Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
J Natl Cancer Inst ; 116(8): 1246-1254, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38569880

ABSTRACT

BACKGROUND: A recent large, well-annotated international cohort of patients with Li-Fraumeni syndrome and early-stage breast cancer was examined for shared features. METHODS: This multicenter cohort study included women with a germline TP53 pathogenic or likely pathogenic variant and nonmetastatic breast cancer diagnosed between 2002 and 2022. Clinical and genetic data were obtained from institutional registries and clinical charts. Descriptive statistics were used to summarize proportions, and differences were assessed using χ2 or Wilcoxon rank sum tests. Metachronous contralateral breast cancer risk, radiation-induced sarcoma risk, and recurrence-free survival were analyzed using the Kaplan-Meier methodology. RESULTS: Among 227 women who met study criteria, the median age of first breast cancer diagnosis was 37 years (range = 21-71), 11.9% presented with bilateral synchronous breast cancer, and 18.1% had ductal carcinoma in situ only. In total, 166 (73.1%) patients underwent mastectomies, including 67 bilateral mastectomies as first breast cancer surgery. Among those patients with retained breast tissue, the contralateral breast cancer rate was 25.3% at 5 years. Among 186 invasive tumors, 72.1% were stages I to II, 48.9% were node negative, and the most common subtypes were hormone receptor-positive/HER2-negative (40.9%) and hormone receptor positive/HER2 positive (34.4%). At a median follow-up of 69.9 months (interquartile range = 32.6-125.9), invasive hormone receptor-positive/HER2-negative disease had the highest recurrence risk among the subtypes (5-year recurrence-free survival = 61.1%, P = .001). Among those who received radiation therapy (n = 79), the 5-year radiation-induced sarcoma rate was 4.8%. CONCLUSION: We observed high rates of ductal carcinoma in situ, hormone receptor-positive, and HER2-positive breast cancers, with a worse outcome in the hormone receptor-positive/HER2-negative luminal tumors, despite appropriate treatment. Confirmation of these findings in further studies could have implications for breast cancer care in those with Li-Fraumeni syndrome.


Subject(s)
Breast Neoplasms , Tumor Suppressor Protein p53 , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Breast Neoplasms/genetics , Breast Neoplasms/epidemiology , Breast Neoplasms/mortality , Middle Aged , Adult , Aged , Tumor Suppressor Protein p53/genetics , Young Adult , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/epidemiology , Li-Fraumeni Syndrome/complications , Germ-Line Mutation , Cohort Studies , Mastectomy , Neoplasm Staging
2.
JCO Precis Oncol ; 8: e2300442, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38394467

ABSTRACT

PURPOSE: The purpose of this article was to provide an overview of syndromic gliomas. DESIGN: The authors conducted a nonsystematic literature review. RESULTS: Cancer predisposition syndromes (CPSs) are genetic conditions that increase one's risk for certain types of cancer compared with the general population. Syndromes that can predispose one to developing gliomas include neurofibromatosis, Li-Fraumeni syndrome, Lynch syndrome, and tuberous sclerosis complex. The standard treatment for sporadic glioma may involve resection, radiation therapy, and/or alkylating chemotherapy. However, DNA-damaging approaches, such as radiation and alkylating agents, may increase the risk of secondary malignancies and other complications in patients with CPSs. In some cases, depending on genetic aberrations, targeted therapies or immunotherapeutic approaches may be considered. Data on clinical characteristics, therapeutic strategies, and prognosis of syndromic gliomas remain limited. CONCLUSION: In this review, we provide an overview of syndromic gliomas with a focus on management for patients with CPSs and the role of novel treatments that can be considered.


Subject(s)
Glioma , Li-Fraumeni Syndrome , Humans , Glioma/genetics , Glioma/therapy , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Prognosis , Genotype
3.
Childs Nerv Syst ; 40(6): 1699-1705, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38316675

ABSTRACT

BACKGROUND: Choroid plexus carcinomas (CPCs) are rare, aggressive grade 3 tumors of the central nervous system associated with Li-Fraumeni syndrome (LFS) in a notable percentage of cases due to TP53 germline mutations. Understanding the correlation between CPCs and LFS is crucial for tailored management strategies. However, distinguishing CPCs from benign choroid plexus papillomas (CPPs) remains challenging, relying largely on histologic features. This study aimed to explore the association between CPCs and LFS, emphasizing the impact of TP53 mutations on diagnosis, treatment, and clinical outcomes. MATERIALS AND METHODS: Scientific databases such as PubMed, Scopus, and Web of Science were systematically searched up to January 2024 using keywords related to CPCs, LFS, TP53 mutation, and central nervous system tumors. Selection criteria included studies investigating the link between CPCs and LFS, their management approaches, and genetic implications of TP53 mutations. Ten relevant studies were selected for analysis after screening titles, abstracts, and full-text articles. Data extraction focused on clinical, genetic, and management factors related to CPCs associated with LFS. RESULTS: The review highlighted the strong association (36%) between CPCs and LFS, primarily due to TP53 germline mutations. Studies emphasized the need for genetic testing in patients with CPCs, especially in pediatric cases, to identify LFS implications. Furthermore, the impact of TP53 mutations on treatment strategies was emphasized, recommending irradiation-sparing therapies due to inferior survival rates associated with radiotherapy in LFS patients with CPCs. Cases illustrated the challenges in diagnosing CPCs and the importance of immunohistochemistry and genetic testing for TP53 mutations. CONCLUSION: CPCs pose challenges in diagnosis and management, particularly in distinguishing them from benign tumors. The association with LFS, often due to TP53 germline mutations, underscores the importance of genetic testing for early detection and tailored treatment strategies. Irradiation-sparing therapies are recommended for LFS-associated CPCs to mitigate the risk of secondary malignancies. Comprehensive profiling of CPC patients, especially in pediatric cases, is crucial for early detection and management of potential secondary cancers associated with LFS.


Subject(s)
Carcinoma , Choroid Plexus Neoplasms , Li-Fraumeni Syndrome , Tumor Suppressor Protein p53 , Female , Humans , Male , Carcinoma/genetics , Carcinoma/therapy , Choroid Plexus Neoplasms/genetics , Choroid Plexus Neoplasms/therapy , Germ-Line Mutation , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/therapy , Li-Fraumeni Syndrome/complications , Mutation , Tumor Suppressor Protein p53/genetics , Child
4.
Am J Med Genet A ; 194(7): e63573, 2024 07.
Article in English | MEDLINE | ID: mdl-38410872

ABSTRACT

Li-Fraumeni syndrome (LFS) is a rare syndrome characterized by an increased lifetime risk of cancer development in multiple organ systems, typically caused by de novo or inherited germline pathogenic variants in the tumor suppressor TP53 gene. LFS is more classically associated with solid tumors; however, it is also associated with hematologic malignancies such as therapy-related acute myeloid leukemia (AML). We present the case of a female patient with a strong family and personal history of cancer who presented to our institution with therapy-related AML with next-generation sequencing showing a pathogenic TP53 mutation. She received several lines of systemic therapy and underwent stem cell transplant using her adult daughter as a haploidentical donor after achieving minimal residual disease (MRD). Her posttransplant bone marrow evaluations demonstrated persistence of the same pathogenic TP53 mutation despite ongoing clinical remission with full donor engraftment and negative MRD. Genetic testing was performed which confirmed the germline origin of the TP53 pathogenic variant in the patient. The patient's adult donor daughter was also identified to have the same pathogenic variant in TP53 consistent with LFS. The presented case highlights the need for increased awareness of LFS in the adult hematologic community, particularly for patients undergoing evaluation for stem cell transplant.


Subject(s)
Li-Fraumeni Syndrome , Tumor Suppressor Protein p53 , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/complications , Female , Tumor Suppressor Protein p53/genetics , Adult , Germ-Line Mutation/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/complications , Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , Hematologic Neoplasms/pathology , Pedigree , Mutation , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing
5.
Diagn Pathol ; 19(1): 16, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38243242

ABSTRACT

BACKGROUND: Epithelioid angiomyolipoma (EAML) is a rare variant of angiomyolipoma that predominantly consists of epithelioid cells and belongs to the perivascular epithelioid cell neoplasm (PEComa) family. The majority of EAMLs arise in the kidneys, and primary hepatic EAML appears to be much less common than renal EAML. Most PEComas arise sporadically, but may be associated with tuberous sclerosis complex (TSC), an autosomal dominant genetic disorder characterized by germline mutations in the TSC1 or TSC2 genes. However, PEComas have previously been reported in five patients with Li-Fraumeni syndrome (LFS), which is an inherited cancer susceptibility disorder resulting from germline mutations in the TP53 tumor suppressor gene. CASE PRESENTATION: We report a 49-year-old female patient with hepatic EAML and pancreatic cancer. Because she had previously been diagnosed with bilateral breast cancer at the age of 30, we performed a comprehensive genetic analysis to identify genetic alterations associated with any cancer predisposition syndrome. Whole-exome sequencing of a blood sample identified a heterozygous germline variant of TP53 (NM_000546.5):c.708C>A, and targeted next-generation sequencing of liver EAML and pancreatic cancer tissue samples demonstrated the same TP53 (NM_000546.5):c.708C>A variant in both. This, plus the patient's history of early-onset breast cancer, met the 2015 version of the Chompret criteria for diagnosis of LFS. CONCLUSIONS: There have been very few case reports regarding the presence of PEComa in LFS, and to the best of our knowledge, this is the first report of EAML of the liver in a patient with LFS.


Subject(s)
Angiomyolipoma , Breast Neoplasms , Kidney Neoplasms , Li-Fraumeni Syndrome , Liver Neoplasms , Pancreatic Neoplasms , Female , Humans , Middle Aged , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Angiomyolipoma/diagnosis , Angiomyolipoma/genetics , Angiomyolipoma/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Kidney Neoplasms/pathology , Genetic Predisposition to Disease
6.
Clin Gastroenterol Hepatol ; 22(3): 662-665.e1, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37714395

ABSTRACT

Li-Fraumeni syndrome (LFS), linked to heterozygous germline pathogenic/likely pathogenic variants in TP53, confers exceptionally high cancer risk, including core cancers (sarcoma, breast, adrenocortical, and brain cancer) among many other cancer types.1 Colorectal cancer (CRC) is most common after the core and hematologic cancers, accounting for ∼2.8% of diagnoses. Stomach and esophageal cancers constitute another 1.3% (TP53 Database; R20, July 2019: https://tp53.isb-cgc.org).2.


Subject(s)
Gastrointestinal Neoplasms , Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/etiology
7.
Pediatr Dermatol ; 41(2): 311-314, 2024.
Article in English | MEDLINE | ID: mdl-38014598

ABSTRACT

Pleomorphic dermal sarcoma (PDS) is an uncommon malignant soft-tissue tumor that occurs mostly in elderly patients, with only 5% of cases occurring in children. However, pediatric patients with Li-Fraumeni syndrome (LFS) can develop several types of cancer, particularly sarcomas. Here, we describe a young LFS patient who presented with early-onset PDS and review the literature.


Subject(s)
Histiocytoma, Malignant Fibrous , Li-Fraumeni Syndrome , Sarcoma , Skin Neoplasms , Soft Tissue Neoplasms , Child , Humans , Aged , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Sarcoma/complications , Sarcoma/diagnosis , Skin Neoplasms/complications , Skin Neoplasms/diagnosis , Genetic Predisposition to Disease
8.
Cell Death Dis ; 14(11): 783, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38030599

ABSTRACT

Li-Fraumeni syndrome (LFS) is characterized by germline mutations occurring on one allele of genome guardian TP53. It is a severe cancer predisposition syndrome with a poor prognosis, partly due to the frequent development of subsequent primary tumors following DNA-damaging therapies. Here we explored, for the first time, the effectiveness of mutant p53 rescue compound in treating LFS-mimicking mice harboring a deleterious p53 mutation. Among the ten p53 hotspot mutations in IARC LFS cohorts, R282W is one of the mutations predicting the poorest survival prognosis and the earliest tumor onset. Among the six clinical-stage mutant p53 rescue compounds, arsenic trioxide (ATO) effectively restored transactivation activity to p53-R282W. We thus constructed a heterozygous Trp53 R279W (corresponding to human R282W) mouse model for the ATO treatment study. The p53R279W/+ (W/+) mice exhibited tumor onset and overall survival well mimicking the ones of human LFS. Further, 35 mg/L ATO addition in drink water significantly extended the median survival of W/+ mice (from 460 to 596 days, hazard ratio = 0.4003, P = 0.0008). In the isolated tumors from ATO-treated W/+ mice, the representative p53 targets including Cdkn1a, Mdm2, and Tigar were significantly upregulated, accompanying with a decreased level of the proliferation marker Ki67 and increased level of apoptosis marker TUNEL. Together, the non-genotoxic treatment of p53 rescue compound ATO holds promise as an alternative for LFS therapeutic.


Subject(s)
Li-Fraumeni Syndrome , Humans , Animals , Mice , Li-Fraumeni Syndrome/drug therapy , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/complications , Tumor Suppressor Protein p53/genetics , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Genetic Predisposition to Disease , Genes, p53
9.
BMC Womens Health ; 23(1): 329, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37344881

ABSTRACT

BACKGROUND: Li-Fraumeni syndrome (LFS) is a rare autosomal dominant disease with high penetrance caused by a germline variant of TP53 gene. We report the first case of endometrial cancer after yolk sac tumor with LFS. CASE PRESENTATION: The presented female patient underwent right adnexectomy at age 23 because of a yolk sac tumor of the ovary. At the age of 27, the patient was diagnosed with endometrial adenocarcinoma, received cytoreductive surgery and chemotherapy. Given that her personal cancer history along with a strong family history of cancer, her father passing away from lung cancer at age 48 and her grandmother dying of ovarian cancer at age 50, the patient was referred for genetic counseling and testing. Genetic screening revealed a heterozygous pathogenic TP53 c.844C > T, p.( R282 W) with NM_000546.5 variant, a class 5 (C5) variant. This is the first reported case of a yolk sac tumor accompanied by subsequent endometrial cancer that is associated with LFS. CONCLUSIONS: We reported a first case of an endometrial cancer after yolk sac tumor patient with a tumor family history of harboring the germline TP53 pathogenic variation which expanded types of tumor that can be presented in patients with LFS. This case highlights the importance of genetic testing for patients with malignant tumors, as well as patients with a family history of malignant tumors. And our case highlights the necessity of screening for gynecologic tumor in LFS patients.


Subject(s)
Endodermal Sinus Tumor , Endometrial Neoplasms , Li-Fraumeni Syndrome , Female , Humans , Young Adult , Adult , Middle Aged , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/diagnosis , Genes, p53 , Endodermal Sinus Tumor/complications , Endodermal Sinus Tumor/genetics , Endometrial Neoplasms/complications , Endometrial Neoplasms/genetics , Germ-Line Mutation , Genetic Predisposition to Disease
10.
Neuro Oncol ; 25(12): 2273-2286, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37379234

ABSTRACT

BACKGROUND: The prognosis for Li-Fraumeni syndrome (LFS) patients with medulloblastoma (MB) is poor. Comprehensive clinical data for this patient group is lacking, challenging the development of novel therapeutic strategies. Here, we present clinical and molecular data on a retrospective cohort of pediatric LFS MB patients. METHODS: In this multinational, multicenter retrospective cohort study, LFS patients under 21 years with MB and class 5 or class 4 constitutional TP53 variants were included. TP53 mutation status, methylation subgroup, treatment, progression free- (PFS) and overall survival (OS), recurrence patterns, and incidence of subsequent neoplasms were evaluated. RESULTS: The study evaluated 47 LFS individuals diagnosed with MB, mainly classified as DNA methylation subgroup "SHH_3" (86%). The majority (74%) of constitutional TP53 variants represented missense variants. The 2- and 5-year (y-) PFS were 36% and 20%, and 2- and 5y-OS were 53% and 23%, respectively. Patients who received postoperative radiotherapy (RT) (2y-PFS: 44%, 2y-OS: 60%) or chemotherapy before RT (2y-PFS: 32%, 2y-OS: 48%) had significantly better clinical outcome then patients who were not treated with RT (2y-PFS: 0%, 2y-OS: 25%). Patients treated according to protocols including high-intensity chemotherapy and patients who received only maintenance-type chemotherapy showed similar outcomes (2y-PFS: 42% and 35%, 2y-OS: 68% and 53%, respectively). CONCLUSIONS: LFS MB patients have a dismal prognosis. In the presented cohort use of RT significantly increased survival rates, whereas chemotherapy intensity did not influence their clinical outcome. Prospective collection of clinical data and development of novel treatments are required to improve the outcome of LFS MB patients.


Subject(s)
Cerebellar Neoplasms , Li-Fraumeni Syndrome , Medulloblastoma , Child , Humans , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/therapy , Medulloblastoma/therapy , Medulloblastoma/drug therapy , Retrospective Studies , Prospective Studies , Cerebellar Neoplasms/therapy , Cerebellar Neoplasms/drug therapy , Germ-Line Mutation , Tumor Suppressor Protein p53/genetics
11.
J Med Case Rep ; 17(1): 195, 2023 May 14.
Article in English | MEDLINE | ID: mdl-37179382

ABSTRACT

INTRODUCTION: Pure androgen-secreting adrenocortical tumors are a rare but important cause of peripheral precocious puberty. CASE PRESENTATION: Here, we report a pure androgen-secreting adrenocortical tumor in a 2.5-year-old boy presenting with penile enlargement, pubic hair, frequent erections, and rapid linear growth. We confirmed the diagnosis through laboratory tests, medical imaging, and histology. Furthermore, genetic testing detected a pathogenic germline variant in the TP53 gene, molecularly confirming underlying Li-Fraumeni syndrome. DISCUSSION: Only 15 well-documented cases of pure androgen-secreting adrenocortical tumors have been reported so far. No clinical or imaging signs were identified to differentiate adenomas from carcinomas, and no other cases of Li-Fraumeni syndrome were diagnosed in the four patients that underwent genetic testing. However, diagnosing Li-Fraumeni syndrome is important as it implies a need for intensive tumor surveillance and avoidance of ionizing radiation. CONCLUSION: In this article, we emphasize the need to screen for TP53 gene variants in children with androgen-producing adrenal adenomas and report an association with arterial hypertension.


Subject(s)
Adrenal Cortex Neoplasms , Li-Fraumeni Syndrome , Puberty, Precocious , Male , Child , Humans , Child, Preschool , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Genes, p53 , Androgens , Puberty, Precocious/etiology , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/genetics
13.
Breast ; 68: 157-162, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36773404

ABSTRACT

INTRODUCTION: Breast cancer (BC) is the most common type of cancer in premenopausal women with germline TP53 pathogenic variants (mTP53) (Li Fraumeni syndrome - LFS). However, little is known about the BC prognosis in these patients. This study analyzed the BC-related oncologic outcomes of patients with LFS. METHODS: We evaluated a cohort of LFS patients with BC in comparison with a control cohort of BC patients with no pathogenic variant in a hereditary cancer panel. The primary endpoint was recurrence-free survival (RFS). Due to the risk of second malignancies in LFS, only locoregional and distant recurrences were considered events for RFS. Secondary endpoints included rates of contralateral BC, overall survival (OS), and breast cancer-specific survival (BCSS). RESULTS: Forty-one patients were evaluated in the mTP53 group and 82 in the control group. Median age at BC diagnosis was 40 and 41 years, respectively. The mTP53 group received less adjuvant radiotherapy than the control group (63.4% vs 93.9%, P < 0.001). Other relevant baseline characteristics and treatment received were similar between groups. 5y-RFS rates were 79.4% in the mTP53 versus 93.6% in the control group (HR 2.43, 95%CI 0.74-8.01, P = 0.143); and were not impacted by the use of adjuvant radiotherapy. 5y-BCSS rates were 92.2% and 98.6%, respectively (HR 1.87, IC95% 0.25-13.48, P = 0.534). CONCLUSIONS: Our results showed no statistically significant difference in BC-related RFS and BCSS between patients with mTP53 and a control group with no pathogenic variant. Larger multicentric studies are warranted to confirm these results.


Subject(s)
Breast Neoplasms , Li-Fraumeni Syndrome , Humans , Female , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Breast Neoplasms/diagnosis , Case-Control Studies , Tumor Suppressor Protein p53/genetics , Germ-Line Mutation , Prognosis , Genetic Predisposition to Disease
15.
J Neurosurg Pediatr ; 31(3): 258-267, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36609372

ABSTRACT

OBJECTIVE: Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome caused by germline mutations in the TP53 gene. CNS tumors are the fourth most common tumor type in LFS, and recent screening guidelines demonstrate that early tumor detection is associated with improved long-term survival. However, there is a paucity of data regarding surgical intervention when lesions are identified in asymptomatic patients on surveillance imaging. The authors investigated this through their cohort and literature review. METHODS: The cohort consisted of children seen in the Pediatric Cancer Genetics Program at Children's National Hospital between August 2012 and August 2021. The authors also include a PubMed (MEDLINE) literature search of articles from 2006 to 2021 related to surveillance and CNS tumors in patients with LFS. Studies in which CNS tumors were not identified or detailed patient information was not provided were excluded. Patients from the selected articles and the authors' cohort were added for further analysis. RESULTS: Between August 2012 and August 2021, 10 children with LFS and CNS tumors were assessed at Children's National Hospital: 4 who were known carriers of the TP53 mutation had CNS lesions found on surveillance imaging, whereas 6 presented with symptomatic CNS lesions and were either known or subsequently found to have germline TP53 mutations. The literature search identified 148 articles, 7 of which were included in this review. Patients from the literature and the present cohort were added for a total of 56 CNS lesions. A majority of the low-grade CNS lesions (22/24, 92%) were found on surveillance protocols in asymptomatic patients, whereas the majority of the high-grade lesions (22/26, 85%) presented in symptomatic patients who were not undergoing routine surveillance or as the initial diagnosis of LFS. The authors noted a significant survival advantage in pediatric patients with low-grade lesions, with an overall survival of 100% at 30 months. Minor limitations of the study include patient sample size and limitations in the patient cohort due to this being a retrospective rather than a prospective study. CONCLUSIONS: Data presented in this study support surveillance protocols in LFS and demonstrate the importance of dedicated CNS imaging and early surgical intervention when lesions are identified. Systematic review registration no.: CRD42022372610 (www.crd.york.ac.uk/prospero).


Subject(s)
Central Nervous System Neoplasms , Li-Fraumeni Syndrome , Child , Humans , Central Nervous System Neoplasms/diagnostic imaging , Central Nervous System Neoplasms/surgery , Genes, p53 , Genetic Predisposition to Disease , Hospitals , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/pathology , Li-Fraumeni Syndrome/surgery , Retrospective Studies , Tumor Suppressor Protein p53/genetics
17.
Am Surg ; 89(11): 4958-4960, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36420590

ABSTRACT

Li-Fraumeni syndrome (LFS) is associated with many different cancers, including early onset breast cancer. Due to an increased risk of radiation-induced malignancy, radiation therapy is often avoided in this patient population. This case study evaluates a 38-year-old female with a history of juvenile granulosa cell tumor of the ovary and malignant phyllodes tumor of right breast, who subsequently developed bilateral invasive ductal carcinoma and was treated with bilateral mastectomies. Studies show that in a high-risk patient, post-mastectomy radiation therapy (PMRT) should not be ruled out due to a history of LFS, as the benefit of PMRT may outweigh the risk of a radiation-induced malignancy.


Subject(s)
Breast Neoplasms , Li-Fraumeni Syndrome , Neoplasms, Radiation-Induced , Female , Humans , Adult , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/surgery , Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Breast Neoplasms/pathology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/surgery , Mastectomy/adverse effects
18.
Diagn Pathol ; 17(1): 93, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36514176

ABSTRACT

BACKGROUND: Germline TP53 mutations have been frequently reported in patients with Li-Fraumeni syndrome (LFS), resulting in a predisposition to various malignancies. Mutations other than germline TP53 mutations can also cause LFS-associated malignancies, but their details remain unclear. We describe a novel c-myc amplification in a unique liposarcoma in a patient with LFS. CASE PRESENTATION: A female patient with LFS developed breast cancer twice at the age of thirty; both were invasive ductal carcinomas harboring HER2 amplifications. Computed tomography revealed an anterior mediastinal mass, which was surgically resected. Histological analysis revealed three different lesions corresponding to myxoid liposarcoma-, pleomorphic liposarcoma-, and well-differentiated liposarcoma-like lesions. Fluorescence in-situ hybridization (FISH) analysis did not detect MDM2 amplification, Rb1 deletion, break apart signals of EWS, FUS, DDIT3, or c-myc, or c-myc-IGH fusion signals, but it did detect more c-myc signals. Further FISH analysis and comprehensive genomic profiling revealed c-myc amplification. We considered two differential diagnoses, dedifferentiated liposarcoma lacking MDM2 amplification and myxoid pleomorphic liposarcoma (MPLPS), and determined that this case is most likely MPLPS. However, definite diagnosis could not be made because a clear-cut differentiation of the case from liposarcomas was not possible. CONCLUSIONS: A previous study demonstrated that c-myc amplification could not be detected in various liposarcomas, but the present unique liposarcoma showed c-myc amplification, so the c-myc amplification may indicate that the present liposarcoma is an LFS-related tumor. The present case further clarifies the pathological features of MPLPS and LFS-related liposarcomas by broadening their histopathological and genetic diversities.


Subject(s)
Li-Fraumeni Syndrome , Lipoma , Liposarcoma, Myxoid , Liposarcoma , Female , Humans , Adult , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Liposarcoma/diagnosis , Liposarcoma/genetics , Liposarcoma/pathology , Liposarcoma, Myxoid/diagnosis , Liposarcoma, Myxoid/genetics , Liposarcoma, Myxoid/pathology , Lipoma/pathology , In Situ Hybridization, Fluorescence , Genomics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/analysis
19.
J Med Case Rep ; 16(1): 468, 2022 Dec 19.
Article in English | MEDLINE | ID: mdl-36529791

ABSTRACT

BACKGROUND: Leiomyosarcoma is a rare malignant tumor of smooth muscle origin and represents 10-20% of all soft tissue sarcomas. Primary colon and rectal sarcomas constitute < 0.1% of all large bowel malignancies. In Li-Fraumeni syndrome, sarcomas are the second most frequent cancer (25%). Li-Fraumeni syndrome is a genetic disease with a familial predisposition to multiple malignant neoplasms. This syndrome has an autosomal dominant pattern of inheritance and high penetrance characterized by germline TP53 mutations. Patients with a history of cancer who do not meet all the "classic" criteria for Li-Fraumeni syndrome are considered to have Li-Fraumeni-like syndrome. To the best of our knowledge, this article is the first report of a patient with rectal leiomyosarcoma as the initial phenotypic manifestation of Li-Fraumeni-like syndrome. The authors also present a literature review. CASE PRESENTATION: A 67-year-old Brazilian woman underwent anterior rectosigmoidectomy and panhysterectomy secondary to rectal leiomyosarcoma. She subsequently developed carcinomatosis and died 2 years after the operation. Her family medical history consisted of a daughter who died at 32 years of age from breast cancer, a granddaughter diagnosed with adrenocortical carcinoma at 6 years of age and two siblings who died from prostate cancer. A genetic study was carried out to identify a pathogenic variant of Li-Fraumeni syndrome. In the DNA extracted from the peripheral blood leukocyte, restriction fragment length polymorphism was analyzed to search for mutations in the TP53 gene. The DNA sequencing identified the germline pathogenic variant p. R337H heterozygous in exon 10 of TP53. The patient was classified as having Li-Fraumeni-like syndrome. CONCLUSION: In patients with rectal leiomyosarcoma, it is advisable to investigate the family history of cancer and perform genetic studies to screen for Li-Fraumeni syndrome.


Subject(s)
Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Leiomyosarcoma , Li-Fraumeni Syndrome , Pelvic Neoplasms , Rectal Neoplasms , Male , Female , Humans , Child , Aged , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Leiomyosarcoma/diagnosis , Leiomyosarcoma/genetics , Leiomyosarcoma/surgery , Tumor Suppressor Protein p53/genetics , Rectal Neoplasms/diagnosis , Rectal Neoplasms/genetics , Rectal Neoplasms/surgery , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/genetics , Genetic Predisposition to Disease
20.
Mol Oncol ; 16(19): 3554-3567, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35981147

ABSTRACT

In humans, germline TP53 mutations predispose carriers to a wide spectrum of cancers, which is known as Li-Fraumeni syndrome (LFS). To date, the association of melanomas with LFS remains unestablished. No melanomas have been reported in any P53-modified mouse models either. In this study, we show that targeted disruption of P53 at the DNA-binding domain in Xenopus tropicalis recapitulates LFS, with the formation of soft-tissue sarcomas and pancreatic ductal adenocarcinoma. Interestingly, 19% of the 14-month-old tp53Δ7/Δ7 homozygotes and 18% of tp53+/Δ7 heterozygotes spontaneously developed small nevi and non-invasive melanomas. Large invasive melanomas were also observed in other older homozygous mutants, with about 7.9% penetrance. Our data suggest that more dermatologic investigation of LFS patients should be able to settle the association of melanoma with LFS in epidemiology. Our model is also valuable for further investigation of the molecular mechanism underlying melanoma progression upon germline alteration of the tp53 locus.


Subject(s)
Li-Fraumeni Syndrome , Nevus , Skin Neoplasms , Animals , DNA , Genetic Predisposition to Disease , Germ-Line Mutation/genetics , Humans , Infant , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Mice , Nevus/complications , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Xenopus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL