Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
BMC Med Genomics ; 17(1): 215, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160519

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a rare autosomal dominant syndrome caused by a germline mutation in the fumarate hydratase (FH) gene that manifests with cutaneous leiomyomas, uterine fibroids, and renal cell cancer (RCC). Patients with HLRCC-associated RCC (HLRCC-RCC) have aggressive clinical courses, but there is no standardized therapy for advanced HLRCC-RCC. In this study, we described a case of aggressive HLRCC in a 33-year-old female who exhibited a novel heterozygous germline insertion mutation in exon 8 of the FH gene (c.1126 C > T; p.Q376*). The patient underwent laparoscopic resection of the right kidney, but metastases appeared within 3 months after surgery. Histological staining of the resected tumor revealed high expression levels of programmed cell death-ligand 1 (PD-L1). Therefore, the patient was treated with immunotherapy. The patient achieved a partial response to immunotherapy, and the treatment of metastatic lesions has continued to improve. A thorough literature review pinpointed 76 historical cases of HLRCC-RCC that had undergone immunotherapy. From this pool, 46 patients were selected for this study to scrutinize the association between mutations in the FH gene and the effectiveness of immunotherapy. Our results indicate that immunotherapy could significantly improve the overall survival (OS) of patients with HLRCC-RCC. However, no influence of different mutations in the FH germline gene on the therapeutic efficacy of immunotherapy was observed. Therefore, our study suggested that immunotherapy was an effective therapeutic option for patients with HLRCC regardless of the type of FH germline mutation.


Subject(s)
Fumarate Hydratase , Immunotherapy , Leiomyomatosis , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms , Humans , Female , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Leiomyomatosis/therapy , Fumarate Hydratase/genetics , Adult , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Uterine Neoplasms/therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Germ-Line Mutation , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy
2.
Zhonghua Bing Li Xue Za Zhi ; 53(6): 605-609, 2024 Jun 08.
Article in Chinese | MEDLINE | ID: mdl-38825907

ABSTRACT

Objective: To investigate the clinicopathological features of children with metachronous or synchronous primary tumors and to identify related genetic tumor syndromes. Methods: The clinicopathological data of 4 children with multiple primary tumors diagnosed in the Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China from 2011 to 2023 were collected. The histological, immunophenotypic and molecular characteristics were examined using H&E staining, immunohistochemical staining, PCR, Sanger sequencing and next-generation sequencing (NGS). The patients were followed up. Results: Case 1 was an 8-year-old boy with the adrenal cortical carcinoma, and 5 years later a poorly differentiated gastric adenocarcinoma was detected. Case 2 was a 2-year-old boy, presented with a left ventricular choroid plexus carcinoma, and a hepatoblastoma was detected 8 months later. Case 3 was a 9-month-old girl, diagnosed with renal rhabdoid tumor first and intracranial atypical teratoid/rhabdoid tumor (AT/RT) 3 months later. Case 4 was a 7-year-old boy and had a sigmoid colon adenocarcinoma 3 years after the diagnosis of a glioblastoma. The morphology and immunohistochemical features of the metachronous or synchronous primary tumors in the 4 cases were similar to the corresponding symptom-presenting/first-diagnosed tumors. No characteristic germ line mutations were detected in cases 1 and 2 by relevant molecular detection, and the rhabdoid tumor predisposition syndrome was confirmed in case 3 using NGS. Case 4 was clearly related to constitutional mismatch repair deficiency as shown by the molecular testing and clinical features. Conclusions: Childhood multiple primary tumors are a rare disease with histological morphology and immunophenotype similar to the symptom-presenting tumors. They are either sporadic or associated with a genetic (tumor) syndrome. The development of both tumors can occur simultaneously (synchronously) or at different times (metachronously). Early identification of the children associated with genetic tumor syndromes can facilitate routine tumor screening and early treatment.


Subject(s)
Hepatoblastoma , Kidney Neoplasms , Liver Neoplasms , Neoplasms, Multiple Primary , Rhabdoid Tumor , Stomach Neoplasms , Humans , Male , Child , Female , Child, Preschool , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/pathology , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Infant , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Hepatoblastoma/genetics , Hepatoblastoma/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/diagnosis , Choroid Plexus Neoplasms/genetics , Choroid Plexus Neoplasms/pathology , Choroid Plexus Neoplasms/diagnosis , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/pathology , Adrenal Cortex Neoplasms/pathology , Adrenal Cortex Neoplasms/genetics , Teratoma/pathology , Teratoma/genetics , Teratoma/surgery , Brain Neoplasms/genetics , Brain Neoplasms/pathology , SMARCB1 Protein/genetics , MutL Protein Homolog 1/genetics , Neoplasms, Second Primary/pathology , Neoplasms, Second Primary/genetics , High-Throughput Nucleotide Sequencing , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology
3.
Eur J Hum Genet ; 32(8): 1027-1031, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38824259

ABSTRACT

BAP1-Tumor Predisposition Syndrome (TPDS) is caused by germline variants in BAP1 and predisposes to solid tumors. After observation of a radiologically malignant-appearing splenic mass with benign pathology in a patient with BAP1-TPDS, we sought to retrospectively characterize splenic lesions in individuals with BAP1-TPDS seen at a comprehensive cancer center. A dedicated radiology review for splenic abnormalities was performed. We identified 37 individuals with BAP1-TPDS, 81% with a history of cancer. Of 33 individuals with abdominal imaging, 10 (30%) had splenic lesions, and none were shown to be malignant on follow-up. Splenectomy in an individual with suspected splenic angiosarcoma showed a benign vascular neoplasm with loss of nuclear staining for BAP1 in a subset of cells. Benign splenic lesions appear to be common and potentially BAP1-driven in individuals with BAP1-TPDS; confirmation of these findings could lead to more conservative management and avoidance of splenectomy.


Subject(s)
Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Humans , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Female , Male , Adult , Middle Aged , Aged , Splenic Neoplasms/genetics , Splenic Neoplasms/pathology , Splenic Neoplasms/metabolism , Germ-Line Mutation , Genetic Predisposition to Disease , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/metabolism , Splenectomy
4.
Sci Rep ; 14(1): 12447, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38822039

ABSTRACT

The innate immune molecule NLR family CARD domain-containing 5 (NLRC5) plays a significant role in endometrial carcinoma (EC) immunosurveillance. However, NLRC5 also plays a protumor role in EC cells. Mismatch repair gene deficiency (dMMR) can enable tumors to grow faster and also can exhibit high sensitivity to immune checkpoint inhibitors. In this study, we attempted to determine whether NLRC5-mediated protumor role in EC is via the regulation of dMMR. Our findings revealed that NLRC5 promoted the proliferation, migration, and invasion abilities of EC cells and induced the dMMR status of EC in vivo and in vitro. Furthermore, the mechanism underlying NLRC5 regulated dMMR was also verified. We first found NLRC5 could suppress nuclear factor-kappaB (NF-κB) pathway in EC cells. Then we validated that the positive effect of NLRC5 in dMMR was restricted when NF-κB was activated by lipopolysaccharides in NLRC5-overexpression EC cell lines. In conclusion, our present study confirmed the novel NLRC5/NF-κB/MMR regulatory mechanism of the protumor effect of NLRC5 on EC cells, thereby suggesting that the NLRC5-mediated protumor in EC was depend on the function of MMR.


Subject(s)
Cell Proliferation , Endometrial Neoplasms , Intracellular Signaling Peptides and Proteins , NF-kappa B , Signal Transduction , Humans , Female , NF-kappa B/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cell Line, Tumor , Animals , Cell Movement/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Mice , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , DNA Mismatch Repair , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Brain Neoplasms
5.
APMIS ; 132(8): 544-552, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38775301

ABSTRACT

Renal cell carcinoma (RCC) accounts for 2% of all cancer cases worldwide, and majority are sporadic. The latest World Health Organization (WHO) classification of renal cell tumors (fifth edition, 2022) has molecularly defined renal tumor entities, which includes fumarate hydratase (FH)-deficient RCC. FH-deficient RCC is an aggressive carcinoma caused by pathogenic alterations in FH gene, seen in 15% of patients with hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC) syndrome. These tumors occur more frequently at a younger age and present at an advanced stage, carrying a dismal prognosis. We report a series of 10 cases of FH-deficient RCC. The mean age was 49.8 years, and all cases presented in advanced stages (III and IV). Morphologically, the cases had varied architectural patterns with characteristic eosinophilic macronucleoli and perinucleolar halo. On immunohistochemistry (IHC), all showed diffuse nucleo-cytoplasmic expression of S-(2-succino)-cysteine (2-SC), with loss of FH in seven cases. FH-deficient RCCs are aggressive neoplasms and can be diagnosed using specific IHC markers (FH and 2-SC). These patients should undergo germline testing for FH gene mutation, genetic counseling, and surveillance of family members.


Subject(s)
Carcinoma, Renal Cell , Fumarate Hydratase , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Middle Aged , Female , Male , Adult , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Immunohistochemistry , Aged , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/pathology , Germ-Line Mutation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/deficiency , Cysteine/analogs & derivatives , Skin Neoplasms , Uterine Neoplasms , Leiomyomatosis
6.
JAMA Dermatol ; 160(8): 838-845, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38759225

ABSTRACT

Importance: BRCA1-associated protein (BAP1) tumor predisposition syndrome (TPDS) is a cancer genodermatosis associated with high risk of uveal and cutaneous melanoma, basal cell carcinoma, and multiple internal malignant neoplasms, including mesothelioma and renal cell carcinoma. Early detection of the syndrome is important for cancer surveillance and genetic counseling of family members who are at risk. Objective: To determine the prevalence of nail abnormalities in individuals with pathogenic germline variants in BAP1. Design, Setting, and Participants: In this prospective cohort study, individuals who were known carriers of pathogenic BAP1 germline variants were consecutively enrolled between October 10, 2023, and March 15, 2024. Dermatologic evaluation for nail abnormalities was performed, including a history of nail abnormalities and associated symptoms, physical examination, medical photography, and nail biopsy for histopathology. This was a single-center study conducted at the National Institutes of Health Clinical Center. Main Outcomes and Measures: Primary outcomes were the prevalence and spectrum of nail changes and histopathologic characterization. Results: Among 47 participants (30 female [63.8%]; mean [SD] age, 46.4 [15.1] years) ranging in age from 13 to 72 years from 35 families, nail abnormalities were detected in 41 patients (87.2%) and included leukonychia, splinter hemorrhage, onychoschizia, and distal nail hyperkeratosis. Clinical findings consistent with onychopapilloma were detected in 39 patients (83.0%), including 35 of 40 individuals aged 30 years or older (87.5%). Nail bed biopsy was performed in 5 patients and was consistent with onychopapilloma. Polydactylous involvement with onychopapillomas was detected in nearly all patients who had nail involvement (38 of 39 patients [97.4%]). Conclusions and Relevance: This study found that BAP1 TPDS was associated with a high rate of nail abnormalities consistent with onychopapillomas in adult carriers of the disease. Findings suggest that this novel cutaneous sign may facilitate detection of the syndrome in family members who are at risk and patients with cancers associated with BAP1 given that multiple onychopapillomas are uncommon in the general population and may be a distinct clue to the presence of a pathogenic germline variant in the BAP1 gene.


Subject(s)
Genetic Predisposition to Disease , Germ-Line Mutation , Skin Neoplasms , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Humans , Ubiquitin Thiolesterase/genetics , Female , Tumor Suppressor Proteins/genetics , Male , Middle Aged , Adult , Prospective Studies , Aged , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Young Adult , Adolescent , Nail Diseases/genetics , Nail Diseases/pathology , Nail Diseases/epidemiology , Nail Diseases/diagnosis , Prevalence , Papilloma/pathology , Papilloma/genetics , Papilloma/epidemiology , Papilloma/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Nails, Malformed/genetics , Nails, Malformed/epidemiology , Nails, Malformed/diagnosis
7.
Appl Immunohistochem Mol Morphol ; 32(6): 285-291, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38721798

ABSTRACT

Knowledge of the molecular pathways of pediatric high-grade gliomas is increasing. Gliomas with mismatch repair deficiency do not currently comprise a distinct group, but data on this topic have been accumulating in recent publications. Immunohistochemistry can effectively determine mismatch repair status, indirectly suggesting the microsatellite instability of the tumor. This study aimed to determine the number of mismatch repair-deficient pediatric high-grade gliomas in a tertiary institution and assess the relationship between the survival and mismatch repair status of the patients. It also aimed to assess the potential for further clinical studies including immunotherapy. Of 24 patients with high-grade gliomas, 3 deceased patients were mismatch repair-deficient. Mismatch repair deficiency was significantly associated with shorter survival ( P =0.004). Immunotherapy trials need to progress, and patients with mismatch repair-deficient pediatric high-grade gliomas are the most suitable candidates for such studies.


Subject(s)
Brain Neoplasms , DNA Mismatch Repair , Glioma , Immunohistochemistry , Humans , Glioma/metabolism , Glioma/pathology , Glioma/genetics , Child , Female , Male , Child, Preschool , Adolescent , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Infant , Neoplasm Grading , Microsatellite Instability , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/metabolism , Colorectal Neoplasms
8.
Br J Cancer ; 131(2): 334-346, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796599

ABSTRACT

BACKGROUND: Identifying the target of natural killer (NK) cells in colorectal cancer (CRC) is critical for optimising the clinical use of NK cell-mediated immunotherapy. Mismatch repair deficiency (dMMR) is associated with high immune cell infiltration and MHC Class I defects. Whether dMMR CRC responses to NK cell therapy remains unclear. METHODS: MLH1, DR4, and DR5 knockout cell lines were established using CRISPR-Cas9 system. NK92-MI or NK cell isolated from BABL/C mice were used as effector cells against tumour cells. Inflammatory cytokines secretion by CRC cells was assessed via cytokine analysis. NK-cell-deficient/proficient animal models were used to validate the NK cell sensitivity. RESULTS: We observed that dMMR CRC cells were more sensitive to NK cell-mediated cytotoxicity than were mismatch-repair-proficient (pMMR) CRC cells. In dMMR CRC, Death receptor (DR)4/5 was upregulated and mediated sensitivity to NK cell-mediated cytotoxicity. DR4/5-mediated secretion of interleukin -12 sustained NK cell viability in dMMR CRC. NK cell depletion induced dMMR CRC tumour growth, and NK cell transfer inhibited lung metastasis of dMMR CRC with DR4/5 expression in vivo. TP53 upregulated DR4/DR5 expression in dMMR CRC. CONCLUSIONS: dMMR associated with increased sensitivity to NK cell-mediated cytotoxicity in CRC. DR4/DR5 sensitise dMMR CRC to NK cell-mediated cytotoxicity.


Subject(s)
Colorectal Neoplasms , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Mice , Humans , Cell Line, Tumor , DNA Mismatch Repair , Cytotoxicity, Immunologic , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/deficiency , MutL Protein Homolog 1/metabolism , Mice, Inbred BALB C , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/immunology , Neoplastic Syndromes, Hereditary/pathology , Brain Neoplasms
9.
Genes Chromosomes Cancer ; 63(2): e23221, 2024 02.
Article in English | MEDLINE | ID: mdl-38682608

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is an autosomal dominant cancer predisposition syndrome characterized by cutaneous leiomyomas, uterine leiomyomas, and aggressive renal cancer. Germline variants in the fumarate hydratase (FH) gene predispose to HLRCC. Identifying germline pathogenic FH variants enables lifetime renal cancer screening and genetic testing for family members. In this report, we present a FH missense variant (c.1039T>C (p.S347P)), initially classified as a variant of uncertain significance. Clinical assessment, histopathological findings, molecular genetic studies, and enzymatic activity studies support the re-classification of the FH c.1039T>C variant to "pathogenic" based on ACMG/AMP criteria. Further insights into pathological recognition of FH-deficient renal cancer are discussed and should be recognized. This study has shown how (a) detailed multi-disciplinary analyses of a single variant can reclassify rare missense variants in FH and (b) careful pathological review of renal cancers is obligatory when HLRCC is suspected.


Subject(s)
Fumarate Hydratase , Leiomyomatosis , Mutation, Missense , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms , Humans , Fumarate Hydratase/genetics , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Female , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Pedigree , Germ-Line Mutation , Male , Adult , Genetic Predisposition to Disease , Middle Aged
10.
Virchows Arch ; 484(5): 723-731, 2024 May.
Article in English | MEDLINE | ID: mdl-38619599

ABSTRACT

A syndromic association between a subset of testicular/paratesticular neoplasms is well established. Such examples include Carney complex and large cell calcifying Sertoli cell tumor, Peutz-Jeghers syndrome and intratubular large cell hyalinizing Sertoli cell neoplasia, and VHL syndrome and clear cell papillary cystadenoma of the epididymis.However, recent studies proposed potential novel links between some testicular and paratesticular neoplasms with certain tumor syndromes. While more studies are still needed to solidify these associations, recent research suggests that a subset of Leydig cell tumors may arise in patients with hereditary leiomyomatosis and renal cell carcinoma syndrome or that some seminomas may occur in Lynch syndrome patients. Additionally, an association between testicular sex cord stromal tumors and paratesticular sarcomas with Familial adenomatous polyposis syndrome and DICER1 syndrome, respectively, has been proposed as well. This review provides a comprehensive overview of the intricate relationship between familial syndromes and associated testicular and paratesticular tumors, shedding light on their clinicopathological and molecular characteristics.


Subject(s)
Neoplastic Syndromes, Hereditary , Testicular Neoplasms , Humans , Male , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/genetics , Genetic Predisposition to Disease , Genital Neoplasms, Male/pathology , Genital Neoplasms, Male/genetics
11.
Eur J Hum Genet ; 32(6): 731-735, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38627541

ABSTRACT

Constitutional loss of SMAD4 function results in Juvenile Polyposis-Hereditary Haemorrhagic Telangiectasia Overlap Syndrome (JP-HHT). A retrospective multi-centre case-note review identified 28 patients with a pathogenic SMAD4 variant from 13 families across all Scottish Clinical Genetics Centres. This provided a complete clinical picture of the Scottish JP-HHT cohort. Colonic polyps were identified in 87% (23/28) and gastric polyps in 67% (12/18) of screened patients. Complication rates were high: 43% (10/23) of patients with polyps required a colectomy and 42% (5/12) required a gastrectomy. Colorectal cancer occurred in 25% (7/28) of patients, at a median age of 33 years. Pulmonary arteriovenous malformations were identified in 42% (8/19) of screened patients. 88% (23/26) and 81% (17/21) of patients exhibited JP and HHT features respectively, with 70% (14/20) demonstrating features of both conditions. We have shown that individuals with a pathogenic SMAD4 variant are all at high risk of both gastrointestinal neoplasia and HHT-related vascular complications, requiring a comprehensive screening programme.


Subject(s)
Intestinal Polyposis , Smad4 Protein , Telangiectasia, Hereditary Hemorrhagic , Humans , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Female , Male , Adult , Middle Aged , Intestinal Polyposis/genetics , Intestinal Polyposis/congenital , Intestinal Polyposis/pathology , Intestinal Polyposis/diagnosis , Adolescent , Scotland , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/diagnosis , Child , Mutation , Retrospective Studies , Aged
12.
J Med Genet ; 61(8): 734-740, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38575304

ABSTRACT

BACKGROUND: Both hereditary haemorrhagic telangiectasia (HHT) and juvenile polyposis syndrome (JPS) are known to be caused by SMAD4 pathogenic variants, with overlapping symptoms for both disorders in some patients. Additional connective tissue disorders have also been reported. Here, we describe carriers of SMAD4 variants followed in an HHT reference centre to further delineate the phenotype. METHODS: Observational study based on data collected from the Clinical Investigation for the Rendu-Osler Cohort database. RESULTS: Thirty-three participants from 15 families, out of 1114 patients with HHT, had an SMAD4 variant (3%).Regarding HHT, 26 out of 33 participants (88%) had a definite clinical diagnosis based on Curaçao criteria. Complication frequencies were as follows: epistaxis (n=27/33, 82%), cutaneous telangiectases (n=19/33, 58%), pulmonary arteriovenous malformations (n=17/32, 53%), hepatic arteriovenous malformations (AVMs) (n=7/18, 39%), digestive angiodysplasia (n=13/22, 59%). No cerebral AVMs were diagnosed.Regarding juvenile polyposis, 25 out of 31 participants (81%) met the criteria defined by Jass et al for juvenile polyposis syndrome. Seven patients (21%) had a prophylactic gastrectomy due to an extensive gastric polyposis incompatible with endoscopic follow-up, and four patients (13%) developed a digestive cancer.Regarding connective tissue disorders, 20 (61%) had at least one symptom, and 4 (15%) participants who underwent echocardiography had an aortic dilation. CONCLUSION: We describe a large cohort of SMAD4 variant carriers in the context of HHT. Digestive complications are frequent, early and diffuse, justifying endoscopy every 2 years. The HHT phenotype, associating pulmonary and hepatic AVMs, warrants systematic screening. Connective tissue disorders broaden the phenotype associated with SMAD4 gene variants and justify systematic cardiac ultrasound and skeletal complications screening.


Subject(s)
Phenotype , Smad4 Protein , Telangiectasia, Hereditary Hemorrhagic , Humans , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/pathology , Female , Male , Adult , Middle Aged , Intestinal Polyposis/genetics , Intestinal Polyposis/congenital , Intestinal Polyposis/pathology , Heterozygote , Aged , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/diagnosis , Adolescent , Genetic Predisposition to Disease , Mutation/genetics , Young Adult
14.
BMJ Case Rep ; 17(2)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38417936

ABSTRACT

Reed's syndrome (RS) is a rare autosomal-dominant disorder characterised by multiple cutaneous and uterine leiomyomas, with a strong tendency for renal cell carcinoma (RCC) development. A woman in her 50s, who had previously undergone total abdominal hysterectomy due to multiple uterine leiomyomas, presented with painful nodules on her trunk and right arm for the past 6 years. These nodules were confirmed as leiomyomas through histopathology. Diagnosis of RS was established through clinicopathological correlation and positive family history, particularly her mother's. Early-onset uterine leiomyomas in patients with a similar family history should raise suspicion for RS, necessitating vigilant long-term follow-up. RCC detection requires mandatory renal imaging. Screening family members and providing genetic counselling are crucial.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Leiomyomatosis , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms , Female , Humans , Carcinoma, Renal Cell/genetics , Uterine Neoplasms/diagnostic imaging , Uterine Neoplasms/surgery , Leiomyomatosis/diagnostic imaging , Leiomyomatosis/surgery , Skin Neoplasms/diagnosis , Skin Neoplasms/surgery , Skin Neoplasms/genetics , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/surgery , Neoplastic Syndromes, Hereditary/pathology , Kidney Neoplasms/genetics , Fumarate Hydratase/genetics
16.
J Imaging Inform Med ; 37(4): 1674-1682, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38378964

ABSTRACT

For molecular classification of endometrial carcinoma, testing for mismatch repair (MMR) status is becoming a routine process. Mismatch repair deficiency (MMR-D) is caused by loss of expression in one or more of the 4 major MMR proteins: MLH1, MSH2, MSH6, PHS2. Over 30% of patients with endometrial cancer have MMR-D. Determining the MMR status holds significance as individuals with MMR-D are potential candidates for immunotherapy. Pathological whole slide image (WSI) of endometrial cancer with immunohistochemistry results of MMR proteins were gathered. Color normalization was applied to the tiles using a CycleGAN-based network. The WSI was divided into tiles at three different magnifications (2.5 × , 5 × , and 10 ×). Three distinct networks of the same architecture were employed to include features from all three magnification levels and were stacked for ensemble learning. Three architectures, InceptionResNetV2, EfficientNetB2, and EfficientNetB3 were employed and subjected to comparison. The per-tile results were gathered to classify MMR status in the WSI, and prediction accuracy was evaluated using the following performance metrics: AUC, accuracy, sensitivity, and specificity. The EfficientNetB2 was able to make predictions with an AUC of 0.821, highest among the three architectures, and an overall AUC range of 0.767 - 0.821 was reported across the three architectures. In summary, our study successfully predicted MMR classification from pathological WSIs in endometrial cancer through a multi-resolution ensemble learning approach, which holds the potential to facilitate swift decisions on tailored treatment, such as immunotherapy, in clinical settings.


Subject(s)
Endometrial Neoplasms , Humans , Female , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , DNA Mismatch Repair/genetics , Immunohistochemistry/methods , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/diagnosis , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Brain Neoplasms , Colorectal Neoplasms
19.
Mol Genet Genomic Med ; 12(1): e2348, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38146137

ABSTRACT

BACKGROUND: Juvenile Polyposis Syndrome (JPS) is a rare autosomal dominant hereditary disorder characterized by the development of multiple hamartomatous gastrointestinal polyps. Here, we present a case of JPS with a mosaic variant in SMAD4. METHODS: Exome sequencing TRIO analysis, using germline DNA from the biological mother and father along with the index case (IC). RESULTS: A 46-year-old male with no family history of cancer presented with chronic iron deficiency anemia and was diagnosed with massive gastric polyposis (≥100 polyps). At the age of 59, he underwent a total gastrectomy, revealing numerous polyps occupying the entire gastric mucosa, including a 5 cm gastric hyperplastic polyp with high-grade dysplasia and focal adenocarcinoma. TRIO analysis identified the c.386A>C p.(Asn129Thr) variant in the SMAD4 gene at an allele frequency (AF) of 22%, suggesting its mosaic origin. Subsequently, the variant was found in heterozygosity in the IC's son, who exhibited two subcentimeter polyps in the colon and seven inflammatory gastric polyps with gastric inflammatory areas and hyperplasia, suggesting that the c.386A>C p.(Asn129Thr) variant in SMAD4 segregated with the phenotype. CONCLUSION: Our study provides evidence supporting the classification of the c.386A>C p.(Asn129Thr) variant in SMAD4 as a likely pathogenic variant. This finding contributes to improved accuracy in the diagnosis and genetic counseling of JPS.


Subject(s)
Adenomatous Polyps , Intestinal Polyposis/congenital , Neoplastic Syndromes, Hereditary , Stomach Neoplasms , Male , Humans , Middle Aged , Stomach Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Smad4 Protein/genetics
20.
Rev. chil. dermatol ; 35(3): 106-109, 2019. ilus, tab
Article in Spanish | LILACS | ID: biblio-1116411

ABSTRACT

El Tricoepitelioma Múltiple Familiar (TMF) constituye una rara enfermedad autosómica dominante, se caracteriza por la aparición de múltiples pápulas color piel, monomorfas, simétricas, ubicadas en la región central de la cara. El diagnóstico es histopatológico, donde se encuentran tricoepiteliomas, los cuales son neoplasias anexiales benignas que se originan en los folículos pilosos. La condición es de comportamiento indolente, pero con una importante repercusión estética y de difícil manejo. Al ser esta una entidad poco frecuente, el objetivo de este artículo es actualizar los aspectos más relevantes de esta enfermedad. Se presenta el caso de una paciente de 23 años con lesiones faciales típicas en quien se confirmó el diagnostico de TMF


Familial Multiple Trichoepithelioma (FMT) is a rare autosomal dominant disease, characte-rized by the appearance of multiple papules of skin color, monomorphic, symmetrical and located in the central region of the face. The diagnosis is based on histopathological features of trichoepitheliomas, which are benign adnexal neoplasms that originate in the hair follicles. The condition has an indolent behavior but it has an important aesthetic repercussion and it's difficult to treat. As this is a rare entity, the objective of this article is to update the most relevant aspects of this disease. We present the case of a 23 year old patient with typical facial lesions in whom the diagnosis of FMT was confirmed.


Subject(s)
Humans , Female , Young Adult , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Facial Neoplasms/genetics , Facial Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL