Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Genet Couns ; 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39189353

ABSTRACT

Hematologic malignancies (HMs) are a heterogeneous group of cancers impacting individuals of all ages that have been increasingly recognized in association with various germline predisposition syndromes. Given the myriad of malignancy subtypes, expanding differential diagnoses, and unique sample selection requirements, evaluation for hereditary predisposition to HM presents both challenges as well as exciting opportunities in the ever-evolving field of genetic counseling. This practice resource has been developed as a foundational resource for genetic counseling approaches to hereditary HMs and aims to empower genetic counselors who encounter individuals and families with HMs in their practice.

2.
Eur J Haematol ; 110(1): 109-116, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36209474

ABSTRACT

Since 2003, more than 15 genes have been identified to predispose to hereditary hematologic malignancy (HHM). Although the yield of germline analysis for leukemia appears like that of solid tumors, genetic referrals in adults with leukemia remain underperformed. We assessed leukemia patients' attitudes toward genetic testing and leukemia-related distress through a survey of 1093 patients diagnosed with acute or chronic leukemia, myelodysplastic syndrome, or aplastic anemia. Principal component analysis (PCA) was used to analyze patient attitudes. Distress was measured through the Impact of Event Scale-Revised (IES-R). Exactly 19.8% of eligible respondents completed the survey. The majority reported interest in (77%) or choosing to have (78%) genetic testing for HHM. Slightly over half identified worry about cost of genetic testing (58%) or health insurance coverage (61%) as possible barriers. PCA identified relevant themes of interest in genetic testing, impact on leukemia treatment, discrimination and confidentiality, psychosocial and familial impacts, and cost of testing. The majority reported low distress. Leukemia patients report high interest in genetic testing, few barriers, and relatively low distress.


Subject(s)
Hematologic Neoplasms , Leukemia , Myelodysplastic Syndromes , Adult , Humans , Genetic Testing , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/genetics , Myelodysplastic Syndromes/diagnosis , Attitude , Leukemia/genetics , Genetic Counseling , Genetic Predisposition to Disease
3.
Epigenetics ; 16(5): 567-576, 2021 05.
Article in English | MEDLINE | ID: mdl-32856987

ABSTRACT

Acute myeloid leukaemia (AML) is a heterogeneous myeloid malignancy characterized by recurrent clonal events, including mutations in epigenetically relevant genes such as DNMT3A, ASXL1, IDH1/2, and TET2. Next-generation sequencing analysis of a mother and son pair who both developed adult-onset diploid AML identified a novel germline missense mutation DNMT3A p.P709S. The p.P709S protein-altering variant resides in the highly conserved catalytic DNMT3A methyltransferase domain. Functional studies demonstrate that the p.P709S variant confers dominant negative effects when interacting with wildtype DNMT3A. LINE-1 pyrosequencing and reduced representation bisulphite sequencing (RBBS) analysis demonstrated global DNA hypomethylation in germline samples, not present in the leukaemic samples. Somatic acquisition of IDH2 p.R172K mutations, in concert with additional acquired clonal DNMT3A events in both patients at the time of AML diagnosis, confirms the important pathogenic interaction of epigenetically active genes, and implies a strong selection and regulation of methylation in leukaemogenesis. Improved characterization of germline mutations may enable us to better predict malignant clonal evolution, improving our ability to provide customized treatment or future preventative strategies.


Subject(s)
DNA Methyltransferase 3A/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute , Adult , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Germ Cells , Humans , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplasm Recurrence, Local
5.
Front Oncol ; 10: 582213, 2020.
Article in English | MEDLINE | ID: mdl-33585199

ABSTRACT

Previously considered rare, inherited hematologic malignancies are increasingly identified. Germline mutations in the RNA helicase DDX41 predispose to increased lifetime risks of myeloid neoplasms with disease often occurring later in life which presents challenges for germline recognition. To improve identification of germline DDX41, individuals presenting with ≥1 DDX41 alteration on an institutional MDS/AML next-generation sequencing based panel with at least one at >40% variant allele frequency were flagged for review and genetic counseling referral. Of 5,801 individuals, 90 (1.5%) had ≥1 DDX41 mutation(s) identified. Thirty-eight (42%) patients with a median age of 66 years were referred for genetic counseling; thirty-one were male (81.5%). Thirty-five (92%) referred patients elected to pursue germline evaluation and in 33/35 (94%) a germline DDX41 variant was confirmed. Twenty-two patients (66%) with germline variants reported antecedent cytopenias, seven (21%) had a prior history of malignancy, and twenty-seven (82%) reported a family history of cancer. Predictive genetic testing for healthy family members under consideration as stem cell transplant donors was successfully performed in 11 family members, taking an average of 15 days. Near-heterozygous DDX41 mutations identified on next-generation sequencing, particularly nonsense/frameshift variants or those at recurrent germline "hot spots" are highly suggestive of a germline mutation. Next-generation sequencing screening is a feasible tool to screen unselected myeloid neoplasms for germline DDX41 mutations, enabling timely and appropriate care.

6.
Cancer Genet ; 235-236: 77-83, 2019 06.
Article in English | MEDLINE | ID: mdl-31101557

ABSTRACT

PURPOSE: Adolescents and young adults with colorectal cancer (CRC) have attracted recent attention, with a hereditary syndrome identified in one-third of patients diagnosed ≤ 35. We aimed to study this population to determine if a CRC-specific gene panel increased the yield of testing. METHODS: Patients with CRC ≤ 35 evaluated from 05/2014-11/2017 were identified from the genetic counseling database. Records were reviewed for personal/family history and genetic counseling outcomes. RESULTS: One hundred forty-three patients with CRC ≤ 35 were included. One hundred four (72.7%) underwent CRC panel testing. Thirty-nine (27.2%) had syndrome-directed testing, declined, or were lost to follow-up. Forty-two patients had a genetic syndrome (29.4%). Twenty-four of the 42 hereditary patients (57.1%) were identified via syndrome-directed testing. Mutations identified via panel testing were consistent with patient personal/family history. Thirty-three patients had at least one variant of uncertain significance. CONCLUSION: Hereditary syndromes were identified in 29.4% of patients. Panel testing in patients without a phenotype did not increase diagnostic yield, but identified variants in one-third. Disease-specific panel testing is of low yield in young patients without a suggestive personal/family history. Testing broader panels may increase the yield of mutation pick-up in this population, although at the expense of identifying variants.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Neoplastic Syndromes, Hereditary/genetics , Adolescent , Adult , Female , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Male , Young Adult
7.
Am J Hematol ; 94(7): 757-766, 2019 07.
Article in English | MEDLINE | ID: mdl-30963592

ABSTRACT

Myeloid neoplasms with germline DDX41 mutations have been incorporated into the 2017 WHO classification. Limited studies describing the clinicopathologic features and mutation profile are available. We searched for myeloid neoplasms with a DDX41 gene mutation tested by an 81-gene next-generation sequencing panel over a 7-month period. We identified 34 patients with myeloid neoplasms with DDX41 abnormalities; 26 (76%) men and 8 women (24%) [median age, 70 years], 20 acute myeloid leukemia (AML), 10 myelodysplastic syndrome (MDS), 1 chronic myelomonocytic leukemia (CMML) and 3 myeloproliferative neoplasms (MPN). Fifty-nine DDX41 variants were detected: 27 (46%) appeared somatic and 32 (54%) were presumably germline mutations. The majority of presumed germline mutations were upstream of the Helicase 2 domain (93%) and involved loss of the start codon (30%). The majority of somatic mutations were within the Helicase 2 domain (78%), with the missense mutation p.R525H being most common (67%). There was a significant difference in the location of germline or somatic mutations (P < .0001). Concomitant mutations were detected involving 19 genes, but only TP53 (n = 11, 32%), ASXL1 (n = 8, 24%), and JAK2 (n = 4, 12%) were recurrent. Twenty (59%) patients showed diploid cytogenetics. Twenty-three (68%) patients presented with AML or MDS-EB-2, suggesting an association with high-grade myeloid neoplasm. Patients with myeloid neoplasms carrying DDX41 mutations show male predominance (3:1), higher age at presentation, association with TP53 mutations, and association with high-grade myeloid neoplasms in our cohort at a referral cancer center setting. These findings support the recognition of myeloid neoplasms with DDX41 mutation as unique, need for germline confirmation, and further assessment of family members.


Subject(s)
DEAD-box RNA Helicases , Germ-Line Mutation , Hematologic Neoplasms , Myeloproliferative Disorders , Sex Characteristics , Tumor Suppressor Protein p53 , Aged , Aged, 80 and over , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Male , Middle Aged , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Retrospective Studies , Risk Factors , Sex Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Article in English | MEDLINE | ID: mdl-30709875

ABSTRACT

Li-Fraumeni syndrome (LFS) is an autosomal dominant condition associated with a high risk of a broad range of childhood- and adult-onset cancers. LFS is related to germline mutations of the tumor-suppressor gene TP53 The most common reported leukemia associated with LFS is hypodiploid acute lymphoblastic leukemia, but myeloid malignancies including acute myeloid leukemia (AML), chronic myeloid leukemia, and myelodysplastic syndrome (MDS) are also reported, often in the setting of therapy-related disease. We reviewed the clinicopathologic characteristics including cytogenetics and molecular analysis for seven adult patients with LFS and hematologic malignancies evaluated at the Hereditary Hematologic Malignancy Clinic (HHMC) at MD Anderson Cancer Center. We present this LFS review series to increase awareness of LFS for the appropriate diagnosis of both patients and potentially affected relatives, as well as provide experience with patient outcomes in this difficult to treat population.


Subject(s)
Germ-Line Mutation , Hematologic Neoplasms/etiology , Li-Fraumeni Syndrome/complications , Tumor Suppressor Protein p53/genetics , Adult , Female , Genetic Predisposition to Disease , Humans , Leukemia, Myeloid, Acute/etiology , Li-Fraumeni Syndrome/genetics , Male , Middle Aged , Myelodysplastic Syndromes/etiology , Pedigree , Young Adult
9.
Leuk Lymphoma ; 60(8): 2025-2033, 2019 08.
Article in English | MEDLINE | ID: mdl-30648453

ABSTRACT

Germline mutations in GATA2 are associated with a complex immunodeficiency and cancer predisposition syndrome. Somatic GATA2mut in myeloid malignancies may impart a similar phenotype. We reviewed adult patients with a diagnosis of GATA2mut hematological malignancy who were referred to our HHMC for genetic testing, and identified to have somatic GATA2mut. Nine patients with a median age of 63 years were included. Six patients (66.7%) were males. Atypical CML and acute myeloid leukemia were the most common initial presentation. The median overall VAF was 47.14%. Monocytopenia was pronounced when the GATA2mut involved the C-terminal ZFD. GATA2 N-terminal ZFD mutations tend to be co-mutated with biCEBPAmut. Unlike germline GATA2 mutations, monocytopenia associated with somatic GATA2 mutations often resolved at remission. We concluded that similar to germline GATA2 mutations, a subset of somatic GATA2 mutations can impart a germline phenotype.


Subject(s)
GATA2 Transcription Factor/genetics , Genetic Predisposition to Disease , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Phenotype , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/etiology , Adult , Aged , Aged, 80 and over , Alleles , Disease Management , Female , Flow Cytometry , GATA2 Transcription Factor/metabolism , Gene Expression , Genetic Association Studies , Genotype , Germ-Line Mutation , Hematologic Neoplasms/complications , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Humans , Leukocyte Count , Male , Middle Aged , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/immunology , Primary Immunodeficiency Diseases/mortality , Primary Immunodeficiency Diseases/therapy , Prognosis , Young Adult
11.
Cancer Prev Res (Phila) ; 11(11): 679-686, 2018 11.
Article in English | MEDLINE | ID: mdl-30274973

ABSTRACT

Introduction: We aimed to determine the prevalence and landscape of germline mutations among patients with young-onset pancreatic ductal adenocarcinoma (PDAC) as well as their influence in prognosis.Methods: Patients from two cohorts were studied, the high-risk cohort (HRC), which included 584 PDAC patients who received genetic counseling at The University of Texas MD Anderson Cancer Center, and a general cohort (GC) with 233 metastatic PDAC patients. We defined germline DNA sequencing on 13 known pancreatic cancer susceptibility genes. The prevalence and landscape of mutations were determined, and clinical characteristics including survival were analyzed.Results: A total of 409 patients underwent genetic testing (277 from HRC and 132 from GC). As expected, the HRC had higher prevalence of germline mutations compared with the GC: 17.3% versus 6.81%. The most common mutations in both cohorts were in BRCA1/2 and mismatch-repair (MMR) genes. Patients younger than 60 years old had significantly higher prevalence of germline mutations in both the HRC [odds ratios (OR), 1.93 ± 1.03-3.70, P = 0.039] and GC (4.78 ± 1.10-32.95, P = 0.036). Furthermore, PDAC patients with germline mutations in the GC had better overall survival than patients without mutations (HR, 0.44; 95% CI of HR, 0.25-0.76, P = 0.030).Discussion: Germline mutations are highly prevalent in patients with PDAC of early onset and can be predictive of better outcomes. Considering emerging screening strategies for relatives carrying susceptibility genes as well as impact on therapy choices, genetic counseling and testing should be encouraged in PDAC patients, particularly those of young onset. Cancer Prev Res; 11(11); 679-86. ©2018 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genetic Predisposition to Disease , Neoplastic Syndromes, Hereditary/epidemiology , Pancreatic Neoplasms/genetics , Adult , Age Factors , Age of Onset , Aged , Aged, 80 and over , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carcinoma, Pancreatic Ductal/mortality , Cohort Studies , DNA Mismatch Repair/genetics , DNA Mutational Analysis , Female , Genetic Counseling , Humans , Male , Medical History Taking , Middle Aged , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Pancreatic Neoplasms/mortality , Prevalence , Risk Factors , Survival Analysis
12.
Cancer ; 124(13): 2704-2713, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29682723

ABSTRACT

Recognizing and referring patients with possible inherited cancer predisposition syndromes for appropriate genetic evaluation and testing provides insights into optimal patient treatment approaches and also can provide education and testing opportunities for family members. Next-generation sequencing (NGS)-based, targeted genotyping for somatic mutations is increasingly used in the diagnosis, prognostication, and treatment selection for patients with hematologic malignancies. However, certain mutations that may be somatically acquired can also be present as germline mutations in some individuals and families. Whether the results of NGS-based leukemia panels can be used to inform decisions and subsequent evaluation of patients with possible inherited cancer predispositions has not been described previously. Because a normal control often is not available when using NGS panels in patients with hematologic malignancies, NGS panel results offer both an opportunity and a challenge to determine the origin and pathogenicity of identified mutations. In the absence of a matched germline control, variant allele frequency (VAF) estimation and data from publically available data sets provide important clues to the possible germline origin of a variant. Careful annotation and review of NGS panels in patients with hematologic malignancies can provide a useful screening tool to systematically improve upon the detection of potentially pathogenic germline variants. Cancer 2018;124:2704-2713. © 2018 American Cancer Society.


Subject(s)
Genetic Testing/methods , Hematologic Neoplasms/diagnosis , Leukemia/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Datasets as Topic , Gene Frequency , Genetic Predisposition to Disease , Genotyping Techniques/methods , Germ-Line Mutation , Hematologic Neoplasms/genetics , High-Throughput Nucleotide Sequencing , Humans , Leukemia/genetics , Molecular Sequence Annotation , Neoplastic Syndromes, Hereditary/genetics , Prognosis
13.
Cancer Prev Res (Phila) ; 10(10): 580-587, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765196

ABSTRACT

Lynch syndrome (LS) is a genetic condition secondary to germline alterations in the DNA mismatch repair (MMR) genes with 30% of changes being variants of uncertain significance (VUS). Our aim was to perform an in silico reclassification of VUS from a large single institutional cohort that will help prioritizing functional validation. A total of 54 VUS were detected with 33 (61%) novel variants. We integrated family history, pathology, and genetic information along with supporting evidence from eight different in silico tools at the RNA and protein level. Our assessment allowed us to reclassify 54% (29/54) of the VUS as probably damaging, 13% (7/54) as possibly damaging, and 28% (15/54) as probably neutral. There are more than 1,000 VUS reported in MMR genes and our approach facilitates the prioritization of further functional efforts to assess the pathogenicity to those classified as probably damaging. Cancer Prev Res; 10(10); 580-7. ©2017 AACR.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Computer Simulation , DNA Mismatch Repair/genetics , DNA-Binding Proteins/genetics , Genetic Variation , Adolescent , Adult , Aged , Cohort Studies , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Female , Humans , Male , Middle Aged , Young Adult
14.
Clin Cancer Res ; 23(17): 5246-5254, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28522602

ABSTRACT

Purpose: Determination of microsatellite instability (MSI) by PCR is the gold standard; however, IHC of mismatch repair (MMR) proteins is frequently performed instead. The reliability of these methods on postneoadjuvant therapy specimens is unknown. We examined the effect of neoadjuvant therapy on MSI results by PCR and IHC.Experimental design: A total of 239 colorectal cancers resected after neoadjuvant therapy were assessed for MSI with PCR and IHC. PCR and IHC results for matched paired pre- and posttreatment specimens were compared. In parallel, 2 isogenic cell lines conditioned for MMR functioning and 2 different patient-derived xenografts (PDXs) were exposed to chemotherapy, radiation, or both. We also examined whether establishment of PDXs induced MSI changes in 5 tumors. IHC and MSI were tested after treatment to assess for changes.Results: We identified paired pre- and posttreatment specimens for 37 patients: 2 with PCR only, 34 with IHC only, and 1 with both. All 3 patients with PCR had microsatellite stable pre- and posttreatment specimens. Of the 35 patients with IHC, 30 had intact MMR proteins in pre- and posttreatment specimens, 1 had equivocal MLH1 staining in the pretreatment and loss in the posttreatment specimen, and 4 had intact pretreatment MSH6 but variable posttreatment staining. In the experimental setting, no changes in MSI status were detected after treatment or tumor implantation in animals.Conclusions: Our findings show that the expression of MMR proteins, commonly MSH6, can change after neoadjuvant therapy and confirm PCR as the gold-standard test for MSI after neoadjuvant therapy. Clin Cancer Res; 23(17); 5246-54. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Microsatellite Instability , Adult , Aged , Aged, 80 and over , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Female , Gene Expression Regulation, Neoplastic , Germ-Line Mutation/genetics , HCT116 Cells , Humans , Male , Mice , Middle Aged , Neoadjuvant Therapy/adverse effects , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
17.
Fam Cancer ; 16(3): 357-361, 2017 07.
Article in English | MEDLINE | ID: mdl-28004223

ABSTRACT

Traditional germline sequencing and deletion/duplication analysis does not detect Lynch syndrome-causing mutations in all individuals whose colorectal or endometrial tumors demonstrate mismatch repair (MMR) deficiency. Unique inversions and other rearrangements of the MMR genes have been reported in families with Lynch syndrome. In 2014, a recurrent inversion of MSH2 exons 1-7 was identified in five families suspected to have Lynch syndrome. We aimed to describe our clinical experience in identifying families with this specific inversion. Four probands whose Lynch syndrome-associated tumors demonstrated absence of MSH2/MSH6 staining and who had negative MMR germline testing were evaluated for the MSH2 inversion of exons 1-7, offered during initial genetic workup or upon routine clinical follow-up. All four probands tested positive for the MSH2 inversion. Proband cancer diagnoses included colon and endometrial adenocarcinoma and sebaceous adenoma. A variety of Lynch syndrome-associated cancers were reported in the family histories, although only one family met Amsterdam II criteria. Thirteen at-risk relatives underwent predictive testing. MSH2 inversion of exons 1-7 was found in four probands previously suspected to have Lynch syndrome based on family history and tumor testing. This testing should be offered routinely to patients with tumors demonstrating loss of MSH2/MSH6 staining.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , MutS Homolog 2 Protein/genetics , Adult , Exons/genetics , Female , Gene Rearrangement/genetics , Humans , Male , Middle Aged , Pedigree
18.
Int J Mol Sci ; 17(6)2016 May 30.
Article in English | MEDLINE | ID: mdl-27248996

ABSTRACT

Myelodysplastic syndromes (MDS) are heterogeneous clonal hematopoietic disorders characterized by ineffective hematopoiesis, bone marrow dysplasia, and peripheral cytopenias. Familial forms of MDS have traditionally been considered rare, especially in adults; however, the increasing availability of somatic and germline genetic analyses has identified multiple susceptibility loci. Bone marrow failure syndromes have been well-described in the pediatric setting, e.g., Fanconi anemia (FA), dyskeratosis congenita (DC), Diamond-Blackfan anemia (DBA), and Shwachman-Diamond syndrome (SBS), hallmarked by clinically-recognizable phenotypes (e.g., radial ray anomalies in FA) and significantly increased risks for MDS and/or acute myeloid leukemia (AML) in the setting of bone marrow failure. However, additional families with multiple cases of MDS or AML have long been reported in the medical literature with little known regarding potential hereditary etiologies. Over the last decade, genomic investigation of such families has revealed multiple genes conferring inherited risks for MDS and/or AML as the primary malignancy, including RUNX1, ANKRD26, DDX41, ETV6, GATA2, and SRP72. As these syndromes are increasingly appreciated in even apparently de novo presentations of MDS, it is important for hematologists/oncologists to become familiar with these newly-described syndromes. Herein, we provide a review of familial MDS syndromes and practical aspects of management in patients with predisposition syndromes.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , Genetic Predisposition to Disease/genetics , Hemoglobinuria, Paroxysmal/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Bone Marrow Failure Disorders , Core Binding Factor Alpha 2 Subunit/genetics , DEAD-box RNA Helicases/genetics , Female , GATA2 Transcription Factor/genetics , Genetic Counseling , Genetic Testing , Heredity , Humans , Intercellular Signaling Peptides and Proteins , Male , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Signal Recognition Particle/genetics , ETS Translocation Variant 6 Protein
19.
Clin Lymphoma Myeloma Leuk ; 16(7): 417-428.e2, 2016 07.
Article in English | MEDLINE | ID: mdl-27210295

ABSTRACT

INTRODUCTION: Although multiple predispositions to hematologic malignancies exist, evaluations for hereditary cancer syndromes (HCS) are underperformed by most hematologist/oncologists. Criteria for initiating HCS evaluation are poorly defined, and results of genetic testing for hereditary hematologic malignancies have not been systematically reported. PATIENTS AND METHODS: From April 2014 to August 2015, 67 patients were referred to the Hereditary Hematologic Malignancy Clinic (HHMC). Referral reasons included (1) bone marrow failure or myelodysplastic syndrome in patients ≤ 50 years, (2) evaluation for germ-line inheritance of identified RUNX1, GATA2, or CEBPA mutations on targeted next-generation sequencing panels, and (3) strong personal and/or family history of malignancy. Cultured skin fibroblasts were utilized for germ-line DNA in all patients with hematologic malignancy. RESULTS: Eight patients (12%) were clinically diagnosed with a HCS: 4 patients with RUNX1-related familial platelet disorder (FPD)/acute myeloid leukemia (AML), and 1 patient each with dyskeratosis congenita, Fanconi anemia, germ-line DDX41, and Li-Fraumeni syndrome (LFS). Two patients with concern for FPD/AML and LFS, respectively, had RUNX1 and TP53 variants of unknown significance. Additionally, 4 patients with prior HCS diagnosis (1 LFS, 3 FPD/AML) were referred for further evaluation and surveillance. CONCLUSION: In this HHMC-referred hematologic malignancy cohort, HCS was confirmed in 12 patients (18%). HCS identification provides insight for improved and individualized treatment, as well as screening/surveillance opportunities for family members. The HHMC has facilitated HCS diagnosis; with increased clinical awareness of hematologic malignancy predisposition syndromes, more patients who may benefit from evaluation can be identified. Mutation panels intended for prognostication may provide increased clinical suspicion for germ-line testing.


Subject(s)
Family/psychology , Genetic Predisposition to Disease/psychology , Hematologic Neoplasms/epidemiology , Neoplastic Syndromes, Hereditary/epidemiology , Patients/psychology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Female , Genetic Counseling , Germ-Line Mutation , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/genetics , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Pedigree , Referral and Consultation , Texas , Workflow , Young Adult
20.
Fam Cancer ; 15(4): 587-91, 2016 10.
Article in English | MEDLINE | ID: mdl-27017610

ABSTRACT

Constitutional mismatch repair deficiency syndrome (CMMRD) is a rare autosomal recessive predisposition to colorectal polyposis and other malignancies, often childhood-onset, that is caused by biallelic inheritance of mutations in the same mismatch repair gene. Here, we describe a patient with a clinical diagnosis of CMMRD based on colorectal polyposis and young-onset endometrial cancer who was identified to have two alterations in trans in PMS2: one known pathogenic mutation (c.1831insA; p.Ile611Asnfs*2) and one novel variant of uncertain significance (c.505C>G; p.Arg169Glu), a missense alteration. We describe the clinical and molecular features in the patient harboring this novel alteration c.505C>G, who meets clinical criteria for CMMRD and exhibits molecular evidence supporting a diagnosis of CMMRD. Although experimental validation is needed to confirm its pathogenicity, PMS2 c.505C>G likely has functional consequences that contributes to our patient's phenotype based on the patient's clinical presentation, tumor studies, and bioinformatics analysis.


Subject(s)
Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , Mismatch Repair Endonuclease PMS2/genetics , Mutation , Neoplastic Syndromes, Hereditary/genetics , Adolescent , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Female , Humans , Male , Microsatellite Instability , Mismatch Repair Endonuclease PMS2/metabolism , Neoplastic Syndromes, Hereditary/pathology , Neoplastic Syndromes, Hereditary/surgery , Pedigree , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL