Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.108
Filtrer
1.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 702-708, 2024 Jul 08.
Article de Chinois | MEDLINE | ID: mdl-38955702

RÉSUMÉ

Objective: To investigate the clinicopathological characteristics and prognostic factors of sporadic mismatch repair deficient (dMMR) colorectal cancer. Methods: A total of 120 cases of sporadic dMMR colorectal cancer from July 2015 to April 2021 were retrospectively collected in Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. Patients with Lynch syndrome; synchronous multiple colorectal cancers; preoperative anti-tumor treatments such as chemotherapy and radiotherapy; and those with incomplete follow-up information were excluded based on family history and next-generation sequencing (NGS) test results. Immunohistochemical stains were used to detect the expression of mismatch repair proteins, methylation-specific PCR for methylation testing, and fluorescent PCR for BRAF V600E gene mutation detection. The clinical and pathological data, and gene mutation status were analyzed. Follow-up was done to assess survival and prognosis including progression-free survival and overall survival rate. Results: Sporadic dMMR colorectal cancer occurred more frequently in the right side of the colon, in females, and in the elderly. Morphologically, it was mostly moderately-differentiated, and most patients had low-grade tumor budding. In terms of immunohistochemical expression, MLH1 and PMS2 loss were dominant, and there were age and location-specificities in protein expression. MLH1 methylation was commonly detected in elderly female patients and rare in young male patients; while MLH1 and PMS2 deficiency, and BRAF V600E mutation occurred more often on the right side (P<0.05). The 3-year and 5-year progression-free survival rates were 90.7% and 88.7% respectively, and the 3-year and 5-year overall survival rates were 92.8% and 90.7% respectively. Tumor budding status was an independent risk factor affecting patient recurrence (hazard ratio=3.375, 95% confidence interval: 1.060-10.741, P=0.039), patients with low-grade tumor budding had better prognosis, and those with medium or high-grade tumor budding had poor prognosis. Conclusion: For dMMR colorectal cancer patients, tumor budding status is an independent risk factor for recurrence.


Sujet(s)
Tumeurs colorectales , Réparation de mésappariement de l'ADN , Protéines proto-oncogènes B-raf , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Mâle , Femelle , Pronostic , Études rétrospectives , Protéines proto-oncogènes B-raf/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Mutation , Taux de survie , Adulte d'âge moyen , Sujet âgé , Méthylation de l'ADN , Adulte , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/métabolisme
2.
Prostate ; 84(10): 945-953, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38629217

RÉSUMÉ

BACKGROUND: Pathogenic germline variants in the mismatch repair (MMR) genes are associated with an increased risk of prostate cancer (PCa). Since 2010 we have recommended MMR carriers annual PSA testing from the age of 40. Prospective studies of the outcome of long-term PSA screening are lacking. This study aimed to investigate the incidence and characteristics of PCa in Norwegian MMR carriers attending annual PSA screening (PSA threshold >3.0 ng/mL) to evaluate whether our recommendations should be continued. METHODS: This is a prospective observational study of 225 male MMR carriers who were recommended annual PSA screening by the Section of Inherited Cancer, Oslo University Hospital from 2010 and onwards. Incidence and tumor characteristics (age, PSA at diagnosis, Gleason score, TNM score) were described. IHC and MSI-analyses were done on available tumors. Standardized incidence ratio (SIR) was calculated based on data from the Cancer Registry of Norway. RESULTS: Twenty-two of 225 (9.8%) had been diagnosed with PCa, including 10/69 (14.5%) MSH2 carriers and 8/61 (13.1%) MSH6 carriers. Ten of 20 (50%) tumors had Gleason score ≥4 + 3 on biopsy and 6/11 (54.5%) had a pathological T3a/b stage. Eight of 17 (47.1%) tumors showed abnormal staining on IHC and 3/13 (23.1%) were MSI-high. SIR was 9.54 (95% CI 5.98-14.45) for all MMR genes, 13.0 (95% CI 6.23-23.9) for MSH2 and 13.74 for MSH6 (95% CI 5.93-27.08). CONCLUSIONS: Our results indicate that the MMR genes, and especially MSH2 and MSH6, are associated with a significant risk of PCa, and a high number of tumors show aggressive characteristics. While the impact of screening on patient outcomes remains to be more firmly established, the high SIR values we observe provide support for continued PSA screening of MSH2 and MSH6 carriers. Studies are needed to provide optimal recommendations for PSA-threshold and to evaluate whether MLH1 and PMS2 carriers should not be recommended screening.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Dépistage précoce du cancer , Antigène spécifique de la prostate , Tumeurs de la prostate , Humains , Mâle , Norvège/épidémiologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/diagnostic , Tumeurs de la prostate/épidémiologie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/sang , Antigène spécifique de la prostate/sang , Adulte d'âge moyen , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/épidémiologie , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Études prospectives , Dépistage précoce du cancer/méthodes , Sujet âgé , Adulte , Protéine-2 homologue de MutS/génétique , Incidence , Réparation de mésappariement de l'ADN/génétique , Grading des tumeurs , Protéines de liaison à l'ADN/génétique
3.
Cancer Prev Res (Phila) ; 17(7): 311-324, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38643981

