Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.011
Filter
3.
Pediatr Blood Cancer ; 71(8): e31077, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38783403

ABSTRACT

BACKGROUND: Li-Fraumeni syndrome (LFS) is a penetrant cancer predisposition syndrome (CPS) associated with the development of many tumor types in young people including osteosarcoma and breast cancer (BC). The McGill Interactive Pediatric OncoGenetic Guidelines (MIPOGG) decision-support tool provides a standardized approach to identify patients at risk of CPSs. METHODS: We conducted a cost-utility analysis, from the healthcare payer perspective, to compare MIPOGG-guided, physician-guided, and universal genetic testing strategies to detect LFS in female patients diagnosed at an age of less than 18 years with osteosarcoma. We developed a decision tree and discrete-event simulation model to simulate the clinical and cost outcomes of the three genetic referral strategies on a cohort of female children diagnosed with osteosarcoma, especially focused on BC as subsequent cancer. Outcomes included BC incidence, quality-adjusted life-years (QALYs), healthcare costs, and incremental cost-utility ratios (ICURs). We conducted probabilistic and scenario analyses to assess the uncertainty surrounding model parameters. RESULTS: Compared to the physician-guided testing, the MIPOGG-guided strategy was marginally more expensive by $105 (-$516; $743), but slightly more effective by 0.003 (-0.04; 0.045) QALYs. Compared to MIPOGG, the universal testing strategy was $1333 ($732; $1953) more costly and associated with 0.011 (-0.043; 0.064) additional QALYs. The ICUR for the MIPOGG strategy was $33,947/QALY when compared to the physician strategy; the ICUR for universal testing strategy was $118,631/QALY when compared to the MIPOGG strategy. DISCUSSION: This study provides evidence for clinical and policy decision-making on the cost-effectiveness of genetic referral strategies to identify LFS in the setting of osteosarcoma. MIPOGG-guided strategy was most likely to be cost-effective at a willingness-to-pay threshold value of $50,000/QALY.


Subject(s)
Cost-Benefit Analysis , Li-Fraumeni Syndrome , Osteosarcoma , Humans , Female , Osteosarcoma/economics , Osteosarcoma/genetics , Osteosarcoma/diagnosis , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/economics , Child , Adolescent , Bone Neoplasms/genetics , Bone Neoplasms/economics , Genetic Testing/economics , Genetic Testing/methods , Practice Guidelines as Topic , Quality-Adjusted Life Years , Breast Neoplasms/genetics , Breast Neoplasms/economics
4.
PLoS Genet ; 20(5): e1011290, 2024 May.
Article in English | MEDLINE | ID: mdl-38805544

ABSTRACT

Li-Fraumeni syndrome is caused by inherited TP53 tumor suppressor gene mutations. MicroRNA miR-34a is a p53 target and modifier gene. Interestingly, miR-34 triple-null mice exhibit normal p53 responses and no overt cancer development, but the lack of miR-34 promotes tumorigenesis in cancer-susceptible backgrounds. miR-34 genes are highly conserved and syntenic between zebrafish and humans. Zebrafish miR-34a and miR-34b/c have similar expression timing in development, but miR-34a is more abundant. DNA damage by camptothecin led to p53-dependent induction of miR-34 genes, while miR-34a mutants were adult-viable and had normal DNA damage-induced apoptosis. Nevertheless, miR-34a-/- compound mutants with a gain-of-function tp53R217H/ R217H or tp53-/- mutants were more cancer-prone than tp53 mutants alone, confirming the tumor-suppressive function of miR-34a. Through transcriptomic comparisons at 28 hours post-fertilization (hpf), we characterized DNA damage-induced transcription, and at 8, 28 and 72 hpf we determined potential miR-34a-regulated genes. At 72 hpf, loss of miR-34a enhanced erythrocyte levels and up-regulated myb-positive hematopoietic stem cells. Overexpression of miR-34a suppressed its reporter mRNA, but not p53 target induction, and sensitized injected embryos to camptothecin but not to γ-irradiation.


Subject(s)
DNA Damage , Hematopoiesis , MicroRNAs , Tumor Suppressor Protein p53 , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Hematopoiesis/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Humans , Li-Fraumeni Syndrome/genetics , Gene Expression Regulation, Developmental , Apoptosis/genetics , Mice , Genes, Tumor Suppressor , Camptothecin/pharmacology
5.
Cancer Genet ; 284-285: 43-47, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677009

ABSTRACT

BACKGROUND: Germline heterozygous TP53 pathogenic variants (PVs) cause Li Fraumeni Syndrome (LFS, OMIM#151623). TP53 PVs at lower-than-expected variant allele frequencies (VAF) may reflect postzygotic mosaicism (PZM) or clonal hematopoiesis (CH); however, no guidelines exist for workup and clinical management. PATIENTS AND METHODS: Retrospective analysis of probands who presented to an academic cancer genetics program with a TP53 PV result on germline genetic testing. RESULTS: Twenty-one of 125 unrelated probands (17 %) were found to harbor a TP53 PV with VAF<30 % or a designation of "mosaic". A diagnosis of PZM was made in nine (43 %) due to a clinical phenotype consistent with LFS with (n = 8) or without (n = 1) positive ancillary tissue testing. Twelve patients (57 %) were diagnosed with presumed CH (pCH) due to a diagnosis of a myeloproliferative neoplasm, negative ancillary tissue testing, clinical phenotype not meeting LFS criteria, no cancer, and/or no first cancer age<50. Of the 19 patients with biological offspring, nine had either partial or complete offspring testing, all negative. CONCLUSIONS: Determining the etiology of low VAF TP53 PVs requires ancillary tissue testing and incorporation of clinical phenotype. Discerning PZM versus CH is important to provide optimal care and follow-up.


Subject(s)
Genetic Testing , Germ-Line Mutation , Li-Fraumeni Syndrome , Mosaicism , Tumor Suppressor Protein p53 , Humans , Genetic Testing/methods , Tumor Suppressor Protein p53/genetics , Female , Male , Li-Fraumeni Syndrome/genetics , Retrospective Studies , Adult , Middle Aged , Young Adult , Adolescent
6.
J Clin Oncol ; 42(18): 2186-2195, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38569124

ABSTRACT

PURPOSE: There exists a barrier between developing and disseminating risk prediction models in clinical settings. We hypothesize that this barrier may be lifted by demonstrating the utility of these models using incomplete data that are collected in real clinical sessions, as compared with the commonly used research cohorts that are meticulously collected. MATERIALS AND METHODS: Genetic counselors (GCs) collect family history when patients (ie, probands) come to MD Anderson Cancer Center for risk assessment of Li-Fraumeni syndrome, a genetic disorder characterized by deleterious germline mutations in the TP53 gene. Our clinical counseling-based (CCB) cohort consists of 3,297 individuals across 124 families (522 cases of single primary cancer and 125 cases of multiple primary cancers). We applied our software suite LFSPRO to make risk predictions and assessed performance in discrimination using AUC and in calibration using observed/expected (O/E) ratio. RESULTS: For prediction of deleterious TP53 mutations, we achieved an AUC of 0.78 (95% CI, 0.71 to 0.85) and an O/E ratio of 1.66 (95% CI, 1.53 to 1.80). Using the LFSPRO.MPC model to predict the onset of the second cancer, we obtained an AUC of 0.70 (95% CI, 0.58 to 0.82). Using the LFSPRO.CS model to predict the onset of different cancer types as the first primary, we achieved AUCs between 0.70 and 0.83 for sarcoma, breast cancer, or other cancers combined. CONCLUSION: We describe a study that fills in the critical gap in knowledge for the utility of risk prediction models. Using a CCB cohort, our previously validated models have demonstrated good performance and outperformed the standard clinical criteria. Our study suggests that better risk counseling may be achieved by GCs using these already-developed mathematical models.


Subject(s)
Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/genetics , Risk Assessment , Female , Male , Neoplasms, Multiple Primary/genetics , Tumor Suppressor Protein p53/genetics , Germ-Line Mutation , Genetic Counseling , Adult , Genetic Predisposition to Disease , Genes, p53 , Middle Aged
7.
Acta Neuropathol Commun ; 12(1): 57, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605367

ABSTRACT

Li-Fraumeni syndrome (LFS) is an autosomal dominant tumor predisposition syndrome caused by heterozygous germline mutations or deletions in the TP53 tumor suppressor gene. Central nervous system tumors, such as choroid plexus tumors, medulloblastomas, and diffuse gliomas, are frequently found in patients with LFS. Although molecular profiles of diffuse gliomas that develop in pediatric patients with LFS have been elucidated, those in adults are limited. Recently, diffuse gliomas have been divided into pediatric- and adult-type gliomas, based on their distinct molecular profiles. In the present study, we investigated the molecular profiles of high-grade gliomas in three adults with LFS. These tumors revealed characteristic histopathological findings of high-grade glioma or glioblastoma and harbored wild-type IDH1/2 according to whole exome sequencing (WES). However, these tumors did not exhibit the key molecular alterations of glioblastoma, IDH-wildtype such as TERT promoter mutation, EGFR amplification, or chromosome 7 gain and 10 loss. Although WES revealed no other characteristic gene mutations or copy number alterations in high-grade gliomas, such as those in histone H3 genes, PDGFRA amplification was found in all three cases together with uniparental disomy of chromosome 17p, where the TP53 gene is located. DNA methylation analyses revealed that all tumors exhibited DNA methylation profiles similar to those of pediatric-type high-grade glioma H3-wildtype and IDH-wildtype (pHGG H3-/IDH-wt), RTK1 subtype. These data suggest that high-grade gliomas developed in adult patients with LFS may be involved in pHGG H3-/IDH-wt. PDGFRA and homozygous alterations in TP53 may play pivotal roles in the development of this type of glioma in adult patients with LFS.


Subject(s)
Brain Neoplasms , Glioma , Li-Fraumeni Syndrome , Adult , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, p53 , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Li-Fraumeni Syndrome/genetics , Mutation/genetics
8.
J Pediatr Health Care ; 38(2): 270-274, 2024.
Article in English | MEDLINE | ID: mdl-38429040

ABSTRACT

The number of youths who identify with a gender different from their sex designated at birth is increasing. Youth account for about 4% of all cancer diagnoses in the United States. Some youths may want gender-affirming medical treatment, such as puberty blockers and/or hormone therapy, which may exacerbate cancer and/or increase cancer development risk. No studies assess the impact of estrogen-based treatment in gender-diverse youth with a history of Li-Fraumeni syndrome. This case report will discuss gender-affirming care and shared decision-making in a youth with a history of Li-Fraumeni syndrome and increased risk for breast cancer.


Subject(s)
Li-Fraumeni Syndrome , Transgender Persons , Infant, Newborn , Female , Adolescent , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/therapy , Gender-Affirming Care , Puberty Inhibitors
9.
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
10.
JCO Clin Cancer Inform ; 8: e2300167, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38346271

ABSTRACT

PURPOSE: LFSPRO is an R library that implements risk prediction models for Li-Fraumeni syndrome (LFS), a genetic disorder characterized by deleterious germline mutations in the TP53 gene. To facilitate the use of these models in clinics, we developed LFSPROShiny, an interactive R/Shiny interface of LFSPRO that allows genetic counselors (GCs) to perform risk predictions without any programming components and further visualize the risk profiles of their patients to aid the decision-making process. METHODS: LFSPROShiny implements two models that have been validated on multiple LFS patient cohorts: a competing risk model that predicts cancer-specific risks for the first primary and a recurrent-event model that predicts the risk of a second primary tumor. Starting with a visualization template, we keep regular contact with GCs, who ran LFSPROShiny in their counseling sessions, to collect feedback and discuss potential improvement. On receiving the family history as input, LFSPROShiny renders the family into a pedigree and displays the risk estimates of the family members in a tabular format. The software offers interactive overlaid side-by-side bar charts for visualization of the patients' cancer risks relative to the general population. RESULTS: We walk through a detailed example to illustrate how GCs can run LFSPROShiny in clinics from data preparation to downstream analyses and interpretation of results with an emphasis on the utilities that LFSPROShiny provides to aid decision making. CONCLUSION: Since December 2021, we have applied LFSPROShiny to over 100 families from counseling sessions at the MD Anderson Cancer Center. Our study suggests that software tools with easy-to-use interfaces are crucial for the dissemination of risk prediction models in clinical settings, hence serving as a guideline for future development of similar models.


Subject(s)
Li-Fraumeni Syndrome , Mobile Applications , Tumor Suppressor Protein p53 , Humans , Genetic Predisposition to Disease , Germ Cells , Germ-Line Mutation , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/epidemiology , Tumor Suppressor Protein p53/genetics
11.
Trials ; 25(1): 103, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38308321

ABSTRACT

BACKGROUND: Li-Fraumeni syndrome (LFS) is a rare autosomal dominant disease caused by inherited or de novo germline pathogenic variants in TP53. Individuals with LFS have a 70-100% lifetime risk of developing cancer. The current standard of care involves annual surveillance with whole-body and brain MRI (WB-MRI) and clinical review; however, there are no chemoprevention agents licensed for individuals with LFS. Preclinical studies in LFS murine models show that the anti-diabetic drug metformin is chemopreventive and, in a pilot intervention trial, short-term use of metformin was well-tolerated in adults with LFS. However, metformin's mechanism of anticancer activity in this context is unclear. METHODS: Metformin in adults with Li-Fraumeni syndrome (MILI) is a Precision-Prevention phase II open-labelled unblinded randomised clinical trial in which 224 adults aged ≥ 16 years with LFS are randomised 1:1 to oral metformin (up to 2 mg daily) plus annual MRI surveillance or annual MRI surveillance alone for up to 5 years. The primary endpoint is to compare cumulative cancer-free survival up to 5 years (60 months) from randomisation between the intervention (metformin) and control (no metformin) arms. Secondary endpoints include a comparison of cumulative tumour-free survival at 5 years, overall survival at 5 years and clinical characteristics of emerging cancers between trial arms. Safety, toxicity and acceptability of metformin; impact of metformin on quality of life; and impact of baseline lifestyle risk factors on cancer incidence will be assessed. Exploratory end-points will evaluate the mechanism of action of metformin as a cancer preventative, identify biomarkers of response or carcinogenesis and assess WB-MRI performance as a diagnostic tool for detecting cancers in participants with LFS by assessing yield and diagnostic accuracy of WB-MRI. DISCUSSION: Alongside a parallel MILI study being conducted by collaborators at the National Cancer Institute (NCI), MILI is the first prevention trial to be conducted in this high-risk group. The MILI study provides a unique opportunity to evaluate the efficacy of metformin as a chemopreventive alongside exploring its mechanism of anticancer action and the biological process of mutated P53-driven tumourigenesis. TRIAL REGISTRATION: ISRCTN16699730. Registered on 28 November 2022. URL: https://www.isrctn.com/ EudraCT/CTIS number 2022-000165-41.


Subject(s)
Li-Fraumeni Syndrome , Metformin , Adult , Humans , Mice , Animals , Li-Fraumeni Syndrome/diagnostic imaging , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/prevention & control , Metformin/adverse effects , Quality of Life , Germ-Line Mutation , Magnetic Resonance Imaging , Genetic Predisposition to Disease , Randomized Controlled Trials as Topic , Clinical Trials, Phase II as Topic
12.
Int J Radiat Oncol Biol Phys ; 119(4): 1086-1091, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38309330

ABSTRACT

PURPOSE: Breast cancer (BC) is the most common malignancy in female patients with Li-Fraumeni syndrome (LFS), a condition associated with an increased risk of various malignancies, including radiation therapy (RT)-induced malignancies (RIM) within previously irradiated areas. Our study aimed to assess the incidence of RIM in patients with LFS and early-stage BC treated with adjuvant RT, including the effect of RT dose and technique. METHODS AND MATERIALS: We examined patients with a germline pathogenic/likely pathogenic TP53 variant diagnosed with early-stage BC and monitored by a hereditary cancer team at a single cancer center. The study endpoints included RIM frequency, the association of RIM with the dose and type of RT (2-dimensional [2D] RT, 3-dimensional [3D] RT, and intensity modulated RT [IMRT]), and BC recurrence. RESULTS: We analyzed 48 patients with a median age of 39 years (range, 21-62). The majority (71%) had the TP53 R337H variant, and 87% were unaware of their LFS diagnosis at the time of BC treatment. Treatment modalities included mastectomy (62%), (neo)adjuvant chemotherapy (66%), and RT (62%), with RT being more common after breast-conserving surgery (87% vs 46% with mastectomy, P = .010). Among the 30 patients treated with RT, 10% developed RIM in the irradiated field, consisting of 3 soft tissue malignancies. RT dose (≤40.8 or >40.8 Gy) did not influence RIM occurrence, but the type of RT did. RIM was observed in 100% of cases with 2D RT (2/2), 50% with IMRT (1/2), and 0% with 3D RT (0/16) (P = .004). CONCLUSIONS: Our study underscores a concerning rate of RIM after adjuvant RT, emphasizing the importance of a thorough risk-benefit evaluation before recommending RT, with preference for its avoidance if possible. Although subgroup sizes were limited, the risk of RIM appeared to be influenced by the RT technique, with higher rates observed with 2D RT and IMRT compared with 3D RT. Early TP53 testing is essential to guide the BC treatment plan.


Subject(s)
Breast Neoplasms , Li-Fraumeni Syndrome , Neoplasms, Radiation-Induced , Humans , Li-Fraumeni Syndrome/genetics , Female , Breast Neoplasms/radiotherapy , Adult , Middle Aged , Young Adult , Neoplasms, Radiation-Induced/etiology , Radiotherapy, Adjuvant/adverse effects , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy Dosage , Germ-Line Mutation , Tumor Suppressor Protein p53/genetics , Neoplasm Recurrence, Local , Incidence
13.
Am J Med Genet A ; 194(7): e63573, 2024 Jul.
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
14.
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
15.
JCO Precis Oncol ; 8: e2300453, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38412388

ABSTRACT

PURPOSE: Establishing accurate age-related penetrance figures for the broad range of cancer types that occur in individuals harboring a pathogenic germline variant in the TP53 gene is essential to determine the most effective clinical management strategies. These figures also permit optimal use of cosegregation data for classification of TP53 variants of unknown significance. Penetrance estimation can easily be affected by bias from ascertainment criteria, an issue not commonly addressed by previous studies. MATERIALS AND METHODS: We performed a maximum likelihood penetrance estimation using full pedigree data from a multicenter study of 146 TP53-positive families, incorporating adjustment for the effect of ascertainment and population-specific background cancer risks. The analysis included pedigrees from Australia, Spain, and United States, with phenotypic information for 4,028 individuals. RESULTS: Core Li-Fraumeni syndrome (LFS) cancers (breast cancer, adrenocortical carcinoma, brain cancer, osteosarcoma, and soft tissue sarcoma) had the highest hazard ratios of all cancers analyzed in this study. The analysis also detected a significantly increased lifetime risk for a range of cancers not previously formally associated with TP53 pathogenic variant status, including colorectal, gastric, lung, pancreatic, and ovarian cancers. The cumulative risk of any cancer type by age 50 years was 92.4% (95% CI, 82.2 to 98.3) for females and 59.7% (95% CI, 39.9 to 81.3) for males. Females had a 63.3% (95% CI, 35.6 to 90.1) cumulative risk of developing breast cancer by age 50 years. CONCLUSION: The results from maximum likelihood analysis confirm the known high lifetime risk for the core LFS-associated cancer types providing new risk estimates and indicate significantly increased lifetime risks for several additional cancer types. Accurate cancer risk estimates will help refine clinical recommendations for TP53 pathogenic variant carriers and improve TP53 variant classification.


Subject(s)
Breast Neoplasms , Li-Fraumeni Syndrome , Male , Female , Humans , United States , Middle Aged , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Genes, p53/genetics , Pedigree , Tumor Suppressor Protein p53/genetics , Genetic Predisposition to Disease/genetics , Breast Neoplasms/genetics , Risk Factors
16.
Cancer Med ; 13(3): e6920, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38230850

ABSTRACT

PURPOSE: Tumor protein p53 (TP53) pathogenic variant (PV) carriers are identified during genetic testing for hereditary causes of cancer. PVs in TP53 are associated with the Li-Fraumeni syndrome (LFS), and thus, surveillance and preventive measures are important for TP53 PV carriers. However, the penetrance of TP53 PVs can be low if the Chompret criteria are not fulfilled. In this study, we compared the phenotypic characteristics of families that did and did not fulfill the LFS criteria according to Chompret. METHODS: The German Consortium for Hereditary Breast and Ovarian Cancer (GC-HBOC) database was used to identify index patients with a likely pathogenic/pathogenic TP53 variant and their family members. The study investigated the type of variant, pedigree, age of onset, number of primary tumors, and histological type of BC. RESULTS: TP53 PV were present in the index cases of 35 families, 57% (20/35) of which fulfilled the Chompret criteria. The median age of onset at first BC diagnosis was lower in families that fulfilled the Chompret criteria compared to those who did not. Four of all diseased individuals were minors (4%; 4/105) when malignancy was first diagnosed. Sarcomas and brain tumors occurred in 10% (10/105) and in 7% (7/105) of all diseased persons, respectively. BC was the most frequently occurring first tumor (60%; 62/105) and additional malignancy (45%; 20/44) in this cohort. Subsequent malignancies developed in 31% (20/65) of the individuals who fulfilled the Chompret criteria compared with 15% (6/40) of those who did not. CONCLUSION: The tumor spectrum and age of onset found in this study showed that tumors other than BC had low disease penetrance in TP53 PV carriers identified using the GC-HBOC criteria for genetic testing.


Subject(s)
Breast Neoplasms , Li-Fraumeni Syndrome , Ovarian Neoplasms , Humans , Female , Tumor Suppressor Protein p53/genetics , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/genetics , Phenotype , Breast , Li-Fraumeni Syndrome/epidemiology , Li-Fraumeni Syndrome/genetics , Germ Cells
18.
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
19.
Cancer Discov ; 14(1): 23-25, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38213298

ABSTRACT

SUMMARY: In the first prospective study evaluating circulating tumor DNA (ctDNA) for early cancer detection, Wong, Luo, and colleauges demonstrate the feasibility of liquid biopsy as an augmentation to current surveillance protocols for patients with Li-Fraumeni syndrome, an inherited cancer predisposition associated with high cancer risk in both pediatric and adult populations. Though additional clinical validation in larger cohorts is needed, this research highlights that a multimodal approach is likely necessary to improve the sensitivity of liquid biopsy assays for early cancer detection. See related article by Wong, Lou et al., p. 104 (9).


Subject(s)
Cell-Free Nucleic Acids , Li-Fraumeni Syndrome , Adult , Child , Humans , Li-Fraumeni Syndrome/diagnosis , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/epidemiology , Tumor Suppressor Protein p53/genetics , Prospective Studies , Germ-Line Mutation , Genetic Predisposition to Disease , Liquid Biopsy
20.
Fam Cancer ; 23(1): 29-33, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38206485

ABSTRACT

Li-Fraumeni Syndrome (LFS), caused by germline pathogenic variants in TP53, predisposes to a wide range of young-onset malignancies, particularly sarcoma, breast and brain cancer. More recently, an increased risk of gastric adenocarcinoma has been recognised, although uptake of surveillance upper endoscopy is unclear. Our retrospective review of 65 patients with LFS, of whom 53.8% had undergone endoscopy, identified four patients (6.2%) with gastro-esophageal junction (GEJ) adenocarcinomas. Two cases were found on asymptomatic screening and were early stage. No cases had family history of gastrointestinal malignancy. Reviewing genomic data from The Cancer Genome Atlas Program, 76.4% of sporadic esophageal adenocarcinomas harboured somatic TP53 pathogenic variants, compared with 39.9% of non-cardia gastric cancers. This similar pattern observed in germline and sporadic cases warrants further investigation. We propose that upper endoscopy be recommended to all patients with LFS, with a focus on appropriate surveillance of the GEJ.


Subject(s)
Adenocarcinoma , Li-Fraumeni Syndrome , Stomach Neoplasms , Humans , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/diagnosis , Genetic Predisposition to Disease , Tumor Suppressor Protein p53/genetics , Germ-Line Mutation , Stomach Neoplasms/genetics , Adenocarcinoma/genetics , Esophagogastric Junction
SELECTION OF CITATIONS
SEARCH DETAIL
...