, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27789085

ABSTRACT

PURPOSE: To assess cost-effectiveness of routine screening for Lynch Syndrome (LS) in endometrial cancer (EC) patients ≤70years of age. METHODS: Consecutive EC patients ≤70years of age were screened for LS by analysis of microsatellite instability, immunohistochemistry and MLH1 hypermethylation. Costs and health benefit in life years gained (LYG) included surveillance for LS carriers among EC patients and relatives. We calculated incremental cost-effectiveness ratios (ICERs) comparing LS screening among EC patients ≤70years with ≤50years and the revised Bethesda guidelines. RESULTS: Screening for LS in 179 EC patients identified 7 LS carriers; 1 was ≤50 and 6 were 51-70years. Per age category 18 and 9 relatives were identified as LS carrier. Screening resulted in 74,7 LYG (45,4 and 29,3 LYG per age category). The ICER for LS screening in EC patients ≤70 compared with ≤50years was €5,252/LYG. The revised Bethesda guidelines missed 4/7 (57%) LS carriers among EC patients. The ICER for LS screening in EC patients ≤70years of age compared with the revised Bethesda guidelines was €6,668/LYG. Both ICERs remained <€16,000/LYG in sensitivity analyses. CONCLUSION: Routine LS screening in EC patients ≤70years is a cost-effective strategy, allowing colorectal cancer prevention in EC patients and their relatives.


Subject(s)
DNA Methylation , DNA Mismatch Repair/genetics , Endometrial Neoplasms/genetics , Genetic Testing/economics , Lynch Syndrome II/diagnosis , Microsatellite Instability , Adult , Age Factors , Aged , Colorectal Neoplasms/diagnosis , Cost-Benefit Analysis , DNA Mutational Analysis , Early Detection of Cancer , Family , Female , Genetic Counseling/economics , Humans , Immunohistochemistry , Lynch Syndrome II/genetics , Mass Screening , Middle Aged , MutL Protein Homolog 1/genetics
10.
Medicina (B Aires) ; 76(3): 180-2, 2016.
Article in Spanish | MEDLINE | ID: mdl-27295708

ABSTRACT

Lynch syndrome is the most frequent syndrome in hereditary colorectal cancer, a family-specific deleterious mutations in genes encoding DNA reparation proteins: MLH1 (mutL homolog 1), MSH2, MSH6 (mutS homolog 2 y 6, respectively), PMS2 (PMS1 homolog 2, mismatch repair system component) y MUTYH (mutY DNA glycosylase). The c.2252_2253delAA, p.Lys751Serfs*3 mutation in MLH1 gene segregates with a haplotype reported in the northern region of Italy and whose origin was attributed to a founder effect. This mutation co-segregates with typical characteristics of Lynch syndrome, including early age at onset and multiple primary tumors in the same individual, a high frequency of pancreatic cancer, high microsatellite instability and lack of PMS2 expression. This report describes a mutation in an Argentinian patient with endometrioid adenocarcinoma of uterus. Her first-degree relatives had a history of colon cancer diagnosed before 50 years, fulfilling the Amsterdam Criteria I and Lynch syndrome II. The high pathogenicity associated to this mutation makes necessary the study of all members from families with hereditary cancer, allowing pre-symptomatic genetic diagnosis, early assessment and the instauration of preventive treatments.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Founder Effect , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , Mutation/genetics , DNA Repair/genetics , Female , Humans , Lynch Syndrome II/genetics , Middle Aged , Pedigree
11.
Surg Pathol Clin ; 9(2): 201-14, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27241104

ABSTRACT

Lynch syndrome is responsible for approximately 5% of endometrial cancers and 1% of ovarian cancers. The molecular basis for Lynch syndrome is a heritable functional deficiency in the DNA mismatch repair system, typically due to a germline mutation. This review discusses the rationales and relative merits of current Lynch syndrome screening tests for endometrial and ovarian cancers and provides pathologists with an informed algorithmic approach to Lynch syndrome testing in gynecologic cancers. Pitfalls in test interpretation and strategies to resolve discordant test results are presented. The potential role for next-generation sequencing panels in future screening efforts is discussed.


Subject(s)
Early Detection of Cancer/methods , Genital Neoplasms, Female/diagnosis , Lynch Syndrome II/diagnosis , DNA Methylation/genetics , DNA Mismatch Repair/genetics , DNA Mutational Analysis/methods , DNA, Neoplasm/genetics , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Germ-Line Mutation , Humans , Lynch Syndrome II/genetics , Lynch Syndrome II/pathology , Microsatellite Instability , MutL Protein Homolog 1/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis
12.
Surg Pathol Clin ; 9(2): 289-99, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27241109

ABSTRACT

This article reviews the main tissue testing modalities for Lynch Syndrome in the pathology laboratory, such as immunohistochemistry and PCR based analyses, and discusses their routine application, interpretation pitfalls, and troubleshooting of common technical performance issues. Discrepancies between laboratory and genetic testing may arise, and are examined in the context of the complexity of molecular abnormalities associated with Lynch Syndrome. The merits of targeted versus universal screening in a changing healthcare climate are addressed. In the absence of comprehensive screening programs, specific tumor topography and histological features that may prompt pathologist-initiated molecular tumor testing are outlined.


Subject(s)
Endometrial Neoplasms/diagnosis , Lynch Syndrome II/diagnosis , Biomarkers, Tumor/analysis , DNA Mismatch Repair , Early Detection of Cancer/methods , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Genetic Testing/methods , Humans , Lynch Syndrome II/genetics , Lynch Syndrome II/pathology , Microsatellite Instability , MutL Protein Homolog 1/genetics , Mutation
13.
Surg Pathol Clin ; 9(2): 307-28, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27241111

ABSTRACT

Hereditary breast ovarian cancer and Lynch/hereditary nonpolyposis colorectal cancer syndrome account for most hereditary gynecologic cancers. In the absence of effective cancer screening and other preventative strategies, risk-reducing surgery in women who are known to be at genetic risk of BRCA-associated or of Lynch syndrome carcinomas is effective in significantly decreasing the lifetime risk of developing malignancy. Reflex genomic testing of high-grade ovarian cancers and reflex immunohistochemistry in endometrial cancers will lead to greater recognition of germline-associated cancers. Approaches to processing surgical specimens, the recognition and classification of cancer precursor lesions, and differentiation from their mimics are discussed.


Subject(s)
Hereditary Breast and Ovarian Cancer Syndrome/prevention & control , Lynch Syndrome II/prevention & control , BRCA2 Protein/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/prevention & control , Female , Genetic Predisposition to Disease , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Hereditary Breast and Ovarian Cancer Syndrome/pathology , Humans , Hysterectomy , Lynch Syndrome II/diagnosis , Lynch Syndrome II/genetics , Lynch Syndrome II/pathology , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/prevention & control , Ovariectomy , Precancerous Conditions/diagnosis , Precancerous Conditions/surgery , Salpingectomy/methods , Ubiquitin-Protein Ligases/genetics
14.
Medicina (B.Aires) ; 76(3): 180-182, June 2016. ilus, tab
Article in Spanish | LILACS | ID: biblio-841567

ABSTRACT

El síndrome de Lynch es la más frecuente de las neoplasias colorrectales hereditarias. Se origina por mutaciones germinales deletéreas familia-específicas en los genes que codifican proteínas de reparación del ADN: MLH1 (homólogo humano de mutL), MSH2 y MSH6 (homólogo humano de mutS 2 y 6, respectivamente), PMS2 (homólogo humano de PMS1 2) y MUTYH (homólogo humano de la ADN-glycosilasa mutY). La mutación c.2252_2253delAA, p.Lys751Serfs*3 en el exón 19 del gen MLH1 segrega con un haplotipo descripto en la región norte de Italia y cuyo origen fue atribuido a un efecto fundador. Esta mutación co-segrega con características típicas del síndrome de Lynch, incluyendo afectación temprana y múltiples tumores primarios en el mismo individuo, una alta frecuencia de cáncer pancreático, elevada inestabilidad microsatelital y falta de expresión de PMS2. En el presente trabajo se comunica dicha mutación en una paciente argentina con adenocarcinoma endometroide de útero en cuya historia familiar existen antecedentes de cáncer de colon diagnosticado antes de los 50 años en familiares de primer grado, reuniendo los criterios de Ámsterdam I y síndrome de Lynch II. Los polimorfismos presentes en la paciente coinciden con el haplotipo descripto en una región del norte de Italia. El alto grado de patogenicidad asociada a esta mutación hace imprescindible el estudio de todos los integrantes de las familias con cáncer hereditario permitiendo el diagnóstico genético pre-sintomático, la instauración de tratamientos o conductas preventivas y su seguimiento.


Lynch syndrome is the most frequent syndrome in hereditary colorectal cancer, a family-specific deleterious mutations in genes encoding DNA reparation proteins: MLH1 (mutL homolog 1), MSH2, MSH6 (mutS homolog 2 y 6, respectively), PMS2 (PMS1 homolog 2, mismatch repair system component) y MUTYH (mutY DNA glycosylase).The c.2252_2253delAA, p.Lys751Serfs*3 mutation in MLH1 gene segregates with a haplotype reported in the northern region of Italy and whose origin was attributed to a founder effect. This mutation co-segregates with typical characteristics of Lynch syndrome, including early age at onset and multiple primary tumors in the same individual, a high frequency of pancreatic cancer, high microsatellite instability and lack of PMS2 expression. This report describes a mutation in an Argentinian patient with endometrioid adenocarcinoma of uterus. Her first-degree relatives had a history of colon cancer diagnosed before 50 years, fulfilling the Amsterdam Criteria I and Lynch syndrome II. The high pathogenicity associated to this mutation makes necessary the study of all members from families with hereditary cancer, allowing pre-symptomatic genetic diagnosis, early assessment and the instauration of preventive treatments.


Subject(s)
Humans , Female , Middle Aged , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Founder Effect , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , Mutation/genetics , Pedigree , DNA Repair/genetics , Lynch Syndrome II/genetics
15.
Duodecim ; 132(3): 233-40, 2016.
Article in Finnish | MEDLINE | ID: mdl-26951027

ABSTRACT

Lynch syndrome (LS) refers to an autosomal dominant genetic predisposition to develop colon cancer or cancers or the uterine corpus, stomach, urinary tract, ovaries, small intestine, mammary gland or bile ducts at a young age. The predisposition to cancer is caused by a germline mutation in one of the genes of the mismatch repair (MMR) system. International recommendations suggest immunohistochemical analysis of tumor tissue from at least those having developed colorectal cancer or endometrial cancer at an age of less than 70 years. This would allow the selection of patients to be referred for gene testing as well as identification of mutation carriers, for whom a regular colonoscopy follow-up is arranged at an interval of 2 to 3 years.


Subject(s)
Lynch Syndrome II/diagnosis , Lynch Syndrome II/therapy , Colonoscopy , Diagnosis, Differential , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Immunohistochemistry , Lynch Syndrome II/genetics
16.
J Pathol ; 234(4): 548-59, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25111426

ABSTRACT

Lynch syndrome (LS) is caused by germline mutations in mismatch repair (MMR) genes, resulting in microsatellite-unstable tumours. Approximately 35% of suspected LS (sLS) patients test negative for germline MMR gene mutations, hampering conclusive LS diagnosis. The aim of this study was to investigate somatic MMR gene aberrations in microsatellite-unstable colorectal and endometrial cancers of sLS patients negative for germline MMR gene mutations. Suspected LS cases were selected from a retrospective Clinical Genetics Department diagnostic cohort and from a prospective multicentre population-based study on LS in The Netherlands. In total, microsatellite-unstable tumours of 40 sLS patients (male/female 20/20, median age 57 years) were screened for somatic MMR gene mutations by next-generation sequencing. In addition, loss of heterozygosity (LOH) of the affected MMR genes in these tumours as well as in 68 LS-associated tumours and 27 microsatellite-unstable tumours with MLH1 promoter hypermethylation was studied. Of the sLS cases, 5/40 (13%) tumours had two pathogenic somatic mutations and 16/40 (40%) tumours had a (likely) pathogenic mutation and LOH. Overall, LOH of the affected MMR gene locus was observed in 24/39 (62%) tumours with informative LOH markers. Of the LS cases and the tumours with MLH1 promoter hypermethylation, 39/61 (64%) and 2/21 (10%) tumours, respectively, demonstrated LOH. Half of microsatellite-unstable tumours of sLS patients without germline MMR gene mutations had two (likely) deleterious somatic MMR gene aberrations, indicating their sporadic origin. Therefore, we advocate adding somatic mutation and LOH analysis of the MMR genes to the molecular diagnostic workflow of LS.


Subject(s)
DNA Mismatch Repair/genetics , Lynch Syndrome II/diagnosis , Lynch Syndrome II/genetics , Adult , Aged , DNA Mutational Analysis , Female , Humans , Loss of Heterozygosity , Male , Microsatellite Instability , Middle Aged , Multiplex Polymerase Chain Reaction , Mutation
17.
Prog Urol ; 24(15): 943-53, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25158326

ABSTRACT

OBJECTIVE: To describe natural history and carcinogenesis of upper tract urothelial carcinoma (UTUC). METHODS: A systematic review of the scientific literature was performed in the Medline database (Pubmed) using different associations of the following keywords: upper tract urothelial carcinoma; clonality; carcinogenesis; mutation; chromosomal instability; Lynch syndrome; genetic polymorphism. RESULTS: Local development of UTUC is characterized by a highly prevalent multifocality that might be explained by the overlap of "field change" and "intraluminal seeding and implantation" theories. UTUC and bladder tumors share common carcinogenesis mechanisms such as mutations of FGFR3 and TP53 defining two distinct pathways of pathogenesis. Epigenetic alterations corresponding to the hypermethylation of different promoters regulating genes expression and chromosomal instability such as chromosome 9 deletions are also involved in UTUC carcinogenesis. Furthermore, specific genetic risk factors fro UTUC including Lynch syndrome and different polymorphisms might explain an individual susceptibility for developing these tumors. CONCLUSIONS: Significant advances have been done in the field of basic research in UTUCs in recent years and have been of particular interest to provide better descriptions of their natural history. Despite these important findings however, some carcinogenic mechanisms remains not elucidated and unknown in the field of UTUC so far.


Subject(s)
Carcinogenesis , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Urologic Neoplasms/genetics , Urologic Neoplasms/pathology , Urothelium/pathology , Chromosomal Instability , Decision Trees , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Humans , Lynch Syndrome II/genetics , Neoplasm Metastasis , Neoplasm Seeding , Oncogenes/genetics , Polymorphism, Genetic
18.
Int J Oncol ; 45(1): 77-81, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24788313

ABSTRACT

Somatic mutations in the POLE gene encoding the catalytic subunit of DNA polymerase ε have been found in sporadic colorectal cancers (CRCs) and are most likely of importance in tumour development and/or progression. Recently, families with dominantly inherited colorectal adenomas and colorectal cancer were shown to have a causative heterozygous germline mutation in the proofreading exonuclease domain of POLE. The highly penetrant mutation was associated with predisposition to CRC only and no extra-colonic tumours were observed. We have identified a mutation in a large family in which the carriers not only developed CRC, they also demonstrate a highly penetrant predisposition to extra-intestinal tumours such as ovarian, endometrial and brain tumours. The mutation, NM_006231.2:c.1089C>A, p.Asn363Lys, also located in the proofreading exonuclease domain is directly involved in DNA binding. Theoretical prediction of the amino acid substitution suggests a profound effect of the substrate binding capability and a more severe impairment of the catalytic activity compared to the previously reported germline mutation. A possible genotype to phenotype correlation for deleterious mutations in POLE might exist that needs to be considered in the follow-up of mutation carriers.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , Lynch Syndrome II/genetics , Adenoma/genetics , Adenoma/pathology , Aged , Amino Acid Substitution , Catalytic Domain , Exodeoxyribonucleases/genetics , Female , Genetic Predisposition to Disease , Humans , Lynch Syndrome II/pathology , Male , Penetrance , Phenotype , Point Mutation
20.
Am J Surg Pathol ; 37(4): 579-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23426126

ABSTRACT

Women with Lynch syndrome (LS) are at increased risk for endometrial (EC) and ovarian carcinoma (OC). Current surveillance recommendations for detection of EC and OC in LS patients are not effective. Small studies have shown that prophylactic hysterectomy and bilateral salpingo-oophorectomy (P-TH-BSO) are the most effective and least expensive preventive measures in these patients. Data regarding histologic findings in prophylactic specimens in these patients are lacking. All LS patients who underwent P-TH-BSO at the Memorial Sloan-Kettering Cancer Center from 2000 to 2011 were identified. Slides were evaluated for the presence of endometrial hyperplasia (EH), EC, OC, or any other recurrent histologic findings. Twenty-five patients were identified, with an age range of 36 to 61 years. Fifteen patients had a synchronous or prior colorectal carcinoma, and 2 patients had a history of sebaceous carcinoma. Focal FIGO grade 1 endometrioid ECs were detected in 2 patients; 1 was 54 years of age (MSH2 mutation; superficially invasive), and the other was 56 years of age (MLH1 mutation; noninvasive). Focal complex atypical hyperplasia, unassociated with carcinoma, was seen in 3 patients, ages 35 and 45 (MLH1 mutations) and 53 years (MSH2 mutation). One patient (44 y, with MSH2 mutation) was found to have a mixed endometrioid/clear cell OC and simple EH without atypia. The OC was adherent to the colon but did not show distant metastasis. In our study, P-TH-BSOs performed because of the presence of LS revealed incidental EC and/or EH in 24% of cases and OC in 4%. The ECs were low grade, confined to the endometrium, and seen in patients older than 50 years. Prophylactic hysterectomy allows detection of early lesions in LS; these lesions appear to be small and focal. This small series of prophylactic hysterectomies may provide some clues about LS-associated endometrial carcinogenesis.


Subject(s)
Hysterectomy , Lynch Syndrome II/pathology , Ovarian Neoplasms/prevention & control , Ovariectomy , Uterine Neoplasms/prevention & control , Adaptor Proteins, Signal Transducing/genetics , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Family Health , Female , Humans , Immunohistochemistry , Lynch Syndrome II/genetics , Lynch Syndrome II/metabolism , Lynch Syndrome II/surgery , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , Mutation , Neoplasms, Multiple Primary , Nuclear Proteins/genetics , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...