RÉSUMÉ

Lynch syndrome (LS) is the most prevalent heritable form of colorectal cancer. Its early onset and high lifetime risk for colorectal cancer emphasize the necessity for effective chemoprevention. NFE2L2 (NRF2) is often considered a potential druggable target, and many chemopreventive compounds induce NRF2. However, although NRF2 counteracts oxidative stress, it is also overexpressed in colorectal cancer and may promote tumorigenesis. In this study, we evaluated the role of NRF2 in the prevention of LS-associated neoplasia. We found increased levels of NRF2 in intestinal epithelia of mice with intestinal epithelium-specific Msh2 deletion (MSH2ΔIEC) compared with C57BL/6 (wild-type) mice, as well as an increase in downstream NRF2 targets NAD(P)H dehydrogenase (quinone 1) and glutamate-cysteine ligase catalytic subunit. Likewise, NRF2 levels were increased in human MSH2-deficient LS tumors compared with healthy human controls. In silico analysis of a publicly accessible RNA sequencing LS dataset also found an increase in downstream NRF2 targets. Upon crossing MSH2ΔIEC with Nrf2null (MSH2ΔIECNrf2null) mice, we unexpectedly found reduced tumorigenesis in MSH2ΔIECNrf2null mice compared with MSH2ΔIEC mice after 40 weeks, which occurred despite an increase in oxidative damage in MSH2ΔIECNrf2null mice. The loss of NRF2 impaired proliferation as seen by Ki67 intestinal staining and in organoid cultures. This was accompanied by diminished WNT/ß-catenin signaling, but apoptosis was unaffected. Microbial α-diversity increased over time with the loss of NRF2 based upon 16S rRNA gene amplicon sequencing of murine fecal samples. Altogether, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. Activation of NRF2 may not be a feasible strategy for chemoprevention in LS. Prevention Relevance: Patients with LS have an early onset and high lifetime risk for colorectal cancer. In this study, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. This suggests that NRF2 may not be a tumor suppressor in this specific context.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Modèles animaux de maladie humaine , Souris de lignée C57BL , Protéine-2 homologue de MutS , Facteur-2 apparenté à NF-E2 , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/métabolisme , Protéine-2 homologue de MutS/génétique , Protéine-2 homologue de MutS/métabolisme , Humains , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Souris knockout , Femelle , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Mâle
5.
Int J Clin Oncol ; 29(7): 944-952, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38642190

RÉSUMÉ

BACKGROUND: Lynch-like syndrome (LLS) has recently been proposed as a third type of microsatellite instability (MSI) tumor after Lynch syndrome (LS) and sporadic MSI colorectal cancer (CRC) without either a germline variant of mismatch repair (MMR) genes or hypermethylation of the MLH1 gene. The present study aimed to clarify and compare the clinicopathological characteristics of LLS with those of the other MSI CRC subtypes. METHODS: In total, 2634 consecutive patients with CRC who underwent surgical resection and subsequently received universal tumor screening (UTS), including MSI analysis were enrolled between January 2008 and November 2019. Genetic testing was performed in patients suspected of having Lynch syndrome. RESULTS: UTS of the cohort found 146 patients with MSI CRC (5.5%). Of these, excluding sporadic MSI CRC, 30 (1.1%) had a diagnosis of LS, and 19 (0.7%) had no germline pathogenic variants of the MMR gene. The CRC type in the latter group was identified as LLS. LLS occurred significantly more often in young patients, was left-sided, involved a KRAS variant and BRAF wild-type, and had a higher concordance rate with the Revised Bethesda Guidelines than sporadic MSI CRC. No significant differences were observed in terms of the clinicopathological factors between LLS and LS-associated MSI CRC; however, LLS had a lower frequency of LS-related neoplasms compared with LS. CONCLUSIONS: Distinguishing clinically between LS and LLS was challenging, but the incidence of neoplasms was higher in LS than in LLS, suggesting the need for different screening and surveillance methods for the two subtypes.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Instabilité des microsatellites , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Protéine-1 homologue de MutL/génétique , Réparation de mésappariement de l'ADN/génétique , Protéines proto-oncogènes B-raf/génétique , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes p21(ras)/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Dépistage génétique
6.
Int J Clin Oncol ; 29(7): 953-963, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38615286

RÉSUMÉ

BACKGROUND: Based on molecular characteristics, deficient DNA mismatch repair (dMMR) solid tumors are largely divided into three categories: somatically MLH1-hypermethylated tumors, Lynch syndrome (LS)-associated tumors, and Lynch-like syndrome (LLS)-associated tumors. The incidence of each of these conditions and the corresponding pathogenic genes related to LLS remain elusive. METHODS: We identified dMMR tumors in 3609 tumors from 9 different solid organs, including colorectal cancer, gastric cancer, small-bowel cancer, endometrial cancer, ovarian cancer, upper urinary tract cancer, urinary bladder cancer, prostate cancer, and sebaceous tumor, and comprehensively summarized the characterization of dMMR tumors. Characterization of dMMR tumors were performed as loss of at least one of MMR proteins (MLH1, MSH2, MSH6, and PMS2), by immunohistochemistry, followed by MLH1 promotor methylation analysis and genetic testing for MMR genes where appropriate. Somatic variant analysis of MMR genes and whole exome sequencing (WES) were performed in patients with LLS. RESULTS: In total, the incidence of dMMR tumors was 5.9% (24/3609). The incidence of dMMR tumors and the proportion of the three categorized dMMR tumors varied considerably with different tumor types. One to three likely pathogenic/pathogenic somatic MMR gene variants were detected in 15 out of the 16 available LLS tumors. One patient each from 12 patients who gave consent to WES demonstrated non-MMR germline variants affect function (POLQ or BRCA1). CONCLUSIONS: Our data regarding the LS to LLS ratio would be useful for genetic counseling in patients who are suspected to have LS, though the genetic backgrounds for the pathogenesis of LLS need further investigation.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Réparation de mésappariement de l'ADN , Mutation germinale , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Réparation de mésappariement de l'ADN/génétique , Femelle , Mâle , Incidence , Adulte d'âge moyen , Sujet âgé , Adulte , Protéine-1 homologue de MutL/génétique , Méthylation de l'ADN ,
7.
ACS Biomater Sci Eng ; 10(5): 3136-3147, 2024 05 13.
Article de Anglais | MEDLINE | ID: mdl-38663028

RÉSUMÉ

Treatment with immune checkpoint inhibitors (ICIs) has shown efficacy in some patients with Lynch syndrome-associated colon cancer, but some patients still do not benefit from it. In this study, we adopted a combination strategy of tumor vaccines and ICIs to maximize the benefits of immunotherapy. Here, we obtained tumor-antigen-containing cell lysate (TCL) by lysing MC38Mlh1 KD cells and prepared liposome nanoparticles (Lipo-PEG) with a typical spherical morphology by thin-film hydration. Anti-PD-L1 was coupled to the liposome surface by the amidation reaction. As observed, anti-PD-L1/TCL@Lipo-PEG was not significantly toxic to mouse intestinal epithelial cells (MODE-K) in the safe concentration range and did not cause hemolysis of mouse red blood cells. In addition, anti-PD-L1/TCL@Lipo-PEG reduced immune escape from colon cancer cells (MC38Mlh1 KD) by the anti-PD-L1 antibody, restored the killing function of CD8+ T cells, and targeted more tumor antigens to bone marrow-derived dendritic cells (BMDCs), which also expressed PD-L1, to stimulate BMDC antigen presentation. In syngeneic transplanted Lynch syndrome-associated colon cancer mice, the combination of anti-PD-L1 and TCL provided better cancer suppression than monoimmunotherapy, and the cancer suppression effect of anti-PD-L1/TCL@Lipo-PEG treatment was even better than that of the free drug. Meanwhile anti-PD-L1/TCL@Lipo-PEG enhanced the immunosuppressive tumor microenvironment. In vivo fluorescence imaging and H&E staining showed that the nanomedicine was mainly retained in the tumor site and had no significant toxic side effects on other major organs. The anti-PD-L1/TCL@Lipo-PEG prepared in this study has high efficacy and good biosafety in alleviating the progression of Lynch syndrome-associated colon cancer, and it is expected to be a therapeutic candidate for Lynch syndrome-associated colon cancer.


Sujet(s)
Antigène CD274 , Tumeurs du côlon , Tumeurs colorectales héréditaires sans polypose , Liposomes , Animaux , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/traitement médicamenteux , Souris , Antigène CD274/métabolisme , Nanomédecine , Lignée cellulaire tumorale , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/immunologie , Humains , Souris de lignée C57BL , Femelle , Cellules dendritiques/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/métabolisme , Nanoparticules/composition chimique , Nanoparticules/usage thérapeutique , Évolution de la maladie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Antigènes néoplasiques/immunologie
8.
Cancer Med ; 13(7): e7041, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38558366

RÉSUMÉ

BACKGROUND: Up to 70% of suspected Lynch syndrome patients harboring MMR deficient tumors lack identifiable germline pathogenic variants in MMR genes, being referred to as Lynch-like syndrome (LLS). Previous studies have reported biallelic somatic MMR inactivation in a variable range of LLS-associated tumors. Moreover, translating tumor testing results into patient management remains controversial. Our aim is to assess the challenges associated with the implementation of tumoral MMR gene testing in routine workflows. METHODS: Here, we present the clinical characterization of 229 LLS patients. MMR gene testing was performed in 39 available tumors, and results were analyzed using two variant allele frequency (VAF) thresholds (≥5% and ≥10%). RESULTS AND DISCUSSION: More biallelic somatic events were identified at VAF ≥ 5% than ≥10% (35.9% vs. 25.6%), although the rate of nonconcordant results regarding immunohistochemical pattern increased (30.8% vs. 20.5%). Interpretation difficulties question the current utility of the identification of MMR somatic hits in the diagnostic algorithm of suspected LS cases.


Sujet(s)
Tumeurs du cerveau , Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Syndromes néoplasiques héréditaires , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Mutation germinale , Réparation de mésappariement de l'ADN/génétique
9.
EBioMedicine ; 103: 105111, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38583260

RÉSUMÉ

BACKGROUND: Lynch syndrome (LS) is one of the most common hereditary cancer syndromes worldwide. Dominantly inherited mutation in one of four DNA mismatch repair genes combined with somatic events leads to mismatch repair deficiency and microsatellite instability (MSI) in tumours. Due to a high lifetime risk of cancer, regular surveillance plays a key role in cancer prevention; yet the observation of frequent interval cancers points to insufficient cancer prevention by colonoscopy-based methods alone. This study aimed to identify precancerous functional changes in colonic mucosa that could facilitate the monitoring and prevention of cancer development in LS. METHODS: The study material comprised colon biopsy specimens (n = 71) collected during colonoscopy examinations from LS carriers (tumour-free, or diagnosed with adenoma, or diagnosed with carcinoma) and a control group, which included sporadic cases without LS or neoplasia. The majority (80%) of LS carriers had an inherited genetic MLH1 mutation. The remaining 20% included MSH2 mutation carriers (13%) and MSH6 mutation carriers (7%). The transcriptomes were first analysed with RNA-sequencing and followed up with Gorilla Ontology analysis and Reactome Knowledgebase and Ingenuity Pathway Analyses to detect functional changes that might be associated with the initiation of the neoplastic process in LS individuals. FINDINGS: With pathway and gene ontology analyses combined with measurement of mitotic perimeters from colonic mucosa and tumours, we found an increased tendency to chromosomal instability (CIN), already present in macroscopically normal LS mucosa. Our results suggest that CIN is an earlier aberration than MSI and may be the initial cancer driving aberration, whereas MSI accelerates tumour formation. Furthermore, our results suggest that MLH1 deficiency plays a significant role in the development of CIN. INTERPRETATION: The results validate our previous findings from mice and highlight early mitotic abnormalities as an important contributor and precancerous marker of colorectal tumourigenesis in LS. FUNDING: This work was supported by grants from the Jane and Aatos Erkko Foundation, the Academy of Finland (330606 and 331284), Cancer Foundation Finland sr, and the Sigrid Jusélius Foundation. Open access is funded by Helsinki University Library.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Instabilité des microsatellites , Mitose , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/complications , Femelle , Mâle , Mitose/génétique , Adulte d'âge moyen , Mutation , Adulte , Sujet âgé , Protéine-1 homologue de MutL/génétique , Analyse de profil d'expression de gènes , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/étiologie , Carcinogenèse/génétique , Réparation de mésappariement de l'ADN/génétique , Transcriptome
10.
Cancer Sci ; 115(5): 1646-1655, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38433331

RÉSUMÉ

The clinical features of sporadic mismatch repair deficiency (MMRd) and Lynch syndrome (LS) in Japanese patients with endometrial cancer (EC) were examined by evaluating the prevalence and prognostic factors of LS and sporadic MMRd in patients with EC. Targeted sequencing of five LS susceptibility genes (MLH1, MSH2, MSH6, PMS2, and EPCAM) was carried out in 443 patients with EC who were pathologically diagnosed with EC at the National Cancer Center Hospital between 2011 and 2018. Pathogenic variants in these genes were detected in 16 patients (3.7%). Immunohistochemistry for MMR proteins was undertaken in 337 of the 433 (77.9%) EC patients, and 91 patients (27.0%) showed absent expression of at least one MMR protein. The 13 cases of LS with MMR protein loss (93.8%) showed a favorable prognosis with a 5-year overall survival (OS) rate of 100%, although there was no statistically significant difference between this group and the sporadic MMRd group (p = 0.27). In the MMRd without LS group, the 5-year OS rate was significantly worse in seven patients with an aberrant p53 expression pattern than in those with p53 WT (53.6% vs. 93.9%, log-rank test; p = 0.0016). These results suggest that p53 abnormalities and pathogenic germline variants in MMR genes could be potential biomarkers for the molecular classification of EC with MMRd.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Réparation de mésappariement de l'ADN , Tumeurs de l'endomètre , Protéine p53 suppresseur de tumeur , Tumeurs de l'utérus , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Adulte d'âge moyen , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Réparation de mésappariement de l'ADN/génétique , Protéines de liaison à l'ADN/génétique , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Molécule d'adhérence des cellules épithéliales/génétique , Molécule d'adhérence des cellules épithéliales/métabolisme , Japon , Mismatch repair endonuclease PMS2/génétique , Protéine-1 homologue de MutL/génétique , Protéine-2 homologue de MutS/génétique , Pronostic , Protéine p53 suppresseur de tumeur/génétique , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie
11.
Crit Rev Oncol Hematol ; 197: 104331, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38521284

RÉSUMÉ

Even with colonoscopy surveillance, Lynch syndromes (LS) carriers still develop colorectal cancer (CRC). The cumulative incidence of CRCs under colonoscopy surveillance varies depending on the affected mismatch repair (MMR) gene. However, the precise mechanisms driving these epidemiological patterns remain incompletely understood. In recent years, several potential mechanisms explaining the occurrence of CRCs during colonoscopy surveillance have been proposed in individuals with and without LS. These encompass biological factors like concealed/accelerated carcinogenesis through a bypassed adenoma stage and accelerated progression from adenomas. Alongside these, various colonoscopy-related factors may contribute to formation of CRCs under colonoscopy surveillance, like missed yet detectable (pre)cancerous lesions, detected yet incompletely removed (pre)cancerous lesions, and colonoscopy-induced carcinogenesis due to tumor cell reimplantation. In this comprehensive literature update, we reviewed these potential factors and evaluated their relevance to each MMR group in an attempt to raise further awareness and stimulate research regarding this conflicting phenomenon.


Sujet(s)
Coloscopie , Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Humains , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/épidémiologie , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Coloscopie/méthodes , Tumeurs colorectales/diagnostic , Tumeurs colorectales/épidémiologie , Tumeurs colorectales/étiologie , Tumeurs colorectales/génétique , Réparation de mésappariement de l'ADN/génétique
12.
Cancer Gene Ther ; 31(6): 842-850, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38332046

RÉSUMÉ

Lynch syndrome (LS) is an inherited condition characterized by an increased risk of developing cancer, in particular colorectal cancer (CRC). Microsatellite instability (MSI) is the main feature of (pre)cancerous lesions occurring in LS patients. Close endoscopic surveillance is the only option available to reduce CRC morbidity and mortality. However, it may fail to intercept interval cancers and patients' compliance to such an invasive procedure may decrease over the years. The development of a minimally invasive test able to detect (pre)cancerous colorectal lesions, could thus help tailor surveillance programs in LS patients. Taking advantage of an endoscopic surveillance program, we retrospectively assessed the instability of five microsatellites (BAT26, BAT25, NR24, NR21, and Mono27) in liquid biopsies collected at baseline and possibly at two further endoscopic rounds. For this purpose, we tested a new multiplex drop-off digital polymerase chain reaction (dPCR) assay, reaching mutant allele frequencies (MAFs) as low as 0.01%. Overall, 78 plasma samples at the three time-points from 18 patients with baseline (pre)cancerous lesions and 18 controls were available for molecular analysis. At baseline, the MAFs of BAT26, BAT25 and NR24 were significantly higher in samples of patients with lesions but did not differ with respect to the grade of dysplasia or any other clinico-pathological characteristics. When all markers were combined to determine MSI in blood, this test was able to discriminate lesion-bearing patients with an AUC of 0.80 (95%CI: 0.66; 0.94).


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Instabilité des microsatellites , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Biopsie liquide/méthodes , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Études rétrospectives , États précancéreux/génétique , États précancéreux/anatomopathologie , États précancéreux/diagnostic , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/diagnostic , Marqueurs biologiques tumoraux/génétique
13.
Zhonghua Yi Xue Za Zhi ; 104(7): 547-551, 2024 Feb 20.
Article de Chinois | MEDLINE | ID: mdl-38317368

RÉSUMÉ

In this study, a case of Lynch syndrome (LS) family line with a novel mutation site in the MLH1 c.463dupC gene was reported and the clinical and pathogenic genetic features of this family were analyzed. A 40-year-old female patient with colon cancer diagnosed at the First Affiliated Hospital of Kunming Medical University on October 2, 2020 was retrospectively included. The clinical data of the family were collected and the family lineage was drawn. The family tumor history met the Amsterdam Criteria Ⅱ and the diagnostic criteria of LS in Chinese, which was a typical LS family lineage. A germline code-shift missense mutation c.463dupC in the MLH1 gene located in exon 6, a possible pathogenic variant, was detected by second-generation sequencing (NGS) in the patient. Subsequently, Sanger sequencing was performed on a total of 20 direct lineage members of the family of the MLH1 gene, 7 cases were found to harbor the mutation and included in the LS high-risk control. Follow-up to October 2023 showed that the patient had endometrial and cervical polyps, one case had colorectal cancer, and two cases had intestinal polyps, all were treated with early intervention and therapy; two cases did not show any clinical symptoms. This study is the first to report a new mutation site for the potentially pathogenic MLH1 c.463dupC, providing a rationale for the pathogenicity of the mutation and standardized health management for familial carriers.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Femelle , Humains , Adulte , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Prédisposition génétique à une maladie , Études rétrospectives , Protéine-1 homologue de MutL/génétique , Mutation
14.
BMC Gastroenterol ; 24(1): 82, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38395750

RÉSUMÉ

BACKGROUND: Deficient DNA mismatch repair (MMR) can cause microsatellite instability (MSI) and is more common in colorectal cancer (CRC) patients. Understanding the carcinogenic mechanism of bacteria and their impact on cancer cells is crucial. Bacteroides fragilis (B. fragilis) has been identified as a potential promoter of tumorigenesis through the alteration of signaling pathways. This study aims to assess the expression levels of msh2, msh6, mlh1, and the relative frequency of B. fragilis in biopsy samples from CRC patients. MATERIALS AND METHODS: Based on the sequence of mlh1, msh2, and msh6 genes, B. fragilis specific 16srRNA and bacterial universal 16srRNA specific primers were selected, and the expression levels of the target genes were analyzed using the Real-Time PCR method. RESULTS: Significant increases in the expression levels of mlh1, msh2, and msh6 genes were observed in the cancer group. Additionally, the expression of these MMR genes showed a significant elevation in samples positive for B. fragilis presence. The relative frequency of B. fragilis in the cancer group demonstrated a significant rise compared to the control group. CONCLUSION: The findings suggest a potential correlation between the abundance of B. fragilis and alterations in the expression of MMR genes. Since these genes can play a role in modifying colon cancer, investigating microbial characteristics and gene expression changes in CRC could offer a viable solution for CRC diagnosis.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Humains , Réparation de mésappariement de l'ADN/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Bacteroides fragilis/génétique , Bacteroides fragilis/métabolisme , Iran , Protéine-2 homologue de MutS/génétique , Protéine-2 homologue de MutS/métabolisme , Instabilité des microsatellites , Protéines de liaison à l'ADN/génétique , Protéine-1 homologue de MutL/génétique , Protéine-1 homologue de MutL/métabolisme , Biopsie
15.
Histopathology ; 84(6): 1056-1060, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38275207

RÉSUMÉ

AIM: Currently, screening of colorectal cancers (CRC) by assessing mismatch repair deficiency (dMMR) or microsatellite instability (MSI) is used to identify Lynch syndrome (LS) patients. Advanced adenomas are considered immediate precursor lesions of CRC. In this study we investigate the relevance of screening of advanced adenomas for LS in population screening. METHODS AND RESULTS: Advanced adenomas (n = 1572) were selected from the Dutch colorectal cancer population screening programme, based on one or more of the criteria: tubulovillous (n = 848, 54%) or villous adenoma (n = 118, 7.5%), diameter ≥ 1 cm (n = 1286, 82%) and/or high-grade dysplasia (n = 176, 11%). In 86 cases (5%), all three criteria were fulfilled at the same time. MMR-IHC and/or MSI analyses were performed on all cases. Only five advanced adenomas (0.3%) showed dMMR and MSI, including two cases with hypermethylation. In at least two patients a germline event was suspected based on allelic frequencies. No pathogenic explanation was found in the last case. CONCLUSION: Timely testing of precursor lesions would be preferable to detect new LS patients before CRC development. However, standard assessment of dMMR of advanced adenomas from the population screening is not effective.


Sujet(s)
Adénomes , Tumeurs du cerveau , Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Syndromes néoplasiques héréditaires , Humains , Réparation de mésappariement de l'ADN/génétique , Dépistage précoce du cancer , Tumeurs colorectales/diagnostic , Tumeurs colorectales/génétique , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Adénomes/diagnostic , Adénomes/génétique , Instabilité des microsatellites
16.
PLoS One ; 19(1): e0292277, 2024.
Article de Anglais | MEDLINE | ID: mdl-38271352

RÉSUMÉ

Colorectal cancer (CRC) is a major global health concern, with microsatellite instability-high (MSI-H) being a defining characteristic of hereditary nonpolyposis colorectal cancer syndrome and affecting 15% of sporadic CRCs. Tumors with MSI-H have unique features and better prognosis compared to MSI-L and microsatellite stable (MSS) tumors. This study proposed establishing a MSI prediction model using more available and low-cost colonoscopy images instead of histopathology. The experiment utilized a database of 427 MSI-H and 1590 MSS colonoscopy images and vision Transformer (ViT) with different feature training approaches to establish the MSI prediction model. The accuracy of combining pre-trained ViT features was 84% with an area under the receiver operating characteristic curve of 0.86, which was better than that of DenseNet201 (80%, 0.80) in the experiment with support vector machine. The content-based image retrieval (CBIR) approach showed that ViT features can obtain a mean average precision of 0.81 compared to 0.79 of DenseNet201. ViT reduced the issues that occur in convolutional neural networks, including limited receptive field and gradient disappearance, and may be better at interpreting diagnostic information around tumors and surrounding tissues. By using CBIR, the presentation of similar images with the same MSI status would provide more convincing deep learning suggestions for clinical use.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Humains , Instabilité des microsatellites , Tumeurs colorectales/imagerie diagnostique , Tumeurs colorectales/génétique , Répétitions microsatellites , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Coloscopie
17.
Mol Genet Genomic Med ; 12(1): e2360, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38284451

RÉSUMÉ

BACKGROUND: Lynch syndrome represents one of the most common cancer predispositions worldwide and is caused by germline pathogenic variants (PV) in DNA mismatch repair (MMR) genes. We repeatedly identified a PV in the MMR gene PMS2, c.1831dup, accounting for 27% of all Swiss PMS2 PV index patients identified. Notably, 2/18 index patients had been diagnosed with colorectal cancer (CRC) before age 30. METHODS: In this study, we investigated if this PV could (i) represent a founder variant by haplotype analysis and (ii) be associated with a more severe clinical phenotype. RESULTS: Haplotype analysis identified a shared common region of about 0.7 Mb/1.3 cM in 13 (81%) out of 16 index patients. Genotype-phenotype correlations, combining data from the 18 Swiss and 18 literature-derived PMS2 c.1831dup PV index patients and comparing them to 43 Swiss index patients carrying other PMS2 PVs, indicate that the PMS2 c.1831dup variant may be associated with earlier (<50 y) age at CRC diagnosis (55% vs. 29%, respectively; p = 0.047). Notably, 30% (9/30) of cancers from c.1831dup carriers displayed atypical MMR protein expression patterns on immunohistochemistry. CONCLUSION: Our results suggest that the PMS2 c.1831dup PV represents a, probably ancient, founder mutation and is possibly associated with an earlier CRC diagnosis compared to other PMS2 PVs.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Humains , Adulte , Tumeurs colorectales/génétique , Mismatch repair endonuclease PMS2/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Génotype , Études d'associations génétiques
18.
World J Surg Oncol ; 22(1): 36, 2024 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-38280988

RÉSUMÉ

BACKGROUND: Lynch syndrome (LS) is the most common hereditary colorectal cancer (CRC) syndrome. This condition is characterized by germline variants in DNA mismatch repair (MMR) genes, including MLH1, MSH2, MSH6, and PMS2. In this study, we analyzed the molecular defects and clinical manifestations of two families affected with CRC and proposed appropriate individual preventive strategies for all carriers of the variant. METHODS: We recruited two families diagnosed with CRC and combined their family history and immunohistochemical results to analyze the variants of probands and those of other family members by using whole exome sequencing. Subsequently, gene variants in each family were screened by comparing them with the variants available in the public database. Sanger sequencing was performed to verify the variant sites. An online platform ( https://www.uniprot.org ) was used to analyze the functional domains of mutant proteins. RESULTS: A novel frameshift variant (NM_001281492, c.1129_1130del, p.R377fs) in MSH6 and a known deleterious variant (NM_000249.4:c.1731G > A, p.S577S) in MLH1 were identified in the two families with CRC. Using bioinformatics tools, we noted that the frameshift variant reduced the number of amino acids in the MSH6 protein from 1230 to 383, thereby leading to no MSH6 protein expression. The silent variant caused splicing defects and was strongly associated with LS. 5-Fluorouracil-based adjuvant chemotherapy is not recommended for patients with LS. CONCLUSIONS: The novel frameshift variant (MSH6, c.1129_1130del, p.R377fs) is likely pathogenic to LS, and the variant (MLH1, c.1731G > A, p.S577S) has been further confirmed to be pathogenic to LS. Our findings underscore the significance of genetic testing for LS and recommend that genetic consultation and regular follow-ups be conducted to guide individualized treatment for cancer-afflicted families, especially those with a deficiency in MMR expression.


Sujet(s)
Tumeurs colorectales héréditaires sans polypose , Syndromes néoplasiques héréditaires , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Réparation de mésappariement de l'ADN/génétique , Mutation germinale , Protéines de liaison à l'ADN/génétique , Chine/épidémiologie , Protéine-1 homologue de MutL/génétique , Mismatch repair endonuclease PMS2/génétique , Mismatch repair endonuclease PMS2/métabolisme
19.
JCO Precis Oncol ; 8: e2300332, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38271656

RÉSUMÉ

PURPOSE: The pan-cancer presence of microsatellite instability (MSI)-positive tumors demonstrates its clinical utility as an agnostic biomarker for identifying immunotherapy-eligible patients. Additionally, MSI is a hallmark of Lynch syndrome (LS), the most prevalent cancer susceptibility syndrome among patients with colorectal and endometrial cancer. Therefore, MSI-high results should inform germline genetic testing for cancer-predisposing genes. However, in clinical practice, such analysis is frequently disregarded. METHODS: A next-generation sequencing (NGS)-based technique was used for MSI analysis in 4,553 patients with various tumor types. Upon request, somatic BRAF gene analysis was conducted. In addition, hereditary testing of cancer-associated genes was performed in MSI-high cases using a capture-based NGS protocol. MLH1 promoter methylation analysis was conducted retrospectively in patients with colorectal and endometrial cancer to further investigate the origin of MSI at the tumor level. RESULTS: The MSI positivity rate for the entire cohort was 5.27%. Endometrial, gastric, colorectal, urinary tract, and prostate cancers showed the highest proportion of MSI-high cases (15.69%, 8.54%, 7.40%, 4.55%, and 3.19%, respectively). A minority of 45 patients (22.73%) among the MSI-high cases underwent germline testing to determine whether the mismatch repair pathway deficiency was inherited. 24.44% of those who performed the genetic test carried a pathogenic variant in an LS-associated gene. Three MSI-high individuals had non-LS gene alterations, including BRCA1, BRCA2, and CDKN2A pathogenic variants, indicating the presence of non-LS-associated gene alterations among MSI-high patients. CONCLUSION: Although MSI analysis is routinely performed in clinical practice, as many as 77% of MSI-high patients do not undergo LS genetic testing, despite international guidelines strongly recommending it. BRAF and MLH1 methylation analysis could shed light on the somatic origin of MSI in 42.50% of the MSI-high patients; however, MLH1 analysis is barely ever requested in clinical practice.


Sujet(s)
Tumeurs du cerveau , Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Tumeurs de l'endomètre , Syndromes néoplasiques héréditaires , Mâle , Femelle , Humains , Tumeurs colorectales héréditaires sans polypose/diagnostic , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Études rétrospectives , Instabilité des microsatellites , Protéines proto-oncogènes B-raf/génétique , Tumeurs colorectales/génétique , Marqueurs biologiques , Tumeurs de l'endomètre/diagnostic , Tumeurs de l'endomètre/génétique
20.
Int J Cancer ; 154(8): 1455-1463, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38175816

RÉSUMÉ

Lynch syndrome (LS) predisposes to cancer in adulthood and is caused by heterozygous germline variants in a mismatch repair (MMR) gene. Recent studies show an increased prevalence of LS among children with cancer, suggesting a causal relationship. For LS-spectrum (LSS) cancers, including high-grade gliomas and colorectal cancer, causality has been supported by typical MMR-related tumor characteristics, but for non-LSS cancers, causality is unclear. We characterized 20 malignant tumors of 18 children with LS, including 16 non-LSS tumors. We investigated second hits, tumor mutational load, mutational signatures and MMR protein expression. In all LSS tumors and three non-LSS tumors, we detected MMR deficiency caused by second hit somatic alterations. Furthermore, these MMR-deficient tumors carried driver variants that likely originated as a consequence of MMR deficiency. However, in 13 non-LSS tumors (81%), a second hit and MMR deficiency were absent, thus a causal link between LS and cancer development in these children is lacking. These findings demonstrate that causality of LS in children with cancer, which can be determined by molecular tumor characterization, seems to be restricted to specific tumor types. Large molecular and epidemiological studies are needed to further refine the tumor spectrum in children with LS.


Sujet(s)
Tumeurs du cerveau , Tumeurs colorectales héréditaires sans polypose , Tumeurs colorectales , Syndromes néoplasiques héréditaires , Enfant , Humains , Tumeurs colorectales héréditaires sans polypose/génétique , Tumeurs colorectales héréditaires sans polypose/anatomopathologie , Tumeurs colorectales/anatomopathologie , Tumeurs du cerveau/génétique , Mutation germinale , Réparation de mésappariement de l'ADN/génétique , Instabilité des microsatellites , Protéine-1 homologue de MutL/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE